You are on page 1of 931

2016 ASCO EDUCATIONAL BOOK

This publication is supported by an


educational donation provided by:

Amgen
Merck & Co., Inc.

AMERICAN SOCIETY OF CLINICAL ONCOLOGY

2016 EDUCATIONAL BOOK

Support for this program is funded through


Collective Wisdom: The Future of Patient-Centered Care and Research

A PEER-REVIEWED, INDEXED PUBLICATION


52nd Annual Meeting | June 37, 2016 | Chicago, Illinois | Volume 36

Vol. 36
American Society of Clinical Oncology Educational Book
The 2016 ASCO Educational Book (Print ISSN: 1548-8748; Electronic ISSN: 1548-8756) is published by American Society of Clinical Oncology,
Inc. (ASCO).

Requests for permission to reprint all or part of any article published in this title should be directed to Permissions, American Society
of Clinical Oncology, Inc., 2318 Mill Road, Suite 800, Alexandria, VA 22314. Tel: (571) 483-1300; fax: (571) 366-9550; or email:
permissions@asco.org.

All other questions should be addressed to ASCO Educational Book Managing Editor, American Society of Clinical Oncology, Inc., 2318
Mill Road, Suite 800, Alexandria, VA 22314. Tel: (571) 483-1300; fax: (571) 366-9550; or email: edbook@asco.org.

Single issues, both current and back, exist in limited quantities and are offered for sale subject to availability. For further information,
email edbook@asco.org or call (888) 273-3508.

Copyright 2016 American Society of Clinical Oncology, Inc. All rights reserved. No part of this publication may be reproduced or transmitted
in any form or by any means, electronic or mechanical, including photocopy, recording, or any information storage and retrieval system,
without permission in writing from ASCO.

Copies of articles in this publication may be made for personal use. This consent is given on the condition, however, that the copier
pay the stated per-copy fee through the Copyright Clearance Center, Inc. (222 Rosewood Drive, Danvers, MA 01923) for any copying
beyond that permitted by Sections 107 or 108 of the U.S. Copyright Law. This consent does not extend to any other kinds of copying, such
as copying for general distribution, for advertising, commercial, or promotional purposes, for creating new collective or derivative works, or
for resale.

ASCO assumes no responsibility for errors or omissions in this publication. The reader is advised to check the appropriate medical
literature and the product information currently provided by the manufacturer of each drug to be administered to verify, among other
matters, the dosage, the method and duration of administration, or contraindications. It is the responsibility of the treating physician
or other health care professional, relying on the independent experience and knowledge of the patient, to determine drug dosages
and the best treatment for the patient.

The ideas and opinions expressed in this publication do not necessarily reflect those of ASCO. The mention of any company, product,
service, or therapy does not constitute an endorsement of any kind by ASCO. ASCO assumes no responsibility for any injury or damage
to persons or property arising out of or related to any use of the material contained in this publication.
American Society of
Clinical Oncology
Educational Book
Editor in Chief: Don S. Dizon, MD

Associate Editor: Nathan Pennell, MD, PhD


Managing Editor: Lindsay Pickell, MFA
Production Manager: Donna Dottellis

2016 American Society of Clinical Oncology, Alexandria, VA


Contents
2016 ASCO Annual Meeting Disclosure xiii

20152016 Cancer Education Committee xiv

2016 Educational Book Expert Panel xvi

2016 Annual Meeting Supporters xx

Letter From the Editor 1

INVITED ARTICLES
Collaborative Practice in an Era of Multidisciplinary Care
Michael P. Kosty, Todd Pickard, and Pamela Viale 3
Value in Oncology: Balance Between Quality and Cost
Derek Raghavan and Mark W. Legnini 9
Womens Health Issues for BRCA Mutation Carriers
Mary E. Sabatini and Leif W. Ellisen 14
Collective Wisdom: Lobular Carcinoma of the Breast
George W. Sledge, Anees Chagpar, and Charles Perou 18

POINTS OF VIEW
Compassionate Use: A Modest Proposal
Arthur L. Caplan, Alison Bateman-House, and Joanne Waldstreicher e2
Less Is More: The Evolving Surgical Approach to Breast Cancer
Laura Esserman, Etienne Gallant, and Michael Alvarado e5
Strategies for Sustainable Cancer Care
David J. Kerr, Anant Jani, and Sir Muir Gray e11
The Role of Nephrectomy for Kidney Cancer in the Era of Targeted and Immune Therapies
Ulka N. Vaishampayan e16

BREAST CANCER
Breast Cancer Survivorship: Strategies for Optimal Care
Issues in Breast Cancer Survivorship: Optimal Care, Bone Health, and Lifestyle Modifications
Michelle E. Melisko, William J. Gradishar, and Beverly Moy e22

The 2016 ASCO Educational Book is published online at asco.org/edbook. Articles can also be accessed
from the Attendee Resource Center at am.asco.org/ARC. Articles that are only available online are
denoted with an e ahead of the page number.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK iii


Less Is More: A Multidisciplinary Conversation on Treatment Options
Tailoring Chemotherapy in Early-Stage Breast Cancer: Based on Tumor Biology or Tumor Burden?
Domen Ribnikar and Fatima Cardoso e31

Targeted Therapies in Hormone ReceptorPositive Breast Cancer


Improving Response to Hormone Therapy in Breast Cancer: New Targets, New Therapeutic Options
Hope S. Rugo, Neelima Vidula, and Cynthia Ma e40

Treatment of Premenopausal Women With Endocrine-Sensitive Breast Cancer


Challenges in Treating Premenopausal Women with Endocrine-Sensitive Breast Cancer
Hatem A. Azim Jr., Nancy E. Davidson, and Kathryn J. Ruddy 23

Triple-Negative Breast Cancer: Is Change on the Horizon?


The Evolution of Triple-Negative Breast Cancer: From Biology to Novel Therapeutics
Carey K. Anders, Vandana Abramson, Tira Tan, and Rebecca Dent 34

Updates and Controversies in HER2-Positive Breast Cancer


A Value-Based Approach to Treatment of HER2-Positive Breast Cancer: Examining the Evidence
Nancy Nixon and Sunil Verma e56
Emerging Therapeutic Options for HER2-Positive Breast Cancer
Miguel Martin and Sara Lopez-Tarruella
e64

CANCER PREVENTION, HEREDITARY GENETICS, AND EPIDEMIOLOGY


Controversies in Genetic Evaluation
How Far Do We Go With Genetic Evaluation? Gene, Panel, and Tumor Testing
Filipa Lynce and Claudine Isaacs e72

Evolving Recommendations on Prostate Cancer Screening


Evolving Recommendations on Prostate Cancer Screening
Otis W. Brawley, Ian M. Thompson Jr., and Henrik Gronberg e80

Refining Breast Cancer Risk Assessment: Additional Genes, Additional Screening


Refining Breast Cancer Risk Stratification: Additional Genes, Additional Information
Allison W. Kurian, Antonis C. Antoniou, and Susan M. Domchek 44

CARE DELIVERY AND PRACTICE MANAGEMENT


Access to Cancer Therapeutics in Low- and Middle-Income Countries
Access to Cancer Therapeutics in Low- and Middle-Income Countries
Sana Al-Sukhun, Charmaine Blanchard, Lawrence N. Shulman, and Paul Ruff 58

iv 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


Integrating Patient-Reported Outcomes Into Real-Life Medical Decisions
Patient-Reported Outcomes in Cancer Clinical Trials: Measuring Symptomatic Adverse Events With
the National Cancer Institutes Patient-Reported Outcomes Version of the Common Terminology
Criteria for Adverse Events (PRO-CTCAE)
Paul G. Kluetz, Diana T. Chingos, Ethan M. Basch, and Sandra A. Mitchell 67

Optimizing Team-Based Oncology Care: Building the Village


Electronic Patient-Reported Outcomes: The Time Is Ripe for Integration Into Patient Care and
Clinical Research
Lee Schwartzberg e89
Oncology Advanced Practitioners Bring Advanced Community Oncology Care
Wendy H. Vogel e97

Quality and Value: Measuring and Utilizing Both in Your Practice


Why the Quality Oncology Practice Initiative Matters: Its Not Just About Cost
Anne C. Chiang e102

The Oncology Care Model: The Man Behind the Curtain


The Oncology Care Model: A Critique
Christian A. Thomas and Jeffrey C. Ward e109

CENTRAL NERVOUS SYSTEM TUMORS


Multidisciplinary Management of Brain Metastases
Immune Checkpoint Inhibitors in Brain Metastases: From Biology to Treatment
Anna S. Berghoff, Vyshak A. Venur, Matthias Preusser, and Manmeet S. Ahluwalia e116
Targeted Therapy in Brain Metastases: Ready for Primetime?
Vyshak A. Venur and Manmeet S. Ahluwalia e123

The Future of Immunotherapy in Glioblastoma


Current State of Immune-Based Therapies for Glioblastoma
Michael Lim, Michael Weller, and E. Antonio Chiocca e132

Treatment of Low-Grade Gliomas in the Era of Genomic Medicine


Treatment of Adult Lower-Grade Glioma in the Era of Genomic Medicine
Susan M. Chang, Daniel P. Cahill, Kenneth D. Aldape, and Minesh P. Mehta 75

DEVELOPMENTAL THERAPEUTICS AND TRANSLATIONAL RESEARCH


Biomarkers, Blood-Based Testing, and the Heterogeneous Tumor
Tumor Evolutionary Principles: How Intratumor Heterogeneity Influences Cancer Treatment
and Outcome
Subramanian Venkatesan and Charles Swanton e141

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK v


Biosimilars: Here and Now
Biosimilars: Here and Now
Steven J. Lemery, Francisco J. Esteva, and Martina Weise e151

Clinical Trial Eligibility Criteria: Tradition Versus Reality


Transforming Clinical Trial Eligibility Criteria to Reflect Practical Clinical Application
Edward S. Kim, Jennifer Atlas, Gwynn Ison, and Jennifer L. Ersek 83

Immunotherapy: Beyond Checkpoint Inhibitors


Arming the Immune System Through Vaccination to Prevent Cancer Recurrence
Diane F. Hale, Timothy J. Vreeland, and George E. Peoples e159
Overcoming Therapeutic Resistance by Targeting Cancer Inflammation
Gregory L. Beatty e168

Pharmacokinetics, Dynamics, and Genomics in the Era of Immunotherapy and Small Molecules
Pharmacogenomics, Pharmacokinetics, and Pharmacodynamics in the Era of Targeted Therapies
Emiliano Calvo, Christine Walko, E. Claire Dees, and Belen Valenzuela e175

GASTROINTESTINAL (COLORECTAL) CANCER


Potentially Resectable Metastatic Colorectal Cancer: A Multidisciplinary Discussion
Liver Metastases in Colorectal Cancer
Gunnar Folprecht e186

Questions on Treatment of Locally Advanced Rectal Cancer


Multimodal Rectal Cancer Treatment: In Some Cases, Less May Be More
Julio Garcia-Aguilar, Rob Glynne-Jones, and Deborah Schrag 92

GASTROINTESTINAL (NONCOLORECTAL) CANCER


Biliary Tract Cancer: The New and the Old
Biliary Tract Cancer: Epidemiology, Radiotherapy, and Molecular Profiling
John A. Bridgewater, Karyn A. Goodman, Aparna Kalyan, and Mary F. Mulcahy e194

Designing Clinical Trials to Achieve Breakthrough Results in Upper Gastrointestinal Malignancies


The Past, Present, and Future of Pancreatic Cancer Clinical Trials
Lynn M. Matrisian and Jordan D. Berlin e205

HER2, VEGFR, and Beyond: Genomic Profiling of Upper Gastrointestinal Tract Cancers and the Future of
Personalized Treatment
Gastric Adenocarcinoma: An Update on Genomics, Immune System Modulations, and Targeted Therapy
Jeeyun Lee, Adam J. Bass, and Jaffer A. Ajani 104

vi 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


Localized Pancreatic Cancer: Multidisciplinary Management With Incorporation of ASCO Guidelines
Localized Pancreatic Cancer: Multidisciplinary Management
Andrew L. Coveler, Joseph M. Herman, Diane M. Simeone, and E. Gabriela Chiorean e217

GENITOURINARY (NONPROSTATE) CANCER


Elderly Patients With Bladder Cancer: Perspectives From a Surgeon, Medical, and Radiation Oncologist
Treatment of Muscle-Invasive Bladder Cancer in Older Patients
Eila C. Skinner e228

Renal Cell Carcinoma: Systemic Treatment, Evolving TKIs, and Immuno-Oncology


The Evolution of Systemic Therapy in Metastatic Renal Cell Carcinoma
Thomas E. Hutson, Gregory R. Thoreson, Robert A. Figlin, and Brian I. Rini 113

GENITOURINARY (PROSTATE) CANCER


Contemporary Active Surveillance for Prostate Cancer: Do We Need Better Imaging and Molecular Testing?
Active Surveillance of Prostate Cancer: Use, Outcomes, Imaging, and Diagnostic Tools
Jeffrey J. Tosoian, Stacy Loeb, Jonathan I. Epstein, Baris Turkbey, Peter L. Choyke,
and Edward M. Schaeffer e235

Oligometastatic Disease in Prostate Cancer: Treating the Patient or the Scan?


Approach to Oligometastatic Prostate Cancer
Brandon Bernard, Boris Gershman, R. Jeffrey Karnes, Christopher J. Sweeney, and Neha Vapiwala 119

Precision Medicine in Advanced Prostate Cancer: Understanding Genomics, Androgen Receptor Splice
Variants, and Imaging Biomarkers
Emerging Molecular Biomarkers in Advanced Prostate Cancer: Translation to the Clinic
Himisha Beltran, Emmanuel S. Antonarakis, Michael J. Morris, and Gerhardt Attard 131

GYNECOLOGIC CANCER
Paradigm Shift in the Management Strategy for Epithelial Ovarian Cancer
Paradigm Shift in the Management Strategy for Epithelial Ovarian Cancer
Keiichi Fujiwara, Jessica N. McAlpine, Stephanie Lheureux, Noriomi Matsumura, and Amit M. Oza e247

Divide and Conquer: Epithelial Gynecologic Cancers Beyond BRCA


Gynecologic Cancers: Emerging Novel Strategies for Targeting DNA Repair Deficiency
Rebecca S. Kristeleit, Rowan E. Miller, and Elise C. Kohn e259

Intraperitoneal Chemotherapy for Ovarian Cancer: Trials and Tribulations


Update on Intraperitoneal Chemotherapy for the Treatment of Epithelial Ovarian Cancer
Charlie Gourley, Joan L. Walker, and Helen J. Mackay 143

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK vii


Neoadjuvant Chemotherapy: Location, Location, Location
Primary Surgery or Neoadjuvant Chemotherapy in Advanced Ovarian Cancer: The Debate
Continues
Alexandra Leary, Renee Cowan, Dennis Chi, Sean Kehoe, and Matthew Nankivell 153

Practical Challenges in Endometrial Cancer


Treatment of Older Women With Endometrial Cancer: Improving Outcomes With Personalized Care
Linda Duska, Armin Shahrokni, and Melanie Powell 164

Symptom Management for the Patient with Gynecologic Cancer


Enhancing Care of the Survivor of Gynecologic Cancer: Managing the Menopause and Radiation
Toxicity
Linda Van Le and Mary McCormack e270

HEAD AND NECK CANCER


Best of the Rest: Top Abstracts on Head and Neck Cancer From 20152016 Oncology Meetings
Whats New in Head and Neck Cancer: Key Findings in 20152016 From ECCO/ESMO, ASTRO,
and the Multidisciplinary Head and Neck Cancer Symposium
Sue S. Yom, Apar K. Ganti, and Andreas Dietz 176

Integrating Immune Checkpoint Inhibitors and Targeted Agents With Surgery and Radiotherapy for Patients
With Head and Neck Cancer
Immunotherapy and Checkpoint Inhibitors in Recurrent and Metastatic Head and Neck Cancer
Nooshin Hashemi Sadraei, Andrew G. Sikora, and David M. Brizel e277

Multimodality Management of Locoregionally Recurrent or Second Primary Head and Neck Cancer
Locoregional Recurrent or Second Primary Head and Neck Cancer: Management Strategies
and Challenges
Stuart J. Wong, Dwight E. Heron, Kerstin Stenson, Diane C. Ling, and John A. Vargo e284

HEALTH SERVICES RESEARCH AND QUALITY OF CARE


Defining and Measuring Quality
Challenges and Opportunities in Delivering High-Quality Cancer Care: A 2016 Update
Patricia A. Ganz, Michael J. Hassett, and David C. Miller e294

Removing Barriers to Clinical Trial Participation


The Role of Clinical Trial Participation in Cancer Research: Barriers, Evidence, and Strategies
Joseph M. Unger, Elise Cook, Eric Tai, and Archie Bleyer 185

Using Social Media, Wearables, and Electronic Medical Records to Improve Quality of Cancer Care
Using Technology to Improve Cancer Care: Social Media, Wearables, and Electronic Health Records
Michael J. Fisch, Arlene E. Chung, and Melissa K. Accordino 200

viii 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


HEMATOLOGIC MALIGNANCIESLEUKEMIA, MYELODYSPLASTIC SYNDROMES,
AND ALLOTRANSPLANT
Evolving Therapies in Acute Myeloid Leukemia: Progress at Last?
Evolving Therapies in Acute Myeloid Leukemia: Progress at Last?
Daniel J. DeAngelo, Eytan M. Stein, and Farhad Ravandi e302

Molecularly and Phenotypically Defined Subtypes of Acute Lymphoblastic Leukemia: Implications


for Management
Advances in the Genetics and Therapy of Acute Lymphoblastic Leukemia
Sabina Chiaretti, Valentina Gianfelici, Susan M. OBrien, and Charles G. Mullighan e314

Progress in Myeloproliferative Neoplasms: Are We Ready?


Individualizing Care for Patients With Myeloproliferative Neoplasms: Integrating Genetics, Evolving
Therapies, and Patient-Specific Disease Burden
Ruben A. Mesa and Francesco Passamonti e324

Why Are Myelodysplastic Syndromes So Difficult to Cure?


Integrating Frailty, Comorbidity, and Quality of Life in the Management of Myelodysplastic
Syndromes
Gregory A. Abel and Rena Buckstein e337
Searching for a Light at the End of the Tunnel? Beyond Hypomethylating Agents in Myelodysplastic
Syndromes
Rami S. Komrokji e345

HEMATOLOGIC MALIGNANCIESLYMPHOMA AND CHRONIC LYMPHOCYTIC LEUKEMIA


Current Management Concepts: Primary Central Nervous System Lymphoma, Natural Killer T-Cell Lymphoma
Nasal Type, Post-transplant Lymphoproliferative Disorder
Current Management Concepts: Primary Central Nervous System Lymphoma, Natural Killer T-Cell
Lymphoma Nasal Type, and Post-transplant Lymphoproliferative Disorder
Tracy T. Batchelor, Lim Soon Thye, and Thomas M. Habermann e354

PET-Directed Strategies in Lymphoma


Functional Imaging Using 18-Fluorodeoxyglucose PET in the Management of Primary Mediastinal
Large B-Cell Lymphoma: The Contributions of the International Extranodal Lymphoma Study Group
Franco Cavalli, Luca Ceriani, and Emanuele Zucca e368
Risk-Adapted Treatment of Advanced Hodgkin Lymphoma With PET-CT
Ryan C. Lynch and Ranjana H. Advani e376

Chemoimmunotherapy Versus Targeted Treatment in Chronic Lymphocytic Leukemia: When, How Long,
How Much, and in Which Combination?
Chemoimmunotherapy Versus Targeted Treatment in Chronic Lymphocytic Leukemia: When,
How Long, How Much, and in Which Combination?
Jennifer R. Brown, Michael J. Hallek, and John M. Pagel e387

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK ix


HEMATOLOGIC MALIGNANCIESPLASMA CELL DYSCRASIA
Consolidation and Maintenance: Moving Beyond Autotransplant
Moving Beyond Autologous Transplantation in Multiple Myeloma: Consolidation, Maintenance,
Allogeneic Transplant, and Immune Therapy
Amrita Krishnan, Ravi Vij, Jesse Keller, Binod Dhakal, and Parameswaran Hari 210

Defining and Redefining Myeloma


Future Directions in the Evaluation and Treatment of Precursor Plasma Cell Disorders
Salomon Manier, Karma Z. Salem, David Liu, and Irene M. Ghobrial e400
The Role of Imaging in the Treatment of Patients With Multiple Myeloma in 2016
Evangelos Terpos, Meletios A. Dimopoulos, and Lia A. Moulopoulos e407
Updated Diagnostic Criteria and Staging System for Multiple Myeloma
S. Vincent Rajkumar e418

Multiple Myeloma: Are We Ready for Personalized Therapy?


Minimal Residual Disease by Next-Generation Sequencing: Pros and Cons
Herve Avet-Loiseau e425
New Targets and New Agents in High-Risk Multiple Myeloma
Ajay K. Nooka and Sagar Lonial e431
Next-Generation Sequencing Informing Therapeutic Decisions and Personalized Approaches
Raphael Szalat and Nikhil C. Munshi e442

LUNG CANCER
Immunotherapy: Beyond AntiPD-1 and AntiPD-L1 Therapies
Immunotherapy: Beyond AntiPD-1 and AntiPD-L1 Therapies
Scott J. Antonia, Johan F. Vansteenkiste, and Edmund Moon e450

Local Therapies in the Management of Oligometastatic and Metastatic NonSmall Cell Lung Cancer
Local Therapy for Limited Metastatic NonSmall Cell Lung Cancer: What Are the Options and Is
There a Benefit?
Puneeth Iyengar, Steven Lau, Jessica S. Donington, and Robert D. Suh e460

Lung Cancer Screening and Prevention


Lung Cancer Screening With Low-Dose CT: Implementation Amid Changing Public Policy at One
Health Care System
Abbie Begnaud, Thomas Hall, and Tadashi Allen e468
Lung Cancer Screening, Cancer Treatment, and Addressing the Continuum of Health Risks Caused
by Tobacco
Graham W. Warren, Jamie S. Ostroff, and John R. Goffin 223

Lung Cancer, Small Cell Lung Cancer: On the Move (Again?)


Seeking New Approaches to Patients With Small Cell Lung Cancer
Marie Catherine Pietanza, Stefan Zimmerman, Solange Peters, and Walter J. Curran Jr. e477

x 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


Treatment of Lung Cancer in Medically Compromised Patients
Treatment of Lung Cancer in Medically Compromised Patients
Jeffrey Crawford, Paul Wheatley-Price, and Josephine Louella Feliciano e484

MELANOMA/SKIN CANCERS
Clinical Conundrums in Melanoma Therapy
Biomarkers for Immunotherapy: Current Developments and Challenges
Kristen R. Spencer, Jianfeng Wang, Ann W. Silk, Shridar Ganesan, Howard L. Kaufman,
and Janice M. Mehnert e493

Curing High-Risk Melanoma: Are We There Yet?


Surgical Management and Adjuvant Therapy for High-Risk and Metastatic Melanoma
Alexander C.J. van Akkooi, Michael B. Atkins, Sanjiv S. Agarwala, and Paul Lorigan e505

PATIENT AND SURVIVOR CARE


Cancer Survivorship: Is Every Patients Journey the Same?
The Cancer Survivorship Journey: Models of Care, Disparities, Barriers, and Future Directions
Michael T. Halpern, Mary S. McCabe, and Mary Ann Burg 231

Cancer Treatment as an Accelerated Aging Process


Cancer Treatment as an Accelerated Aging Process: Assessment, Biomarkers, and Interventions
Arti Hurria, Lee Jones, and Hyman B. Muss e516

New and Improved? Use of White Blood Cell Growth Factors in Oncology Practice
Real-World Conundrums and Biases in the Use of White Cell Growth Factors
Thomas J. Smith and Bruce E. Hillner e524
Issues on the Use of White Blood Cell Growth Factors in Oncology Practice
Gary H. Lyman e528

Optimizing Palliative and End-of-Life Care: Evidence-Based Practice Improvement


Discussing the Evidence for Upstream Palliative Care in Improving Outcomes in Advanced Cancer
Myles S. Nickolich, Areej El-Jawahri, Jennifer S. Temel, and Thomas W. LeBlanc e534

Rehabilitation of Patients and Survivors: Seizing the Opportunity


Developing High-Quality Cancer Rehabilitation Programs: A Timely Need
Catherine M. Alfano, Andrea L. Cheville, and Karen Mustian 241

PEDIATRIC ONCOLOGY
Controversies in Germline Genetic Testing and Disclosure in Pediatric Oncology
The Advantages and Challenges of Testing Children for Heritable Predisposition to Cancer
Chimene Kesserwan, Lainie Friedman Ross, Angela R. Bradbury, and Kim E. Nichols 251

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK xi


Exploiting Laboratory Insights to Improve Outcomes of Pediatric Central Nervous System Tumors
Exploiting Laboratory Insights to Improve Outcomes of Pediatric Central Nervous System Tumors
Giles W. Robinson, Hendrik Witt, and Adam Resnick e540

The State of the Art in Neuroblastoma: From Biology to Survivorship


Neuroblastoma: A Tough Nut to Crack
Frank Speleman, Julie R. Park, and Tara O. Henderson e548

PROFESSIONAL DEVELOPMENT
Curing Burnout in Oncology: Mindful Self-Compassion, Communication, and Practice
Addressing Burnout in Oncology: Why Cancer Care Clinicians Are At Risk, What Individuals Can Do,
and How Organizations Can Respond
Fay J. Hlubocky, Anthony L. Back, and Tait D. Shanafelt 271

SARCOMA
Is There a Future for Immunotherapy in Sarcoma?
Immunotherapy for Soft Tissue Sarcoma: Tomorrow Is Only a Day Away
Alex Lee, Paul Huang, Ronald P. DeMatteo, and Seth M. Pollack 281
Manipulating the Immune System With Checkpoint Inhibitors for Patients With Metastatic Sarcoma
Sandra P. DAngelo e558

Noncytotoxic Approaches to the Treatment of Metastatic Soft Tissue Sarcoma


Local Ablative Therapies to Metastatic Soft Tissue Sarcoma
Alessandro Gronchi, B. Ashleigh Guadagnolo, and Joseph Patrick Erinjeri e566

TUMOR BIOLOGY
Expanding the Clinically Actionable Cancer Genome
Interrogating the Cancer Genome to Deliver More Precise Cancer Care
Joaquin Mateo and Johann S. de Bono e577

Molecular Diagnostics in Cancer


Considerations for Implementation of Cancer Molecular Diagnostics Into Clinical Care
Daniel F. Hayes 292

Tumor Heterogeneity and Therapeutic Resistance


Tumor Heterogeneity and Therapeutic Resistance
Christine M. Lovly, April K.S. Salama, and Ravi Salgia e585

Whats Next in Cancer Immunotherapy?


Basic Overview of Current Immunotherapy Approaches in Cancer
Vamsidhar Velcheti and Kurt Schalper 298

xii 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


2016 ASCO Annual Meeting Disclosure
As the CME provider for the 2016 Annual Meeting, ASCO is committed to balance, objectivity, and scientific rigor in the management of
financial interactions with for-profit health care companies that could create real or perceived conflicts of interest. Participants in the
Meeting have disclosed their financial relationships in accordance with ASCOs Policy for Relationships with Companies; review the policy at
asco.org/rwc.

ASCO offers a comprehensive disclosure management system, using one disclosure for all ASCO activities. Members and participants in
activities use coi.asco.org to disclose all interactions with companies. Their disclosure is kept on file and can be confirmed or updated with
each new activity.

Please email coi@asco.org with specific questions or concerns.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK xiii


20152016 Cancer Education Committee
The Cancer Education Committee assesses the need for, plans, develops, and initiates the education programs for the Annual Meeting.

Apar Kishor Ganti, MD, MBBSChair GASTROINTESTINAL (NONCOLORECTAL) CANCER


Michael A. Thompson, MD, PhDChair-Elect Vincent J. Picozzi, MDTrack Leader
John Vernon Cox, DO, FACP, FASCO, MBAImmediate Past Chair Tanios S. Bekaii-Saab, MD
Lillian L. Siu, MDBoard Liaison Jimmy J. Hwang, MD
Se Hoon Park, MD
BREAST CANCER Manish A. Shah, MD
Hope S. Rugo, MDTrack Leader William Small, MD
Judy Caroline Boughey, MD
Jennifer A. Brown, MD
Raquel Nunes, MD GENITOURINARY (NONPROSTATE) CANCER
Debra A. Patt, MD, MPH, MBA Stephen Boyd Riggs, MDTrack Leader
Rinaa S. Punglia, MD Thomas E. Hutson, DO, PharmD
Elizabeth C. Reed, MD Jose A. Karam, MD
Tallal Younis, MBBCh, FRCP Ulka N. Vaishampayan, MD

CANCER PREVENTION, HEREDITARY GENETICS, GENITOURINARY (PROSTATE)


AND EPIDEMIOLOGY CANCER
P. Kelly Marcom, MDTrack Leader Himisha Beltran, MDTrack Leader
Margreet Ausems, MD, PhD Chris Parker, BA, BM Bchir, MD, FRCR, FRCP
Monique A. De Bruin, MD Edward M. Schaeffer, MD, PhD
Jeri Kim, MD Nicholas J. Vogelzang, MD, FASCO
Howard L. McLeod, PharmD
Electra D. Paskett, PhD GERIATRIC ONCOLOGY
Surendra Srinivas Shastri, MD, MBBS
Andrew S. Artz, MD, MSTrack Leader
William P. Tew, MD
CARE DELIVERY AND PRACTICE MANAGEMENT William Tse, MD
Jeffery C. Ward, MDTrack Leader
Aditya Bardia, MBBS, MPH
GYNECOLOGIC CANCER
Kelly Bugos, RN, ANP, MS
Helen Mackay, MDTrack Leader
Moshe C. Chasky, MD
Elise C. Kohn, MD
John Emmett Hennessy, CMPE, MBA
Linda Duska, MD
Lee Steven Schwartzberg, MD, FACP
Linda Van Le, MD

CENTRAL NERVOUS SYSTEM TUMORS


Manmeet Singh Ahluwalia, MDTrack Leader HEAD AND NECK CANCER
Eric L. Chang, MD Joseph Kamel Salama, MDTrack Leader
Nicole A. Shonka, MD John Truelson, MD
Roger Stupp, MD John A. Ridge, MD, PhD
Irina Veytsman, MD
CLINICAL TRIALS
Sumithra J. Mandrekar, PhDTrack Leader HEALTH SERVICES RESEARCH AND QUALITY OF CARE
Janet Dancey, MD Bruce Lee Jacobs, MDTrack Leader
Ethan M. Basch, MD
DEVELOPMENTAL THERAPEUTICS AND TRANSLATIONAL Daniel Jacob Becker, MD
RESEARCH Dawn L. Hershman, MD
Howard A. Burris, MD, FASCOTrack Leader Nicole Maria Kuderer, MD
Julia A. Beaver, MD Jennifer W. Mack, MD
Francisco J. Esteva, MD, PhD
John Charles Morris, MD HEMATOLOGIC MALIGNANCIESLEUKEMIA,
MYELODYSPLASTIC SYNDROMES,
GASTROINTESTINAL (COLORECTAL) CANCER AND ALLOTRANSPLANT
Donald A. Richards, MD, PhDTrack Leader Jorge E. Cortes, MDTrack Leader
Chris R. Garrett, MD Steven M. Devine, MD
Chi Lin, MD, PhD Jonathan Michael Gerber, MD
Ashwin Reddy Sama, MD Hanna Khoury, MD
Henry Q. Xiong, MD David Leibowitz, MD

xiv 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


HEMATOLOGIC MALIGNANCIESLYMPHOMA PEDIATRIC ONCOLOGY
AND CHRONIC LYMPHOCYTIC LYMPHOMA Tara O. Henderson, MD, MPHTrack Leader
Grzegorz S. Nowakowski, MDTrack Leader Gregory T. Armstrong, MD, MSCE
Matthew Alexander Lunning, DO Stewart Goldman, MD
Jeffrey Matous, MD Joel A. Weinthal, MD
John M. Pagel, MD, PhD
Michael A. Thompson, MD, PhD PROFESSIONAL DEVELOPMENT
Anne S. Tsao, MDTrack Leader
HEMATOLOGIC MALIGNANCIESPLASMA Kristin Anderson, MD, MPH
Kelly J. Cooke, DO
CELL DYSCRASIA Jill Gilbert, MD
Sagar Lonial, MDTrack Leader Laura Goff, MD
Asher Alban Chanan-Khan, MD Roberto Leon-Ferre, MD
Parameswaran Hari, MD
Irene M. Ghobrial, MD SARCOMA
Ravi Vij, MD Gary K. Schwartz, MDTrack Leader
L. Johnetta Blakely, MD
LUNG CANCER Robin Lewis Jones, MD, BSc, MBBS, MRCP
Martin J. Edelman, MDTrack Leader Min S. Park, MD, MS
Arkadiusz Z. Dudek, MD, PhD
Shirish M. Gadgeel, MD TUMOR BIOLOGY
Puneeth Iyengar, MD, PhD Kimryn Rathmell, MD, PhDTrack Leader
Craig H. Reynolds, MD Ravi Salgia, MD, PhD
David R. Spigel, MD Eliezer Mendel Van Allen, MD
Vamsidhar Velcheti, MD
MELANOMA/SKIN CANCERS
Sanjiv S. Agarwala, MDTrack Leader LIAISONS
Jason John Luke, MD, FACP Sana Al-Sukhun, MD, MScInternational Affairs Committee
Janice M. Mehnert, MD Howard Bailey, MDCancer Prevention Committee
Ahmad A. Tarhini, MD, PhD Gregg Franklin, MD, PhDClinical Practice Committee
Alexander Christopher Jonathan Van Akkooi, MD, PhD Melissa Johnson, MDCancer Research Committee
Thomas Leblanc, MD, MAEthics Committee
PATIENT AND SURVIVOR CARE Leonard Saltz, MDValue Task Force
Nagendra Tirumali, MDTrack Leader Manish Shah, MDClinical Practice Guidelines Committee
Deborah Mayer, PhD, RN, AOCN, FAAN Charles Shapiro, MDCancer Survivorship Committee
Kathi Mooney, PhD, RN William Tew, MDGeriatrics Special Interest Group
Maria Alma Rodriguez, MD Gina Villani, MDClinical Practice Guidelines Committee
Louise Christie Walter, BS, MD Ann Von Gehr, MDClinical Practice Guidelines Committee

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK xv


2016 Educational Book Expert Panel
The Expert Panel is a group of well-recognized physicians and researchers in oncology and related fields who have served as peer
reviewers of the ASCO Educational Book articles.

Ghassan K. Abou-Alfa, MD Emily K. Bergsland, MD Franco Cavalli, MD


Memorial Sloan Kettering Cancer Center, University of California, San Francisco University of Bellinzona
Weill Cornell Medical College
Axel Bex, MD, PhD Andrea Cercek, MD
Donald I. Abrams, MD The Netherlands Cancer Institute Memorial Sloan Kettering Cancer Center
San Francisco General Hospital
William Blum, MD Anthony Chan, MBBS, MD, FRCP, FHKCP,
Manmeet S. Ahluwalia, MD The Ohio State University FHKAM
Cleveland Clinic Sir YK Pao Centre for Cancer, Prince of Wales
Sharon Bober, PhD Hospital
Fabrice Andre, MD, PhD Dana-Farber Cancer Institute
Institut Gustave Roussy Alice Chen, MD
Diane Bodurka, MD, MPH National Cancer Institute at the National
Christina Annunziata, MD, PhD The University of Texas MD Anderson Institutes of Health
National Cancer Institute at the National Cancer Center
Allen M. Chen, MD
Insitutes of Health
Joann Bodurtha, MD, MPH University of California, Los Angeles
Stephen Maxted Ansell, MD Johns Hopkins University School of Medicine Stephen K.L. Chia, MD
Mayo Clinic BC Cancer Agency
George J. Bosl, MD
Frederick Appelbaum, MD Memorial Sloan Kettering Cancer Center Laura Quan Man Chow, MD
Fred Hutchinson Cancer Research Center University of Washington
Alba Brandes, MD
Saro Armenian, DO, MPH Bellaria Hospital Jessica Clement, MD
City of Hope University of Connecticut Health Center
Eduardo Bruera, MD
Gregory Armstrong, MD, MSCE The University of Texas MD Anderson Cancer Anthony Cmelak, MD
St. Jude Childrens Research Hospital Center Vanderbilt-Ingram Cancer Center

Christina Baik, MD, MPH Alan Haruo Bryce, MD Ezra Cohen, MD


Seattle Cancer Care Alliance Mayo Clinic University of California, San Diego

Lodovico Balducci, MD Jan C. Buckner, MD Robert Coleman, MD


Moffitt Cancer Center Mayo Clinic The University of Texas MD Anderson Cancer
Center
Carlos H. Barrios, MD Howard A. Burris, MD, FASCO
Pontifical Catholic University of Rio Grande Sarah Cannon Research Institute, Tennessee Frances Collichio, MD
do Sul School of Medicine Oncology The University of North Carolina at Chapel
Hill
Brigitta G. Baumert, MD, PhD, MBA Harold Burstein, MD, PhD, FASCO
Roisin Connolly, MBBCh
University of Bonn Medical Center Dana-Farber Cancer Institute
The Sidney Kimmel Comprehensive Cancer
Tanios S. Bekaii-Saab, MD Toby Christopher Campbell, MD Center at Johns Hopkins
The Ohio State University Comprehensive University of Wisconsin Carbone Cancer Rena M. Conti, PhD
Cancer Center Center The University of Chicago
Himisha Beltran, MD Beverly E. Canin Daniel Costa, MD, PhD
Weill Cornell Medical College Breast Cancer Options Beth Israel Deaconess Medical Center
Robert S. Benjamin, MD Lisa Carey, MD Carien L. Creutzberg, MD, PhD
The University of Texas MD Anderson The University of North Carolina at Leiden University Medical Center
Cancer Center Chapel Hill
Sia Daneshmand, MD
Michael F. Berger, PhD William L. Carroll, MD University of Southern California
Memorial Sloan Kettering Cancer Center NYU Langone Medical Center
Molly S. Daniels, MS, CGC
P. Leif Bergsagel, MD, FASCO Richard Carvajal, MD The University of Texas MD Anderson Cancer
Mayo Clinic Columbia University Medical Center Center

xvi 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


Jonas De Souza, MD James M. Foran, MD Neil N. Hayes, MD, MPH
The University of Chicago Mayo Clinic The University of North Carolina at Chapel
Hill
H. Joachim Deeg, MD James Ford, MD
Fred Hutchinson Cancer Research Center Stanford University Medical Center Bryan Hennessy, MD
The University of Texas MD Anderson Cancer
Egidio Del Fabbro, MD William Foulkes, MBBS, PhD Center
VCU Massey Cancer Center Jewish General Hospital
John Emmett Hennessy, MBA, CMPE
Pierre-Yves Dietrich, MD Jonathan Freidberg, MD, MMSc Sarah Cannon Research Institute
Hopitaux Universitaires de Geneve
` University of Rochester
Norah Henry, MD, PhD
Mary L. Disis, MD Richard R. Furman, MD University of Michigan
University of Washington Weill Cornell Medical College, New York-
Presbyterian Hospital Peter Hillmen, PhD
Tanya B. Dorff, MD
Leed University
USC Norris Comprehensive Cancer Center Leena Gandhi, MD, PhD
Dana-Farber Cancer Institute Jeffrey Hoch, PhD
Mitchell Dowsett, PhD
Cancer Care Ontario
The Royal Marsden NHS Foundation Trust David E. Gerber, MD
The University of Texas Southwestern Christine Holmberg, DPhil, MPH
Martin Dreyling, MD Medical Center Charite Universitatsmedizin Berlin
University Hospital Grosshadern
Peter Gibbs, MBBS, FRACP, MD Gabriel Hortobagyi, MD, FACP
Dan Duda, DMD, PhD The Royal Melbourne Hospital The University of Texas MD Anderson Cancer
Massachusetts General Hospital
Center
Sharon Hermes Giordano, MD, MPH
Reinhard Dummer, MD The University of Texas MD Anderson Cancer Clifford A. Hudis, MD, FACP
University Hospital Zurich Center Memorial Sloan Kettering Cancer Center
Linda R. Duska, MD Robert G. Gish, MD
University of Virginia Health System Kevin S. Hughes, MD, FACS
Stanford Medicine Harvard Medical School, Massachusetts
Grace K. Dy, MD Bernardo H.L. Goulart, MD, MS General Hospital
Roswell Park Cancer Institute Fred Hutchinson Cancer Research Center Kelly Hunt, MD
Craig Earle, MD, MSc Ramaswamy Govindan, MD The University of Texas MD Anderson Cancer
Institute for Clinical Evaluative Sciences Washington University School of Medicine in Center
St. Louis Maha Hussain, MD, FACP, FASCO
Martin J. Edelman, MD
University of Maryland School of Medicine Timothy Graubert, MD University of Michigan
Massachusetts General Hospital
Lawrence H. Einhorn, MD Thomas Hutson, DO, PharmD, FACP
Indiana University Melvin and Bren Simon Gordon Hafner, MD, FACS Texas Oncology, Baylor Charles A. Sammons
Cancer Center Inova Health System Cancer Center

Hatem El Halabi, MD Peter Hammerman, MD, PhD David H. Ilson, MD, PhD
Cancer Treatment Centers of America Dana-Farber Cancer Institute Memorial Sloan Kettering Cancer Center,
Weill Cornell Medical College
Marwan Fakih, MD Hans Hammers, MD, PhD
City of Hope Johns Hopkins University School of Medicine Syma Iqbal, MD
University of Southern California
Lesley Fallowfield, DPhil, BSc Nasser H. Hanna, MD
Sussex Health Outcomes Research and Indiana University Melvin and Bren Simon David Jablons, MD
Education in Cancer Cancer Center University of California, San Francisco
Michelle A. Fanale, MD Michelle Harvie, PhD Sekwon Jang, MD
The University of Texas MD Anderson Cancer Nightingale and Genesis Prevention Centre, Inova Comprehensive Cancer and Research
Center Wythenshawe Hospital Institute
Adele K. Fielding, MBBS, PhD Hege Haugnes, MD, PhD Michalina Janiszewska, PhD
University College London University Hospital of North Norway Dana-Farber Cancer Institute
Paul Graham Fisher, MD Axel Hauschild, MD Connie Jimenez,
PhD
Stanford University University of Kiel Vanderbilt University Medical Center
Gini F. Fleming, MD Gillian Haworth, MBBS Maxine S. Jochelson, MD
University of Chicago Medical Center Great Ormond Street Hospital for Children Memorial Sloan Kettering Cancer Center

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK xvii


Joseph Jurcic, MD Rami Manochakian, MD Ray D. Page, DO, PhD
Columbia University Medical Center Case Western Reserve University, Louis The Center for Cancer and Blood Disorders
Stokes Cleveland VA Medical Center
Matthew Katz, MD, FACS Sumanta Pal, MD
The University of Texas MD Anderson Cancer Mara-Victoria Mateos, MD, PhD City of Hope
Center University Hospital of Salamanca-IBSAL
Alberto Pappo, MD
Steven Katz, MD, MPH Rana McKay, MD St. Jude Childrens Research Hospital
University of Michigan Medical School Dana-Farber Cancer Institute
Chris Parker, FRCR, BA, MBBCh, MD
Stanley B. Kaye, MBBS, MRCP, MD, FRCP, Usha Menon, MD The Royal Marsden NHS Foundation Trust
FRCPS, FRSE, FMedSci University College London
Donald Williams Parsons, MD, PhD
The Institute of Cancer Research, The Royal
Paul A. Meyers, MD Texas Childrens Cancer Center, Baylor
Marsden NHS Foundation Trust
Memorial Sloan Kettering Cancer Center College of Medicine
Andrew Kennedy, MD, FACRO Lynne Penberthy, MD, MPH
Linda R. Mileshkin, MBBS, MD, MBioeth
Sarah Cannon Research Institute National Cancer Institute at the National
Peter MacCallum Cancer Centre
Institutes of Health
Gretchen Kimmick, MD
Matthew Milowsky, MD
Duke University Medical Center Vincent J. Picozzi, MD
The University of North Carolina at Chapel
Hill Virginia Mason Medical Center
Hedy Kindler, MD
The University of Chicago Blase N. Polite, MD, MPP
Shanu Modi, MD
Memorial Sloan Kettering Cancer Center The University of Chicago
Heidi D. Klepin, MD, MS
Wake Forest University School of Medicine Sandro V. Porceddu, MD
Ana Molina, MD
Weill Cornell Medical College Princess Alexandra Hospital
Theresa Koppie, MD
Oregon Health & Science University Michael Andrew Postow, MD
Alison Moliterno, MD
Johns Hopkins University Memorial Sloan Kettering Cancer Center
Thomas Krivak, MD
Magee-Womens Hospital of UMPC Thomas Powles, MD
Silvio Monfardini, MD
Barts Health
Istituto Palazzolo Fondazione Don Gnocchi
Ian Krop, MD, PhD
Dana-Farber Cancer Institute Kumar Prabhash, MD, DM
Halle C.F. Moore, MD
Tata Memorial Centre
Cleveland Clinic
Geoff Ku, MD, MBA
Memorial Sloan Kettering Cancer Center Amanda Psyrri, MD, PhD
Kathleen N. Moore, MD
Attikon Hospital National Kapodistrian
The University of Oklahoma Health Sciences
Shaji Kumar, MD University of Athens
Center
Mayo Clinic
Cornelis J.A. Punt, MD, PhD
Philippe Moreau, MD University of Amsterdam
Stephen Yenzen Lai, MD, PhD
CHU de Nantes, Hotel DieuHME
The University of Texas MD Anderson Cancer
Mark J. Ratain, MD
Center Lindsay Morton, PhD The University of Chicago
National Cancer Institute at the National
James M.G. Larkin, MD, PhD Institutes of Health John Ridge, MD, PhD
The Royal Marsden NHS Foundation Trust
Fox Chase Cancer Center
Michael Neuss, MD
Jonathan Ledermann, MD, FRCP Vanderbilt-Ingram Cancer Center Danny Rischin, MBBS, MD, FRACP
University College London Cancer Institute Peter MacCallum Cancer Centre
Lee Nisley Newcomer, MD
Stuart M. Lichtman, MD United Health Group Miriam B. Rodin, MD, PhD
Memorial Sloan Kettering Cancer Center Saint Louis University School of Medicine
Grzegorz S. Nowakowski, MD
Nancy Lin, MD Mayo Clinic Julia Rowland, PhD
Dana-Farber Cancer Institute National Cancer Institute at the National
Olatoyosi Odenike, MD Institutes of Health
Tracy Lively, PhD The University of Chicago
National Cancer Institute at the National David P. Ryan, MD
Institutes of Health Francesco Onida, MD Massachusetts General Hospital
University of Milan
Ravi Madan, MD Gilles A. Salles, MD, PhD
National Cancer Institute at the National Eric Padron, MD Hospices Civils de Lyon, Universite Claude
Institutes of Health Moffitt Cancer Center Bernard

xviii 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


Hanna Kelly Sanoff, MD, MPH Zsofia K. Stadler, MD Catherine H. Van Poznak, MD
University of North Carolina at Chapel Hill Memorial Sloan Kettering Cancer Center University of Michigan Comprehensive
School of Medicine Cancer Center
Stephan Stilgenbauer, MD
Gary K. Schwartz, MD University Hospital Ulm Brian Van Tine, MD, PhD
Columbia University Medical Center Washington University School of Medicine in
Richard Sullivan, PhD St. Louis
David W. Scott, MBCHB, PhD
Kings Health Partners Integrated Cancer
BC Cancer Agency Anna Varghese, MD
Centre
Memorial Sloan Kettering Cancer Center
Tanguy Y. Seiwert, MD
The University of Chicago Joel Tepper, MD Dian Wang, MD, PhD
The University of North Carolina at Chapel Rush University Medical Center
Charles L. Shapiro, MD Hill
Ichan School of Medicine at Mount Sinai Lari B. Wenzel, PhD
William P. Tew, MD University of California, Irvine
Liran Shlush, MD, PhD Memorial Sloan Kettering Cancer Center,
Princess Margaret Cancer Centre Weill Cornell Medical College William Wierda, MD, PhD
The University of Texas MD Anderson Cancer
Marc Shuman, MD Center
Mike Thompson, MD, PhD
USCF Helen Diller Comprehensive Cancer
Aurora Clinic
Center Peter Yu, MD
Palo Alto Medical Foundation
Sonali Smith, MD Andreas Ullrich, MD, MPH
The University of Chicago World Health Organization Andrew Zelenetz, MD, PhD
Memorial Sloan Kettering Cancer Center
John David Sprandio, MD Saad Zafar Usmani, MD
Consultants in Medical Oncology and Levine Cancer Institute/Carolinas Healthcare Clive Zent, MD
Hematology System University of Rochester

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK xix


2016 Annual Meeting Supporters*
MISSION ENDOWMENT FOUNDING DONORS
Genentech BioOncology
GlaxoSmithKline Oncology
Novartis Oncology
Sanofi Oncology

MISSION ENDOWMENT SUSTAINING DONORS


AbbVie, Inc.
Amgen
Astellas
AstraZeneca
Celgene Corporation
Eisai Inc.
Gilead Sciences, Inc.
HELSINN
Incyte Corporation
Lilly
Onyx Pharmaceuticals
Sanofi Oncology
Takeda Oncology

ENDOWED FUND DONORS


Anonymous
Sally Gordon YIA Endowment Fund
Conquer Cancer Foundation of ASCO Young Investigator Award, in memory of Sally Gordon
American Society of Clinical Oncology
Jane C. Wright, MD, YIA Endowment Fund
Jane C. Wright, MD, Young Investigator Award
Friends, Family, and Colleagues of Dr. Ronald Beller and Mrs. Judith Beller
Bradley Stuart Beller Endowment Fund
Bradley Stuart Beller Merit Award
Breast Cancer Research Foundation
Evelyn H. Lauder YIA Endowment Fund
Conquer Cancer Foundation of ASCO Young Investigator Award, in memory of Evelyn H. Lauder
Celgene Corporation
John R. Durant, MD, YIA Endowment Fund
Conquer Cancer Foundation of ASCO Young Investigator Award, in memory of John R. Durant, MD
GlaxoSmithKline Oncology
Gianni Bonadonna Award and Lecture Endowment Fund
Gianni Bonadonna Breast Cancer Award and Fellowship
HELSINN
Anna Braglia YIA Endowment Fund
Anna Braglia Young Investigator Award in Cancer Supportive Care, supported by HELSINN
Friends, Family, and Colleagues of Dr. James B. Nachman
James B. Nachman Pediatric Oncology Fund
James B. Nachman ASCO Junior Faculty Award in Pediatric Oncology
Aaron and Barbro Sasson
Bertil Eriksson YIA Endowment Fund
Ake
Bertil Eriksson Endowed Young Investigator Award
Ake

* This list reflects commitments as of April 18, 2016.

xx 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


CANCERLINQ MAJOR SUPPORTERS Bayer HealthCare Pharmaceuticals Inc. Merck & Co., Inc.
Amgen Genitourinary Cancer Tracks Bundle Annual Meeting Proceedings and
Astellas Educational Book Bundle
Boehringer Ingelheim Pharmaceuticals, Inc.
AstraZeneca Best of ASCO Meetings
Best of ASCO Meetings
Bayer HealthCare Pharmaceuticals Inc. Breast Cancer Track
Lung Cancer Track
Boehringer Ingelheim Pharmaceuticals, Inc. Development Therapeutics and Translational
Cancer Treatment Centers of America Bristol-Myers Squibb Research/Clinical Trials Tracks Bundle
Chan Soon-Shiong Family Foundation Pre-Annual Meeting Seminar: Hematology Gastrointestinal (Colorectal) Cancer Track
Genentech BioOncology for the Oncologist Gastrointestinal (Noncolorectal) Cancer Track
HELSINN Genitourinary Cancer Tracks Bundle
Celgene Corporation
Janssen Oncology Head and Neck Cancer Track
Breast Cancer Track
Lilly Hematologic Malignancies Tracks Bundle
Gastrointestinal (Noncolorectal) Cancer
Raj Mantena, RPh Lung Cancer Track
Track
Novartis Oncology Melanoma/Skin Cancers Track
Hematologic Malignancies Tracks Bundle
Pfizer Oncology Pre-Annual Meeting Seminar: How to
Lung Cancer Track
Thomas G. Roberts Jr., MD, and Susan M. Integrate Tumor Immunotherapy Into Your
DaSilva Conquer Cancer Foundation & Conquer Clinical Practice
Susan G. Komen Cancer Foundation Mission Endowment Merrimack Pharmaceuticals
Cancer Survivorship Symposium Gastrointestinal (Noncolorectal) Cancer
EDUCATIONAL SUPPORT
Ferring Pharmaceuticals Track
Ambry Genetics Genitourinary Cancer Tracks Bundle Novartis Oncology
Cancer Prevention, Hereditary Genetics, Best of ASCO Meetings
and Epidemiology Track Genentech BioOncology
Breast Cancer Track
Pre-Annual Meeting Seminar: Genetics Breast Cancer Track
Gastrointestinal (Noncolorectal) Cancer
and Genomics for the Practicing Lung Cancer Track
Track
Clinician Gilead Sciences, Inc. Hematologic Malignancies Tracks Bundle
Best of ASCO Meetings Melanoma/Skin Cancers Track
Amgen
Development Therapeutics and Translational Pre-Annual Meeting Seminar: Hematology
Annual Meeting Proceedings and
Research/Clinical Trials Tracks Bundle for the Oncologist
Educational Book Bundle
Hematologic Malignancies Tracks Bundle Pre-Annual Meeting Seminar: How to
Best of ASCO Meetings
Integrate Tumor Immunotherapy Into Your
Gastrointestinal (Colorectal) Cancer HELSINN
Clinical Practice
Track Patient and Survivor Care/Health Services
Pre-Annual Meeting Seminar: New Drugs in
Hematologic Malignancies Tracks Research and Quality of Care Tracks Bundle
Oncology
Bundle
Incyte Corporation Patient Access Network Foundation
Melanoma/Skin Cancers Track
Pre-Annual Meeting Seminar: How to Annual Meeting Patient Advocate
Pre-Annual Meeting Seminar: How to
Integrate Tumor Immunotherapy Into Your Scholarship Program (2)
Integrate Tumor Immunotherapy Into
Clinical Practice
Your Clinical Practice Pharmacyclics LLC
Pre-Annual Meeting Seminar: New Drugs Janssen Oncology Hematologic Malignancies Tracks Bundle
in Oncology Patient and Survivor Care/Health Services
Pre-Annual Meeting Seminar: The TAIHO Oncology, Inc.
Research and Quality of Care Tracks Bundle
Economics of Cancer Care Gastrointestinal (Colorectal) Cancer Track
Kidney Cancer Association Pre-Annual Meeting Seminar: New Drugs in
Astellas Genitourinary Cancers Track Oncology
Best of ASCO Meetings
Lilly TESARO
Genitourinary Cancer Tracks Bundle
Best of ASCO Meetings Best of ASCO Meetings
Astellas and Genentech BioOncology Breast Cancer Track Gynecologic Cancer Track
Lung Cancer Track Development Therapeutics and Translational
Research/Clinical Trials Tracks Bundle GENERAL SUPPORT
AstraZeneca Gastrointestinal (Colorectal) Cancer Track
Best of ASCO Meetings Head and Neck Cancer Track AbbVie, Inc.
Lung Cancer Track Lung Cancer Track Annual Meeting iPlanner Mobile Application
Pre-Annual Meeting Seminar: How to Pre-Annual Meeting Seminar: New Drugs in and Website Bundle
Integrate Tumor Immunotherapy Into Your Oncology ASCO Live
Clinical Practice Sarcoma Track Young Investigator Award

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK xxi


Adenoid Cystic Carcinoma Research Bristol-Myers Squibb EMD Serono Inc.
Foundation Annual Meeting Onsite Connectivity Young Investigator Award
Career Development Award in Adenoid Resources Bundle
Fibrolamellar Cancer Foundation
Cystic Carcinoma Research Patient Advocate Scholarship Program Bundle
Young Investigator Award in Fibrolamellar
American Cancer Society Celgene Corporation Research
ASCO/American Cancer Society Award and Merit Awards
Patient Advocate Scholarship Program Bundle Florida Society of Clinical Oncology
Lecture
Young Investigator Award Florida-Based
Amgen The Cholangiocarcinoma Foundation Recipient
Career Development Award Young Investigator Award in
Diversity in Oncology Bundle Gateway for Cancer Research
Cholangiocarcinoma Research (1)
International Innovation Grant Young Investigator Award
Long-Term International Fellowship Coalition of Cancer Cooperative Groups
Genentech BioOncology
Merit Awards Clinical Trials Participation Award
Career Development Award (4)
Oncology Trainee Travel Award Patient Advocate Scholarship Program
CRDF Global, National Cancer Institute, and
Patient Advocate Scholarship Program Bundle Bundle
National Science Foundation
Young Investigator Award (2) Young Investigator Award (7)
International Development and Education
ASCO and Conquer Cancer Foundation Award in Palliative Care Gilead Sciences, Inc.
Boards of Directors Annual Meeting Onsite Connectivity
Young Investigator Award, in memory of Conquer Cancer Foundation
Resources Bundle
Jane C. Wright, MD Cancer.Net
Merit Awards
Career Development Award
Young Investigator Award (2)
ASCO State Affiliate Council and ASCO Guidelines USB
Clinical Practice Committee Journal of Global Oncology Fellows Program Guardant Health, Inc.
Young Investigator Award Long-Term International Fellowship (2) Annual Meeting iPlanner Mobile Application
Medical Student Rotation for and Website Bundle
Astellas
Underrepresented Populations
ASCO Meeting Library Bundle HELSINN
Merit Awards (2)
Career Development Award Annual Meeting iPlanner Mobile Application
Patient Advocate Scholarship Program
AstraZeneca and Website Bundle
Patient Materials
Merit Awards Foundation General Mission Support
Survivorship Forum
Young Investigator Award (2) Young Investigator Award Infinity Pharmaceuticals Inc.
Avon Foundation for Women Annual Meeting iPlanner Mobile Application
Conquer Cancer Foundation Mission and Website Bundle
International Development and Education
Endowment
Award in Breast Cancer Janssen Oncology
ASCO Community Research Forum Award
ASCO Policy Fellowship Diversity in Oncology Bundle
Bayer HealthCare Pharmaceuticals Inc.
ASCO University Maintenance of Merit Awards
International Development and Education
Certification Application Young Investigator Award (2)
Award
Cancer.Net
Janssen Scientific Affairs, LLC
Boehringer Ingelheim Pharmaceuticals, Inc. International Development and Education
Career Development Award in Deep Vein
Annual Meeting Onsite Connectivity Award in Palliative Care
Thrombosis
Resources Bundle Journal of Clinical Oncology Figures Program
Young Investigator Award in Deep Vein
Annual Meeting Special Attendees Lounge Journal of Global Oncology
Thrombosis
Bundle Journal of Oncology Practice, Special Series
Patient Advocate Scholarship Program Bundle Long-Term International Fellowship (2) Journal of Clinical Oncology
Young Investigator Award (2) NCI-ASCO Teams in Cancer Care Delivery Young Investigator Award
Breast Cancer Research Foundation Project
Kidney Cancer Association
Advanced Clinical Research Award in Breast patientACCESS
Annual Meeting Merit Awards in Kidney
Cancer Quality Measurement System
Cancer
Career Development Award in Breast Cancer (2) Resident Travel Award for
Young Investigator Award in Kidney
Comparative Effectiveness Research Underrepresented Populations
Cancer
Professorship in Breast Cancer Eisai Inc.
Young Investigator Award in Breast Cancer, Annual Meeting Onsite Connectivity The John and Elizabeth Leonard Family
in honor of Susan Hirschhorn and in Resources Bundle Foundation
memory of her mother Young Investigator Award (2) Young Investigator Award

xxii 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


Lilly Strike 3 Foundation
Annual Meeting Special Attendees Lounge Young Investigator Award in Pediatric
Bundle Oncology (2)
Career Development Award
Takeda Oncology
Medical Student Rotation for
ASCO Meeting Library Bundle
Underrepresented Populations
Career Development Award
Merit Awards
International Development and Education
Patient Advocate Scholarship Program
Award
Bundle
Merit Awards
Young Investigator Award (2)
Oncology Trainee Travel Award
Lung Cancer Alliance Young Investigator Award (3)
Young Investigator Award in Lung Cancer
Teva Oncology
J. Edward Mahoney Foundation Patient Advocate Scholarship Program
Young Investigator Award Bundle

McHenry and Lisa Tichenor Fund of Triple Negative Breast Cancer Foundation
Communities Foundation of Texas Career Development Award
Young Investigator Award in Sarcoma Young Investigator Award
Research, in memory of Willie Tichenor
The WWWW Foundation Inc. (QuadW) and
Medivation The Sarcoma Fund of the QuadW
Annual Meeting Special Attendees Lounge Foundation of Communities Foundation of
Bundle Texas
Young Investigator Award in Sarcoma
Merck & Co., Inc.
Research, in memory of Willie Tichenor
Annual Meeting Program Publications
Bundle
ASCO Meeting Library Bundle
Young Investigator Award (4)

NF Nimeh, MD
International Development and Education
Award in Palliative Care
Novartis Oncology
Merit Awards
Young Investigator Award

Susan K. Parsons, MD, and Walter


Armstrong
Young Investigator Award

Pfizer Oncology
Young Investigator Award

Roche
Career Development Award
Long-Term International Fellowship

Reid R. Sacco Adolescent and Young Adult


Alliance
Young Investigator Award

Sanofi Foundation for North America


Foundation General Mission Support

Sanofi Oncology
Drug Development Research Professorship

Spectrum Pharmaceuticals
Merit Awards

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK xxiii


Letter From the Editor
O n behalf of my Associate Editor, Dr. Nate Pennell, I welcome you to the 2016 American Society of Clinical Oncology (ASCO)
Annual Meeting. It is also my distinct honor to present the 36th volume of the NLM-indexed ASCO Educational Book. This
years theme, Collective Wisdom: The Future of Patient-Centered Care and Research, sits at the heart of what we aim to do as
oncologists: improve the lives of patients with cancer. Harnessing our combined knowledge, not just the expertise of one, is how
we must deliver the highest quality of personalized care to meet the diverse needs of our patients.

Dr. Julie Vose, 20152016 ASCO President, challenged us to bring our readers a volume that reflected the multidisciplinary
aspects of cancer care, representing not only the roles of the many different types of health care providers and their specialties,
but also topics such as quality, cost, and survivorship. We answered by requesting articles authored with this interdisciplinary
view in mind, and I am pleased to say that we have more co-authored articles than ever before represented in this years
volume. To be sure, the level of effort required for this impressive collaboration of minds was no small feat, and I humbly thank
the more than 100 authors who generously took the time to write and, in some cases, revise the articles in this volume.

In the same vein, the Invited Articles in this volume illustrate how, as our knowledge of cancer evolves, we must include
various areas of expertise to transform how we deliver care to our patients, such as enlisting genetic counselors and enhancing
the role of advanced practitioners. Beyond that, applying our collective wisdom into practice also means expanding com-
munication; we must incorporate results of molecular biology into treatment plans and converse honestly with patients about
treatment costs and care expectations, among other strategies. I graciously thank the authors of these articles for their
contributions to this volume.

This year, our readers will also find a new type of article that we are calling Points of View. Articles published under this
banner describe emerging and/or controversial topics in oncology care. I believe these articles are a most valuable addition to
the volume and highlight important care considerations for our patients as well as for the field of oncology.

Lastly, I want to recognize and thank our truly remarkable expert panel who dedicated their time and effort to careful,
thorough, and thoughtful reviews. Your commitment to education and ASCOs mission cannot be highlighted enough and is
tremendously underscored by this years theme.

It is my honor to invite you to read through the exceptional contributions that comprise the 2016 volume, only a selection of
which are found in the print edition. The print edition includes a curated selection of articles that most embody the power our
collective wisdom. For access to all of the 2016 ASCO Educational Book articles, as well as access to past volumes, please visit
www.asco.org/edbook.

Nate and I welcome your feedback and suggestions on how we can improve the content, so please contact us at edbook@
asco.org with your comments.

Sincerely,

Don S. Dizon, MD
Editor in Chief

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 1


INVITED ARTICLES
This years invited articles represent the 2016 ASCO Annual Meeting theme, Collective Wisdom: The Future
of Patient-Centered Care and Research. These important contributions to the 36th volume of the ASCO
Educational Book focus on the significance of a collaborative approach to how we provide care for our
patients. The authors represent a diverse, multidisciplinary set of expertise and backgrounds.

AUTHORS
Michael P. Kosty, MD, FACO, FASCO
Scripps Clinic
La Jolla, CA

Derek Raghavan, MD, PhD, FACP, FRACP, FASCO


Levine Cancer Institute, Carolinas HealthCare System
Charlotte, NC

Mary E. Sabatini, MD, PhD


Massachusetts General Hospital
Boston, MA

George W. Sledge, MD, FASCO


Stanford University School of Medicine
Stanford, CA
INVITED ARTICLES COLLECTIVE WISDOM

Collaborative Practice in an Era of Multidisciplinary Care


Michael P. Kosty, MD, FACP, FASCO, Todd Pickard, MMSc, PA-C, and Pamela Viale, RN, MS, CNS, ANP

T he clinical practice of oncology has become increasingly


complex. An explosion of medical knowledge, increased
demands on provider time, and involved patients have
Clear communication and transparent, defined roles and
responsibilities help ensure that care needs are addressed
and timely decisions are made. Lasting change can only come
necessitated changes in the way many oncologists practice. from explicitly helping to transform individual clinicians and
What was an acceptable practice model in the past may separate groups into a team that works together.
now be relatively inefficient. The American Society of Clinical Teams are defined as two or more people who interact
Oncologys (ASCOs) annual National Oncology Census dem- dynamically, interdependently, and adaptively to accom-
onstrates that with each successive year there is a shift plish a shared goal.7 The field of primary care has actively
away from small independent group practice to larger in- engaged in reinventing care to form a patient-centered
dependent groups or hospital-based affiliations.1-3 It is medical home.8-10 Experiments are underway to apply the
unclear whether there will be further consolidation and/or same concepts to specialty care delivery.11-16 At the same
shift, or whether the rate of change will accelerate or time, public and private payers and ASCO are proposing
continue at its current pace. Ultimately, understanding the payment models that would move away from payment
practice environment surrounding providers who care for based on specific procedures and physician contributions
patients with cancer is important to ensure that increasingly toward an approach that provides bundled payments for
complex therapies are delivered in an efficient, value-based comprehensive care and allows greater flexibility in how
setting and that all patients have access to high-quality care is organized and delivered.17 A team-based approach
cancer care. can potentially leverage these changes, provide an oppor-
The 2015 ASCO State of Cancer Care in America report tunity to reexamine clinician roles and responsibilities, and
emphasized practice trends: workforce composition, health may enable the most efficient delivery of high-quality health
systems innovation, regulatory compliance, and the financial care services. Ongoing education, training, mentorship,
realities of cancer care today.4 Among the most pressing networking, and communication are necessary to cultivate
issues the report highlighted was a growing population of and maintain a collaborative team-based practice model.
patients with cancer, increased complexity of care provided, Integration of resources from each practice setting, com-
and an oncology workforce projected to fall short of the munity organizations, e-health technologies, and advocacy
expected demand (Fig. 1). In a recent survey of 22,000 groups is essential. Human factors, health system factors,
oncologists, 11,700 medical oncologists were estimated to situational factors, and socioeconomic factors are constantly
provide direct care, managing the majority of patients with changing within the continuum of care, and they must be
cancer over extended periods of time.5 Factors contributing considered in designing tailored patient and caregiver
to this predicted shortfall of providers and increasing support (Fig. 2).6 The National Cancer Institute (NCI) and
complexity of cancer care delivery are shown in Sidebar 1.6 ASCO have collaborated to bring clinicians, patient advo-
Efficient multimodality therapy requires coordinated care cates, and researchers together to explore application of the
delivery among many diverse groups of clinical providers as evidence of team effectiveness to clinical practice. This
patients move along the continuum of cancer carefrom project fits into the larger context of the transformation of
risk assessment, prevention, diagnosis, treatment, and sur- health care delivery and payment models. The NCI-ASCO
veillance to survivorship and care of advanced cancer. Teams in Cancer Care Delivery project was presented at
Within any given clinic, increasingly complex therapies the NCI-ASCO Teams in Cancer Care Delivery Workshop
require clinicians with advanced, specialized training. in February 2016. Their findings will be published in the

From the Scripps Green Cancer Center, Scripps Clinic, La Jolla, CA; The University of Texas MD Anderson Cancer Center, Houston, TX; Department of Physiological Nursing, University of
California, San Francisco, San Francisco, CA.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Michael P. Kosty, MD, FACP, FASCO, Scripps Green Cancer Center, Scripps Clinic, 10666 North Torrey Pines Rd., MS217, La Jolla, CA 92037; email: mkosty@
scripps.edu.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 3


KOSTY, PICKARD, AND VIALE

FIGURE 1. Baseline Projected Supply of and Demand Shulman18 suggested several goals for integrating advanced
for Oncologist Visits, 2005 to 20205 practice providers (APPs) into collaborative practice
in oncology: (1) improved patient care, (2) increased
clinical productivity, (3) improved access for patients,
(4) urgent care patient treatment, (5) long-term care of
patients with cancer, and (6) coverage for the academic
physician. 6 The oncology APP is a licensed health care
professional who has completed advanced training in
nursing or pharmacy or has completed training as a physician
assistant.
The use of APPs to help meet the workforce demands and
improve the quality and coordination of care has been one of
the proposed solutions since the ASCO board of directors
approved the initial workforce strategic plan in 2008. The
utilization of APPs in oncology practices can increase
practice efficiency, productivity, and professional satisfac-
tion of collaborating oncologists.19 The APPs role is diverse
and includes both inpatient and outpatient settings. Recent
Journal of Oncology Practice. In the context of increasingly publications have highlighted the practice of physician as-
complex care requirements, increasing demand, and de- sistants who provide intensive palliative care on inpatient
creasing physician and economic resources, how do we create a units through an interdisciplinary team model.20 Similarly,
collaborative team-based practice model that allows for the nurse practitioners have published their experiences as an
provision of high-quality, value-based oncology care? inpatient malignant hematology nurse practitioner service
working within a collaborative and multidisciplinary model.21
The utilization of APPs on an allogeneic stem cell transplant unit
INTEGRATING ADVANCED PRACTICE PROVIDERS resulted in decreased length of stay and reduced hospital
INTO ONCOLOGY PRACTICE charges with similar patient outcomes.22 Towle and col-
The cost of care is a primary concern in oncology today. leagues23 suggested similar roles for the APP in a collaborative
Collaborative practice models that provide mecha- practice model. These included (1) assisting patients during
nisms for revenue generation and reduce unnecessary treatment visits; (2) pain and symptom management; (3)
costs to patients through application of clinical practice follow-up care for patients in remission (survivorship care); (4)
guidelines will promote patient and provider satisfaction. patient education and counseling; (5) end-of-life care; and (6)

SIDEBAR 1. Factors Affecting the Demand and Complexity of Cancer Care6


1. Implementation of the Affordable Care Act has resulted in an increasing number of individuals gaining access to health care.
2. An aging population has Medicare as their primary insurance.
3. The number of cancer survivors is growing related to improvements in cancer detection, risk-adapted treatment strategies,
supportive care, and palliative care.
4. Increasing costs of care are resulting in a shift in practice models and the integration of formalized programs for preauthorization
and reimbursement.
5. The oncology workforce is aging (50% over the age of 50), with a shift toward group practices in urban settings (. 90%).
6. Cancer care initiatives are being set as standards of care or required for certification necessary to achieve designation or improve
revenue.
7. Meaningful Use benchmarks are being developed for the use of electronic health records and patient-reported outcomes and their
impact on physician productivity.
8. The American College of Surgeons Commission on Cancer published Cancer Program Standards 2012: Ensuring Patient-Centered
Care, which established new requirements around patient-centered needs and is expanding the focus on improving the quality of
care and patient outcomes. More recently, the Commission on Cancer has set a standard for distress screening for every patient
with cancer and their caregivers across the continuum of care.
9. Survivorship care: The Institute of Medicine, ASCO, and the Commission on Cancer have set guidelines for survivorship care. Cancer
survivors are projected to exceed 19 million by 2024.
10. Palliative care: The Institute of Medicine released its report, Improving Palliative Care for Cancer, in 2000. ASCO published
a provisional clinical opinion in 2012, recommending that palliative care be integrated into the care of every patient with cancer at
the time of diagnosis. The National Consensus Project put forth its Clinical Practice Guidelines for Palliative Care, a set of nationally
recognized guidelines that include quality measures and the eight domains of palliative care. The National Comprehensive Cancer
Network published the first clinical practice guidelines for palliative care in 2013.

4 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


COLLABORATIVE PRACTICE IN AN ERA OF MULTIDISCIPLINARY CARE

FIGURE 2. Collaborative Practice Relationships can help meet future demand is unclear because the current
and projected workforce of APPs in oncology has been
challenging to determine. Based on census data from 2013,
the American Academy of Physician Assistants (AAPA) re-
ported that there were an estimated 2,140 practicing
physician assistants in adult medical, surgical, and radiation
oncology subspecialties.2 This represented a 25% increase
compared with 2010 census data.3 However, important
characteristics such as age, geographic distribution, education,
years of experience, and years to expected retirement are un-
known. Similar challenges are faced when trying to describe
the nurse practitioner workforce in oncology. The American
Association of Nurse Practitioners (AANP) publically reports
survey data for licensed nurse practitioners. In 2013, of the
more than 205,000 licensed nurse practitioners, approxi-
mately 2,050 worked in oncology and had been in practice for
an average of 7.7 years and had a median age of 48.25
Varied collaborative practice models are currently in use
based on the needs of the practice, patient volume, skills, and
training of the physician and the APP. Each has implications for
billing and productivity. The key to efficient integration of an
APP in oncology into a collaborative practice model is a careful
Collaborative practice in oncology is a dynamic process focused on assessment of skills and knowledge about oncology practice.
interdisciplinary support of patients and their caregivers with a broad range of
health care providers. The AP in oncology plays a critical role in the collaborative At a high level, potential roles of an APP include enabling
treatment of patients and their caregivers. Ongoing education, training, physicians to focus on more complex and higher acuity
mentorship, networking, and communication are necessary to cultivate and
maintain a collaborative practice model. Integration of resources from each patient needs; extending the range of high-level services for
practice setting, community organizations, e-health technologies, and advocacy patients without using additional physician time; and pro-
groups is essential. Human factors, health system factors, situational factors, and
socioeconomic factors are ever-changing within the continuum of care and must viding follow-up, symptom management, and survivorship
be considered in designing tailored patient and caregiver support.6 care to patients. To accomplish this, three models of care
Abbreviations: AP, advanced practice provider.
Reprinted with permission from the Advanced Practitioner Society for utilize APPs in the team-based care setting: (1) the auton-
Hematology and Oncology. omous or independent visit model, in which APPs pre-
dominantly see patients independently in a clinic but under a
ordering chemotherapy. The underlying theme in these collaborative practice agreement with their physicians; (2)
publications is that a collaborative practice model, with on- the shared visit model, in which patients are seen by both
cologists and APPs in oncology working together to the extent the APP and the physician during the same clinical en-
of their training and licensure, can improve patient and pro- counter; and (3) the mixed visit model, in which both the
vider satisfaction and safety and increase productivity and independent and shared visit models are used to manage the
revenue.24,25 However, the degree to which the use of APPs clinical volume but neither is the predominant encounter

FIGURE 3. Collaborative Practice Models Represented by the APSHO Practice Survey (192 patients)6,27

Abbreviation: APSHO, Advanced Practitioner Society for Hematology and Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 5


KOSTY, PICKARD, AND VIALE

used.26 The independent visit practice model goal is not to APPs as the most common reason not to use them in their
supplant or replace physician involvement but to allow practice.23 Exploring potential reasons for this lack of in-
physicians to delegate these tasks and medically direct terest is important to determine how to best motivate at-
without a required physical presence at the time of service. titudinal change. Because the ASCO report was primarily
The shared visit model goal is to allow for combining physician-owned private practices (73%) with only 8% sur-
the individual efforts of the APPs and physicians into a veyed in academic practice, it is possible this lack of interest
comanaged service that allows physicians to increase patient was based on the fear of decreased physician compensation.
volumes and the medical services they provide. This model It has been shown that the private practice model has
allows physicians to be continuously involved and to dele- significantly more oncologists compensated on an incentive-
gate aspects of the services provided to the APP. The col- based model compared with academic models (39.3% vs.
laborative practice models represented by the Advanced 3.1%). 28 Therefore, it may be important to focus on the
Practitioner Society for Hematology and Oncology (APSHO) increased practice productivity when using APPs to
practice survey are shown in Figure 3. encourage utilization in private practice. Furthermore,
To examine the differences between the different models, because a pure incentive-based model is associated with the
Buswell et al26 reported the effect of these practice models highest rate of burnout, increased professional satisfaction
on productivity, fees, and provider and patient satisfaction when working with APPs can be another educational point to
in an academic cancer center. They found that productivity change perceptions.
for the independent, mixed, and shared visit models, as Other challenges to incorporating APPs into clinical
measured by the number of new and established patients, practice are largely historical or based more on personal bias
was similar for all models (6.8, 6.7, and 7.0 patients seen than fact.29 For example, data do not substantiate the belief
per 4-hour session, respectively). Both physician and APPs that utilizing APPs will negatively affect the physician-
were very satisfied with the independent visit model and provider relationship or that patients will not accept APPs
reported patient-centered and productivity-based reasons as part of the care team. Studies have demonstrated high
influencing the decision to use their chosen model. The levels of patient and provider satisfaction with the collab-
shared visit model physicians were still very satisfied with orative practice model with increased utilization nation-
the model whereas APPs were only moderately satisfied. ally.23 It is likely that the portion of the workforce nearing
Reasons for utilizing the shared visit model were more retirement is also the same group that has less experience
physician-centered, focusing on physician preferences and and understanding of the physician assistant and nurse
perceptions. Importantly, there were extremely high levels practitioner professions and, therefore, more perceived
of patient satisfaction for both models (100% satisfaction bias. This attitude is changing as new oncologists entering
with care received from either model). the workforce come with experience working with APPs
In a much larger study in the private practice setting, the during their fellowship. In a survey of fellowship program
results of the ASCO study of collaborative practice arrange- directors in 2011, 90% of medical directors reported that
ments similarly noted high levels of patient and provider their fellows work with physician assistants and nurse
satisfaction with the different APP models.23 The most com- practitioners.30 What is unclear is how well prepared these
mon model in the survey was the independent visit model. The oncologists are to lead a medical team that incorporates
independent visit model was 19% more productive (based on APPs. It will be important moving forward for oncologists to
relative value units, RVUs) when the APP worked with the understand the different models for APP utilization, as well
entire group of physicians as compared with an independent as the regulatory and reimbursement requirements to lead
visit model when the APP worked exclusively with a limited the medical team effectively. Ideally, this educational need
number of physicians. However, it may not be correct to will be incorporated into fellowship training programs be-
conclude that the most productive RVU model is the ideal fore new oncologists enter the workforce and then will be
model in which to use APPs. Further insight into measures of further refined at the practice level based on state laws and
quality and continuity of care of the two models would be institutional policies.
important to distinguish. In addition, using RVUs as the sole
productivity measure is a limited assessment of the value an
APP might add to a practice. This study did not take into ac- Productivity and Reimbursement
count the nonrevenue-generating activity performed for each It is generally accepted that practices that incorporate APPs
model, which could be important in defining the preferred are more productive and efficient in providing quality care to
practice model. Value is not only about revenue generation; patients than practices that do not. However, as practices
quality of care and clinical productivity also are important and work to integrate APPs into clinical practice, they have been
might enhance physician quality of life.6,27 challenged with accurately assessing the productivity and
value of individual APPs. Practices that use a system strictly
BARRIERS TO INTEGRATION based on RVUs will likely underestimate the productivity and
Provider and Patient value of APPs because of the inability to accurately measure
ASCOs study of the collaborative practice arrangements of RVUs. For example, global surgical visits and the shared visit
APPs identified physicians lack of interest in working with model will render the time and effort of the APP invisible.31

6 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


COLLABORATIVE PRACTICE IN AN ERA OF MULTIDISCIPLINARY CARE

Even in the independent visit model, all incident-to visits and 30% to 50% higher than APPs, incident-to billing reimbursed
visits for many commercial payers are billed under the at 100%, and the savings on APP reduced recruitment and
physicians national provider identifier, despite all care being retention costs.
provided by the APP. In addition, numerous clinical care Currently, ASCO is exploring a project to assess the con-
activities that APPs provide are not billable encounters, but tribution of APPs in oncology practices. This project has
they bring quality and value to the practice. Importantly, if several goals: (1) count the number of APPs working in
the APP does not complete these nonrevenue-generating oncology practices; (2) identify the mix of direct patient care
activities, the physician would have to complete them. The services and other work that APPs conduct in oncology
challenges in assessing the productivity of APPs and the practices; (3) investigate the clinical, economic, and patient
limited benchmarking data available affect the ability to not effect of APPs in oncology practices; and (4) estimate how
only improve productivity of the APPs, but it also will hinder current and future APP workforces will affect the shortage
increased utilization. Practices will struggle to determine that exists between demands for services and the number of
when to hire new APPs and decide how their time should be physician oncology providers.
allocated to support the clinical enterprise. Also, practice
managers inherently will be unable to determine equitable
compensation, comparison, and accountability of APPs CONCLUSION
within a practice. Collaborative, multidisciplinary teambased care is es-
As to reimbursement, several common myths and sential if patients are to receive the highest quality value-
misconceptions can stymie the expansion of APPs in based oncology care. Achieving this goal will require ef-
oncology. Impressions such as APPs cant see new pa- fective integration of APPs into all aspects of patient care.
tients or APPs cant bill above a certain level are easily Although barriers remain, many are perceptual and rel-
debunked with a little education and, if needed, support atively easy to overcome. Understanding the APP work-
from national advocacy organizations. However, one of force, pipeline, and current utilizationas well as the
the more challenging misconceptions is the over- clinical, economic, and patient impact of APPs in oncology
estimation that the 85% reimbursement rate of the APPs practiceswill facilitate achieving the goal of optimal
compared with the physician rate will have on the cost- patient-centered cancer care. More information on the
effectiveness of APPs. Numerous studies on physician scope of practice of APPs, state regulations, payer policy,
assistants and nurse practitioners have demonstrated and information on integration into oncology practices
that APPs are cost-effective health care providers. This can be found on the AAPA, Association of Physician As-
can be explained by physician salaries being consistently sistants in Oncology, AANP, and APSHO websites.

References
1. Forte GJ, Hanley A, Hagerty K, et al. American Society of Clinical On- 9. Carrier E, Gourevitch MN, Shah NR. Medical homes: challenges in
cology National Census of Oncology Practices: preliminary report. translating theory into practice. Med Care. 2009;47:714-722.
J Oncol Pract. 2013;9:9-19. 10. Jackson GL, Powers BJ, Chatterjee R, et al. Improving patient care: the
2. Yang W, Williams JH, Hogan PF, et al. Projected supply of and demand for patient-centered medical home: a systematic review. Ann Intern Med.
oncologists and radiation oncologists through 2025: an aging, better- 2013;158:169-178.
insured population will result in shortage. J Oncol Pract. 2014;10:39-45. 11. National Committee for Quality Assurance. Patient-Centered Spe-
3. Hanley A, Hagerty K, Towle EL, et al. Results of the 2013 American cialty Practice Recognition. http://www.ncqa.org/Programs/Recognition/
Society of Clinical Oncology National Oncology Census. J Oncol Pract. Practices/PatientCenteredSpecialtyPracticePCSP.aspx. Accessed March
2014;10:143-148. 26, 2015.
4. American Society of Clinical Oncology. The state of cancer care in 12. Robeznieks A. At home with the specialist: oncologists and other
America, 2015: a report by the American Society of Clinical Oncology. specialists launching patient-centered medical homes. Mod Healthc.
J Oncol Pract. 2015;11:79-113. 2014;44:24-25.
5. Erikson C, Salsberg E, Forte G, et al. Future supply and demand for 13. Innovative Oncology Business Solutions. COME HOME Model. www.
oncologists: challenges to assuring access to oncology services. J Oncol comehomeprogram.com/index.php/come-home-practices/. Accessed
Pract. 2007;3:79-86. March 26, 2015.
6. Kurtin S, Peterson M, Goforth P, et al. The advanced practitioner and 14. Oncology Management Services. Transforming Oncology. www.opcmh.
collaborative practice in oncology. J Adv Pract Oncol. 2015;6:515-527. com/. Accessed March 26, 2015.
7. Taplin SH, Weaver S, Chollette V, et al. Teams and teamwork during a 15. Community Oncology Alliance. Oncology Medical Home: Improved Quality
cancer diagnosis: interdependency within and between teams. J Oncol and Cost of Care. http://coaadvocacy.org/2014/09/oncology-medical-
Pract. 2015;11:231-238. home-improved-qualityand-cost-of-care/. Accessed March 26, 2015.
8. Kilo CM, Wasson JH. Practice redesign and the patient-centered medical 16. Medical Home Oncology. Commission on Cancer Accreditation. www.
home: history, promises, and challenges. Health Aff (Millwood). 2010; medicalhomeoncology.org/coa/commission-on-cancer-accreditation.htm.
29:773-778. Accessed March 26, 2015.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 7


KOSTY, PICKARD, AND VIALE

17. Kosty MP, Bruinooge SS, Cox JV. Intentional approach to team-based 24. Brown CG. Commentary: new findings substantiate the successful use
oncology care: evidence-based teamwork to improve collaboration and of nurse practitioners and physician assistants in collaborative practice
patient engagement. J Oncol Pract. 2015;11:247-248. models. J Oncol Pract. 2011;7:285-286.
18. Shulman, LN. Efficient and effective models for integrating advanced 25. Welch WP, Cuellar AE, Stearns SC, et al. Proportion of physicians in large
practice professionals into oncology practice. Am Soc Clin Oncol Educ group practices continued to grow in 2009-11. Health Aff (Millwood).
Book. 2013;33:e377-e379. 2013;32:1659-1666.
19. Kosty M, Acheson A, Tetzlaff E. Clinical Oncology Practice 2015: 26. Buswell LA, Ponte PR, Shulman LN. Provider practice models in am-
preparing for the future. Am Soc Clin Oncol Educ Book. 2015;35: bulatory oncology practice: analysis of productivity, revenue, and
e622-e627. provider and patient satisfaction. J Oncol Pract. 2009;5:188-192.
20. Drury L, Baccari K, Fang A, et al. Providing intensive palliative care 27. Kurtin SE, Viale PH, Hylton HM, et al. The APSHO practice survey. Paper
on an inpatient unit: a full-time job. J Adv Pract Oncol. 2016;7: presented at: 3rd Annual Meeting of JADPRO Live, Advanced Practitioner
60-64. Society for Hematology and Oncology; November 2015; Phoenix, AZ.
21. McNally GA, Florence KJ, Logue AC. The evolution of a malignant 28. Shanafelt TD, Gradishar WJ, Kosty M, et al. Burnout and career satis-
hematology nurse practitioner service. Clin J Oncol Nurs. 2015;19: faction among US oncologists. J Clin Oncol. 2014;32:678-686.
367-369. 29. Tetzlaff E, Polansky M, Carr K, et al. Physician assistants in oncology.
22. Shah NN, Kucharczuk CR, Mitra N, et al. Implementation of an advanced J Oncol Pract. 2007;3:283.
practice provider service on an allogeneic stem cell transplant unit: 30. Kosty MP. Informational itemsWAG, classic references, QOPI for
impact on patient outcomes. Biol Blood Marrow Transplant. 2015;21: fellows. Paper presented at: Annual Meeting of the American Society of
1692-1698. Clinical Oncology; June 2011; Chicago, IL.
23. Towle EL, Barr TR, Hanley A, et al. Results of the ASCO Study of Col- 31. Pickard T. Calculating your worth: understanding productivity and
laborative Practice Arrangements. J Oncol Pract. 2011;7:278-282. value. J Adv Pract Oncol. 2014;5:128-133.

8 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


INVITED ARTICLES COLLECTIVE WISDOM

Value in Oncology: Balance Between Quality and Cost


Derek Raghavan, MD, PhD, FACP, FRACP, FASCO, and Mark W. Legnini, DrPH

I t is a disturbing fact that the costs of health care are


spiraling upwards and that an exemplar of this troubling
trend is the domain of oncology treatment and research. By
were publicly funded (34.1% Medicare and 5.3% Medicaid);
resulting federal scrutiny of health care spending meant
Washington policy makers measures of success often
2020, it is projected that cancer care will cost more than became political and ideological footballs. Lobbying by the
$150 billion annually in the United States. Although this re- health care industry to protect economic interests has
presents only a relatively small fraction of total health care produced self-serving success stories, some from the
cost, cancer care is escalating more rapidly than most other pharmaceutical and device industries and others from
specialties. Many factors contribute to this unhappy situa- providers of care locked in competition for patients and
tion, including the aging of the population, persistence of revenues. The advocacy research behind these success
risk-taking by the community at large (smoking, excessive stories is frequently cited but often unvalidated and diverts
sun exposure, and lack of attention to industrial pollution, attention from a proper focus on the patient. Thus, oncol-
to name a few), increasingly expensive research, diagnostic ogists must address the challenges of responding to the
tests, surgical approaches, radiotherapy techniques, and communitys evolving requirements for greater value, with-
novel systemic therapies as well as some unrealistic expec- out loss of innovation, in a more proactive, creative, and
tations of patients, families, and the community at large (often structured fashion to preserve quality while attempting to
predicated on false claims from our profession and/or hype contain expense.
from the Fourth Estate). Of clear relevance is the consid- At its simplest, the value proposition in health care has
eration that another political declaration of war on cancer, been defined by Porter and Teisberg2,3 with the following
the third in 40 years, this time an amalgam of the State of the equation:
Union address and some opportunism from the biomedical
Value Outcomes=Cost
community, is unlikely to fix the problem. Although this type
of hyperbole might increase focus, and even funding, it will This equation makes sense and is routinely used in planning
create a renewed sense that death is optional and will en- the strategy of cancer care for our health care systems in the
courage patients and their physicians to continue futile at- Carolinas and Michigan, as it helps us to consider what is
tempts at active treatment of truly resistant disease, rather contributing to poor value in cancer care and allows reso-
than considering palliative care with its associated improve- lution of those elements that are accessible. Of key impor-
ment in quality of life and reduction in cost. tance is to critically consider the true benefits of the use of
A more sensible approach is to weave the concept of our diagnostic and treatment modalities, defining whether
value-based medical care into the equation, thus leveraging they contribute to prolongation of survival, increased quality
resource expenditure in a more productive and sensible way of life, or reduced morbidity of disease or its treatment, and
than has characterized much of modern medicine. However, also whether the most cost-efficient options are chosen.
one of the more substantial challenges in modern oncology In addition, Porter and Teisburg2,3 have emphasized the
practice is to provide true increase in value when so many important principles of transparency, provision of com-
new parameters of success are being introduced into the prehensive disease management and prevention services,
equation. Health care spending to treat cancer rose 55% in organization around medical conditions, and redefinition of
the decade from 2001 to 2011, and annual retail prescription the health plan/subscriber relationship (with an end to cost-
drug expenditures for cancer quintupled over the same shifting practices) as potential solutions to the current
period.1 Almost 40% of expenditures for cancer care in 2011 dysfunction in the provision of health care.

From the Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC; University of Michigan Medical School, Institute for Healthcare Policy & Innovation, Ann Arbor, MI.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Derek Raghavan, MD, PhD, FACP, FRACP, FASCO, Levine Cancer Institute, Carolinas HealthCare System, 1021 Morehead Medical Dr., Charlotte, NC 28204;
email: derek.raghavan@carolinas.org.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 9


RAGHAVAN AND LEGNINI

The prospect of cancer treatment is daunting for most EXPERIMENTS IN THE DELIVERY OF
patients when one considers the nature of the treatment VALUE-BASED CANCER CARE
being offered and its potential consequences. In addition, Toward Symmetry of Care in the Carolinas
asymmetry of medical information, potential fiscal toxicity, Charlotte, the largest city in North Carolina, with a pop-
and dislocation of patients and their families for treatment ulation approaching 1 million (and a referral base of around
far from their homes mean patients lose control over their 2 million people) has not had local access to a comprehensive
lives, compounding their anxiety over a cancer diagnosis and cancer center. Specifically, there has been paucity of locally
the treatment that follows. The provision of symmetrical available bone marrow transplant facilities and of phase I
(i.e., evenly distributed) access to high-quality care, by and investigator-initiated clinical trials, although the latter
overcoming barriers of poverty, language, geographical have been intermittently available on a small scale. Qua-
isolation, and information while providing the key advances ternary cancer services have been provided by outstanding
in modern oncology, with a greater emphasis on palliative cancer centers at Emory, Duke, Wake Forest, and The
care optimization, should improve national cancer survival University of North Carolina at Chapel Hill, but have often
and morbidity figures without continuing to increase the required travel of more than 3 to 4 hours for geographically
strain on federal and state budgets.4,5 A focus on universally isolated patients and for those living in Charlotte who
distributed access contrasts with the continued reluc- have needed more specialized cancer services. Carolinas
tance of some medical institutions to accept indigent, un- HealthCare System, an amalgam of more than 40 hospitals
derinsured, Medicaid, or even Medicare patients, thus and medical centers spanning North Carolina and South
protecting their bottom lines from loss, an approach that is Carolina, employs more than 2,000 physicians and 50,000
unfair to the community.3 In addition to creating maldis- staff and sees more than 15,000 new cancer cases per year.
tribution of the costs of providing care, cherry-picking of the As the system has expanded to be one of the largest
wealthiest, most educated, and robust patients (who also nonprofit health systems in North America, it made sense
usually have the strongest health insurance coverage) may to establish a tertiary and quaternary referral cancer
lead to biased representation of outcomes. These centers center that would serve its network of smaller centers
publish data drawn from treatment of star patients, who throughout the Carolinas, but which would attempt to
have fewer of the adverse prognosticators and comorbid- provide a system of cancer care closer to the patients
ities of unselected patient populations, leading to inflated home, contrary to some of the more conventional models.
survival figures and artificially reduced morbidity and tox- In design, several criteria were deemed crucial and in-
icity data. Sadly, many politicians and community leaders corporated into the creation of this multisite institute, as
may fail to understand this nuance when they translate the summarized in the Sidebar.
resulting highly selected data into expectations for the Our strategy for the system throughout the Carolinas has
population at large, thus distorting the concept of true value. been to link with selected hospitals and their staffs,
Successful provision of symmetrical, high-quality out- establishing contractual relationships with the hospital
comes also requires easily accessible tertiary and quaternary leadership and personal and professional relationships with
level cancer care close to home, with access to clinical trials, their physicians, which includes the provision of centrally
extensive patient support and navigation, optimal palliative controlled Levine Cancer Institute clinical trial units. Where
and supportive care, and the benefits of the genomic rev- needed, additional faculty members have been recruited
olution as elements of routine cancer care.1,5 It also has to be from a range of other academic cancer centers. Academic
refined by the establishment of an evidence-based, system- and programmatic leadership has been drawn from National
wide set of standards for diagnosis and treatment, which are Cancer Institutedesignated comprehensive cancer centers,
routinely used by physicians. and trained clinical investigators have been placed at out-
In many major centers, following the innovative approach reach centers to ensure symmetry of access to trials. We
of Fox Chase Cancer Center in the Delaware Valley, outreach have thus created a hybrid academic-practice model, and
clinics have been established as a mechanism to provide this has led to dramatic increments in cancer trial accrual and
easier intake to attract new patients and provide access to maintenance of patient care in distant geographic sites, with
cancer trials. However, many of these ventures have not improved access to tertiary and quaternary facilities, often
been designed to provide all of the key resources, such as via telemedicine.
access to translational trials with laboratory and bio-
repository support, genetic counseling, and patient-family
support, for these geographically isolated patients. Strategies in Play in Michigan
In the United States in recent times, less than 5% of pa- The Michigan Value Collaborative provides 65 hospitals
tients have been enrolled in cancer clinical trials,6,7 and it is statewide with data and tools to help them control costs for
clear that there are noteworthy disparities of access to high- episodes of care, including colorectal cancer resection,
level cancer programs.4 We are now attempting to address esophagectomy, lung cancer resection, and prostatectomy,
these challenging issues in the Carolinas8 and in Michigan, up to 90 days postdischarge. This includes the initial
two areas challenged by considerable geographical and hospitalization plus skilled nursing, rehabilitation, home
demographic constraints. health, transfers/readmissions, etc. The Michigan Value

10 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


VALUE IN ONCOLOGY: BALANCE BETWEEN QUALITY AND COST

SIDEBAR. Criteria for a Symmetrical System of Cancer Care


Multidisciplinary clinics with standard operating procedures and protocols of management, embedded in electronically linked,
evidence-based clinical pathways
System-wide interdisciplinary tumor boards and conferences, augmented by electronic two-way video conferencing to connect
geographically isolated oncology teams
Easy availability of clinical trials, with access close to home throughout North Carolina and South Carolina
A single, central institutional review board for cancer trials covering the whole system, facilitating swift and synchronous, system-
wide activation of studies
Central protocol review and monitoring system, with initial protocol submissions via tumor-specific teams
Central oversight of a tightly controlled clinical trials unit, with central training and monitoring of staff
Centralized connections and data capture for each hospital tumor registry with ability to measure outcomes and costs of care
Extensive patient support services, including patient navigation linked throughout the system, standard operating procedures for
emergency departments throughout the system, palliative care and pain management services, cancer-focused pastoral care, and live
or video-linked genetic counseling and financial counseling
Availability of subspecialty services, such as bone marrow transplantation, phase I clinical trial units, and sophisticated radiation
techniques and equipment in as accessible a fashion as possible
A focus on translational bench research that is focused specifically on the clinical emphases of the instituteearly programs have
focused on cancer pharmacology, stem cell biology of hematologic disorders, and molecular prognostication with availability of a
cost-effective, molecular testing platform where appropriate
Optimal informatics technology support and electronic and video linkage for conferencing, tumor boards, and creation of electronic
pathways in tumor-specific team meetings

Collaborative creates a unique window for hospitals to see discontinuation of these diagnostic or management ap-
charges generated for a patients care outside their four proaches would not result in reduced outcomes and in some
walls, an essential tool for cost-effective care coordination. cases (e.g., screening for prostate cancer in elderly men with
In addition, Collaborative Quality Improvement initia- intercurrent diseases and limited life expectancy) could
tives around the state share clinical process and outcomes actually improve survival and/or morbidity. As can easily be
data to improve outcomes for patients receiving surgical, appreciated from Table 1, nationwide implementation of
radiation, and medical oncology services. Together, these these recommendations has achieved dramatic cost savings,
programs provide clinical and claims-based registries (price- without measurable loss of quality or outcomes.
standardized and risk-adjusted), web-based analytic tools, These items were chosen because they reflected clinical
and forums across the state for practicing physicians and practice among most oncologists and thus would avoid
hospital quality improvement staff to work on individ- partisan subspecialty rivalries and concerns, were broadly
ual improvement and collectively to make Michigan a applicable to large numbers of patients, and also clearly
better place for patients with cancer to receive treatment. reflected the absence of a robust evidence base un-
Combining episode costs (the value equations denominator) derpinning patterns of standard clinical practice.
with clinical outcomes data from collaborative quality im- With the growing evidence on producing high value for
provements (the numerator) gives Michigans hospitals one health expenditure, we will increasingly need to consider
of the countrys best opportunities to improve the value of clinical practices that have evolved without evidence to
cancer care. support a survival or quality-of-life benefit and must focus on
whether the treatments selected have equi-active but less
costly alternatives (e.g., use of generic drugs, biosimilars,
CHOOSING WISELY
and lower doses of cytotoxics, reduced treatment duration
One of the most important aspects to reduction of un-
of radiotherapy, and elimination of heroic but unproven
necessary expenditure in oncology is the consideration of
surgical procedures). In each case, the driver is cost con-
what is gained by the use of the available management
tainment or avoidance of profligate expenditure without
approaches. The Institute of Medicine has encouraged
loss of quality or outcomes.
physicians to carefully consider the benefits and drawbacks
associated, in particular, with expensive management op-
tions in the so-called Choosing Wisely campaign. In line MEASUREMENT OF VALUE
with this philosophy, the American Society of Clinical On- In 2015, the ASCO Value Task Force published the Conceptual
cology (ASCO) created the Value Task Force and developed Framework for Assessing Value in Cancer Care.11 The basic
two consensus documents,9,10 based on the best available construct is as follows:
evidence, that proposed discontinuation of standard
clinical practices that provided no patient benefit (Table 1).
Two major contextual categories are created, advanced
disease (e.g., treatment of metastases) and curative
In each instance, level 1 to 2 evidence clearly showed that intent (e.g., adjuvant therapy).

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 11


RAGHAVAN AND LEGNINI

Clinical benefit is given a score of 1 to 5, and then


numerical weightings are added to reflect the impact of
position paper.12 What is quite remarkable is the extraor-
dinary similarity in concept and execution, achieved by two
treatment on overall survival, progression-free survival, committees working in the United States and in Europe, both
or response rate. The weightings reflect the thought without knowledge of the work in progress of the other
that overall survival is more important than progression- team. The ESMO team also has focused on separate domains
free survival, which is more important than response of curative and palliative settings and attributed levels of
rate when attempting to attribute value for the patient. benefit predicated on absolute or relative increments in
Negative scores are added to reflect the extent of survival and levels of toxicity. Of importance, they have set
toxicity. In the advanced disease setting, bonus points time-dependent criteria, such that the value points allocated
are awarded for palliation of symptoms and for longer for absolute survival increments differ for those with sus-
treatment-free intervals. Finally, the cost of treatment is tained increases versus the less fortunate patients with only
short-term increments in survival.
listed to broaden the context.
Interestingly, both committees used similar sets of level 1
In the adjuvant setting, instead of the response-survival
parameters above, hazard ratio is incorporated into the
data to develop their models and came to quite similar
interpretations of value, notwithstanding different com-
algorithm, and disease-free interval is substituted, but munity pressures, health care costs, and medical traditions
with a slightly lower weighting. on different sides of the Atlantic.
The overall construct of net health benefit is incorpo-
rated to allow the patient to attribute likely value to the
These efforts are important and represent crucial first
steps by members of our profession in attempting to in-
treatment, balancing benefit(s) and toxicities. crease the level of self-discipline and value-based self-
Unbeknownst to the ASCO Task Force, the European criticism of what we do and increased transparency in
Society for Medical Oncology (ESMO) had also created a what we tell our patients about the benefits of treatment.
similar task force, which produced the Magnitude of Clinical We would have preferred a higher bar with regard to at-
Benefit Scale. Led by Dr. Nathan Cherny, the ESMO position tribution of value pointsit is obvious that a patient with a
paper was published at about the same time as the ASCO prognosis of only 3 to 6 months will sustain greater relative

TABLE 1. ASCO Choosing Wisely List

Topic Rationale Level of Cost Savings*


Do not use cancer Rx if PS 3 to 4, no prior benefit, Logical, maximizes QOL, no loss of proven survival Extensive at individual and national level
ineligible for trial, and no evidence to support benefit
more Rx
No extensive staging for low-risk early prostate Overview of published data shows extremely low Extensive at national level
cancer positive yield within false-positive rate.
No extensive staging for low-risk early breast Overview of published data shows extremely low Extensive at national level
cancer positive yield within false-positive rate.
No surveillance via biomarkers or imaging for No survival benefit from early diagnosis of metastatic Extensive at individual and national level
breast cancer after curative Rx (excludes relapse
mammography)
No white cell stimulatory prophylaxis for Risk of febrile neutropenia is predictable for low-risk Extensive at national level
patients with < 20% risk of febrile patients; in high-risk patients, prophylaxis should
neutropenia be considered
Avoid starting antinausea prophylaxis with most Effective options are available for control of regimens Extensive at national level
expensive high-risk drugs for low-risk at low-risk of emesis; crossover can easily be
emetogenic regimens affected if vomiting occurs
Avoid combination chemotherapy for salvage Single-agent Rx is an effective salvage option and Extensive at individual and national level
Rx of metastatic breast cancer unless urgent usually associated with less toxicity
response is required
Avoid post-Rx routine surveillance with PET or No data to support survival benefit of this approach Extensive at individual and national level
CT-PET scanning after completion of Rx unless for most clinical settings
evidence shows survival benefit
Avoid screening asymptomatic men with PSA Randomized data do not support survival benefit Extensive at national level
for prostate cancer if < 10 years of life from screening for prostate cancer and may cause
expectancy harm in elderly
Do not use targeted therapy against specific Very low response rate to most targeted Rx unless Extensive at individual and national level
gene aberration unless biomarker predicts specific target is expressed by tumor
likely response
Abbreviation: Rx, prescription; PS, pain scale; QOL, quality of life; PSA, prostate-specific antigen.
*Individual reflects potential for substantial copays or expensive self-pay. National reflects costs associated with specific service delivery as well as incidence/frequency figures at
national level.

12 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


VALUE IN ONCOLOGY: BALANCE BETWEEN QUALITY AND COST

benefit from a 2-month increment than someone treated professional lives. One way to transform training is to ensure
with curative or aggressive palliative intent; that said, a that teaching programs are located in care settings in which
2-month survival increment, offset by considerable physical residents see high-value care delivered every day. This will
or fiscal toxicity, needs to be taken into context. This is require that funding for graduate medical education be
complex, paradigm-shifting work and will require careful value-based to provide incentives for training programs to
and critical attention and feedback. change. Together, the federal Medicare program and a
majority of state Medicaid programs contribute approxi-
TRAINING THE NEXT GENERATION OF mately $14 billion annually to fund graduate medical edu-
PHYSICIANS TO INCORPORATE THE VALUE cation, based primarily on counts of trainees and residency
PROPOSITION program accreditation. If these payments were value-based,
Training oncology residents to provide high-value care is key that would be a strong incentive for programs to ensure that
to transforming the practice of oncology in the United succeeding cohorts of oncologists emerge from training
States.13 Current policies to improve the performance of imbued with the concept of value in health care and
health care practitioners and the institutions in which they equipped with the tools to provide safe, high-quality, cost-
practice depend primarily on financial incentives aimed at effective cancer care.
changing the behavior of fully-trained physicians. This is
much less effective than intervening earlier in the workforce
pipeline to influence physicians during their training and SUMMARY
professional socialization. The exponential increase in costs of health care is un-
Expressed simply, residents who do not train in high-value necessary and reflects many avoidable factors at a com-
care settings are less likely to become high-value physi- munity level, including poor health practices, unrealistic
cians.13 Residency faculty must be skilled in the organization expectations, corporate profiteering, and a poor medical
and delivery of high-value care and in how to teach those decision process (which often contravenes level 1 to 2 ev-
skills. idence). Physicians must increasingly consider true value
Exhortations without systems transformation will not (outcome/cost ratio) when creating management plans and
break the cycle of teaching each generation of oncologists include these considerations in transparent and realistic
outmoded ways of thinking about safety, quality, and cost- conversations with patients. Attention to these issues will
effectiveness during residency, when their decision making dramatically reduce the burgeoning costs of cancer care in
habits, professional values, ways of interacting with pa- our community while improving the quality and value of
tients and colleagues, etc., are shaped for the rest of their care.

References

1. Soni A. Trends in Use and Expenditures for Cancer Treatment among 8. Raghavan D. Costs of cancer care: rhetoric, value, and steps forward.
Adults 18 and Older, U.S. Civilian Noninstitutionalized Population, 2001 Semin Oncol. 2013;40:659-661.
and 2011. Medical Expenditure Panel Survey, Statistical Brief #443. 9. Schnipper L, Smith TJ, Raghavan D, et al. American Society of Clinical
http://meps.ahrq.gov/mepsweb/data_files/publications/st443/ Oncology identifies five key opportunities to improve care and reduce
stat443.pdf. Accessed February 23, 2016. costs: the top five list for oncology: ASCOs top five list. J Clin Oncol.
2. Porter ME, Teisberg EO. How physicians can change the future of health 2012;30:1715-1724.
care. JAMA. 2007;297:1103-1111. 10. Schnipper LE, Lyman GH, Blayney DW, et al. American Society of Clinical
3. Porter ME, Teisberg EO. Redefining Health Care. Boston, MA: Harvard Oncology 2013 top five list in oncology. J Clin Oncol. 2013;31:
Business School Press; 2006;97-283. 4362-4370.
4. Raghavan D. Slow progress in cancer care disparities: HIPAA, PPACA, 11. Schnipper LE, Davidson NE, Wollins DS, et al; American Society of
and CHEWBACCA... but were still not there! Oncologist. 2011;16: Clinical Oncology. American Society of Clinical Oncology statement:
917-919. a conceptual framework to assess the value of cancer treatment op-
5. Goss E, Lopez AM, Brown CL, et al. American Society of Clinical Oncology tions. J Clin Oncol. 2015;33:2563-2577.
policy statement: disparities in cancer care. J Clin Oncol. 2009;27: 12. Cherny NI, Sullivan R, Dafni U, et al. A standardised, generic, validated
2881-2885. approach to stratify the magnitude of clinical benefit that can be an-
6. Lara PN Jr, Higdon R, Lim N, et al. Prospective evaluation of cancer ticipated from anti-cancer therapies: the European Society for Medical
clinical trial accrual patterns: identifying potential barriers to enroll- Oncology Magnitude of Clinical Benefit Scale (ESMO-MCBS). Ann Oncol.
ment. J Clin Oncol. 2001;19:1728-1733. 2015;26:1547-1573.
7. Raghavan D. An essay on rearranging the deck chairs: whats wrong with 13. Legnini, MW. Can low-performing hospitals train high-performing
the cancer trials system? Clin Cancer Res. 2006;12:1949-1950. residents? Am J Med Qual. 2011;26:408-410.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 13


INVITED ARTICLES COLLECTIVE WISDOM

Womens Health Issues for BRCA Mutation Carriers


Mary E. Sabatini, MD, PhD, and Leif W. Ellisen, MD, PhD

I n 2015, it was estimated that there were 1.6 million new


cases of cancer diagnosed in the United States.1 Remarkably,
more than 86,000 were diagnosed in women under the age
25. Breast screening should begin by age 25 with MRI.
Earlier MRIs are also considered if any family member
was diagnosed with breast cancer before age 25. Annual
of 45. The majority of cancers are thought to be a result of mammography and MRI starts at age 30. Risk-reducing
bad luck; that is, random mutations arising during DNA mastectomy should be offered as an option to all BRCA
replication in normal stem cells,2 whereas inherited muta- mutation carriers, as it has been shown to reduce the risk
tions are thought to be causal in 5%10% of cancers. Thus, of cancer by as much as 90%, although definitive studies
the number of cancer cases that are attributable to inherited showing either an overall or breast cancerspecific sur-
mutations may seem small, however, they are clinically vival advantage are lacking. In contrast, risk-reducing bi-
important. Cancers in patients with hereditary cancer syn- lateral salpingo-oophorectomy (rrBSO) at age 35 to 40
dromes tend to occur at a younger age, and most hereditary or whenever childbearing is completed is associated with a
cancer syndromes are also associated with more than one lower risk of breast cancer and reduces all-cause, ovarian
type of cancer.3 cancerspecific, and breast cancerspecific mortality.9
Approximately 20% of women with inherited breast and/ rrBSO results in an 80% risk reduction for ovarian, fallopian
or ovarian cancers harbor a harmful mutation of one of the tube, and peritoneal cancers. There has been recent interest
breast cancer susceptibility genes 1 or 2, known as BRCA1 and in the possibility of performing salpingectomy only, as it
BRCA2.3,4 Mutations in BRCA1/2 are inherited in an autosomal seems that some ovarian cancers may arise primarily in the
dominant fashion. Families that carry germline mutations may fallopian tube, but this is not recommended other than in
have several women affected by breast and possibly ovarian the setting of a clinical trial. Hysterectomy is not routinely
cancer and men with breast cancer and prostate cancer. These recommended given that the absolute risk of endometrial
mutations also predispose carriers to cancers such as fallopian cancer is low enough that the benefits do not outweigh the
tube cancer, peritoneal cancer, endometrial cancer, pancreatic risks of surgery. For those who do not elect for rrBSO,
cancer, colon cancer, and cancers in other sites. Although there ovarian cancer screening with transvaginal ultrasound and
are many issues for BRCA mutation carriers who have been carbohydrate antigen 125 (CA-125) is suggested every
diagnosed with cancer, this article will focus on the care of 6 months starting at age 30. Notably, however, there are no
women who are known carriers but who have not been di- data to support the efficacy of this practice.
agnosed with cancer. The NCCN guidelines are established by incorporating
For BRCA1 mutation carriers, cumulative estimates of the best evidence available to date. However, in caring for
developing cancer by age 70 vary by population, ranging women who do carry a deleterious BRCA mutation, one of
from 40% to 85% for breast cancer and 10% to 59% for the major challenges is trying to help an individual patient to
ovarian cancer. For BRCA2 mutation carriers, cumulative make decisions using data obtained from studying pop-
estimates of developing cancer are 45% to 57% for breast ulations. Like most autosomal-dominant mutations, pene-
cancer and 11% to 18% for ovarian cancer.5-7 Because of trance of a BRCA mutation is not 100%. The probability of
these risks, the National Comprehensive Cancer Network developing cancer for carriers is alarmingly high, yet the
(NCCN) has established guidelines for women with BRCA morbidity associated with surveillance and/or prevention
mutations to reduce the risk of cancer.8 This includes breast is also not negligible. Furthermore, taking steps toward
awareness and breast self-examinations starting at age 18, prevention does not absolutely eliminate the risk of
with annual to biannual clinical examinations starting at age cancer development. Decision making can be empowering,

From the Department of Obstetrics and Gynecology, Harvard Medical School, Massachusetts General Hospital, Boston, MA; Breast Medical Oncology, Harvard Medical School,
Massachusetts General Hospital Cancer Center, Boston, MA.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Mary E. Sabatini, MD, PhD, Massachusetts General Hospital, 55 Fruit St., Boston, MA 02114; email: msabatini@mgh.harvard.edu.

2016 by American Society of Clinical Oncology.

14 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


WOMENS HEALTH ISSUES FOR BRCA MUTATION CARRIERS

as indicated by the New York Times opinion article written women with BRCA mutations. One recent survey study
by Angelina Jolie.10 Nonetheless, decisions can be anxiety described the reaction of 25 male partners about their fe-
provoking, particularly when the consequences of the male partners mutation status.16 Twenty of the men were
wrong decision are so large and life altering. married or engaged at the time of the survey, and 19 had
Unfortunately, knowledge of ones mutation and the NCCN been in the relationship at least 5 years. Nine participants
guidelines does not necessarily enable an individual to plan reported changes regarding intimacy levels, and two re-
ones life accordingly. In an ideal world, perhaps every woman spondents reported changes in their level of attraction.
who carried a BRCA mutation would have completed child- Almost all participants noted changes in their commu-
birth prior to age 35. Indeed, younger age at first birth and nication after mutation disclosure, with changes in the
breast-feeding are protective for development of breast and topics discussed and more discussions about the future.
ovarian cancers overall. Additionally, in an ideal world, there Interestingly, of the 14 men whose partners had not un-
would be no repercussions to mastectomy and early surgical dergone a risk-reducing mastectomy, five expressed concerns
menopause. However, these ideals run counter to the realities about their level of attraction to their partner, but of the 11
of life for many. In high-income countries, the median age of men whose partner had had a mastectomy, all said that they
first birth is rising,11 with many women not starting a family were as attracted to their partner after surgery as they were
until after their 35th birthday. Surgery and surgical menopause before surgery. It is difficult to generalize the results of this
can cause morbidity and can be life altering. study broadly, as most of these men were in long-term or
A retrospective study including 305 BRCA mutation carriers committed relationships at the time of the survey.
seen in a California integrated health system between 1995 In general, reproductive options for all women have vastly
and 2012 showed that the uptake of options for BRCA mu- expanded in the past several years, and this may also benefit
tation carriers who were informed of the NCCN guidelines was those who carry a BRCA mutation. For women who are not
lacking in some areas. The median follow-up during the yet ready to have children, in vitro fertilization (IVF) can be
study was 41 months. The median time from diagnosis to risk- used to freeze either oocytes or embryos for future use.
reducing surgery was 6 months, with 74% of patients choosing Embryo cryopreservation has been available since the 1980s,
to undergo rrBSO and 44% of patients choosing to undergo and outcomes have improved dramatically in the last several
risk-reducing mastectomy. Of those choosing surveillance, years. For couples with infertility and a female partner
compliance was 45% in the first year but decreased to younger than age 35, expected pregnancy rates after a single
approximately 10% at 3 years and 2% at 5 years.12 IVF are upwards of 50%.17 For couples with unknown fertility
Decisions about prophylactic surgery are complicated interested in banking embryos for future use, there are no
by concerns about appearance, sexuality, control of re- numbers to guide us, but presumably the success rate would
production, and relationships. A retrospective study by be at least as good if not higher. Oocyte freezing is available
Stukey et al13 highlights that these decisions among carriers to women who are unpartnered, unwilling to use donor
are not, as might be anticipated, based on risk alone. Ninety sperm, or who wish to maintain complete autonomy of their
women, all of whom were carriers of BRCA mutation, were reproductive potential. Historically, oocyte freezing has been
offered prophylactic surgery. Fifty-one percent of the technically more challenging. Although embryo freezing
women underwent surgery, 85% underwent BSO, and 28% remains the gold standard, this will likely change in the near
underwent mastectomy. Women who elected to have future as success rates with frozen oocytes are approaching
surgery were more likely to be parous, married, employed, those of frozen embryos.17
and had a prior history of breast cancer. This implies that Although embryo and oocyte preservation provide op-
those who did not elect for prophylactic surgery may be tions, IVF is expensive, arduous, and time consuming. It
interested in future fertility. BRCA2 mutation carriers were comes with risks including hemorrhagic cysts, ovarian tor-
more likely than BRCA1 carriers to pursue surgery. If sion, ovarian hyperstimulation syndrome, blood clots, in-
avoidance of developing disease were the only factor in- fections, medication reactions, and surgical risks associated
volved in decision making, BRCA1 carriers should be more with oocyte extraction. For these reasons, it is not univer-
likely to undergo surgery. sally available or used. Overall, IVF has not been associated
Hoskins et al14 described 11 cases of unmarried women with an increased risk of development of breast or ovarian
age 26 to 35 who carry a BRCA mutation. These women cancer18-22 in the general population or in BRCA mutation
relayed challenges regarding their perception of their own carriers.23,24 The data for the general population are quite
desirability and anxiety about partners reactions to dis- robust as a result of the length of availability of IVF and the
closure of mutation status. Another web-based survey of 44 number of people who have used IVF. For those with BRCA
women with a BRCA mutation included 13 women who were mutations, however, the numbers are smaller and not as
unmarried, and all of them expressed concerns about how robust, so the conclusions are not as definitive.
and when to disclose their status to future partners. Most of One of the most challenging issues for women who carry
them expressed a sense of urgency to have children.15 a BRCA mutation surrounds the choice about the potential
Considering the anxiety expressed about the effect of their to eliminate this mutation from future offspring. Pre-
own BRCA mutation on their relationships, very little data implantation genetic diagnosis (PGD) is a procedure in which
exist regarding the attitudes of the sexual partners of a cell or cells from a human embryo are removed and tested

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 15


SABATINI AND ELLISEN

for diseases and/or mutations that can result in disease (for surgical menopause happens at an earlier age than does
example, cystic fibrosis, Huntington disease, BRCA mutation). natural menopause. As such, menopausal symptoms, the
PGD requires IVF. The resulting embryos are allowed to grow effects of hormonal changes, and their treatment should be
for 3 to 5 days so there are enough cells to be biopsied for PGD. discussed with women prior to surgery. Menopause is as-
Embryos found by biopsy to be unaffected (on average ap- sociated with hot flashes, sleep disturbances, and changes in
proximately 50%) can be transferred into the womans uterus mood. Women often have a decline in libido and sexual
with the hope that a normal pregnancy will be established. satisfaction and have vaginal changes that result in difficulty
Affected embryos generally are not transferred and can be with intercourse; early menopause may also dramatically
disposed per a couples wishes. impact bone and cardiovascular health.35,36
The topic of PGD can raise a number of social and ethical There are several medical and alternative treatments that
debates. A recent large survey study of the general population can be used to mitigate the effects of menopause; the most
showed that approximately 70% of respondents were sup- effective of these is hormone replacement therapy (HRT)
portive of PGD for diseases that were lethal in early childhood or containing estrogen. The use of HRT in women with BRCA
caused lifelong disability (e.g., cystic fibrosis), yet only 48% mutation can be somewhat of a contentious subject. Currently,
supported it for screening embryos for diseases that strike later most of the literature does not show an increased risk of breast
in life (e.g., BRCA-associated cancers or Huntington disease).25 cancer following rrBSO for BRCA carriers who use HRT.37,38
Another survey assessed the attitudes about PGD among Women who use HRT report less discomfort with intercourse
women with BRCA mutations and/or hereditary cancer syn- and a higher quality of life than nonusers.39 Again, because of
dromes. Of the 52 respondents, 75% thought that PGD was an the absence of long-term studies, the lack of randomization,
acceptable option for those with BRCA mutation.26 Interestingly, and the variations in treatments used, it is still difficult to
though, of the respondents who had completed family building, counsel patients about the safety of HRT. Any woman who
only 37.5% would have used this option for themselves, and for has a uterus should also be counseled about the use of pro-
those who did not yet have children, only 14% stated they would gestin for endometrial protection. This also may impart a higher
consider using PGD. Thirty-five percent of the respondents risk of breast cancer based on previous data,40,41 but how this
stated they worried about the use of PGD because they assume applies to those with BRCA mutations is unclear.
that the effectiveness of screening and prevention for women In a recent article about her BRCA status and medical
with BRCA mutation will improve with time. decisions, Angelina Jolie said, Life comes with many chal-
Choices about the use of birth control and/or any other lenges. The ones that should not scare us are the ones we can
hormone treatments can also be complicated for BRCA take on and take control of.10 In that article, Jolie speaks
carriers. Oral birth control pills (OCPs; containing either much about her family, her partner, and her children, which
estrogen and progestin or just progestin) are the most no doubt gave her clarity on her health decisions. She did not
common form of reversible contraception used world- mention anything about PGD, so we are unclear about her
wide.27 A very large cohort study showed that in the general determination regarding this issue. Jolie also has vast access
population OCPs are not associated with an increased risk to medical care and, thus, has many treatment options. For
overall for cancer, and OCP users had a reduced risk of many women, lifes circumstances do not make decisions so
ovarian, endometrial, and colorectal cancers.28 OCP use has, clear. Decisions can be empowering, but they can also bring
however, also been linked to a trend toward increased risk of about stress and anxiety. Fortunately, most of the studies
breast cancer, but studies are conflicting.29-32 The literature show that women who undergo risk-reducing surgery are
regarding OCP use for BRCA carriers is similar.33,34 Therefore, satisfied with their decision.42,43 Unfortunately, there are
the choice about use of OCPs must be individualized and very few studies that assess womens satisfaction with their
account not only for BRCA status but also other health issues choices about nonintervention or appraise womens feelings
and reproductive goals. over long periods of time. Much additional work is needed to
Surgical menopause causes a more dramatic drop in help patients and the medical community deal effectively
systemic levels of ovarian hormones than the gradual de- with these complex issues. Even with strong data, decision
cline that occurs with natural menopause. Additionally, for making can be complicated; but without data, it is all the
BRCA carriers who undergo rrBSO per NCCN guidelines, more difficult.

References
1. American Cancer Society. Cancer Facts & Figures 2015. www.cancer.org/ 3. American Cancer Society. Family Cancer Syndromes. www.cancer.org/
research/cancerfactsstatistics/cancerfactsfigures2015. Accessed Novem- cancer/cancercauses/geneticsandcancer/heredity-and-cancer. Accessed
ber 12, 2015. January 8, 2016.
2. Tomasetti C, Vogelstein B. Cancer etiology. Variation in cancer risk 4. Couch FJ, Nathanson KL, Offit K. Two decades after BRCA: setting
among tissues can be explained by the number of stem cell divisions. paradigms in personalized cancer care and prevention. Science. 2014;
Science. 2015;347:78-81. 343:1466-1470.

16 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


WOMENS HEALTH ISSUES FOR BRCA MUTATION CARRIERS

5. Antoniou A, Pharoah PD, Narod S, et al. Average risks of breast and 25. Winkelman WD, Missmer SA, Myers D, et al. Public perspectives on the
ovarian cancer associated with BRCA1 or BRCA2 mutations detected in use of preimplantation genetic diagnosis. J Assist Reprod Genet. 2015;
case series unselected for family history: a combined analysis of 22 32:665-675.
studies. Am J Hum Genet. 2003;72:1117-1130. 26. Menon U, Harper J, Sharma A, et al. Views of BRCA gene mutation
6. Chen S, Parmigiani G. Meta-analysis of BRCA1 and BRCA2 penetrance. carriers on preimplantation genetic diagnosis as a reproductive option
J Clin Oncol. 2007;25:1329-1333. for hereditary breast and ovarian cancer. Hum Reprod. 2007;22:
7. Mavaddat N, Peock S, Frost D, et al; EMBRACE. Cancer risks for BRCA1 1573-1577.
and BRCA2 mutation carriers: results from prospective analysis of 27. United Nations. Population Facts. www.un.org/en/development/desa/
EMBRACE. J Natl Cancer Inst. 2013;105:812-822. population/publications/pdf/popfacts/popfacts_2013-9.pdf. Accessed
8. National Comprehensive Cancer Network Clinical Practice Guidelines in January 14, 2016.
Oncology. Genetic/Familial High-Risk Assessment. Version 2. 2015. 28. Hannaford PC, Selvaraj S, Elliott AM, et al. Cancer risk among users of
www.nccn.org/professionals/physician_gls/pdf/genetics_screening. oral contraceptives: cohort data from the Royal College of General
pdf. Accessed January 8, 2016. Practitioners oral contraception study. BMJ. 2007;335:651.
9. Domchek SM, Friebel TM, Singer CF, et al. Association of risk-reducing 29. Oral-contraceptive use and the risk of breast cancer. The Cancer and
surgery in BRCA1 or BRCA2 mutation carriers with cancer risk and Steroid Hormone Study of the Centers for Disease Control and the
mortality. JAMA. 2010;304:967-975. National Institute of Child Health and Human Development. N Engl J
10. Jolie A. My Medical Choice. www.nytimes.com/2013/05/14/opinion/ Med. 1986;315:405-411.
my-medical-choice.html. Accessed February 6, 2016. 30. Hankinson SE, Colditz GA, Manson JE, et al. A prospective study of oral
11. Finer LB, Philbin JM. Trends in ages at key reproductive transitions in contraceptive use and risk of breast cancer (Nurses Health Study,
the United States, 1951-2010. Womens Health Issues. 2014;24: United States). Cancer Causes Control. 1997;8:65-72.
e271-e279. 31. Marchbanks PA, McDonald JA, Wilson HG, et al. Oral contraceptives and
12. Garcia C, Wendt J, Lyon L, et al. Risk management options elected by the risk of breast cancer. N Engl J Med. 2002;346:2025-2032.
women after testing positive for a BRCA mutation. Gynecol Oncol. 2014; 32. Vessey M, Yeates D. Oral contraceptive use and cancer: final report from
132:428-433. the Oxford-Family Planning Association contraceptive study. Contra-
13. Stuckey A, Dizon D, Scalia Wilbur J, et al. Clinical characteristics and ception. 2013;88:678-683.
choices regarding risk-reducing surgery in BRCA mutation carriers. 33. McLaughlin JR, Risch HA, Lubinski J, et al; Hereditary Ovarian Cancer
Gynecol Obstet Invest. 2010;69:270-273. Clinical Study Group. Reproductive risk factors for ovarian cancer in
14. Hoskins LM, Roy K, Peters JA, et al. Disclosure of positive BRCA1/2- carriers of BRCA1 or BRCA2 mutations: a case-control study. Lancet
mutation status in young couples: the journey from uncertainty Oncol. 2007;8:26-34.
to bonding through partner support. Fam Syst Health. 2008;26: 34. Iodice S, Barile M, Rotmensz N, et al. Oral contraceptive use and breast
296-316. or ovarian cancer risk in BRCA1/2 carriers: a meta-analysis. Eur J Cancer.
15. Hamilton R. Being young, female, and BRCA positive. Am J Nurs. 2012; 2010;46:2275-2284.
112:26-31, quiz 46, 32. 35. Mercuro G, Zoncu S, Saiu F, et al. Menopause induced by oophorectomy
16. Mauer C, Spencer S, Dungan J, et al. Exploration of male attitudes on reveals a role of ovarian estrogen on the maintenance of pressure
partnerships and sexuality with female BRCA1/2 mutation carriers. homeostasis. Maturitas. 2004;47:131-138.
J Genet Couns. Epub 2015 Aug 8. 36. Lobo RA. Surgical menopause and cardiovascular risks. Menopause.
17. SART CORS. Clinic Summary Report 2013. www.sartcorsonline.com/ 2007;14:562-566.
rptCSR_PublicMultYear.aspx?ClinicPKID=0. Accessed January 21, 37. Madalinska JB, Hollenstein J, Bleiker E, et al. Quality-of-life effects of
2016. prophylactic salpingo-oophorectomy versus gynecologic screening
18. Rizzuto I, Behrens RF, Smith LA. Risk of ovarian cancer in women treated among women at increased risk of hereditary ovarian cancer. J Clin
with ovarian stimulating drugs for infertility. Cochrane Database Syst Oncol. 2005;23:6890-6898.
Rev. 2013;8:CD008215. 38. Kotsopoulos J, Huzarski T, Gronwald J, et al. Hormone replacement
19. Sergentanis TN, Diamantaras AA, Perlepe C, et al. IVF and breast cancer: therapy after menopause and risk of breast cancer in BRCA1 mutation
a systematic review and meta-analysis. Hum Reprod Update. 2014;20: carriers: a case-control study. Breast Cancer Res Treat. 2016;155:
106-123. 365-373.
20. Gennari A, Costa M, Puntoni M, et al. Breast cancer incidence after 39. Johansen N, Liavaag AH, Tanbo TG, et al. Sexual activity and functioning
hormonal treatments for infertility: systematic review and meta- after risk-reducing salpingo-oophorectomy: impact of hormone re-
analysis of population-based studies. Breast Cancer Res Treat. 2015; placement therapy. Gynecol Oncol. 2016;140:101-106.
150:405-413. 40. Chlebowski RT, Hendrix SL, Langer RD, et al; WHI Investigators. Influ-
21. Zhao J, Li Y, Zhang Q, et al. Does ovarian stimulation for IVF increase ence of estrogen plus progestin on breast cancer and mammography in
gynaecological cancer risk? A systematic review and meta-analysis. healthy postmenopausal women: the Womens Health Initiative Ran-
Reprod Biomed Online. 2015;31:20-29. domized Trial. JAMA. 2003;289:3243-3253.
22. Kessous R, Davidson E, Meirovitz M, et al. The risk of female malig- 41. Chlebowski RT, Rohan TE, Manson JE, et al. Breast cancer after use of
nancies after fertility treatments: a cohort study with 25-year follow-up. estrogen plus progestin and estrogen alone: analyses of data from 2
J Cancer Res Clin Oncol. 2016;142:287-293. Womens Health Initiative Randomized Clinical Trials. JAMA Oncol.
23. Gronwald J, Glass K, Rosen B, et al; Hereditary Breast Cancer Clinical 2015;1:296-305.
Study Group. Treatment of infertility does not increase the risk of 42. Borreani C, Manoukian S, Bianchi E, et al. The psychological impact of
ovarian cancer among women with a BRCA1 or BRCA2 mutation. Fertil breast and ovarian cancer preventive options in BRCA1 and BRCA2
Steril. Epub 2015 Dec 14. mutation carriers. Clin Genet. 2014;85:7-15.
24. Perri T, Lifshitz D, Sadetzki S, et al. Fertility treatments and invasive 43. Finch A, Narod SA. Quality of life and health status after prophylactic
epithelial ovarian cancer risk in Jewish Israeli BRCA1 or BRCA2 mutation salpingo-oophorectomy in women who carry a BRCA mutation: a re-
carriers. Fertil Steril. 2015;103:1305-1312. view. Maturitas. 2011;70:261-265.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 17


INVITED ARTICLES COLLECTIVE WISDOM

Collective Wisdom: Lobular Carcinoma of the Breast


George W. Sledge, MD, FASCO, Anees Chagpar, MD, and Charles Perou, PhD

B reast cancer researchers have a longstanding fascination


for infiltrating lobular carcinomas. Second in frequency
only to ductal adenocarcinomas, these tumors are charac-
decreased E-cadherin mRNA levels were uniformly observed
in ILC cases. In total, 63% of ILC cases had an E-cadherin
mutation, and 95% of ILC cases had loss when mutation, DNA
terized by unique histopathology and (among breast can- copy number, and low gene and/or protein expression were
cers) distinctive clinical biology, both in the primary and summed together. It is reassuring that this modern study
metastatic settings. Early hints regarding the underlying reaffirmed the primacy of E-cadherin loss in lobular breast
sources of this peculiar cancer, in particular the important cancer, and if a biomarker were to be chosen to identify ILC
role of E-cadherin loss, have now been confirmed through on a molecular level, it would likely be low E-cadherin
comprehensive molecular portraits of the disease. These protein expression and/or DNA mutation of CDH1. Lobu-
molecular observations, in turn, go far to explain how lobular lar carcinomas unique histopathologic features and its
carcinomas play out in the clinic, both as regards local metastatic patterns undoubtedly have their origins in their
control and therapeutic response to systemic therapy. To- impoverished E-cadherin status.
gether, the collective wisdom of the laboratory and the Beyond previously reported CDH1 and PIK3CA mutations
clinic paint an interesting portrait of this fascinating disease. (which occur at a 48% frequency in ILC), the TCGA study
identified a number of novel ILC-enriched recurrent muta-
tions targeting FOXA1, PTEN, RUNX1, and TBX3. FOXA1
MOLECULAR BIOLOGY OF LOBULAR function is particularly intriguing, as it works with the es-
CARCINOMAS trogen receptor (ER) to drive the transcriptional output of
Invasive lobular breast carcinoma (ILC) is the second most ER. Interestingly, FOXA1 also plays a similar role in prostate
prominent histologic form of breast cancer and accounts for cancer, but in these cancers its cofactor is the androgen
10%15% of invasive breast tumors. On a molecular level, receptor.2 In ILC, we find an increased incidence (9% in ILC vs.
lobular breast cancers are a distinct disease type and should 2% in IDC) of FOXA1 mutations, whereas in IDC, we find that
be considered as a unique disease. These molecular un- GATA3 mutations are considerably enriched in IDC luminal
derpinnings were recently intensely studied as part of The tumors (19% IDC vs. 5% ILC). Within ILC tumors, FOXA1
Cancer Genome Atlas (TCGA) effort on breast cancer, mutations were found to cluster into a specific region of the
including a recent publication focused on lobular breast forkhead (FK) domain. A broader analysis of FOXA1 muta-
cancers.1 In this publication, 817 breast tumors from the tions in breast and prostate cancer, in which it is also re-
TCGA project, including 490 invasive ductal cancers (IDCs), currently mutated, confirms two specific hotspots in the FK
127 ILCs, and 88 samples with a mixed IDC-ILC histology, domain and the C-terminal transactivation domain. In-
were molecularly profiled on six genomic platforms to de- terestingly, these mutational classes were associated with
velop a comprehensive portrait of the genetic, epigenetic, higher FOXA1 messenger RNA and protein expression, and
transcriptional, and proteomic landscape of lobular breast with unique transcriptional changes suggesting different
cancers. Comprehensive multiplatform analyses, both su- functional effects.3 More work into why FOXA1 is mutated in
pervised and unsupervised, of ILC tumors and across his- ILC, but GATA3 is mutated in IDC, is needed and could
tologic subtypes were performed to identify genomic possibly reveal some important underlying biology.
drivers of ILC oncogenesis. Another important finding concerning ILC was the in-
As expected, low expression of E-cadherin protein, as creased incidence of phosphatase and tensin homologue
determined by reverse-phase protein array (RPPA), and (PTEN)-inactivating events. When including both mutations

From the Division of Oncology, Stanford University School of Medicine, Stanford, CA; Yale University, New Haven, CT; The University of North Carolina at Chapel Hill, Chapel
Hill, NC.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: George W. Sledge, MD, Stanford University School of Medicine, 269 Campus Dr., CCSR-1115, Stanford, CA 94305; email: gsledge@stanford.edu.

2016 by American Society of Clinical Oncology.

18 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


LOBULAR CARCINOMA OF THE BREAST

and DNA copy number changes, ILC-luminal A showed a 13% TABLE 1. Imaging Concordance With Pathologic
PTEN altered phenotype compared with 3% in IDC-luminal A Tumor Size (Correlation Coefficient)
tumors. This increased mutation frequency of PTEN loss in
ILC correspond with decreased PTEN protein expression and Study MRI Mammography Ultrasound Clinical Exam
was largely mutually exclusive with PIK3CA mutations. Anal- Boetes et al25 0.81 0.34 0.24
yses of RPPA protein and phosphor-protein expression data Francis et al 26
0.87 0.79 0.56 0.89
demonstrated increased phosphoinositide-3 kinase/Akt signal- Kepple et al27 0.88 0.71
ing as evident by increased levels of phosphorylated Akt (pS473 Kneeshaw et al28 0.86 0.47
and pT308) and downstream Akt substrates including p-p27 and 29
Munot et al 0.97 0.66 0.67
p-p70S6 kinase in ILC tumors; these findings may represent a
potential therapeutic opportunity for patients with ILC.
In terms of gene-expression patterns, ILC was found to be conservation are more likely to result in local recurrences13;
of the luminal A subtype in 83% of the cases. Within this ILC- however, this has not been borne out in other studies,9,10,14
luminal A subset, additional expression subtypes were also and long-term survival rates are no different between breast
identified. These included a subset enriched for stromal and conservation and mastectomy.15 Hence, both are consid-
extracellular matrix features, often referred to as the re- ered appropriate in terms of surgical management for this
active subtype.4 Another ILC expression subtype was en- disease. Of note, invasive lobular histology falls into the
riched for immune cell features/infiltrates, whereas the American Society for Radiation Oncologys cautionary
third group showed a more proliferative expression signa- subgroup for the use of accelerated partial breast irradia-
ture and a concomitant worse patient outcome. Finally, a tion,16 given concerns regarding higher ipsilateral breast
multiplatform analysis of the mixed histology tumors tumor recurrences in this population.
showed that approximately 80% of these samples could
clearly be molecularly classified as either ILC or IDC, with Lymph Node Evaluation
only a few showing a possibly hybrid phenotype. This could Besides tumor extirpation from the breast, the other key task
have important implications for treatment of patients with of the breast surgeon is lymph node evaluation. Although it is
mixed ILC-IDC histology, as it suggests they are not a unique clear that sentinel node biopsy is feasible and accurate in
group, but instead that their molecular features could be patients with ILC, the ability to find micrometastatic deposits
used to classify them as either ILC or IDC. particularly on intraoperative evaluation with hematoxylin and
eosin staining alone may be challenging given the discohesive
SURGICAL ASPECTS OF LOBULAR CARCINOMA nature of the neoplastic cells. Some pathologists have not
ILC is a distinct subtype of breast cancer that is deserving of found this to be problematic.17 Others, however, may rec-
particular attention by surgeons. ILCs tend to be insidious ommend deferral of final diagnosis to permanent sections, at
as a result of their lack of E-cadherin, causing noncohesive which time immunohistochemistry can be used to draw at-
neoplastic cells that permeate through tissue in a single-file tention to deposits that otherwise could be missed.18
pattern. Given its biology, a few areas are of particular
concern for surgeons. SYSTEMIC THERAPY FOR LOBULAR CARCINOMA:
CHEMOTHERAPY
Response to Neoadjuvant Therapy
Preoperative Imaging Increasingly, neoadjuvant chemotherapy is part of the mul-
It is clear that the extent of ILCs tend to be underestimated tidisciplinary approach to breast cancer, as more novel ther-
by conventional imaging (Table 1). Some have suggested apeutics are being evaluated in this setting. However,
that MRI may be useful in this context5; however, a recent physicians and patients should be aware that response rates to
meta-analysis found that MRI did not significantly reduce neoadjuvant chemotherapy are lower for ILC than for its ductal
positive margin rates in patients with ILC undergoing breast counterpart, as illustrated by the lower rates of pathologic
conservation.6 The concept that MRI could also find occult complete response and breast conservation (Table 2).
contralateral disease has also been raised; however, ILCs are Why is neoadjuvant chemotherapy relatively ineffective for
no more likely to have a synchronous contralateral cancer lobular carcinomas? Is it something intrinsic to the biology of
than are IDCs.7 MRI is therefore not routinely recommended lobular carcinomas per se? Mathieu et al19 have argued that
in the presurgical workup of patients with ILCs.8 this relative futility is predictable, in that histologic and bi-
ologic factors predicting a poor response to chemotherapy
Breast-Conserving Surgery Versus Mastectomy (low histologic grade, high ER content and bcl-2 expression,
Regardless of whether neoadjuvant chemotherapy is used or and low proliferative rates as measured by Ki67 and negative
not, ILCs are more often associated with positive margins p53 staining) are all more frequent in lobular rather than
after breast-conserving surgery.911 Patients with this his- ductal carcinomas. These, in turn, reflect the luminal A
tologic subtype are more likely to require re-excision, and nature of most lobular carcinomas, discussed above.
potentially mastectomy, for margin clearance.12 Some have There is a much smaller body of data evaluating neoadju-
suggested that invasive lobular cancers treated with breast vant hormonal therapy for lobular carcinoma. Dixon et al20

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 19


SLEDGE, CHAGPAR, AND PEROU

TABLE 2. Response to Neoadjuvant Chemotherapy


Study No. Patients pCR (%) p Value BCS (%) p Value
Truin et al30 IDC: 3,622 20.2 , .0001 39.4 , .0001
ILC: 466 4.9 24.4
Loibl et al31 Non-ILC: 7,969 17.4 , .001 71.1 , .0001
ILC: 1,051 6.2 59.1
Lips et al32 IDC: 601 25 .01 46 .037
ILC: 75 11 33
Wenzel et al33 IDC: 124 20 .009 79 .001
ILC: 37 3 51
Tubiana-Hulin et al34 IDC: 742 9 .002 48 .0004
ILC: 118 1 30
Cocquyt et al35 IDC: 101 15 .0066 50 NS
ILC: 26 0 38
Abbreviations: pCR, pathologic complete response; BCS, breast-conserving surgery; IDC, invasive ductal carcinoma; ILC, invasive lobular carcinoma; NS, not significant.

evaluated the responsiveness of lobular carcinomas to neo- TEAM trial design in that early switch (2 to 3 years of ta-
adjuvant letrozole in 61 patients treated for 3 months. Mean moxifen followed by 2 to 3 years with an aromatase inhibitor)
tumor volume reduction was 66%, with a high rate of breast and up-front (5 years with an aromatase inhibitor) strategies
conservation (81%). Although we lack any head-to-head com- were compared for relative benefit in lobular and invasive
parisons of neoadjuvant endocrine therapy with neoadjuvant ductal cancers. The TEAM analysis suggested that endocrine
chemotherapy (and may never see such a comparison), a pri- therapy efficacy was similar for IDC and ILC once one had
mary endocrine approach does not seem unreasonable. adjusted for ER content. The early switch strategy arm might
well muzzle the treatment interactions seen in the BIG 1-98 trial.
If, as suggested by the BIG 1-98 analysis, lobular carci-
RESPONSE TO HORMONAL THERAPY nomas are relatively less sensitive to tamoxifen than ductal
If lobular carcinoma is relatively resistant to standard chemo- carcinomas, what molecular changes might underlie these
therapeutic agents, and if this is largely as a result of predictable findings? Sikora et al23 have recently evaluated the response
biology (i.e., higher ER and lower proliferative rates), then what of lobular carcinoma cell lines in vitro. Their work suggests
about adjuvant hormonal therapy? Recent data suggest that that the ER drives a unique program of gene expression in
not all hormonal therapies are equal where lobular cancer is lobular cancers when compared with ductal carcinomas.
concerned. In particular, tamoxifen appears to be considerably Indeed, tamoxifen appears to drive the growth of these cell
less effective than aromatase inhibition for lobular cancers. lines, rather than inhibiting them, although this limited cell
Metzger Filho et al21 compared the relative efficacies of line work cannot be safely extrapolated to the clinic.
tamoxifen and letrozole for lobular and ductal carcinomas in In contrast to the preclinical results seen with tamoxifen,
the BIG 1-98 trial. This trial was among the first randomized Arthur et al24 have recently demonstrated in the neo-
controlled trials of aromatase inhibitor therapy in the ad- adjuvant hormonal therapy setting that changes in gene
juvant setting and now has relatively long follow-up (median expression in response to letrozole were highly similar be-
8.1 years). Comparing patients by histologic subtype, patients tween responding ILC and IDC tumors.
with ILC were far more likely to benefit from letrozole than
tamoxifen, regardless of whether patients were luminal A CONCLUSION
or luminal B like. The 8-year disease-free survival estimate Infiltrating lobular carcinoma of the breast represents a bi-
was 66% for tamoxifen compared with 82% for letrozole in ologically distinct subset of breast cancer, a biology defined
the ILC subset (hazard ratio [HR] 0.48) and was 75% for by specific genetic aberrations in E-cadherin, the high
tamoxifen and 82% for letrozole in the IDC subset (HR 0.80). prevalence of ER-positive disease, the relatively low fre-
The test for interaction was significantly positive (p = .006). quency of HER2-positive disease, and specific mutational
These seem, on the face of it, to represent a clinically sig- events revealed by deep sequencing of ILC genomes. This
nificant difference and are paralleled by overall survival distinctive biology, in turn, affects the presentation, treat-
differences (74% for tamoxifen compared with 89% for ment, andpotentiallythe prognosis of ILC. Biology af-
letrozole in the ILC subset [HR 0.40]; 84% for tamoxifen and fects surgery and preoperative chemotherapy results, and,
88% for letrozole in the IDC subset [HR 0.73]). as recent data suggest, it also affects adjuvant hormonal
In contrast, van de Water et al22 have examined the ad- therapy benefits. Ultimately, an improved understanding of
juvant TEAM (Tamoxifen and Exemestane Adjuvant Multi- ILC biology should also lead to novel targeted approaches to
national) trial. This analysis differed considerably in that the the conquest of the disease.

20 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


LOBULAR CARCINOMA OF THE BREAST

References

1. Ciriello G, Gatza ML, Beck AH, et al; TCGA Research Network. Comprehensive 19. Mathieu MC, Rouzier R, Llombart-Cussac A, et al. The poor re-
molecular portraits of invasive lobular breast cancer. Cell. 2015;163:506-519. sponsiveness of infiltrating lobular breast carcinomas to neoadjuvant
2. Jozwik KM, Carroll JS. Pioneer factors in hormone-dependent cancers. chemotherapy can be explained by their biological profile. Eur J Cancer.
Nat Rev Cancer. 2012;12:381-385. 2004;40:342-351.
3. Cancer Genome Atlas Research Network. The molecular taxonomy of 20. Dixon JM, Renshaw L, Dixon J, et al. Invasive lobular carcinoma: re-
primary prostate cancer. Cell. 2015;163:1011-1025. sponse to neoadjuvant letrozole therapy. Breast Cancer Res Treat. 2011;
4. Cancer Genome Atlas Network. Comprehensive molecular portraits of 130:871-877.
human breast tumours. Nature. 2012;490:61-70. 21. Metzger Filho O, Giobbie-Hurder A, Mallon E, et al. Relative effec-
5. Mann RM, Hoogeveen YL, Blickman JG, et al. MRI compared to con- tiveness of letrozole compared with tamoxifen for patients with lobular
ventional diagnostic work-up in the detection and evaluation of invasive carcinoma in the BIG 1-98 trial. J Clin Oncol. 2015;33:2772-2779.
lobular carcinoma of the breast: a review of existing literature. Breast 22. van de Water W, Fontein DB, van Nes JG, et al. Influence of semi-
Cancer Res Treat. 2008;107:1-14. quantitative oestrogen receptor expression on adjuvant endocrine
6. Houssami N, Turner R, Morrow M. Preoperative magnetic resonance therapy efficacy in ductal and lobular breast cancer - a TEAM study
imaging in breast cancer: meta-analysis of surgical outcomes. Ann Surg. analysis. Eur J Cancer. 2013;49:297-304.
2013;257:249-255. 23. Sikora MJ, Cooper KL, Bahreini A, et al. Invasive lobular carcinoma cell
7. Langlands F, White J, Kearins O, et al. Contralateral breast cancer: lines are characterized by unique estrogen-mediated gene expression
incidence according to ductal or lobular phenotype of the primary. Clin patterns and altered tamoxifen response. Cancer Res. 2014;74:
Radiol. 2016;71:159-163. 1463-1474.
8. Pilewskie M, King TA. Magnetic resonance imaging in patients with 24. Arthur LM, Turnbull AK, Webber VL, et al. Molecular changes in lobular
newly diagnosed breast cancer: a review of the literature. Cancer. 2014; breast cancers in response to endocrine therapy. Cancer Res. 2014;74:
120:2080-2089. 5371-5376.
9. Fortunato L, Mascaro A, Poccia I, et al. Lobular breast cancer: same 25. Boetes C, Veltman J, van Die L, et al. The role of MRI in invasive lobular
survival and local control compared with ductal cancer, but should both carcinoma. Breast Cancer Res Treat. 2004;86:31-37.
be treated the same way? analysis of an institutional database over a 26. Francis A, England DW, Rowlands DC, et al. The diagnosis of invasive
10-year period. Ann Surg Oncol. 2012;19:1107-1114. lobular breast carcinoma. Does MRI have a role? Breast. 2001;10:38-40.
10. Molland JG, Donnellan M, Janu NC, et al. Infiltrating lobular carcinoma 27. Kepple J, Layeeque R, Klimberg VS, et al. Correlation of magnetic
a comparison of diagnosis, management and outcome with infiltrating resonance imaging and pathologic size of infiltrating lobular carcinoma
duct carcinoma. Breast. 2004;13:389-396. of the breast. Am J Surg. 2005;190:623-627.
11. Moore MM, Borossa G, Imbrie JZ, et al. Association of infiltrating lobular 28. Kneeshaw PJ, Turnbull LW, Smith A, et al. Dynamic contrast enhanced
carcinoma with positive surgical margins after breast-conservation magnetic resonance imaging aids the surgical management of invasive
therapy. Ann Surg. 2000;231:877-882. lobular breast cancer. Eur J Surg Oncol. 2003;29:32-37.
12. Kryh CG, Pietersen CA, Rahr HB, et al. Re-resection rates and risk 29. Munot K, Dall B, Achuthan R, et al. Role of magnetic resonance imaging
characteristics following breast conserving surgery for breast cancer in the diagnosis and single-stage surgical resection of invasive lobular
and carcinoma in situ: a single-centre study of 1575 consecutive cases. carcinoma of the breast. Br J Surg. 2002;89:1296-1301.
Breast. 2014;23:784-789. 30. Truin W, Vugts G, Roumen RM, et al. Differences in response and
13. Hussien M, Lioe TF, Finnegan J, et al. Surgical treatment for invasive surgical management with neoadjuvant chemotherapy in invasive
lobular carcinoma of the breast. Breast. 2003;12:23-35. lobular versus ductal breast cancer. Ann Surg Oncol. 2016;23:51-57.
14. Ott OJ, Hildebrandt G, Potter R, et al. Accelerated partial breast 31. Loibl S, Volz C, Mau C, et al. Response and prognosis after neoadjuvant
irradiation with interstitial implants: risk factors associated with chemotherapy in 1,051 patients with infiltrating lobular breast carci-
increased local recurrence. Int J Radiat Oncol Biol Phys. 2011;80: noma. Breast Cancer Res Treat. 2014;144:153-162.
1458-1463. 32. Lips EH, Mukhtar RA, Yau C, et al; I-SPY TRIAL Investigators. Lobular
15. Fodor J, Major T, Toth
J, et al. Comparison of mastectomy with breast- histology and response to neoadjuvant chemotherapy in invasive breast
conserving surgery in invasive lobular carcinoma: 15-year results. Rep cancer. Breast Cancer Res Treat. 2012;136:35-43.
Pract Oncol Radiother. 2011;16:227-231. 33. Wenzel C, Bartsch R, Hussian D, et al. Invasive ductal carcinoma and
16. Smith BD, Arthur DW, Buchholz TA, et al. Accelerated partial breast invasive lobular carcinoma of breast differ in response following
irradiation consensus statement from the American Society for Radi- neoadjuvant therapy with epidoxorubicin and docetaxel + G-CSF. Breast
ation Oncology (ASTRO). Int J Radiat Oncol Biol Phys. 2009;74:987-1001. Cancer Res Treat. 2007;104:109-114.
17. Horvath JW, Barnett GE, Jimenez RE, et al. Comparison of intraoperative 34. Tubiana-Hulin M, Stevens D, Lasry S, et al. Response to neoadjuvant
frozen section analysis for sentinel lymph node biopsy during breast chemotherapy in lobular and ductal breast carcinomas: a retrospective
cancer surgery for invasive lobular carcinoma and invasive ductal study on 860 patients from one institution. Ann Oncol. 2006;17:
carcinoma. World J Surg Oncol. 2009;7:34. 1228-1233.
18. Cserni G, Bianchi S, Vezzosi V, et al. The value of cytokeratin immu- 35. Cocquyt VF, Blondeel PN, Depypere HT, et al. Different responses to
nohistochemistry in the evaluation of axillary sentinel lymph nodes in preoperative chemotherapy for invasive lobular and invasive ductal
patients with lobular breast carcinoma. J Clin Pathol. 2006;59:518-522. breast carcinoma. Eur J Surg Oncol. 2003;29:361-367.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 21


POINTS OF VIEW
The section, new for 2016, contains articles describing emerging, highly debated, or controversial topics
in cancer research, treatment, and care to benefit patients and the field of oncology.

AUTHORS
Arthur L. Caplan, PhD
NYU Langone Medical Center
New York, NY

Laura Esserman, MD, MBA


University of California, San Francisco
San Francisco, CA

David J. Kerr, MD, PhD


University of Oxford
Oxford, United Kingdom

Ulka N. Vaishampayan, MD
Karmanos Cancer Institute
Detroit, MI
CAPLAN, BATEMAN-HOUSE, AND WALDSTREICHER

Compassionate Use: A Modest Proposal


Arthur L. Caplan, PhD, Alison Bateman-House, PhD, MPH, MA, and Joanne Waldstreicher, MD

Editors Note: The following article is based on the 2016 ASCO Annual Meeting Education Session Expanded Access and the
Right to Try: Navigating the Intersection of Drug Development and Patient Access to Investigational Agents. The authors
present the approach to compassionate use through use of an independent committee to vet and review requests for one
pharmaceutical company.

P atient requests for rapid access to agents that are still in


the research pipeline fall into two categories: requests
from groups of persons with the same malady and requests
who own the drug or agent. Regardless of the approach, this
process is, at best, hit or miss and one that favors patients
with a compelling personal interest story or who are oth-
by individuals. The former are often described as requests erwise well-connected.
for expanded access, the latter as requests for compas- Many have claimed that the key obstacle for patients
sionate use. Regulatory bodies in many countries have seeking compassionate use access is the U.S. Food and Drug
created programs for providing greater access to requests Administration (FDA). However, estimates are that the FDA
from groups including the creation of expanded access approves over 99% of the compassionate use protocols it
programs and emergency use waivers for patients who do receives.1 Furthermore, the FDA plays no role in responding
not qualify for clinical trials. Compassionate use requests for to requests for compassionate use until the company de-
individuals have proven to be more difficult to resolve. veloping the agent has indicated its willingness to provide
Compassionate use requests can come at any time during the product. In contrast, companies encounter multiple
the research processfrom when a drug is being tested in issues when faced with compassionate use requests. These
animals, to its first-in-human studies, to dose-finding trials, include the fact that they have no legal duty to offer access
to when the agent under investigation nears the end of and that they must balance requests with ongoing clinical
clinical trials. Requests can come from patients nearing the development programs. In addition, even if the company
end of life, but they may also come from those facing serious wanted to respond, there may be uncertainty as to what the
disability and pain for which no approved agent has been response should be and how best to respond to all requests
proven effective. Requests can come from around the world fairly, particularly when there are those who are well-
and from parents for their children, patients on their connected and/or using social media campaigns.
deathbeds, those in the midst of lethal disease outbreaks, In this article, we review the approach taken by one of
those newly infected as well as the chronically ill, the very many companies (Janssen) that receives such requests,
poor, and those for whom money is no object. which involved the formation of a third party. Internal
Until very recently, the main strategy for patients seeking discussions at Janssen had focused on how to best handle
compassionate use was to try to locate a possible treatment, compassionate use requests consistent with its commit-
frequently online, and often, though not always, with the ment to obtain regulatory approval for investigational
help (or approval) of their physician. Once a possible treat- agents, thereby making products available to the greatest
ment was found, patients and/or their physicians seeking number of persons. Ultimately, Janssen decided to un-
compassionate use would try to contact the researcher or dertake a first-of-its-kind partnership that enlisted a third
leader of a group involved with testing the agentalmost party to review requests made to the company for com-
always at a company. Some are able to use personal con- passionate use. This novel approach was initiated as a pilot
nections to initiate a request, whereas others reach out program focused on a single investigational medicine,
to their elected officials, shareholders in the company, or daratumumab, which was being evaluated in late-stage
others thought to wield influence. Sometimes patients try to phase III trials at the time and had shown evidence of
interest the traditional media in their plight. Alternatively, efficacy in patients with refractory multiple myeloma.
they may launch campaigns using social media with the hope Janssen approached the director of the Division of Medical
that public pressure might be brought to bear on the parties Ethics at NYU Langone Medical Center, Arthur Caplan, PhD,

From the New York University School of Medicine, New York, NY; NYU Langone Medical Center, New York, NY; Johnson & Johnson, New Brunswick, NJ.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Arthur L. Caplan, PhD, New York University School of Medicine, 227 East 30th St., New York, NY 10016; email: arthur.caplan@nyumc.org.

2016 by American Society of Clinical Oncology.

e2 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


COMPASSIONATE USE REQUESTS

to propose a method for reviewing compassionate use re- 1. worked to improve navigation of Janssens worldwide
quests that would be transparent, fair, beneficent, evidence- websites so that those seeking compassionate use for
based, and patient-focused. Drawing upon his prior work multiple myeloma would be able to more easily find
involving the allocation of cadaver organs for transplan- clear information on how to request daratumumab,
tation, Caplan formed a 10-person committee, the Com- 2. insisted that requests came from physicians and be
passionate Use Advisory Committee (CompAC), that consists standardized so that all requests would have the same
of physicians, bioethicists, patients, and patient advocates information, unadorned by letters of support from
from around the world to advise Janssen on compassionate prominent individuals,
use requests for daratumumab. The CompAC was created 3. decided to rotate voting on requests among three of
and staffed by the New York University School of Medicine its 10 members weekly to minimize any effort to lobby
(NYUSOM) and was independent of Janssen. CompAC the committee members,
members contracted with NYUSOM to remain indepen-
4. directed Janssen to redact information pertaining to
dent and reduce the risk of conflicts of interest. Some
the requesters name, location, nationality, and gen-
CompAC members accepted payment for their time;
der to prevent these factors from being taken into
the chair and deputy chair, however, received no direct
consideration by voting members,
compensation.
5. created an appeal process by which a physician whose
request was denied could reintroduce further stan-
HOW THE COMPAC WORKS dardized information about his or her patient (i.e., new
Compassionate use requests for daratumumab are received
laboratory values), and
directly at Janssen through a standardized patient intake
6. committed to respond to all requests within 5 business
form on the companys website. Janssen physicians screen
days, thereby ensuring that appropriate concern was
the intake forms for medical appropriateness, and, if the
shown to all patients and that no one would be left
patient is eligible for a clinical trial, expanded access pro-
gram, or has not yet tried an available drug or program, without an expeditious answerthe opposite of which
Janssen informs the submitting physician of this and is commonly a source of great patient and physician
does not route the request to CompAC. For all other frustration about compassionate use requests.
cases, CompAC receives requests on a weekly basis and Soon after the creation of the CompAC in May 2015, these
returns its recommendation to Janssen within 5 business policies were implemented. Further discussion among
days of receiving each request. CompAC is an advisory CompAC members led to an agreement on a set of principles
committee, and, legally, Janssen must remain in control that would be used to guide the committees recommen-
of the ultimate decision to provide access; however, dations in response to requests. These included: preventing
Janssen made a good faith commitment to follow CompAC harm to patients, requiring that patients had exhausted all
recommendations. available approved treatments before trying unapproved
Although CompAC spent a good deal of time discussing agents, estimating the likelihood of obtaining an efficacious
what, if any, criteria it would use in deciding who to rec- response, and assessing patient functionality, and, at a lower
ommend to receive access, it quickly became evident that order of priority, prior participation in a clinical trial, direct
the key ethical requirement that the CompAC had to achieve support for dependents, suffering from the disease, and age.
was fairness. The existing pathways for making requests of Decision making in response to requests commenced in July
Janssen and other companies were not always fair, some- 2015, with an average of four to five requests per week.
times favoring the rich, celebrities, and those who could Supply varied from week to week including some weeks in
command attention. To work, CompAC had to ensure that it which there was none.
would strengthen the fairness of compassionate use de-
cisions to better meet the needs of patients, doctors, re-
searchers, and the public. To do so, the CompAC: LESSONS TO DATE
Although there is obvious interest both in who was se-
lected, who was rejected, and on what basis, the major
lesson learned from this pilot program is that it is fairness,
KEY POINTS rather than justice, that is the key to the success of the
CompAC. By having a committee with broad expertise that
This article describes the creation of a dedicated is insulated from what ought to be morally irrelevant
committee to respond to compassionate use requests
considerations of wealth, privilege, and media interest,
for an investigational drug for multiple myeloma.
This article describes the importance of creating
the CompAC has been able to institute strategies that
principles of fair opportunity for those making requests minimize the ability of any patient, family, advocacy
for compassionate use. group, or advantaged party to sway decisions. Patients,
This article suggests that this model may have their doctors, and Janssen officials report that they be-
application to other agents and settings. lieve that the pilot approach is more equitable than what
existed beforehand.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e3


CAPLAN, BATEMAN-HOUSE, AND WALDSTREICHER

The CompAC model may be applied to other novel agents certainly not beyond criticism as to its composition, prin-
across Johnson & Johnson. Other companies, both large and ciples, and decisions. However, the CompAC has identified
small, patient advocacy organizations, and government of- ways to make very hard decisions more equitable. Those
ficials have all expressed interest in learning about or deserve close attention and debate in a world with many
extending the CompAC model to other settings. more unapproved agents that will be sought by the des-
The CompAC pilot program has been resource intensive, perate who face constraints upon the resources available
involving many different groups to support its activities, and is to them.

References

1. Silverman E. The FDA Says Its More Compassionate Than You Think.
Healthcare Blog, The Wall Street Journal. 2014. http://blogs.wsj.com/
corporate-intelligence/2014/05/05/the-fda-says-its-more-compassionate-than-
you-think/. Accessed February 24, 2016.

e4 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


THE EVOLVING SURGICAL APPROACH TO BREAST CANCER

Less Is More: The Evolving Surgical Approach to Breast Cancer


Laura Esserman, MD, MBA, Etienne Gallant, and Michael Alvarado, MD

Editors Note: The following article is based on the 2016 ASCO Annual Meeting Education Session Less Is More: A Multi-
disciplinary Conversation on Treatment Options. The authors review the indications for reducing the treatment burden for
women with breast cancer by capitalizing on the emerging opportunity to identify and recognize more indolent forms of the
disease using advanced tools, therapies, and screening methods.

OVERVIEW

Personalized medicine is emerging as an important guiding principle in diagnosis and treatment. This means not just doing
more for some, but safely doing less for others. The lessons learned about the biology of breast cancer over the last 2 decades
have enabled us to understand the incredible heterogeneity of breast cancer and its associated behavior. Although much
work remains, there is an emerging opportunity to identify and recognize more indolent forms of breast cancer, made more
prevalent through the widespread adoption of screening. With our improving systemic therapies and improved molecular
tools, we now have the opportunity to reduce the burden of treatment in women with lower-risk tumors. Our surgical
treatments have evolved, with less morbid and more cosmetic procedures. In this article, we review the indications for
further reducing local therapy, including adjuvant radiation.

T he history of breast surgery is one in which we have gone


from more to less. Radical mastectomies, when first
introduced by Halsted, were an improvement over leaving
reason is that the benefit from an intervention and its side
effects may not outweigh the risk of the disease. Thus, a
postmenopausal woman with favorable-risk hormone
women with fungating masses. However, the field evolved receptorpositive breast cancer has several options after
through a series of randomized trials to minimize the breast conservation. In addition to what has been the gold
morbidity of our surgical procedures. Modified radical standard, standard dose external beam radiation therapy
mastectomies have replaced radical mastectomies. Imme- (EBRT), there are several alternatives including hypo-
diate reconstruction and skin-sparing mastectomy, and now fractionated EBRT, intraoperative radiation therapy (IORT),
total skinsparing mastectomies, have been shown to be or no radiation, which are all supported by the literature as
oncologically safe and cosmetically better.1,2 We have re- at least equivalent, if not superior, strategies to EBRT.5,6 And
duced extended axillary procedures, and sentinel node yet, trial results have not significantly influenced practice in
dissections have fortunately successfully reduced the the United States.
complications of lymphedema. Even in the setting of stage II Over time, the local recurrence rates have been decreasing
and III disease, with axillary disease, surgeons are finding from the original reported risk of 10% EBRT. Recent studies
safe ways to reduce the burden of axillary surgery in the show rates largely less than 5% and even lower when en-
setting of excellent responses to neoadjuvant chemother- docrine therapy is used. Data from four trials with pop-
apy. Especially because we are increasingly more successful ulations of similar biology (postmenopausal women with
in preventing recurrence and death from breast cancer and node-negative, hormone receptorpositive, early-stage breast
as women lead longer lives, we must pay more attention to cancer) are shown in Table 1. Local recurrence rates are very
identifying the opportunities to reduce the burden of low in the EBRT arms or in the complete absence of radiation.
treatment. We now have the opportunity to focus on the Fyles et al found that women over age 50 in the low-risk
advances in molecular biology to help us to identify tumors groupimmunohistochemistry subtype luminal Ahad a
with indolent behavior and to adjust our local therapy local recurrence of 4.9% at 10 years with or without EBRT.7
accordingly.3,4 Intraoperative radiation therapy results, which also show a
Decision making for patients with low-risk disease is often very low locoregional recurrence, are consistent with other
more complicated than for those with high-risk disease. The modern trial results. It should be noted that there was no

From the University of California, San Francisco, San Francisco, CA.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Laura Esserman, MD, MBA, University of California, San Francisco, 1600 Divisadero St., 2nd Floor, Box 1710, San Francisco, CA; email: laura.esserman@ucsf.edu.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e5


ESSERMAN, GALLANT, AND ALVARADO

difference in metastatic rates or overall survival for any of When the CALGB 9343 results were presented in 2002,
the trial arms. they met a similar reaction to the results of the TARGIT-A
The question of how to approach patients with early- results in 2010. Many demanded 10 years of data prior to
stage, favorable-risk breast cancer illustrates the complexity adoption. However, even with 10 years of data demon-
of forces influencing decision making in regard to adoption strating that mortality is not impacted by radiation in women
of new approaches. Whole-breast EBRT remains the stan- over age 70, or even with the publication of the PRIME 2 trial
dard of care following breast-conserving surgery. However, results,13 the standard of care after breast-conserving sur-
multidose partial breast radiation is increasingly offered as gery has remained EBRT.
an alternative for eligible women. Emerging technologies Partial breast radiation as an alternative to EBRT has been
have provided impetus for shifts in radiation approaches on the market for 10 years with variable uptake. There was
despite lack of randomized clinical trial data for these de- a large international randomized trial comparing single
vices. The frequency of brachytherapy use increased from intraoperative radiation using low-energy photons delivered
approximately 1% in 2001 to 10% in 2006, despite concerns by the TARGIT device. The results of the TARGIT A trial were
about long-term efficacy.8 In fact, over 30,000 women had first published in 2010, followed by a second publication in
been treated with accelerated partial breast irradiation 2014.14 The results showed noninferiority, specifically for
(APBI) by the time the American Society for Radiation On- the prepathology stratum (TARGIT given at time of lump-
cology published its first consensus statement.9 ectomy). There was a great deal of criticism and arguments
The finding from a randomized trial (CALGB 9343) that that it was too early to adopt the findings,15 with bias from
older women with hormone receptorpositive breast cancer many radiation oncologists not expecting that a treatment
could be effectively treated with tamoxifen without radia- with low-energy photons would work. Indeed, early adop-
tion therapy has yet to be adopted into clinical practice.10 tion of technology that turns out to be inferior to the status
The results demonstrated that with or without radiation, quo can be harmful, but late adoption of technology that
distant recurrence, breast cancer mortality, and mastectomy turns out to be equivalent or superior to the status quo can
rates are the same and very low in the two arms.11 Despite be a missed opportunity that also harms individuals and
these results with more than 10 years of follow-up and society. Using a framework to evaluate the risks and benefits
corroborating evidence from a similar Canadian trial in all of early versus late adoption, this technology is one where
postmenopausal women,12 as well as the PRIME 2 trial,13 early adoption would be preferable to EBRT for all eligible
radiation is rarely omitted and postlumpectomy radiation is cases.16
considered a quality measure by the American College of Breast cancer is now recognized as being comprised of
Surgeons for older women. Fear of omitting therapy and several distinct diseases. One of the reasons that the
being less aggressive often makes both physicians and pa- locoregional recurrence appears to be so much lower is likely
tients uncomfortable, even in the face of supporting evi- due to the impact of mammography screening and the
dence to the contrary. The cultural bias that more aggressive identification of tumors that are biologically more
treatment of cancer is better, fear of malpractice, and fi- indolent.17-19 The results from older trials in which there
nancial rewards all conspire to encourage intervention. was a 40% locoregional recurrence in the absence of radi-
ation applied to younger women with tumors with more
aggressive biology.20 Thus, reframing risk and options must
occur as we have the ability to better characterize the bi-
ology of newly diagnosed breast cancers21 and offer better
KEY POINTS options for women with low-risk disease. This is even more
Surgery for breast cancer has dramatically changed in
important where local recurrence is not life threatening.
recent years, moving away from extensive surgical The risk for distant breast cancer recurrence for women
resections for all patients to more individualized, limited with hormone receptorpositive disease does extend over a
surgery for early stages of disease. 20-year period.22 This well-known fact leads to an erroneous
Treatment decisions for patients with early-stage conclusion that long-term follow-up for 10 or more years is
disease are complex because decision making typically needed to assess outcomes. However, the peak hazard rate
involves multiple options that likely have little effect on for local recurrence is early, and then is low and stable, thus,
overall survival. the overall conclusions are extremely unlikely to change.
The widespread use of screening has resulted in This has been demonstrated by the overview analyses as
identification of a higher fraction of low-risk cancers. well as many trials,23,24 the Canadian trial, the CALGB trial,
Women undergoing breast-conserving therapy have
and the TARGIT A trial, which showed that early results
numerous options that include surgery alone or surgery
and limited fractionated radiation and IORT.
predicted the later results.12,25-27
Molecular classifiers of indolent tumors can guide a less The demonstration that less-aggressive interventions are
aggressive initial strategy and should inform treatment equally effective for women with lower-risk tumors is a
of both invasive disease and help to reclassify some critical breakthrough for women and a major advance in our
types of DCIS. ability to tailor treatments for women according to the
biology of their tumors. If we can safely accomplish the same

e6 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


THE EVOLVING SURGICAL APPROACH TO BREAST CANCER

TABLE 1. Recent Studies of Breast Conservation, Radiation, and Local Therapy Outcomes
Study Accrual Dates No. of Patients Study Arms 5-Year LRR 10-Year LRR 12.6-Year LRR
TARGIT-A14 20002012 2,298 prepathology IORT 2.1% NA NA
EBRT 1.1%
Studies Comparing XRT With No XRT
CALGB C934311 19941999 636 Tam 4% 7% 10%
Tam + RT 1% 1% 2%
Luminal A7 19922000 611 (all T1 patients) Tam 5.5% 13.8% NA
Tam + RT 0.4% 5.3%
114 (subset of G1/2, Tam 2% 4.9% NA
luminal A patients)
Tam + RT 5.5%
PRIME II13 20032009 1,326 No RT 4.1% NA NA
RT 1.3%
Studies Comparing Hypofractionated 3-Week EBRT With 5-Week EBRT
Hypofractionated 19931996 1,234 3-week EBRT 2.33% 6.2% NA
Radiation5
5-week EBRT 2.17% 6.7%
START-B6 19992001 2,215 3-week EBRT 1.9% 5.2% NA
5-week EBRT 3.3% 3.8%
Abbreviations: LRR, locoregional recurrence; NA, not available; IORT, intraoperative radiation therapy; EBRT, external beam radiation therapy; Tam, tamoxifen; RT, radiation therapy.

goal (preventing cancer recurrence) in a much more effi- fraction of tumors with low-risk biology.28 An important way
cient, less invasive, and less personally time-consuming to mitigate that risk is to recognize that lower-risk biology
manner for women, the physician community should be tumors are more likely to be detected with screening and to
the first to embrace this therapy. The complaint about IORT have tools to identify them at the time of diagnosis and
seems to be that the data are immature; however, clinical enable less-aggressive treatment. Using a data set from a
practitioners can switch to IORT and watch the data mature. randomized trial of no treatment compared with treatment
The chance that the results will change has a very small with tamoxifen for 2 years, we sought to leverage a validated
probability, and practitioners could switch back to EBRT or molecular classifier to determine if we could define a group
switch to another treatment that has been shown to be of women whose prognosis was excellent even 20 years after
more effective than EBRT in the interim. As our analysis diagnosis.29 We used a test that was originally designed to
indicates, little will have been lost for any patient who re- identify a population of patients with breast cancer, in the
ceives IORT, as many are also eligible for hormone therapy absence of systemic therapy, with a good prognosis at
alone. 5 years. The 70-gene test (MammaPrint) has been used to
Daily radiation treatments over many weeks are bur- identify women who do not benefit from adjuvant che-
densome to patients, especially those that live far from an motherapy and is currently being evaluated in the MINDACT
adequate radiation treatment center. It is reasonable to clinical trial (6,600 patients; results expected late spring
assume that patients would prefer no radiation, IORT, or 2016). We created a new threshold with the purpose of
shorter courses of EBRT, even simply on the basis of con- specifically evaluating long-term (. 20 years) breast can-
venience. Women concerned only about distant recurrence cerspecific mortality, which we have designated indolent
should be advised about the options of no radiation for threshold.
node-negative disease. If a 5% to 10% difference in local The women in the STO1 study presented with palpable
recurrence is important to avoid, IORT is an excellent al- tumors, so by definition, they had tumors that had come to
ternative to no radiation. clinical attention. These are not cancers detected by
The difficulty in adoption of less-aggressive therapy may screening. When the indolent threshold is applied to the STO
best be approached by using biomarkers that characterize study in which women presented with palpable, clinically
tumors as indolent. One biomarker, specifically adapted for detected tumors, we were able to identify women whose
the express purpose of identifying indolent disease in the tumors, after surgery and a short course (2 to 5 years) of
absence of all treatment, could have the potential to provide tamoxifen, posed no risk for breast cancerspecific death for
reassurance to providers and patients alike that a less- 15 years.29
aggressive initial treatment is appropriate. An indolent tumor should signal that a more minimal
The widespread prevalence of screening has resulted in a treatment approach would be sufficient. Although there
shift in the kinds of cancers detected. The objective of may be a small fraction of patients who progress, that
screening is to identify cancers at a lower stage, however, a progression is many years later, and early treatments are
consequence of screening is the identification of a higher unlikely to impact that progression. The critical distinction is

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e7


ESSERMAN, GALLANT, AND ALVARADO

FIGURE 1. Indolent Threshold of the 70-Gene Assay The indolent threshold could therefore be used to identify
women for whom lumpectomy alone and a short course
of adjuvant endocrine therapy is sufficient. Many women
are unable to tolerate 5 years of endocrine therapy, and as
many as 60% have discontinued aromatase inhibitors by
3 years.30,31 The ability to identify a population of women
with little benefit of extended endocrine risk-reducing
therapy would be of huge benefit. The indolent threshold
does not allow us to say that women would never experience
recurrence over their lifetime (we have not found the
classification for truly IDLE [indolent lesions of epithelial
origin] conditions), but we can say that the chance of any
recurrence is low and that more aggressive therapy can
be administered at the time of recurrence, sparing many
women treatments that will not be of benefit.
It is estimated that over a 10-year period, five to seven
cancers per 1,000 might be diagnosed that might not oth-
An indolent threshold of the 70-gene assay has been validated in a randomized erwise come to clinical attention.32 These represent ap-
trial of postmenopausal women with node-negative palpable tumors.29 These proximately 17% of the patients diagnosed with breast
tumors are essentially curable at the time of presentation. Thus, there would be no
value to identifying the precursors of such tumors. Such precursors would therefore cancer. Interestingly, approximately 15% of the STO1 trial
fit the definition of IDLE conditions18 and should not be called cancer nor be the target population had tumors that met the indolent threshold. In
of screening. The most likely candidates for these precursors are low-grade ductal
carcinoma in situ lesions. Studies are ongoing to investigate the relationship of the MINDACT study, a modern cohort of patients in which
low-grade ductal carcinoma in situ and indolent invasive breast cancer. over 80% of women are screened, the fraction of women
Abbreviations: IDLE, indolent lesions of epithelial origin; DCIS, ductal carcinoma in
situ; Dx, diagnosis; TAM, tamoxifen. with screen-detected tumors with indolent cancers may be
in the range of 37%, or about 20% more than what was
that additional treatment should be reserved for when and observed in the STO trial, which was conducted prior to the
if that disease progresses, and additional treatment should widespread use of screening. This is consistent with the
be administered at the time of progression (Fig. 1). additional low-risk cancers diagnosed with screening. Over a
We can reflect on the three randomized studies that 10-year period in the United States, where 50 million women
demonstrate that women with relatively low-risk biology are screened for 10 years, this might amount to 300,000
have a low risk of recurrence in the setting of hormone cancers with extremely low-risk of recurrence. Character-
therapy alone and without radiation.11-13 The risk of local ization of all screen-detected cancers could provide critical
recurrence, as stated earlier, has dropped over the years information about how to further personalize treatment.
likely because of the dramatic increase in the number of Even in the setting of presentation of palpable disease,
women diagnosed who have indolent cancers.16 In the about 10% of tumors fit the molecular definition of indolent
CALGB 9343 trial of women treated with lumpectomy alone
and 5 years of tamoxifen, local recurrence was uncommon
and successfully treated at the time of diagnosis without an FIGURE 2. Identification and Therapy of the Indolent
impact on mortality or the mastectomy rate. Recurrence Spectrum of Breast Cancers
rates in the absence of radiation are low,11-13 and, in par-
ticular, the event of recurrence appears to be salvageable
without an adverse impact on mastectomy rates or mor-
tality. In particular, the Canadian trial of radiation compared
with no radiation demonstrates that luminal A tumors have
only a 5% risk of local therapy with 5 years of tamoxifen
therapy and absence of radiation. The indolent threshold
described here is more stringent than the luminal A cate-
gory, with only 31% of luminal A tumors meeting the in-
dolent designation. Importantly, in all of the radiation versus
no radiation studies, the chance of dying of other causes is
considerably higher than the chance of dying of breast
cancer. The analysis of the Stockholm 1 study confirms the
existence of a tumor type that has an indolent course, which
can be treated less aggressively at the time of diagnosis. In
this population of women, the indolent threshold did not
Identification of the indolent spectrum of breast cancers and tailoring therapy
identify women who had zero risk absent therapy, but al- accordingly (local and systemic) should be considered part of the evolution
most no risk with a minimal course of tamoxifen. toward personalized breast cancer treatment.

e8 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


THE EVOLVING SURGICAL APPROACH TO BREAST CANCER

disease. In the setting of screening, that number is more Such precursors would better be reclassified as IDLE, instead
likely to be in the range of 35% or higher33 in keeping with of carcinoma in situ.18 If the indolent invasive tumors can be
observations from screening trials.34 If the biology of these treated without radiation, then certainly women with risk
tumors can be recognized at the time of diagnosis, and factors or precursors of IDLE conditions such as low and
women can be reassured that their prognosis is excellent, intermediate DCIS should not be routinely offered radia-
simpler interventions can be used such as excision alone, tion.35 Evidence to support this concept was recently
sentinel node, and hormone therapy for 2 to 5 years. published by Sagara et al.36 The investigators looked at
Registry studies for using less intervention for post- surgery or no surgery for DCIS in a large SEER database of
menopausal women with lower-risk tumors are under 57,000 patients. For low-grade DCIS, the 10-year breast
development. The University of Michigan is leading a multi- cancerspecific survival for no surgery was 98.8% whereas
institutional study of surgical excision alone for women age the survival for the surgery group was 98.6%. Surely, we can
50 to 69 with T1NO tumors with Oncotype Dx results lower start advocating for less treatment for these women.
than 18. LUMINA is a Canadian Ontario Cancer Group study Learning how to safely do less should be considered part of
of lumpectomy alone for women older than age 55 with the evolution of personalized breast cancer treatment
T1NO tumors that are luminal A. There are several ductal (Fig. 2).
carcinoma in situ (DCIS) trials starting, two in Europe, in- All treatments have side effects. Mastectomy, even with
cluding the LORD trial and the LORIS trial, and three in the reconstruction, can be disfiguring and debilitating to
United States (a registry trial through the ATHENA network, women. They can experience postmastectomy pain. Radi-
the COMET trial recently funded by PCORI, and the ECOG- ation also has side effects.37,38 Extended endocrine therapy
ACRIN MRI and Oncotype prospective study) exploring op- can also be challenging to take for women, and it has been
tions of using less intervention. reported that over 40% of women stop taking their medicine
Overdiagnosis is much less of a problem if we accept and after 3 years.30,31 The STO data suggest that 2 years of ta-
recognize that we will identify indolent disease with moxifen should be sufficient for women with tumors that
screening and if we have tools to characterize it and be less meet the molecular definition of indolent disease. Although
aggressive with our interventions. To do so, we have to have it would be difficult to test, the persistence of a small risk for
confidence in the tools that we use to identify such indolent recurrence after 15 years over so many years suggests that
conditions. perhaps 2 years of hormonal therapy could be repeated 7 to
These data should also make us reflect on the dilemma of 10 years after diagnosis if we are able to identify those
DCIS. If there are indolent cancers that can be successfully women with late risk for recurrence. The opportunity to
treated upon diagnosis, then certainly there would not be know when such treatments are not necessary would be a
any value to finding the precursors of these indolent cancers. great boon to patients.

References
1. Wang F, Peled AW, Garwood E, et al. Total skin-sparing mastectomy and 7. Fyles A, McCready D, Pintilie M. Luminal A subtype predicts radiation
immediate breast reconstruction: an evolution of technique and as- response in patients with T1N0 breast cancer enrolled in a randomized
sessment of outcomes. Ann Surg Oncol. 2014;21:3223-3230. trial of tamoxifen with or without breast radiation. Cancer Res. 2011;71:
2. Peled AW, Wang F, Foster RD, et al. Expanding the indications for total S2-2.
skin-sparing mastectomy: is it safe for patients with locally advanced 8. Smith BD, Arthur DW, Buchholz TA, et al. Accelerated partial breast
disease? Ann Surg Oncol. 2016;23:87-91. irradiation consensus statement from the American Society for Ra-
3. Boughey JC, Suman VJ, Mittendorf EA, et al; Alliance for Clinical Trials in diation Oncology (ASTRO). Int J Radiat Oncol Biol Phys. 2009;74:
Oncology. Sentinel lymph node surgery after neoadjuvant chemo- 987-1001.
therapy in patients with node-positive breast cancer: the ACOSOG 9. Smith BD, Arthur DW, Buchholz TA, et al. Accelerated partial breast
Z1071 (Alliance) clinical trial. JAMA. 2013;310:1455-1461. irradiation consensus statement from the American Society for Radi-
4. Caudle AS, Yang WT, Krishnamurthy S, et al. Improved axillary ation Oncology (ASTRO). J Am Coll Surg. 2009;209:269-277.
evaluation following neoadjuvant therapy for patients with node- 10. Soulos PR, Yu JB, Roberts KB, et al. Assessing the impact of a cooperative
positive breast cancer using selective evaluation of clipped nodes: group trial on breast cancer care in the medicare population. J Clin
implementation of targeted axillary dissection. J Clin Oncol. Epub Oncol. 2012;30:1601-1607.
2016 Jan 25. 11. Hughes KS, Schnaper LA, Bellon JR, et al. Lumpectomy plus tamoxifen
5. Whelan TJ, Pignol JP, Levine MN, et al. Long-term results of hypo- with or without irradiation in women age 70 years or older with early
fractionated radiation therapy for breast cancer. N Engl J Med. 2010; breast cancer: long-term follow-up of CALGB 9343. J Clin Oncol. 2013;
362:513-520. 31:2382-2387.
6. Haviland JS, Owen JR, Dewar JA, et al; START Trialists Group. The UK 12. Fyles AW, McCready DR, Manchul LA, et al. Tamoxifen with or without
Standardisation of Breast Radiotherapy (START) trials of radiotherapy breast irradiation in women 50 years of age or older with early breast
hypofractionation for treatment of early breast cancer: 10-year follow- cancer. N Engl J Med. 2004;351:963-970.
up results of two randomised controlled trials. Lancet Oncol. 2013;14: 13. Kunkler IH, Williams LJ, Jack WJ, et al; PRIME II investigators. Breast-
1086-1094. conserving surgery with or without irradiation in women aged 65 years

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e9


ESSERMAN, GALLANT, AND ALVARADO

or older with early breast cancer (PRIME II): a randomised controlled 26. Vaidya JS, Wenz F, Bulsara M, et al. Targeted intraoperative radio-
trial. Lancet Oncol. 2015;16:266-273. therapy for early breast cancer: TARGIT-A trial- updated analysis of local
14. Vaidya J, Wenz F, Bulsara M, et al; TARGIT trialists group. Risk- recurrence and first analysis of survival. Cancer Res. 2012;72:S4-2.
adapted targeted intraoperative radiotherapy versus whole-breast 27. Hughes KS, Schnaper LA, Berry D, et al; Cancer and Leukemia Group B;
radiotherapy for breast cancer: 5-year results for local control and Radiation Therapy Oncology Group; Eastern Cooperative Oncology
overall survival from the TARGIT-A randomised trial. Lancet. 2014;383: Group. Lumpectomy plus tamoxifen with or without irradiation in
603-613. women 70 years of age or older with early breast cancer. N Engl J Med.
15. Woloshin S, Schwartz LM. Whats the rush? The dissemination and adoption 2004;351:971-977.
of preliminary research results. J Natl Cancer Inst. 2006;98:372-373. 28. Esserman LJ, Thompson IM, Reid B, et al. Addressing overdiagnosis and
16. Esserman LJ, Alvarado MD, Howe RJ, et al. Application of a decision overtreatment in cancer: a prescription for change. Lancet Oncol. 2014;
analytic framework for adoption of clinical trial results: are the data 15:e234-e242.
regarding TARGIT-A IORT ready for prime time? Breast Cancer Res Treat. 29. Esserman LJ, Thompson CK, Yau C, et al. Identification of tumors with an
2014;144:371-378. indolent disease course: MammaPrint ultralow signature validation in a
17. Esserman LJ, Shieh Y, Rutgers EJ, et al. Impact of mammographic retrospective analysis of a Swedish randomized tamoxifen trial. Cancer
screening on the detection of good and poor prognosis breast cancers. Res. 2016;76:P6-09-01.
Breast Cancer Res Treat. 2011;130:725-734. 30. Cuzick JF, Sestak I, Howell A, et al. Anastrozole versus tamoxifen for the
18. Esserman L, Shieh Y, Thompson I. Rethinking screening for breast cancer prevention of loco-regional and contralateral breast cancer in post-
and prostate cancer. JAMA. 2009;302:1685-1692. menopausal women with locally excised ductal carcinoma in-situ (IBIS-II
19. Bleyer A, Welch HG. Effect of three decades of screening mammography DCIS). Lancet. 2016;387:866-873.
on breast-cancer incidence. N Engl J Med. 2012;367:1998-2005. 31. Ganz PA, Cecchini RS, Julian TB, et al. Patient-reported outcome (PRO)
20. Fisher B, Montague E, Redmond C, et al. Findings from NSABP Protocol results, NRG Oncology/NSABP B-35: a clinical trial of anastrozole (A) vs
No. B-04-comparison of radical mastectomy with alternative treat- tamoxifen (tam) in postmenopausal patients with DCIS undergoing
ments for primary breast cancer. I. Radiation compliance and its relation lumpectomy plus radiotherapy. Cancer Res. 2016;76:S6-04.
to treatment outcome. Cancer. 1980;46:1-13. 32. Marmot MG, Altman DG, Cameron DA, et al. The benefits and harms of
21. Buyse M, Loi S, vant Veer L, et al; TRANSBIG Consortium. Validation and breast cancer screening: an independent review. Br J Cancer. 2013;108:
clinical utility of a 70-gene prognostic signature for women with node- 2205-2240.
negative breast cancer. J Natl Cancer Inst. 2006;98:1183-1192. 33. Drukker CA, Schmidt MK, Rutgers EJ, et al. Mammographic screening
22. Esserman LJ, Moore DH, Tsing PJ, et al. Biologic markers determine both detects low-risk tumor biology breast cancers. Breast Cancer Res Treat.
the risk and the timing of recurrence in breast cancer. Breast Cancer Res 2014;144:103-111.
Treat. 2011;129:607-616. 34. Miller AB, Wall C, Baines CJ, et al. Twenty five year follow-up for breast
23. Early Breast Cancer Trialists Collaborative Group (EBCTCG). Effects of cancer incidence and mortality of the Canadian National Breast
chemotherapy and hormonal therapy for early breast cancer on re- Screening Study: randomised screening trial. BMJ. 2014;348:g366.
currence and 15-year survival: an overview of the randomised trials. 35. Narod SA, Iqbal J, Giannakeas V, et al. Breast cancer mortality after a
Lancet. 2005;365:1687-1717. diagnosis of ductal carcinoma in situ. JAMA Oncol. 2015;1:888-896.
24. Clarke M, Collins R, Darby S, et al; Early Breast Cancer Trialists Col- 36. Sagara Y, Mallory MA, Wong S, et al. Survival benefit of breast surgery
laborative Group (EBCTCG). Effects of radiotherapy and of differences in for low-grade ductal carcinoma in situ: a population-based cohort
the extent of surgery for early breast cancer on local recurrence and study. JAMA Surg. 2015;150:739-745.
15-year survival: an overview of the randomised trials. Lancet. 2005; 37. Darby SC, Ewertz M, McGale P, et al. Risk of ischemic heart disease in
366:2087-2106. women after radiotherapy for breast cancer. N Engl J Med. 2013;368:
25. Liu F, Shi W, Done SJ, et al. Identification of a low-risk luminal A breast 987-998.
cancer cohort that may not benefit from breast radiotherapy. J Clin 38. Esserman L, Yau C. Rethinking the standard for ductal carcinoma in situ
Oncol. 2015;33:2035-2040. treatment. JAMA Oncol. 2015;1:881-883.

e10 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


STRATEGIES FOR SUSTAINABLE CANCER CARE

Strategies for Sustainable Cancer Care


David J. Kerr, MD, Anant Jani, PhD, and Sir Muir Gray, CBE, FRCPSGlas, FCLIP

Editors Note: The following article is based on the 2016 ASCO Annual Meeting Education Session NICE or Not? Evaluating
Global Strategies for Sustainable Cancer Care. The authors review strategies for sustainable cancer care and the National
Institute for Health and Care Excellences role in the assessment of clinical and cost-effectiveness of new pharmaceutical and
biopharmaceutical products in the United Kingdom.

OVERVIEW

There is an increasing focus on the relative cost-effectiveness and sustainability of delivering high-quality cancer care,1 with
most emphasis, debatably, given to cost control of innovative treatments. It is difficult to calculate all the direct and indirect
contributors to the total cost of cancer treatment, but it is estimated that cancer drugs constitute 10% to 30% of the total cost
of cancer care. A 2007 study in France2 showed the contribution of drug costs was less than 20%, with approximately 70% of
the total expenditure on cancer accounted for by health care resource use, such as hospitalization. The U.K. government
established the National Institute for Health and Care Excellence (NICE)the dominant function of which is technology
appraisalto assess the clinical and cost-effectiveness of new pharmaceutical and biopharmaceutical products. This is to
ensure that all National Health Service (NHS) patients have equitable access to the most clinically effective and cost-effective
treatments that are viable. NICE has developed a transparent, public process to judge incremental cost-effectiveness using
the quality-adjusted life year (QALY), which allows comparisons of cost-effectiveness across medical specialties. NICE has
been both lauded and criticizedespecially when it passes judgment on marginally effective but expensive anticancer
drugsbut it provides a route to rational rationing and, therefore, may contribute to sustainable cancer care by
highlighting the issue of affordable medicine. This implies a challenge to the wider oncology community as to how we might
cooperate to introduce the concept of value-driven cancer care.

M ichael Porter, a leading professor at the Harvard


Business Institute, has developed the following defi-
nition of value:
and the resources used. In turn, the outcome is determined
by the quality and safety of the intervention or service.

Value in any field must be defined around the customer, not STRATEGIES FOR SUSTAINABLE CANCER CARE:
the supplier. Value must also be measured by outputs, not VALUE
inputs. Hence it is patient health results that matter, not the Value Versus Quality
volume of services delivered. But results are achieved at some Services become high quality for several reasons, many of
cost. Therefore, the proper objective is patient health which are factors that have been shown to reduce the in-
outcomes relative to the total cost (inputs). Efficiency, then, cidence of errors, notably: (1) good leadership, (2) devel-
is subsumed in the concept of value.3(p. 12) opment of systems, and (3) the creation of a strong culture.
To maximize the probability of benefit and minimize the
Porter and Teisberg put this another way: Achieving high
probability of harm and errors, professionals need standard
value for patients must become the overarching goal of
operating procedures (SOPs), commonly defined as a pro-
health care delivery, with value defined as the health out- cedure, or set of procedures, to perform a given operation
comes achieved per dollar spent.4 Value, therefore, de- or evolution in reaction to a specific event. To improve
pends on results, not input, and so value in health care is quality and safety, it is necessary to have a style of gen-
measured by the outcomes achieved, not the volumes de- eral management that will increase effectiveness and re-
livered, and the relative costs of achieving those outcomes.5 duce harmfor example, promotion of safety culture; use
Put simply, value is the relationship between the outcome of systems, including clinical guidelines; clear lines of

From the Nuffield Division of Clinical and Laboratory Science, Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, United Kingdom.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: David J. Kerr, MD, Nuffield Division of Clinical and Laboratory Sciences, Level 4, Academic Block, John Radcliffe Infirmary, Headington, Oxford, OX3 9DU
United Kingdom; email: david.kerr@ndcls.ox.ac.uk.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e11


KERR, JANI, AND GRAY

accountability for risk management and safety; and data The second problem is underuse of effective and cost-
collection about risk factors, errors, and near misses. effective interventions for cancer, principally because of poor
Certainly there is concern about low-value, low-quality organization in the management of cancer services. One other
cancer care, and the Institute of Medicine report, Crossing aspect of underuse that should be considered is inequity, where
the Quality Chasm, on improving the quality of health care certain subgroups of the populationfor example, distin-
has been welcome, but high-quality health care need not be guished by age, ethnic background, or classreceive lower
synonymous with high-value health care. Low-quality health levels of high-value treatment than the general population.
care is certainly of lower value, but there are many examples Therefore, a focus on value is needed. Three aspects of
of health care providing high quality that is of low value. The value are:
semantics of this are important because consumers often
associate value (e.g., in supermarkets) at the lower end of
allocative value, determined by how the assets are
distributed to different subgroups in the population;
the market (e.g., Attention Kmart shoppers, as Beetlejuice
once said!).
technical value, determined by how well resources are
used for all the people in need in the population; and
The assessment of cost-effectiveness by organizations
such as NICE is of vital importance, but cost-effectiveness
personalized value, determined by how well the de-
cisions relate to the values of each individual.
refers to an intervention, whereas value refers to a pop-
ulations services. Perhaps populations will have to make
decisions between different degrees of cost-effectiveness, Triple-Value Health Care
worrying not about the financial cost of one particular in- Practitioners providing cancer care should continue to cam-
tervention but about its opportunity costnamely what else paign for more resources for cancer, but they should also be
can be done with resources within a finite health budget. If prepared to defend the allocative value of everything they are
there is an investment in cancer drugs, there may have to be currently doing. There are ways in which reallocation can be
disinvestment elsewhere within the cancer program or the facilitated. For example, in Spain, there was a bid for immu-
wider health service. notherapy resources for malignant melanoma, the total cost
being about V2 million for the population served. The payers
said they were unable to find this resource, but actually, in that
Choosing Wisely same population, the annual expenditure on skin disease was
There is increasing recognition of unwarranted variation not approximately V170 million per year. Practitioners in cancer
only in investment in cancer care but also in quality, cost, and care should learn to argue that consideration should be given
outcome. This variation highlights two other problems that to shifting V2 million from lower-value care for other skin
should be addressed not only by policy makers but also by diseases to malignant melanoma.
every clinician providing cancer services. The main challenge for people providing cancer services is
The first of these is overuse, highlighted in the Choosing to consider the allocation of resources between different
Wisely Campaign in the United States and in the Too Much cancer groups or between different modalities. The balance
Medicine campaign of the British Medical Journal. Overuse of investment in different patient groups is often a matter of
always wastes resources and sometimes does harm (e.g., historical accident, with one particular clinical group having
prescribing chemotherapy to patients when palliative care been more successful than others, but this should be
would be more appropriate, where the concern is not to save reviewed to see if the balance is optimal between the dif-
costs, but that the prescription of chemotherapy can be ferent types of common cancer, not excluding rare cancers.
classified as inappropriate or futile). As the path becomes more difficult, providers of cancer care
and patient representatives should consider whether the
balance is right; for example, should the interval between
KEY POINTS breast cancer screening be increased to free up resources
that can be switched to chemotherapy for women with
There is a need to balance cost-effectiveness and breast cancer?
sustainability of delivering high-quality cancer care. Technical value is different from efficiency. Technical value
It is estimated that cancer drugs constitute 10% to 30% has to take into account not only the use of resources for
of the total cost of cancer care. the patients seen, but also the possibility of overuse and
The U.K. government established NICE to assess the underuse. Every cancer service should conduct a review of
clinical effectiveness and cost-effectiveness of new inappropriate or futile care, which relates to the third type of
pharmaceutical and biopharmaceutical products.
value, personalized value
NICE has been praised as well as criticized, but it
contributes to sustainable cancer care by highlighting
In making decisions about personalized value, the model
the issue of affordable medicine. of decision making at the Centre of Evidence-Based Medi-
The worldwide oncology community must cooperate to cine is useful (Fig. 1). Cancer services should ensure that
introduce the concept of value-driven cancer care for every patient is fully informed about not only the benefits of
patients. treatment, but also the risks, and help them reflect on how
these odds reflect on values.

e12 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


STRATEGIES FOR SUSTAINABLE CANCER CARE

FIGURE 1. Center of Evidence-Based Medicines of course plays to the current model of precision med-
Personalized Value Model of Decision Making icine, selecting the right drug for the right patient.
Payers also should be explicit about health care condi-
tions that will or will not be given treatment from
publicly/insurance-funded health services.
In addition, services must make explicit decisions and
publish the decisions to not provide interventions they
deem to be of low value, such as expensive but mar-
ginally effective cancer therapeutics.
Management of demand may not be possible if left
solely to individual clinicians faced with the distress of
the individual patient. Funders should make decisions
at a population level. This is the implicit tension be-
tween the practice of population and clinical medicine
should practicing clinicians always stand outside of
decisions on drug rationing to maintain the integrity of
Managing Clinical Demand their relationship with the patients entrusted to their
There is a fashion in operations, as there is in sleeves and care?
skirts: the triumph of some surgeon who has at last found out
how to make a once desperate operation fairly safe is usually
followed by a rage for that operation not only among the National Institute for Health and Care Excellence
doctors, but actually among their patients. Since 1999, NICE has provided the United Kingdoms NHS
and those who rely on it for their care, with an increasing
George Bernard Shaw, preface to The Doctors Dilemma, 1906 range of advice on effective, good-value health care, and
Enthusiastic medical professionals, altruistic as we are and it has gained a reputation for rigor, independence, and
keen to do good, are armed with information provided by objectivity (Fig. 3). This has been described by some as
enthusiastic promoters of new technology, expressed in terms rational rationing. Available guidance takes several forms:
of relative benefit rather than absolute benefit, and are, not
surprisingly, the people who may fuel the growth in demand.
NICE guidelines make evidence-based recommenda-
tions that aim to promote integrated care.
Clinical demand may be overtly expressedfor example by
clinicians leading a campaign for a new drug or a new facility
Technology appraisal guidance to assess the clinical and
cost-effectiveness of health technologies, such as new
such as a PET scanneror demand can arise through what pharmaceutical and biopharmaceutical products, but
is called creep (i.e., an inexorable increase in testing and
treatment). Innovation should decrease cost as well as increase
it, and there is now a drive to innovate for better value, FIGURE 2. Causes of Cost Inflation
harnessing the power of new technology and improved ways
of organizing services. A classic study by Eddy6 in the United
States showed that, in a health care system in which ex-
penditure is not finite, changes in the volume and intensity
of clinical practice are the main factors driving increases
in the cost of care that can be controlled by those who
pay for or manage health care. The other causes of in-
creasing costsaging population, medical and general price
inflationare beyond the power of health service managers
to control (Fig. 2).
Managing innovation usually focuses on a small number of
potentially high-cost interventions; however, the conclusion
from the work of Eddy,6 which is as true today as when it was
published, is that every innovation, no matter how small, should
have its introduction to, or removal from, the health care system
carefully managed, particularly if it is for a common disease.
Managing demand is not easy, but some steps can be
taken; for example:
Be very clear and explicit about the subgroups of cancer
patients most likely to benefit from an intervention and,
as a corollary, those who are least likely to benefit. This

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e13


KERR, JANI, AND GRAY

FIGURE 3. Kerr Led a Review of Scotlands Health care. These are derived from the best available evi-
ServiceThe Resemblance Is Evident! dence, particularly NICEs own guidance.

Technology Appraisals
NICE recommendations review clinical and economic evi-
dence, balancing the clinical data in relation to how much it
costs the NHSdoes it represent value for money? This advice
ends the uncertainty and helps to standardize access to health
care across the country. This is reflected in the NHS Consti-
tution, which states that patients have the right to drugs and
treatments that have been recommended by NICE for use in
the NHS, if their doctor believes they are clinically appropriate.
Sir Andrew Dillon, Chief Executive of NICE, said:
also include procedures, devices, and diagnostic agents. We support the general principle that the NHS should pay a
This is to ensure that all NHS patients have equitable price which reflects the additional therapeutic benefit of new
drugs. We also share the Governments ambition to ensure
access to the most clinically effective and cost-effective
that the option exists for all new licensed drugs to be offered
treatments that are viable. to those patients who can benefit from them, provided the
Quality standards are concise sets of statements with
accompanying metrics designed to drive and measure
price is a fair reflection of their value. We are confident that
the Government will want to take advantage of NICEs
priority quality improvements within a particular area of expertise and experience as it develops value-based pricing.

FIGURE 4. Estimating Cost and Cost-effectiveness

e14 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


STRATEGIES FOR SUSTAINABLE CANCER CARE

The dominant drivers of NICEs approach are incremental


cost-effectiveness and the QALY, which allows comparisons
The use of information technology to seamlessly in-
tegrate clinical and translational research and patient
of cost-effectiveness across all of medicine. How else would care.
we compare the relative benefits of cataract or hip surgery
with cancer therapy? The following hypothetical example
Greater use of adaptive (Bayesian) design techniques in
the design and analysis of randomized controlled trials
gives a clear idea of how the arithmetic is worked out. could reduce trial duration and the number of patients
As is described in Fig. 4, the model is dependent on inputs. needed.
If drug A had an incremental QALY of only 0.2 with an in-
cremental cost of 6,500, then the incremental cost- Oncologists and patient advocacy organizations should
challenge regulatory authority data requirements.
effectiveness would be 32,500 and possibly outside the
value range. Rather than criticize organizations such as NICE for de-
clining reimbursement on grounds of cost-effectiveness,
clinicians and patient advocates should start challenging
Operationalizing Strategies for Sustainable Cancer pharmaceutical companies about the high prices they
Care: Affordable Drugs seek for products with modest benefits.
Almost every time that NICE makes a negative pro-
nouncement on a new cancer drug, there is a volubly
Address the difficulties facing low- and middle-income
countries in accessing affordable cancer care, rather
negative press reaction, often driven, understandably, by than only focusing on problems facing high-income
disease-oriented interest groups. The NICE leadership has countries.
pushed back on this somewhat and has set a series of
challenges for the wider oncology community,7 namely: (1) CONCLUSION
the industry must operate in a much more efficient manner, Research and technologic innovation continue to produce
(2) the costs of drug development should be slashed, and (3) novel therapies, but these are often of marginal clinical
oncologists and patient advocacy groups must ask tough value, while managing to be very costly. Similarly, there is
questions of regulators and the pharmaceutical industry. increasing dependence on expensive new imaging modali-
They also made suggestions for practical implementation ties to assess tumor burden throughout treatment, despite
of effective solutions: the seeming paradox that patients in the United Kingdom, at
They suggest that clinical trial costs could be decreased
by 40%60% without detriment to their quality using
least, present with later-stage disease. There is no doubt
that a programmatic approach to budgeting the totality of
measures such as electronic data capture, reduction in cancer care for a specific population will highlight areas that
the length of case management forms, and modified represent low value and that, therefore, could be targets for
site management practices. disinvestment to make way for more effective treatment.

References

1. Sullivan R, Peppercorn J, Sikora K, et al. Delivering affordable cancer care 4. Porter ME, Teisberg EO. Redefining Health Care. Creating value-based
in high-income countries. Lancet Oncol. 2011;12:933-980. competition on results. Boston, MA: Harvard Business School Press;
2. Amalric F. Analyse e conomique des Couts de Cancer en France. Impact Sur 2006.
la Qualite de Vie, Prevention, Depistage, Soin, Recherche. Paris: Institut 5. Porter ME. What is value in health care? N Engl J Med. 2010;363:
National Du Cancer; 2007. 2477-2481.
3. Porter ME. Defining and introducing value in health care. In Evidence- 6. Eddy DM. Clinical decision making: from theory to practice. Three battles
based Medicine and the Changing Nature of Health Care: 2007 Institute to watch in the 1990s. JAMA. 1993;270:520-526.
of Medicine (IOM) Annual Meeting Summary. Washington, DC: Institute 7. Rawlins MD, Chalkidou K. The opportunity cost of cancer care: a state-
of Medicine; 2008. ment from NICE. Lancet Oncol. 2011;12:931-932.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e15


ULKA VAISHAMPAYAN

The Role of Nephrectomy for Kidney Cancer in the Era of


Targeted and Immune Therapies
Ulka N. Vaishampayan, MD

Editors Note: The following article is based on the 2016 ASCO Annual Meeting Education Session Cytoreductive Nephrectomy in
Renal Cell Carcinoma: A Debate. The author reviews the pros and cons of cytoreductive nephrectomy and whether recent
advances in systemic therapy warrant physicians to proceed directly to systemic therapy as in other metastatic solid tumors,
without the integral step of cytoreductive nephrectomy.

OVERVIEW

Although two phase III trials support the recommendation of nephrectomy followed by interferon alpha in metastatic renal
cell carcinoma (RCC), this procedure cannot be applied to every patient with this condition. Systemic therapy has changed
from interferon alpha to antiangiogenic-targeted therapy, and the clinical impact of nephrectomy in the era of targeted
therapy has not been proven. The SEER database shows that only 35% of patients with advanced RCC undergo nephrectomy
as their initial treatment. Retrospective studies showed improved overall survival (OS) outcomes with nephrectomy and
interleukin-2 (IL-2) therapy; however, the inherent selection bias of younger and healthier patients receiving IL-2 likely
accounts for this finding. Neoadjuvant therapy has demonstrated only modest efficacy in unresectable disease, and if
remission is obtained with systemic therapy, it is unclear whether nephrectomy has any incremental benefit. In the absence
of proven benefit of nephrectomy in the setting of targeted therapy, it seems advisable for patients with RCC with severely
symptomatic disease, competing comorbidities, poor performance status, or unresectable disease to avoid nephrectomy
and proceed directly to systemic therapy. The clinical implications of deferred cytoreductive nephrectomy for patients with
metastatic RCC are poorly understood, and patient cohorts that do not undergo this procedure are likely to be comprised of
patients with unfavorable disease characteristics. Unfortunately, the completed trials of targeted therapy were 90%
comprised of patients with prior nephrectomy (the majority of trials incorporate prior nephrectomy as an eligibility re-
quirement) and hence may not reflect the outcomes of the majority of the patients with advanced RCC who have not
undergone nephrectomy. Newer therapies such as nivolumab and cabozantinib have also been evaluated for a population in
which 90% of the patients underwent nephrectomy. Future clinical trials and registry studies must focus on the therapeutic
treatment and overall outcome of patients without nephrectomy and treated with contemporary systemic therapy.

K idney cancer is one of the few malignancies in which


surgical resection of the primary disease, or cytore-
ductive nephrectomy, has held tremendous importance.
without the integral step of cytoreductive nephrectomy.
The pros and cons of this procedure are summarized in
Table 1.
Historically, systemic therapy was underdeveloped and in-
effective, and surgical resection was considered the only
hope of remission. However, an analysis of the Surveillance, NEPHRECTOMY IN THE CONTEXT OF IMMUNE
Epidemiology, and End Results Program (SEER) database THERAPY
revealed that only 539 of the 1,537 (35%) cases diagnosed Renal cell carcinoma is unique in the fact that randomized
with advanced RCC between 2000 and 2013 had nephrec- trials have actually been conducted to establish the role of
tomy as their initial therapy.1 nephrectomy in metastatic disease.24 The results revealed
The major advances in systemic therapy of RCC in recent that patients subjected to nephrectomy followed by in-
years beg the fundamental question whether a large enough terferon alpha in the presence of metastatic disease ex-
impact has been made to consider proceeding directly to perienced OS benefit (Table 2). The reasons for this
systemic therapy as in other metastatic solid tumors, benefit remain unclear. It does not appear that nephrectomy

From the Karmanos Cancer Institute, Wayne State University, Detroit, MI.

Disclosures of potential conflicts of interest provided by the author are available with the online article at asco.org/edbook.

Corresponding author: Ulka N. Vaishampayan, MD, Karmanos Cancer Institute 4100 John R St., 4233 HWRC, Detroit, MI 48201; email: vaishamu@karmanos.org.

2016 by American Society of Clinical Oncology.

e16 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


ROLE OF NEPHRECTOMY IN THE TARGETED THERAPY ERA

TABLE 1. Pros and Cons of Nephrectomy in Metastatic nephrectomy; median OS was 6.9 months in the cytore-
Renal Cell Carcinoma ductive nephrectomy arm and 4.8 months in the interferon-
alone arm.5
Pros of Nephrectomy in Cons of Nephrectomy in There are numerous reports indicating that cytoreductive
Metastatic RCC Metastatic RCC
nephrectomy improves host immune reaction to the
Control of symptoms from Complications of surgery metastases and is likely to decrease the levels of immu-
primary tumor
nosuppressive factors. It also normalized the nuclear factor-
Phase III trials showing OS benefit No benefit proven with targeted
therapy
kappa B and various other immune defects.6,7 Clinically
No change in response to systemic spontaneous regressions have been observed. A peri-
therapy operative (neoadjuvant) checkpoint inhibitor trial has
Resection of resistant clones Delay of systemic therapy been proposed based on the finding that peripheral
Long-term remissions noted with Impaired creatinine clearance, blood PD-1 expression is reduced significantly after cyto-
surgical resection of metastatic increased risk of hypertension reductive nephrectomy.8 Expression of PD-1 in tumor-
RCC
infiltrating lymphocytes was associated with a more
Nephrectomy patients comprise Unlikely to benefit patients with aggressive phenotype of RCC (larger tumors, higher nu-
90% of clinical trial patient comorbidities and impaired
population performance status clear grade, and sarcomatoid differentiation) and increased
Abbreviations: RCC, renal cell carcinoma; OS, overall survival risk of cancer-specific death, as reported in a study led
by the Mayo Clinic (risk ratio of 2.24; p = .004).9 However,
a recent report evaluating tumor PD-L1 expression in
boosted the response rates to interferon therapy, as the 417 cases of clear cell RCC from The Cancer Genome
response rate remained at a dismal 3.3% and 3.6% in the Atlas database reported a lack of association between
cytoreductive nephrectomy and interferon-alone arms, PD-L1 expression and unfavorable tumor characteristics
respectively, regardless of whether the patients were and an improved OS outcome (hazard ratio [HR] 0.59;
randomly selected to undergo nephrectomy or not. Retro- p = .006).10
spective case series have been reported favoring cytore- Unfortunately, modern immunotherapy trials cannot in-
ductive nephrectomy prior to immune therapy, such as IL-2, form the benefits of treatment for patients who have not
because of OS benefit.5 However, the inherent biases of a undergone nephrectomy. For immune checkpoint inhibitors,
retrospective study are prominently prevalent in the IL-2 the recently reported randomized trial of nivolumab versus
population, as this is a stringently selected group of patients everolimus mainly included patients who had undergone
with excellent cardiopulmonary status and no brain me- nephrectomy.11 Similarly for patients on antiangiogenic or
tastases, which predicts a better OS regardless of choice or mTOR inhibitor therapies, approximately 90% of the patients
sequence of therapy. In fact, even the patients with a included had nephrectomy prior to study enrollment.1216
performance status of 1 within the IL-2treated patient co- Studies are ongoing to evaluate the impact of PD-1 inhibition
hort had a minimal benefit in OS favoring cytoreductive on biomarkers of immune response pre- and post-nephrectomy.
Phase II trials such as ADAPTeR (NCT02446860) are assessing
the response and changes in immune-related markers after
the neoadjuvant administration of nivolumab for 8 weeks
KEY POINTS followed by nephrectomy.
Two phase III trials support the recommendation of
nephrectomy in metastatic renal cancer when
compared with interferon therapy alone; however, the
NEPHRECTOMY IN THE CONTEXT OF VEGF
clinical applicability of this finding is not clear in the era INHIBITOR THERAPY
of targeted therapy. The clinical implications of deferred nephrectomy for pa-
A delay in systemic therapy (because of nephrectomy) tients with metastatic renal cancer are not well understood.
can have deleterious effects on the survival of patients As in the case of immunotherapy trials, the percentage of
with metastatic renal cancer. patients who had undergone nephrectomy in a majority of
Patients with poor-risk disease or predicted survival less clinical trials evaluating VEGF inhibitors is greater than 90%.
than 12 months are unlikely to benefit from The report by Choueiri et al,17 which suggested that patients
nephrectomy. The clinical implications of deferred treated with targeted therapy had a Karnofsky performance
nephrectomy for patients with metastatic renal cancer status less than 80% or poor-risk disease as gauged by the
are not well understood.
Memorial Sloan Kettering Cancer Center criteria, experi-
Surveillance, Epidemiology, and End Results Program
database analysis revealed that 65% of patients with
enced no benefit from cytoreductive nephrectomy (p = .08
advanced renal cancer do not undergo nephrectomy as and .06, respectively; Table 2). A separate report used data
their initial treatment. This patient population warrants from the International Metastatic Renal Cell Carcinoma
further study, especially as it is not included in current Database Consortium database, which included 982 patients
clinical trials. treated with nephrectomy and targeted therapy and 676
patients treated with targeted therapy alone.18 Although

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e17


ULKA VAISHAMPAYAN

TABLE 2. Summary of Study Data on Nephrectomy and Systemic Therapy


First Author, Reference Type of Study No. of Patients Results
Flanigan and Yonover2 Phase III 241 Median OS: CN + IFN: 11.1 months; IFN: 8.1 months; PFS 0; p = .05;
Median OS: CN + IFN: 17.4 months; IFN: 11.7 months; PFS 1; p = .08;
Median OS: CN + IFN: 6.9 months; IFN: 4.8 months
Mickisch et al3 Phase III 84 Median OS: CN + IFN: 17 months; IFN: 7 months (HR 0.54; 95% CI, 0.310.94)
4
Flanigan et al Meta-analysis 331 Median OS: CN + IFN: 13.6 months; IFN: 7.8 months (HR 0.69; p = .002)
Pantuck et al5 Retrospective 89 CN + IL-2: median OS, 16.7 months; 5-year OS, 19.6%
Choueiri et al17 Retrospective 314 (CN: 201; Median OS: 19.8 months (CN) vs. 9.4 months (no CN); HR 0.44; p , .01;
targeted-therapy era no CN: 113) multivariate analysis: no benefit; p = .08; poor risk: no benefit; p = .06
Heng et al18 Retrospective 1,658 (CN: 982; Median OS: 20.6 months (CN) vs. 9.5 months (no CN); p , .0001
targeted-therapy era no CN: 676)
Abbreviation: CN, cytoreductive nephrectomy; OS, overall survival; IFN, interferon; PFS, progression-free survival; N/A, not applicable.

patients treated with cytoreductive nephrectomy had sig- those without nephrectomy.5 Neoadjuvant therapy has also
nificantly better OS compared with those treated with shown only modest benefit in advanced RCC.19 In summary,
targeted agents (median OS, 20.6 vs. 9.5 months, re- until the results of randomized trials are available, no
spectively; HR 0.60; 95% CI, 0.520.69), it is important to synergy has been proven to date between targeted
recognize that selection bias is at play; patients with poor- antiangiogenic therapy and cytoreductive nephrectomy
risk disease comprised only 28% of those treated with in RCC.
cytoreductive nephrectomy versus 54% of the group who did
not undergo the procedure. In addition, only 1% of patients
who did not undergo cytoreductive nephrectomy had fa- THE IMPORTANCE OF MULTIDISCIPLINARY
vorable risk characteristics. TEAM INPUT: COLLECTIVE WISDOM AND THE
Several trials are seeking to prospectively evaluate the role FUTURE OF PATIENT-CENTERED CARE AND
of cytoreductive nephrectomy for patients being treated RESEARCH
with targeted therapy. The CARMENA trial studying ne- Typically, patients who are not offered cytoreductive ne-
phrectomy followed by sunitinib versus sunitinib alone phrectomy have poor performance status, major comor-
will likely shed light on the question of whether cyto- bidities, and limited access to care. Racial disparity in rates of
reductive nephrectomy remains relevant in the context nephrectomy has also been shown to have an impact on
of anti-VEGF therapy for metastatic renal cancer. The patient outcomes. In a SEER database analysis of advanced
primary endpoint is OS, and secondary endpoints are RCC, patients of African-American origin were noted to
progression-free survival and operative complication rates. have a worse OS outcome as compared with white patients
The study has completed accrual, and results are awai- with metastatic RCC, and an interaction effect was noted
ted. The SURTIME study is randomly selecting patients with the disparity in rates of nephrectomy.20 However,
with metastatic RCC to receive sunitinib followed by whether this finding reflects other inherent biases noted
nephrectomy versus sunitinib alone. The study will ad- above is unknown. This observation highlights the need for
dress whether patients with adequate response to sys- data collection regarding the factors commonly considered
temic therapy should receive surgical consolidation with toward the decision-making process of nephrectomy.
nephrectomy. In addition, cytoreductive nephrectomy carries a certain
risk that has to be factored into the decision. The major
complication rates of cytoreductive nephrectomy have been
SYNERGY BETWEEN NEPHRECTOMY AND reported to be about 5%, and 11% of the patients did not
SYSTEMIC THERAPY: REALITY OR PERCEPTION? start systemic therapy within 60 days after nephrectomy
Despite a hypothesized synergy between systemic therapy because of surgical complications. Overall, 61% of patients
and nephrectomy, it has not been borne out in clinical did not start systemic therapy in a timely fashion. The
practice. For example, the randomized trials of nephrectomy presence of liver metastases, intraoperative transfusion, and
and interferon versus interferon alone reported identical pathologic nodal involvement were noted to be predictors of
response rates of approximately 3% in both arms. An ad- delay of systemic therapy more than 60 days after cytore-
juvant trial of sunitnib versus sorafenib versus placebo ductive nephrectomy.21
(ASSURE/ECOG 2805) reported lack of benefit for either of It is not advisable to reflexively send the patient for ne-
the agents postnephrectomy.18 The only data that suggest phrectomy upon diagnosis of advanced kidney cancer.
potential synergy come from retrospective trials of patients Multidisciplinary evaluation to carefully weigh the risk and
treated with IL-2 showing an improved outcome achieved benefit ratio factoring in whether the bulk of the patients
for patients who underwent nephrectomy as compared with symptoms are related to the cancer or to other comorbidities

e18 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


ROLE OF NEPHRECTOMY IN THE TARGETED THERAPY ERA

would be important. Renal cancer is a heterogeneous dis- Risk of Surgical Complications


ease,22 and to date, the resection of resistant clones is still Patients with impaired performance status and comorbid-
considered an ideal way to manage the problem. Stage IV ities are likely to have a higher complication rate from
disease within RCC spans a wide spectrum of outcomes, and cytoreductive nephrectomy.
risk prognostication is critically important before nephrec-
tomy is considered.
The reported efficacy of targeted therapy for patients with CASE ILLUSTRATIONS
metastatic renal cancer is mainly known only in the setting of Deferral of Nephrectomy as a Result of Disease
cytoreductive nephrectomy. This means that current clinical Symptoms and Burden
trialbased data regarding efficacy of systemic therapy in Case 1. A 68-year-old man presented with malena, anemia
RCC do not apply to a large magnitude of the patient (hemoglobin of 4.0 g/dL), and hip pain. Imaging revealed
population treated. As systemic therapy options and effi- a large renal mass, hip metastases, and lung metastases.
cacies evolve, the role of nephrectomy requires re-evaluation. Colonoscopy showed large mucosal lesions, the biopsy of
It does not seem appropriate to subject every patient with which revealed clear cell RCC. This patient, given the life-
metastatic renal cancer to the morbidity, adverse events, threatening nature of his metastatic disease, would not be a
cost, and potential delay of systemic therapy that result candidate for nephrectomy. The patient was started on an
from the nephrectomy procedure. In addition, special oral anti-VEGF tyrosine kinase inhibitor. His hemoglobin
efforts must be made to collect information regarding the improved within 2 weeks of starting therapy, and his lesions
efficacy of specific systemic therapies for the patients who were clinically responding. Clearly, this case needed to
are not candidates for nephrectomy. It is likely that the proceed to systemic therapy rapidly, and attempting cyto-
presence of nephrectomy is an objective measure that reductive nephrectomy for this patient was likely to have
reflects a subgroup within RCC that has a distinctly favorable a deleterious effect. This case also raises the dilemma of
outcome. whether to consider a nephrectomy at a later date after initial
response to targeted therapy. There is no evidence to support
FACTORS TO BE CONSIDERED PRIOR TO that cytoreductive nephrectomy would be beneficial at this
CYTOREDUCTIVE NEPHRECTOMY time in this case. The concerns comprise discontinuing VEGF
Histology (Clear Cell or Non-clear Cell) inhibitor therapy, the regrowth potential of his disease, and
Non-clear cell histology has suboptimal systemic therapy patient safety during surgery. In a clinical trial of preoperative
options, and hence cytoreductive nephrectomy maybe a sunitinib followed by cytoreductive nephrectomy, about 36%
more important component of the management. However, (17 of 47) of patients had disease progression during the
poor prognostic characteristics such as liver metastases, break from systemic therapy, confirming the concerns re-
impaired performance status, and the unresectable nature garding this approach.19 The patient continues to be treated
of the disease should be considered as relative contrain- without nephrectomy and with systemic therapy alone.
dications to cytoreductive nephrectomy.
Case 2. A 58-year-old man presented with severe dyspnea,
noted to be related to pleural effusion. A 6-cm left-sided
Patient Performance Status renal mass was noted. Thoracentesis was performed, and
Retrospective studies show that patients with impaired lung and pleural metastases were noted. Cytology revealed
performance status failed to benefit from cytoreductive malignant cells, and biopsy of kidney revealed clear cell
nephrectomy. cancer. The patient was treated with targeted therapy and
demonstrated a response with complete resolution of the
Current/Impending Symptoms From Disease pleural effusion and stable renal mass. In this case, ne-
Delay in systemic therapy because of cytoreductive ne- phrectomy was considered advisable to render the patient in
phrectomy is a concern for symptomatic patients with RCC. clinical complete remission. However, there were valid
concerns of holding systemic therapy and risking progres-
sion of disease during cytoreductive nephrectomy and the
Burden of Metastatic Disease healing period after the procedure.
Patients with liver metastases or nodal involvement were
likely to have a higher complication rate with cytoreductive
Case 3. A 52-year-old man was hospitalized with hyper-
nephrectomy that resulted in more than a 60-day delay in
calcemia; his corrected calcium was 15 mg/dL, and workup
systemic therapy.
revealed a renal mass and bilateral lung and bone lesions,
including one in the sacrum. Biopsy of bone showed clear cell
Memorial Sloan Kettering Cancer Center Risk Criteria cancer consistent with renal primary. This patient needs to
Multivariate analysis revealed no benefit from cytoreduc- proceed directly to systemic therapy, as attempting a ne-
tive nephrectomy for patients with poor risk characteristics phrectomy in the presence of uncontrolled metastatic dis-
per the Memorial Sloan Kettering Cancer Centers risk ease would result in worsening patient condition and
classification. compromise survival outcome.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e19


ULKA VAISHAMPAYAN

Deferral of Nephrectomy as a Result of Comorbid CONCLUSION


Conditions Absence of cytoreductive nephrectomy is a very common oc-
Case. An 85-year-old woman with coronary artery dis- currence in advanced RCC (65% of cases) and warrants further
ease and angioplasty with stent placement 4 months clinical and research investigation. The efficacy and toxicity
ago presented with microscopic hematuria. Workup reports from clinical trials of targeted and immune therapy
revealed a 7-cm right renal mass and bilateral lung predominantly represent the patient population that has had
masses. Biopsy of the lung lesion demonstrated clear cytoreductive nephrectomy and need to be interpreted and
cell carcinoma consistent with renal primary. Cytore- applied in the appropriate context. The complication rates of
ductive nephrectomy was considered risky because of nephrectomy range from 7% to 21%19 and may have impli-
the patients cardiac condition, and, hence, systemic cations in the delay of or inability to administer systemic therapy.
therapy was started with clinical response. Currently, Outcome-based research and interventional trials focusing on
the patient remains free of progression for the last the clinical treatment of patients with metastatic RCC not un-
10 months. dergoing nephrectomy represent a critical unmet need.

References

1. National Cancer Institute. SEER*Stat Databases: November 2014 Submission. 13. Motzer RJ, Hutson TE, Tomczak P, et al. Sunitinib versus interferon
http://seer.cancer.gov/data/seerstat/nov2014/. Accessed March 21, 2016. alfa in metastatic renal-cell carcinoma. N Engl J Med. 2007;356:
2. Flanigan RC, Yonover PM. The role of radical nephrectomy in metastatic 115-124.
renal cell carcinoma. Semin Urol Oncol. 2001;19:98-102. 14. Hudes G, Carducci M, Tomczak P, et al; Global ARCC Trial. Temsirolimus,
3. Mickisch GH, Garin A, van Poppel H, et al; European Organisation for interferon alfa, or both for advanced renal-cell carcinoma. N Engl J Med.
Research and Treatment of Cancer (EORTC) Genitourinary Group. Radical 2007;356:2271-2281.
nephrectomy plus interferon-alfa-based immunotherapy compared with 15. Sternberg CN, Davis ID, Mardiak J, et al. Pazopanib in locally advanced or
interferon alfa alone in metastatic renal-cell carcinoma: a randomised metastatic renal cell carcinoma: results of a randomized phase III trial.
trial. Lancet. 2001;358:966-970. J Clin Oncol. 2010;28:1061-1068.
4. Flanigan RC, Mickisch G, Sylvester R, et al. Cytoreductive nephrectomy 16. Rini BI, Halabi S, Rosenberg JE, et al. Phase III trial of bevacizumab plus
in patients with metastatic renal cancer: a combined analysis. J Urol. interferon alfa versus interferon alfa monotherapy in patients with
2004;171:1071-1076. metastatic renal cell carcinoma: final results of CALGB 90206. J Clin
5. Pantuck AJ, Belldegrun AS, Figlin RA. Nephrectomy and interleukin-2 for Oncol. 2010;28:2137-2143.
metastatic renal-cell carcinoma. N Engl J Med. 2001;345:1711-1712. 17. Choueiri TK, Xie W, Kollmannsberger C, et al. The impact of cytore-
6. Dadian G, Riches PG, Henderson DC, et al. Immunological parameters in ductive nephrectomy on survival of patients with metastatic renal cell
peripheral blood of patients with renal cell carcinoma before and after carcinoma receiving vascular endothelial growth factor targeted
nephrectomy. Br J Urol. 1994;74:15-22. therapy. J Urol. 2011;185:60-66.
7. Uzzo RG, Clark PE, Rayman P, et al. Alterations in NFkappaB activation in 18. Heng DY, Wells JC, Rini BI, et al. Cytoreductive nephrectomy in patients
T lymphocytes of patients with renal cell carcinoma. J Natl Cancer Inst. with synchronous metastases from renal cell carcinoma: results from
1999;91:718-721. the International Metastatic Renal Cell Carcinoma Database Consor-
8. MacFarlane AW IV, Jillab M, Plimack ER, et al. PD-1 expression on tium. Eur Urol. 2014;66:704-710.
peripheral blood cells increases with stage in renal cell carcinoma 19. Haas NB, Manola J, Uzzo R, et al. Initial results from ASSURE (E2805):
patients and is rapidly reduced after surgical tumor resection. Cancer adjuvant sorafenib or sunitinib for unfavorable renal carcinoma, an
Immunol Res. 2014;2:320-331. ECOG-ACRIN-led, NCTN phase III trial. J Clin Oncol. 2015;33;(suppl; abstr
9. Thompson RH, Dong H, Lohse CM, et al. PD-1 is expressed by tumor- 403).
infiltrating immune cells and is associated with poor outcome for 20. Vaishampayan U, Vankayala H, Vigneau FD, et al. The effect of targeted
patients with renal cell carcinoma. Clin Cancer Res. 2007;13:1757-1761. therapy on overall survival in advanced renal cancer: a study of the
10. Peters I, Tezval H, Kramer MW, et al. Implications of TCGA network data national surveillance epidemiology and end results registry database.
on 2nd generation immunotherapy concepts based on PD-L1 and PD-1 Clin Genitourin Cancer. 2014;12:124-129.
target structures. Aktuelle Urol. 2015;46:481-485. 21. Gershman B, Moreira DM, Boorjian SA, et al. Comprehensive charac-
11. Motzer RJ, Escudier B, McDermott DF, et al; CheckMate 025 In- terization of the perioperative morbidity of cytoreductive nephrec-
vestigators. Nivolumab versus everolimus in advanced renal-cell car- tomy. Eur Urol. 2016;69:84-91.
cinoma. N Engl J Med. 2015;373:1803-1813. 22. Gerlinger M, Rowan AJ, Horswell S, et al. Intratumor heterogeneity and
12. Escudier B, Eisen T, Stadler WM, et al; TARGET Study Group. Sorafenib in branched evolution revealed by multiregion sequencing. N Engl J Med.
advanced clear-cell renal-cell carcinoma. N Engl J Med. 2007;356:125-134. 2012;366:883-892.

e20 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


BREAST CANCER

Breast Cancer Survivorship:


Strategies for Optimal Care

CHAIR
Beverly Moy, MD, MPH
Massachusetts General Hospital Cancer Center
Boston, MA

SPEAKERS
Debra L. Barton, PhD, AOCN, RN
University of Michigan
Ann Arbor, MI

William J. Gradishar, MD
Robert H. Lurie Comprehensive Cancer Center of Northwestern University
Chicago, IL

Michelle E. Melisko, MD
UCSF Helen Diller Family Comprehensive Cancer Center
San Francisco, CA
MELISKO, GRADISHAR, AND MOY

Issues in Breast Cancer Survivorship: Optimal Care, Bone


Health, and Lifestyle Modifications
Michelle E. Melisko, MD, William J. Gradishar, MD, and Beverly Moy, MD, MPH

OVERVIEW

There are an estimated 3.1 million survivors of breast cancer in the United States. The predominant reasons for this
substantially large population are that breast cancer is the most common noncutaneous malignancy among women and that
5-year survival rates after breast cancer treatment are approximately 90%. These patients have many medical consid-
erations, including the need to monitor for disease recurrence and to manage complications of their previous cancer
treatments. Most patients remain at risk indefinitely for local and systemic recurrences of their breast cancers and have an
increased risk of developing contralateral new primary breast cancers. Therefore, optimizing care for this patient population
is critical to the overall health care landscape in the United States. Here, we summarize survivorship care delivery and its
challenges, the optimization of bone health in breast cancer survivors, and opportunities for risk reduction through lifestyle
modifications.

T here are an estimated 3.1 million survivors of breast


cancer in the United States.1 The predominant reasons
for this substantially large population are that breast cancer
From an economic perspective, breast cancer is the
costliest of all cancers, with an estimated $18.1 billion of
national expenditures in the United States in 2014. Almost
is the most common noncutaneous malignancy among $8 billion of these expenditures are for the costs of survi-
women and that 5-year survival rates after breast cancer vorship care.3 This proportional cost of breast cancer care
treatment are approximately 90%.2 In recognition of the exceeds the annual expenditures for the entire treatment of
importance of breast cancer survivorship care, multiple other types of cancer, such as leukemia and ovarian, brain,
medical organizations, including the Institute of Medicine, head and neck, or pancreatic cancer.
American Society of Clinical Oncology (ASCO), American Despite the calls for action from multiple prestigious
Cancer Society, and the Commission on Cancer, have high- medical societies and the reality of the various medical and
lighted the importance of addressing the many issues and economic challenges, to date, there is no universally agreed
concerns of breast cancer survivors following treatment. The upon standardized follow-up model for patients with early-
medical community recognizes the multiple challenges for stage breast cancer who have completed active cancer therapy.
breast cancer survivors that follow-up care creates. In addi- Barriers to survivorship care implementation are well docu-
tion to the significant logistics required to care for a large mented and include challenges with staffing and resources.4,5
population of breast cancer survivors, reasons for these Many societies have recommended that every patient receive a
challenges include the long course of disease, a relentless risk survivorship care plan that details follow-up care plans, a sur-
of recurrence even 15 to 20 years after the original diagnosis, veillance plan, and general health recommendations.6 However,
the need for endocrine therapy for 5 to 10 years in most survivorship care plan adoption in oncology clinics is highly
patients, and the need to continue screening because of a 1% variable, and there is insufficient evidence that the plans
risk per year of new contralateral primary breast cancers. improve outcomes for patients.4,7
Clinicians must address the physical, emotional, social, and There are three models of comprehensive survivorship
long-term effects faced by many breast cancer survivors after care delivery found in academic medical centers.8 The first
they complete active treatment. These long-term effects model consists of consultative clinics that offer a one-time
include anxiety and depression, vasomotor symptoms, cog- survivorship visit for patients when active therapy is com-
nitive dysfunction, fatigue, pain, sexual dysfunction, loss of plete. The second model consists of advanced care
bone density, infertility, neuropathy, and cardiac toxicity. practitionerled survivorship clinics, during which patients

From the UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL; Mas-
sachusetts General Hospital Cancer Center, Boston, MA.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Beverly Moy, MD, Massachusetts General Hospital Cancer Center, 55 Fruit St., Boston, MA 02114; email: bmoy@partners.org.

2016 by American Society of Clinical Oncology.

e22 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


ISSUES IN BREAST CANCER SURVIVORSHIP

meet with nurse practitioners or physician assistants either reassurance, and, oncologists, driven by a sense of duty to
in a stand-alone clinic or embedded within the oncology their patients, may be unwilling to relinquish control of their
practice. The patient is then transitioned back to primary care patients medical care after developing close relationships
as appropriate. The third model is a multidisciplinary survivor during active cancer treatment. Furthermore, primary care
program that relies on collaboration between oncologists, physicians may be uncomfortable taking the lead in caring for
advanced care practitioners, social workers, and psychologists these patients because they may feel ill-prepared to manage
to provide comprehensive care. the many issues faced by breast cancer survivors. Regarding
Some policymakers would argue that follow-up care of these specific guidance for medical care, we lack evidence about
patients can be transitioned to primary care physicians on the whether certain types of breast cancer, such as locally ad-
basis of randomized trial results that show equivalent out- vanced or hormone receptornegative disease, would benefit
comes with follow-up by either the oncologist or primary care from more intensive follow-up and imaging. Finally, with the
physician.9-12 The most compelling data are from a study in lack of interinstitutional standards, providers may worry about
which 968 women with early-stage breast cancer were ran- the risk of being sued if they decrease their follow-up visits or
domly assigned to follow-up care with either a family physician imaging studies.
or an oncologist.9 Recurrence rates, mortality rates, and rates There have been multiple efforts to provide clinical
of serious clinical events, such as spinal cord compression, guidance on how best to care for breast cancer survivors.
pathologic fractures, brachial plexopathy, or hypercalcemia, The National Comprehensive Cancer Network (NCCN) issued
were equivalent in both the family physician and the oncol- guidelines for the treatment of breast cancer, which in-
ogist groups. Health-related quality of life was also similar in cluded information on recommended surveillance for cancer
both groups. Lending additional support to simplifying follow- recurrence or new cancers.17 Both ASCO and NCCN have
up for breast cancer survivors, a British study revealed that symptom-specific survivorship care guidelines to address
twice as many patients preferred simpler, less frequent follow- issues such as cardiac toxicity and neuropathy.18,19 Recently,
up by telephone.13 Finally, patients are less likely to demand ASCO and the American Cancer Society jointly issued a
medically inappropriate testing, such as imaging or tumor breast cancer survivorship care guideline to provide con-
markers, if they are educated about the specificity, sensitivity, crete recommendations to assist clinicians, both oncologists
and usefulness of the available tests.14 and primary care physicians, in the care of these patients.20
These studies beg the question: Are we providing exces- Among the concrete recommendations, there is a recom-
sively redundant care to breast cancer survivors? In the current mendation patients receive a history and physical every 3 to
health care climate with finite resources, are oncology pro- 6 months for the first 3 years after primary therapy, every 6
viders obligated to be more discriminating about the to 12 months for the next 2 years, and annually thereafter.
amount of care we provide?15,16 There are myriad reasons Supported by the recommendations made by the ASCO
why oncology providers are not prepared to relinquish the Choosing Wisely campaign, clinicians should not offer rou-
care of breast cancer survivors. Providers and patients often tine laboratory tests or imaging, except mammography if
feel emotionally attached to each other. It is not uncommon indicated, for the detection of disease recurrence in the
for patients to want to see their oncologists more often for absence of symptoms.21 The guideline also recommends that
the cancer treatment team provides a treatment summary
and survivorship care plan to primary care clinicians.
These and other similar guidelines are helpful. However,
KEY POINTS they still provide ambiguity about the ideal models for care
and the appropriate intervals between follow-up exami-
Care for the estimated 3.1 million survivors of breast
cancer is logistically and medically complex, given the
nations. A history and physical every 3 to 6 months amounts
long course of disease, continued risk of recurrence, and to a difference of two to four clinician visits per year. Can
treatment-related side effects. these visits be shared between primary care physicians,
Additional research to provide evidence about the medical oncologists, surgical oncologists, and radiation
optimal way to deliver survivorship care to this large and oncologists? Would patients benefit more from increased
growing population is needed. care coordination? From a purely economical point of view,
To date, there is no uniform consensus about the role of one could argue that biannual visits by any oncology pro-
bone-modifying agents in the treatment of early-stage vider or by a primary care physician in an otherwise healthy
breast cancer. asymptomatic patient would be supported by the evidence
Data on breast cancer risk reduction are based on and may provide the most value in terms of containing
multiple epidemiologic studies.
medical costs. However, certain higher-risk patient pop-
Independent of the potential for reducing risk of breast
cancer recurrence, strong evidence exists that lifestyle
ulations may benefit from more intensive follow-up care.
factors, such as modification of diet, increasing physical Unfortunately, we lack clear evidence to be able to identify
activity, weight management, and smoking cessation, these patient subgroups. There is a pressing need for ad-
influence other medical comorbidities that ultimately ditional research to provide evidence about the optimal way
impact overall survival. to deliver care to a large and growing breast cancer survi-
vorship population.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e23


MELISKO, GRADISHAR, AND MOY

BONE HEALTH AGENTS IN BREAST CANCER The phase III ABSCG-18 trial was presented by Gnant et al at
The role of bone agents in the treatment of patients with the 2015 San Antonio Breast Cancer Symposium, in which
early-stage breast cancer has fallen under two broad cate- postmenopausal women with nonmetastatic breast cancer
gories: the established role of preventing loss of bone density were randomly assigned to receive the RANK ligand inhibitor,
and the more contentious role of reducing the risk of re- denosumab, or placebo in addition to aromatase inhibitor
currence of breast cancer.22-25 The two classes of bone agents therapy. 37 Previous results from the ABCSG-18 trial de-
used are bisphosphonates, of which several agents currently monstrated a reduced fracture rate with adjuvant denosumab
are in use, and RANK ligand inhibitors, of which there is one, compared with placebo in postmenopausal women with
denosumab. Cancer treatmentinduced bone loss is an iat- hormone receptorpositive breast cancer who were treated
rogenic disease, a condition created by breast cancer treat- with aromatase inhibitors.16 In the most recent presentation,
ments. It is well known that various cancer therapies (e.g., the primary outcomes were related to bone health, and the
aromatase inhibitors, chemotherapy, and surgery) decrease secondary outcomes included DFS. In an intention-to-treat
bone mineral density and increase the risk of fracture.26 analysis, the impact of denosumab on DFS reached borderline
Treatment-related fractures lead to decreased quality of life significance (hazard ratio [HR] 0.82; 95% CI, 0.661.00). The
and shorter survival times, which translate into clinical, social, benefit was approximately 1% after 3 years, 2% after 5 years,
and economic consequences.27,28 and 3% after 7 years of follow-up. Exploratory subgroup an-
The ABCSG-12,26,29,30 Z-FAST,31,32 and ZO-FAST33,34 trials alyses suggested that the benefit increases when denosumab
have shown that bone-targeted agents can prevent cancer is started early, along with aromatase inhibitor therapy, and
that the benefit is greater in patients who have larger tumors
treatmentinduced bone loss in early breast cancer. In the
and ductal histology. Overall, the DFS benefit seen with
ZO-FAST trial,29,33,34 up-front zoledronic acid increased bone
denosumab was comparable to that reported in the meta-
mineral density in both the lumbar spine and hip. Gnant
analysis of bisphosphonates.16
et al26,29,30 reported that the addition of zoledronic acid to
The debate about these data largely revolves around how
both tamoxifen and anastrozole yielded no treatment-
to translate the apparent benefit into day-to-day manage-
induced bone loss compared with treatment without the
ment decisions. Different bone agents were used in the
bone-strengthening agent. Additionally, denosumab pro-
trials, the trial designs were meant to evaluate primarily
duced similar results, with increases in bone mineral density
bone health endpoints with DFS as a secondary endpoint,
seen over 24 months in trabecular and cortical bone in women
duration of therapy is an open question, and which post-
with nonmetastatic breast cancer and low bone mass who
menopausal patients should be considered for this type of
were receiving adjuvant aromatase inhibitor therapy.35 adjuvant therapy remains unclear. To date, though the data
The issue of adjuvant bisphosphonate therapy in breast
are compelling, the NCCN guideline panel could not reach
cancer has been an area controversy. The results from several
consensus for supporting these bone agents as a compo-
long-term, large clinical trials have added to the confusion. nent of adjuvant therapy of breast cancer. That said, indi-
Improvements in disease-free survival (DFS) were reported vidual patients who have any issues with bone density or
in postmenopausal women who were treated with letrozole increased risks of fracture may reap the benefits on bone
and zoledronic acid compared with letrozole alone (ZO- health as well as some additional reduction in the risk of
FAST).29,33,35 In contrast, the findings of the AZURE trial35 did disease recurrence.
not support the routine use of zoledronic acid in the ad-
juvant management of breast cancer; no significant benefits
in DFS and overall survival (OS) were reported. In both the RISK REDUCTION THROUGH LIFESTYLE
ABCSG-1226,30 and AZURE35 trials, adjuvant bisphosphonates MODIFICATIONS
offered an advantage to postmenopausal women, regardless of After a breast cancer diagnosis, most patients have great
whether their postmenopausal status occurred naturally or was interest in adoption of lifestyle changes that might reduce
drug induced. In the results of the 9-year update of the ABCSG- their risk of cancer recurrence. Despite significant advances
12 trial, there was a persistent benefit in DFS with zoledronic in breast cancer therapies that are supported by randomized
acid in a low-estrogen environment (e.g., a postmenopausal clinical trials demonstrating benefits in DFS and OS, the data
state). The meta-analysis of 36 trials by Coleman et al,36 which on risk reduction through lifestyle modifications are more
evaluated nearly 23,000 women, focused on the use of limited and are primarily based on epidemiologic studies.
bisphosphonates in the adjuvant setting. Although no effects on Patients, the lay press, and physicians alike often confuse the
disease outcomes in premenopausal women were observed, data about which lifestyle factors may impact risk of de-
adjuvant bisphosphonates reduced bone metastases and im- veloping breast cancer compared with those factors that
proved survival in women who had low levels of reproductive have an influence on recurrence after diagnosis. Strong
hormones (which included women older than age 55 if men- evidence exists that, independent of the potential for re-
opausal status was unknown). There was a 34% reduction in the ducing risk of breast cancer recurrence, lifestyle factors that
risk of bone recurrence (p = .00001) and a 17% reduction in the include modification of diet, increasing physical activity,
risk of breast cancer death (p = .004). No significant reduction weight management, and smoking cessation influence other
was reported in first distant tumor recurrence outside of bone. medical comorbidities that ultimately impact OS. Similarly,

e24 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


ISSUES IN BREAST CANCER SURVIVORSHIP

lifestyle and treatment factors also contribute to an in- maintenance (# 5.0% weight change) was associated with
creased risk of second primary malignancies seen among increased all-cause mortality (HR 1.12). In a secondary
breast survivors.38 analysis to evaluate effects of the amount of weight gain,
moderate-level weight gain (5%10% compared with main-
Management of Comorbidities tenance of baseline weight) was not associated with a hazard
Cancer survivors face many health challenges, including of overall mortality (HR 0.97; 95% CI, 0.861.11; p = .70), but
morbidity from residual treatment toxicity and increased higher weight gain (. 10% from baseline compared with
mortality from cardiovascular and respiratory diseases. The maintenance) was associated with increased mortality (HR
impact of comorbidities on survival after a diagnosis of 1.23; 95% CI, 1.091.39; p , .001). Among studies for which
breast cancer cannot be underestimated. Among 63,566 data were available, weight gain among patients who were
women diagnosed with breast cancer who were older than already overweight at baseline (body mass index [BMI] .
age 66 and identified through the Surveillance, Epidemiol- 25.0 kg/m2) did not result in worse mortality. In terms of breast
ogy, and End Results database, age and comorbidities at the cancer outcomes, moderate weight gain (5%10%) was not
time of diagnosis had the largest effects on mortality from associated with breast cancerspecific mortality, whereas
causes other than breast cancer.39 The presence of car- there was a suggestion that weight gain of greater than 10%
diovascular disease, chronic obstructive pulmonary disease, was associated with breast cancerspecific mortality (HR 1.17;
or diabetes increased breast cancerspecific mortality by 95% CI, 1.001.38; p = .05). Interestingly, in the studies within
between 10% and 24%. Among this study population, car- this meta-analysis that included data, no amount of weight
diovascular disease was the primary cause of death in 15.9% gain was associated with an increase in breast cancer recur-
of women, followed closely by breast cancer in 15.1%. Al- rences.47,48 The reason for this may be that a larger number of
though this study focused on an older population with breast patients would be required to demonstrate a relatively small
cancer, exposure to radiation, anthracyclines, and trastu- contribution of weight gain on breast cancer recurrences.
zumab are all associated with a small risk of cardiac toxicity Alternatively, the negative impact of weight gain on mortality
that may subsequently result in excess morbidity and, in and possibly breast cancerspecific mortality after diagnosis of
some cases, mortality.39 In addition, the long-term impacts breast cancer may not be due to increased rate of recurrence
of inducing premature menopause and of treatment with but, rather, to the overall health, comorbidities, treatment
an aromatase inhibitor on cardiovascular health in younger patterns, and responses to treatment after recurrence among
breast cancer survivors are still unknown. Studies to in- patients with significant weight gain.
vestigate beta blockers, angiotensin-converting enzyme
inhibitors, and statins to reduce the risk of cardiac toxicity Weight Loss Interventions
during and after treatment with anthracyclines and tras- No matter what the underlying cause, avoidance of signif-
tuzumab have shown some promise, but none of these icant weight gain after a diagnosis of breast cancer is a de-
pharmacologic interventions have become standard of sirable goal to improve OS. The challenge of avoiding weight
care.40-42 Attention to reducing the risk of subsequent gain is substantial for breast cancer survivors, many of whom
cardiovascular disease should be a priority after the di- enter menopause abruptly as a result of chemotherapy and
agnosis and treatment of breast cancer. some of whom remain on hormonal therapy as long as 10
years. Additionally, many patients suffer ongoing toxicities
Weight Gain from their treatment, including fatigue, sleep disturbance,
Weight gain is common during and after treatment of breast neuropathy, and arthralgias, all which may interfere with the
cancer and may be even more pronounced in patients who ability to initiate and maintain an exercise program supportive
are overweight or obese prior to diagnosis.43 Factors con- of weight control.
tributing to weight gain include use of corticosteroid pre- There have been numerous weight loss intervention
medications with chemotherapy, decreased physical activity studies among breast cancer survivors. A recent systematic
during treatment, transition into menopause, and ongoing review included 15 studies, among which were eight ran-
hormonal therapy.44 Weight gain after diagnosis has been domized controlled trials, four single-cohort experimental
associated with a higher rate of breast cancer recurrences intervention studies, two randomized parallel-intervention
and with worse OS. In an analysis of a cohort of 3,993 women trials, one randomized crossover trial, and one controlled
identified through state registries who were diagnosed with nonrandomized trial.49 The primary objective of each of
stage I to III breast cancer, each 5-kg gain was associated these studies was weight loss intervention with a focus on
with a 12% increase in all-cause mortality, a 13% increase in changing body weight (measured as change in weight, BMI,
breast cancerspecific mortality, and a 19% increase in percentage body fat, or percent overweight). Studies de-
cardiovascular disease mortality.45 A systematic review and signs included multicomponent interventions that in-
meta-analysis that included a total of 12 cohort studies and corporated diet, physical activity, and behavior modification,
clinical trials with a total of 23,832 patients measured weight whereas other studies combined personalized lifestyle
change after breast cancer diagnosis and investigated breast telephone counseling with face-to-face group-based edu-
cancerspecific mortality, all-cause mortality, and recur- cation. Several studies used a commercial weight loss
rence outcomes.46 Weight gain ($ 5.0%) compared with programs, such as Weight Watchers and/or Curves,50,51 and

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e25


MELISKO, GRADISHAR, AND MOY

one study incorporated group teleconferencing for breast The Womens Healthy Eating and Living (WHEL) randomized
cancer survivors in rural locations.52 trial tested whether increasing vegetable, fruit, and fiber intake
Of the 15 studies included in this systematic review, 14 and decreasing dietary fat intake could reduce the risk of re-
resulted in significant weight loss; weight loss of 5% or greater currence or new primary breast cancer and impact mortality in
from baseline was reported in 10 of the studies (p , .001). patients with early-stage breast cancer.55 A total of 3,088
Unfortunately, weight regain was observed in some women were randomly assigned to an intervention arm to
studies that included longer follow-up. For example, in the receive telephone counseling, cooking classes, and newsletters
Exercise and Nutrition to Enhance Recovery and Good promoting high fruit and vegetable intake, increasing fiber, and
Health for You (ENERGY) study, 692 overweight or obese reducing energy intake from fat, or to a comparison group that
breast cancer survivors were randomly assigned to either a was provided only with print materials. Analysis validated by
group-based behavioral intervention, supplemented with plasma carotenoid concentrations indicated that the inter-
telephone counseling and tailored newsletters, or to a less- vention group maintained significant differences compared
intensive control intervention to encourage weight loss. with the control group for increased fruit, vegetable, and fiber
Greater weight loss was observed in the intervention than in intake and reduction in fat through 4 years of follow-up. After a
the control group at 12 months (6% vs. 1.5%) and was still mean follow-up time of 7.3 years, there were no differences
maintained but was less pronounced at 24 months (3.7% vs. between the intervention and control groups for recurrence of
1.3%). Improvements in blood pressure control and physical invasive or noninvasive breast cancer or death. Explanations of
activity were also observed in the intervention arm.53 this lack of treatment effect included the fact that many WHEL
participants had already changed their dietary patterns after
Dietary Interventions diagnosis of breast cancer, so 75% of patients were already
The evidence to support specific dietary recommendations consuming at least five servings of vegetables and fruit a day at
after a diagnosis of breast cancer is surprisingly limited and is baseline; results suggest that intake of fruits and vegetables
backed by only a handful of randomized clinical trials, few of beyond that amount may not be beneficial. Multiple sub-
which achieved the desired endpoints of either reduction in sequent endocrinologic and behavioral findings and subset
breast cancer recurrences or improvement in DFS or OS. analyses have been reported from the data collected from this
The Womens Intervention Nutrition Study (WINS) was a very well-conducted trial, which demonstrates the importance
randomized prospective multicenter clinical trial that of this study despite its negative results.
included a total of 2,437 patients with early-stage breast
cancer to test the effect of a dietary intervention designed to Exercise
reduce fat intake.54 The goal of the dietary intervention was Physical activity has been shown to improve quality of life
to reduce percentage of calories from fat to 15%, such that after a breast cancer diagnosis.56 Multiple cohort studies
there would actually be a sustained reduction in fat intake to have investigated the impact of physical activity after di-
approximately 20% of calories in the intervention arm. The agnosis on breast cancerspecific and overall mortalities.
low-fat eating plan included self-monitoring (fat gram Most of these studies describe exercise frequency and in-
counting and recording), goal setting, modeling, and relapse tensity in metabolic equivalent (MET) hours, for which 3 MET
support and involved multiple in-person counseling sessions hours is equivalent to walking at average pace of 2 to 2.9 mph
initially and then subsequent contact with a dietician every for 1 hour. Among 2,987 patients with early-stage breast cancer
3 months. The patients in the control group had a baseline in the Nurses Health Study, the relative risk of death from
dietician visit and then visits every 3 months, during which breast cancer, compared with women who engaged in fewer
general dietary guidelines were reviewed. At a median than 3 MET hours per week of physical activity, for patients with
follow-up time of 60 months, an interim analysis demon- 9 to 14.9 MET hours per week of exercise was 0.50 (95% CI,
strated that dietary fat intake was significantly lower in the 0.310.82); for patients with 15 to 23.9 MET hours per week
intervention than in the control group (p , .001), and the was 0.56 (95% CI, 0.380.84); and for patients with 24 or more
effect was maintained out to 60 months. Additionally, there MET hours per week was 0.60 (95% CI, 0.400.89). Absolute
was a 6-pound lower mean body weight in the intervention mortality risk was also reduced by 6% at 10 years for women
group. The hazard of relapse events (local, regional, distant, who engaged in 9 or more MET hours per week.57
or ipsilateral breast cancer recurrence or new contralateral In the Collaborative Womens Longevity Study (CWLS),
breast cancer) in the intervention group compared with the 4,482 women completed questionnaires on postdiagnosis
control group was 0.76 (95% CI, 0.600.98). Subgroup an- physical activity and other lifestyle factors. Compared
alyses were conducted on the basis of BMI, hormone re- with women engaging in fewer than 2.8 MET hours per
ceptor status, and nodal status, and the dietary intervention week of physical activity, women who engaged in greater
had a greater effect on relapse-free survival in women with levels of activity ($ 21 MET hours per week) had a sig-
estrogen receptornegative cancer (HR 0.58; 95% CI, nificantly lower risk of dying as a result of breast cancer
0.370.91) than in women with estrogen receptorpositive (HR 0.51; 95% CI, 0.290.89; p = .05) and had improved OS
disease (HR 0.85; 95% CI, 0.631.14). In this analysis, with a (HR 0.44; 95% CI, 0.320.60; p , .001). These results were
60-month median follow-up time, there was no difference in consistent independent of age, stage of disease, and
OS between the dietary intervention and control groups. BMI.58

e26 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


ISSUES IN BREAST CANCER SURVIVORSHIP

In two smaller-sized cohorts (1,970 women in The Life After with no drinking was associated with an increased risk of
Cancer Epidemiology [LACE] study and 933 women in the breast cancer recurrence (HR 1.35; 95% CI, 1.001.83) and
Health, Eating, Activity and Lifestyle [HEAL] study), greater death as a result of breast cancer (HR 1.51; 95% CI,
physical activity after diagnosis was associated with a de- 1.002.29). The increased risk of recurrence was most
crease in overall mortality but not in breast cancerspecific pronounced in postmenopausal and overweight/obese
mortality.59,60 women. Alcohol intake was not associated with overall
A longitudinal study of 4,643 women diagnosed with in- mortality, possibly because of a cardioprotective effect
vasive breast cancer after entry into the Womens Health and a reduction in noncancer deaths.61
Initiative study evaluated the association between change in In contrast to the findings in the LACE cohort, alcohol
physical activity from before diagnosis to after diagnosis with intake was not significantly associated with breast cancer
all-cause and breast cancerspecific mortalities. Physical ac- recurrence in 3,088 patients participating in the WHEL
tivity was reported at baseline (before breast cancer diagnosis) trial.62 Among this group of patients, moderate alcohol
and after diagnosis (3 or 6 years after baseline visit). Women intake (. 300 g/month or seven drinks a week) was as-
who engaged in moderate levels of physical activity (fast walking sociated with improved all-cause mortality (HR 0.69; 95%
for 3 hours/week) before diagnosis had lower all-cause mortality CI, 0.490.97) when analysis was adjusted for obesity. One
(HR 0.61; 95% CI, 0.440.87; p = .01) than inactive women. explanation for this may be that obese women were more
Women engaging in similar moderate physical activity levels likely to be nondrinkers, and patients with moderate al-
after diagnosis had both lower breast cancer mortality (HR 0.61; cohol consumption may engage in protective behaviors,
95% CI, 0.350.99; p = .049) and lower all-cause mortality (HR such as increased physical activity.
0.54; 95% CI, 0.380.79; p , .01). Women who increased or In the After Breast Cancer Pooling Project, which included
maintained physical activity of 9 or more MET hours per week 9,329 patients with breast cancer (more than half of whom
after diagnosis had lower all-cause mortality (HR 0.67; 95% CI, were part of the LACE and WHEL cohorts), regular alcohol
0.460.96) even if they were inactive before diagnosis.59 intake ($ 6.0 g/day) had no effect on breast cancer recurrence
With the abundance of epidemiologic data demonstrating or overall mortality among the entire population. However,
the benefit of exercise after diagnosis of breast cancer, nu- among women who were postmenopausal, alcohol con-
merous randomized prospective trials are ongoing to better sumption of 6.0 or more g/day was associated with increased
understand which interventions are most effective to increase breast cancer recurrence (HR 1.19; 95% CI, 1.011.40).61
physical activity, what levels and types of physical activity are
optimal, and if associated lifestyle factors have an interaction
with the benefit of exercise. CONCLUSION
Breast cancer survivorship care has emerged as a high-
Alcohol Consumption priority issue in the oncology community, given its large
There is reasonably strong evidence that moderate alcohol and growing population. Breast cancer survivors face
consumption increases the risk of developing breast cancer, multiple complex issues, including optimization of bone
but the effect of ongoing alcohol intake after diagnosis is health and lifestyle considerations. Clinicians caring for
less clear. In the LACE cohort consisting of 1,897 patients this substantial population must be knowledgeable on
with early-stage breast cancer who enrolled on average 2 many medical issues and must be prepared to counsel
years after diagnosis, self-reported drinking of 6 g/day or their patients on the best health promotion strategies to
more of alcohol (approximately 2.5 ounces of wine, 6 minimize risks of recurrence and long-term side effects of
ounces of beer, or 1.5 ounces of hard liquor) compared their treatments.

References
1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin. 6. Hewitt M, Greenfield S, and Stovall E (eds). From Cancer Patient to
2015;65:5-29. Cancer Survivor: Lost in Transition. Washington, DC: The National
2. Howlader N, Noone AM, Krapcho M, et al (eds). SEER Cancer Statistics Academies Press; 2005.
Review, 1975-2012. http://seer.cancer.gov/csr.1975-2012/. Bethesda, 7. Salz T, McCabe MS, Onstad EE, et al. Survivorship care plans: is there buy-in
MD: National Cancer Institute; 2015. from community oncology providers? Cancer. 2014;120:722-730.
3. National Institutes of Health. Cancer trends progress report. http:// 8. Oeffinger KC, McCabe MS. Models for delivering survivorship care. J Clin
progressreport.cancer.gov. Accessed January 12, 2016. Oncol. 2006;24:5117-5124.
4. Mayer DK, Birken SA, Check DK, et al. Summing it up: an integrative 9. Grunfeld E, Levine MN, Julian JA, et al. Randomized trial of long-term
review of studies of cancer survivorship care plans (2006-2013). Cancer. follow-up for early-stage breast cancer: a comparison of family phy-
2015;121:978-996. sician versus specialist care. J Clin Oncol. 2006;24:848-855.
5. Birken SA, Mayer DK, Weiner BJ. Survivorship care plans: prevalence 10. Grunfeld E, Mant D, Yudkin P, et al. Routine follow up of breast cancer in
and barriers to use. J Cancer Educ. 2013;28:290-296. primary care: randomised trial. BMJ. 1996;313:665-669.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e27


MELISKO, GRADISHAR, AND MOY

11. Grunfeld E, Fitzpatrick R, Mant D, et al. Comparison of breast cancer patient 31. Brufsky AM, Harker WG, Beck JT, et al. Final 5-year results of Z-FAST trial:
satisfaction with follow-up in primary care versus specialist care: results adjuvant zoledronic acid maintains bone mass in postmenopausal
from a randomized controlled trial. Br J Gen Pract. 1999;49:705-710. breast cancer patients receiving letrozole. Cancer. 2012;118:
12. Kerrigan D, Waters P, Ryan M, et al. Follow-up arrangements for breast 1192-1201.
cancer patients: is it appropriate to transfer surveillance to general 32. Coleman R, de Boer R, Eidtmann H, et al. Zoledronic acid (zoledronate)
practioners? Ir Med J. 2014;107:273-275. for postmenopausal women with early breast cancer receiving adjuvant
13. Gulliford T, Opomu M, Wilson E, et al. Popularity of less frequent follow letrozole (ZO-FAST study): final 60-month results. Ann Oncol. 2013;24:
up for breast cancer in randomised study: initial findings from the 398-405.
hotline study. BMJ. 1997;314:174-177. 33. Eidtmann H, de Boer R, Bundred N, et al. Efficacy of zoledronic acid in
14. Loprinzi CL, Hayes D, Smith T. Doc, shouldnt we be getting some tests? postmenopausal women with early breast cancer receiving adjuvant
J Clin Oncol. 2000;18:2345-2348. letrozole: 36-month results of the ZO-FAST Study. Ann Oncol. 2010;21:
15. Jagsi R. Debating the oncologists role in defining the value of cancer 2188-2194.
care: we have a duty to society. J Clin Oncol. 2014;32:4035-4038. 34. Ellis GK, Bone HG, Chlebowski R, et al. Randomized trial of denosumab in
16. Sulmasy D, Moy B. Debating the oncologists role in defining the value of patients receiving adjuvant aromatase inhibitors for nonmetastatic
cancer care: our duty is to our patients. J Clin Oncol. 2014;32:4039-4041. breast cancer. J Clin Oncol. 2008;26:4875-4882.
17. Gradishar WJ, Anderson BO, Balassanian R, et al. Breast Cancer, Version 35. Coleman R, Cameron D, Dodwell D, et al; AZURE investigators. Adjuvant
1.2016. J Natl Compr Canc Network. 2015;13:1475-1485. zoledronic acid in patients with early breast cancer: final efficacy
18. Denlinger CS, Ligibel JA, Are M, et al. Survivorship: screening for cancer analysis of the AZURE (BIG 01/04) randomised open-label phase 3 trial.
and treatment effects, version 2.2014. J Natl Compr Canc Netw. 2014; Lancet Oncol. 2014;15:997-1006.
12:1526-1531. 36. Coleman R, Powles T, Paterson A, et al; Early Breast Cancer Trialists
19. Khatcheressian JL, Hurley P, Bantug E, et al; American Society of Clinical Collaborative Group (EBCTCG). Adjuvant bisphosphonate treatment in
Oncology. Breast cancer follow-up and management after primary early breast cancer: meta-analyses of individual patient data from
treatment: American Society of Clinical Oncology clinical practice randomised trials. Lancet. 2015;386:1353-1361.
guideline update. J Clin Oncol. 2013;31:961-965. 37. Gnant M, Pfeiler G, Dubsky PC, et al; Austrian Breast and Colorectal
20. Runowicz CD, Leach CR, Henry NL, et al. American Cancer Society/ Cancer Study Group. Adjuvant denosumab in breast cancer (ABCSG-
American Society of Clinical Oncology breast cancer survivorship care 18): a multicentre, randomised, double-blind, placebo-controlled trial.
guideline. J Clin Oncol. 2016; 34:611-635. Lancet. 2015;386:433-443.
21. Schnipper LE, Smith TJ, Raghavan D, et al. American Society of Clinical 38. Curtis RE, Freedman DM, Ron E, et al. New Malignancies among
Oncology identifies five key opportunities to improve care and reduce Cancer Survivors: Seer Cancer Registries, 1973-2000. NIH Publication
costs: the top five list for oncology. J Clin Oncol. 2012;30:1715-1724. No. 05-5302 181-186. Washington, DC: National Cancer Institute;
22. Ben-Aharon I, Vidal L, Rizel S, et al. Bisphosphonates in the adjuvant 2006.
setting of breast cancer therapyeffect on survival: a systematic review 39. Zagar TM, Cardinale DM, Marks LB. Breast cancer therapy-associated
and meta-analysis. PLoS One. 2013;8:e70044. cardiovascular disease. Nat Rev Clin Oncol. 2016;13:172-184.
23. Hadji P, Coleman RE, Wilson C, et al. Adjuvant bisphosphonates in early 40. Cardinale D, Colombo A, Sandri MT, et al. Prevention of high-dose
breast cancer: consensus guidance for clinical practice from a European chemotherapy-induced cardiotoxicity in high-risk patients by
Panel. Ann Oncol. 2016;27:379-390. angiotensin-converting enzyme inhibition. Circulation. 2006;114:
24. Strobl S, Korkmaz B, Devyatko Y, et al. Adjuvant bisphosphonates and 2474-2481.
breast cancer survival. Annu Rev Med. 2016;67:1-10. 41. Seicean S, Seicean A, Plana JC, et al. Effect of statin therapy on the risk
25. Gnant M, Mlineritsch B, Luschin-Ebengreuth G, et al; Austrian Breast for incident heart failure in patients with breast cancer receiving
and Colorectal Cancer Study Group (ABCSG). Adjuvant endocrine anthracycline chemotherapy: an observational clinical cohort study.
therapy plus zoledronic acid in premenopausal women with early-stage J Am Coll Cardiol. 2012;60:2384-2390.
breast cancer: 5-year follow-up of the ABCSG-12 bone-mineral density 42. Pituskin E, Mackey JR, Koshman S, et al. Prophylactic beta blockade
substudy. Lancet Oncol. 2008;9:840-849. preserves left ventricular ejection fraction in HER2-overexpressing
26. Ezat SW, Syed Junid SM, Noraziani K, et al. Skeletal-related events breast cancer patients receiving trastuzumab: primary results of the
among breast and prostate cancer patients: towards new treatment MANTICORE randomized controlled trial. Presented at: San Antonio
initiation in Malaysias hospital setting. Asian Pac J Cancer Prev. 2013; Breast Cancer Symposium; December 8-12, 2015; San Antonio, TX.
14:3357-3362. Abstract S1-05.
27. Hagiwara M, Delea TE, Chung K. Healthcare costs associated with 43. Chlebowski RT, Aiello E, McTiernan A. Weight loss in breast cancer
skeletal-related events in breast cancer patients with bone metastases. patient management. J Clin Oncol. 2002;20:1128-1143.
J Med Econ. 2014;17:223-230. 44. Goodwin PJ, Ennis M, Pritchard KI, et al. Adjuvant treatment and onset
28. Gnant M, Eidtmann H. The anti-tumor effect of bisphosphonates ABCSG- of menopause predict weight gain after breast cancer diagnosis. J Clin
12, ZO-FAST and more. Crit Rev Oncol Hematol. 2010;74:S2-S6 (suppl 1). Oncol. 1999;17:120-129.
29. Gnant M, Mlineritsch B, Stoeger H, et al; Austrian Breast and Colorectal 45. Nichols HB, Trentham-Dietz A, Egan KM, et al. Body mass index before
Cancer Study Group, Vienna, Austria. Adjuvant endocrine therapy plus and after breast cancer diagnosis: associations with all-cause, breast
zoledronic acid in premenopausal women with early-stage breast cancer, and cardiovascular disease mortality. Cancer Epidemiol Bio-
cancer: 62-month follow-up from the ABCSG-12 randomised trial. markers Prev. 2009;18:1403-1409.
Lancet Oncol. 2011;12:631-641. 46. Playdon MC, Bracken MB, Sanft TB, et al. Weight gain after breast
30. Brufsky AM, Bosserman LD, Caradonna RR, et al. Zoledronic acid ef- cancer diagnosis and all-cause mortality: systematic review and meta-
fectively prevents aromatase inhibitor-associated bone loss in post- analysis. J Natl Cancer Inst. 2015;107:djv275.
menopausal women with early breast cancer receiving adjuvant 47. Caan BJ, Emond JA, Natarajan L, et al. Post-diagnosis weight gain and
letrozole: Z-FAST study 36-month follow-up results. Clin Breast Cancer. breast cancer recurrence in women with early-stage breast cancer.
2009;9:77-85. Breast Cancer Res Treat. 2006;99:47-57.

e28 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


ISSUES IN BREAST CANCER SURVIVORSHIP

48. Jeon YW, Lim ST, Choi HJ, et al. Weight change and its impact on prognosis 55. Pierce JP, Natarajan L, Caan BJ, et al. Influence of a diet very high in
after adjuvant TAC (docetaxel-doxorubicin-cyclophosphamide) chemo- vegetables, fruit, and fiber and low in fat on prognosis following
therapy in Korean women with node-positive breast cancer. Med Oncol. treatment for breast cancer: the Womens Healthy Eating and Living
2014;31:849. (WHEL) randomized trial. JAMA. 2007;298:289-298.
49. Playdon M, Thomas G, Sanft T, et al. Weight loss intervention for breast 56. Daley AJ, Crank H, Saxton JM, et al. Randomized trial of exercise therapy
cancer survivors: a systematic review. Curr Breast Cancer Rep. 2013;5: in women treated for breast cancer. J Clin Oncol. 2007;25:1713-1721.
222-246. 57. Holmes MD, Chen WY, Feskanich D, et al. Physical activity and survival
50. Djuric Z, DiLaura NM, Jenkins I, et al. Combining weight-loss counseling after breast cancer diagnosis. JAMA. 2005;293:2479-2486.
with the weight watchers plan for obese breast cancer survivors. Obes 58. Holick CN, Newcomb PA, Trentham-Dietz A, et al. Physical activity and
Res. 2002;10:657-665. survival after diagnosis of invasive breast cancer. Cancer Epidemiol
51. Greenlee HA, Crew KD, Mata JM, et al. A pilot randomized controlled Biomarkers Prev. 2008;17:379-386.
trial of a commercial diet and exercise weight loss program in minority 59. Irwin ML, McTiernan A, Manson JE, et al. Physical activity and survival in
breast cancer survivors. Obesity (Silver Spring). 2013;21:65-76. postmenopausal women with breast cancer: results from the womens
52. Befort CA, Klemp JR, Austin HL, et al. Outcomes of a weight loss in- health initiative. Cancer Prev Res (Phila). 2011;4:522-529.
tervention among rural breast cancer survivors. Breast Cancer Res 60. Sternfeld B, Weltzien E, Quesenberry CP Jr, et al. Physical activity and
Treat. 2012;132:631-639. risk of recurrence and mortality in breast cancer survivors: findings from
53. Rock CL, Flatt SW, Byers TE, et al. Results of the Exercise and Nutrition to the LACE study. Cancer Epidemiol Biomarkers Prev. 2009;18:87-95.
Enhance Recovery and Good Health for You (ENERGY) trial: a behavioral 61. Kwan ML, Chen WY, Flatt SW, et al. Postdiagnosis alcohol consumption
weight loss intervention in overweight or obese breast cancer survivors. and breast cancer prognosis in the after breast cancer pooling project.
J Clin Oncol. 2015;33:3169-3176. Cancer Epidemiol Biomarkers Prev. 2013;22:32-41.
54. Chlebowski RT, Blackburn GL, Thomson CA, et al. Dietary fat reduction 62. Flatt SW, Thomson CA, Gold EB, et al. Low to moderate alcohol intake is
and breast cancer outcome: interim efficacy results from the Womens not associated with increased mortality after breast cancer. Cancer
Intervention Nutrition Study. J Natl Cancer Inst. 2006;98:1767-1776. Epidemiol Biomarkers Prev. 2010;19:681-688.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e29


BREAST CANCER

Less Is More: A Multidisciplinary


Conversation on Treatment
Options

CHAIR
Fatima Cardoso, MD
Champalimaud Cancer Centre
Lisbon, Portugal

SPEAKERS
Eun-Sil Shelley Hwang, MD, MPH
Duke University
Durham, NC

Laura Esserman, MD, MBA


University of California, San Francisco
San Francisco, CA
TAILORING CHEMOTHERAPY IN EARLY-STAGE BREAST CANCER

Tailoring Chemotherapy in Early-Stage Breast Cancer: Based


on Tumor Biology or Tumor Burden?
Domen Ribnikar, MD, and Fatima Cardoso, MD

OVERVIEW

The question of whether to offer adjuvant chemotherapy to patients with early-stage breast cancer has always been
challenging to answer. It is well known that a substantial proportion of patients with early-stage breast cancer are over
treated, especially when staging and hormonal and HER2 receptors are solely taken into consideration. The advances in our
knowledge of breast cancer biology and its clinical implications were the basis for the discovery of additional reliable
prognostic markers to aid decision making for adjuvant treatment. Gene expression profiling is a molecular tool that more
precisely defines the intrinsic characteristics of each individual tumor. The application of this technology has led to the
development of gene signatures/profiles with relevant prognosticand some predictivevalue that have become im-
portant tools in defining which patients with early-stage breast cancer can be safely spared from chemotherapy. However,
the exact clinical utility of these tools will only be determined after the results of two large prospective randomized trials,
MINDACT and TailorX, evaluating their role become available. Notwithstanding the existence of these genomic tools, tumor
burden (defined as tumor size and nodal status) still has independent prognostic value and must be incorporated in decision
making. In addition, these gene signatures have limited predictive value, and new biomarkers and new targets are needed.
Therefore close collaboration between clinicians and scientists is crucial. Lastly, issues of cost-effectiveness, reimbursement,
and availability are crucial and widely variable around the globe.

H istorically, breast cancer was viewed as one disease with


different histopathological characteristics and responses
to systemic treatment. The choice of treatment was solely
The advent of genomic signatures allowed us to better
understand the heterogeneity of breast cancer and led to
more individualized systemic treatment approaches. In this
based on clinicopathologic parameters that are prognostic, article, we describe traditional clinicopathologic factors and
such as tumor size, nodal status, and histologic grade, and the new molecular tools that may be used to more accurately
three predictive markers of response to either endocrine tailor adjuvant treatment decisions for patients with early-
therapy and/or trastuzumab therapy (estrogen and pro- stage breast cancer. Additionally, we discuss some studies
gesterone receptor expression for endocrine therapy and showing that response to a specific type of treatment is
HER2 for trastuzumab). These factors are combined with mainly determined by the intrinsic molecular characteristics
different algorithms for treatment decision making, such as of each individual tumor rather than by anatomic prognostic
those used by Adjuvant! Online1 and the Nottingham parameters. However, these anatomic parameters have
Prognostic Index, and represent the basis of international important risk implications and effects on the relative
treatment guidelines, including those from the National magnitude of benefit of treatment.
Comprehensive Cancer Network,2 National Cancer Institute,
and European Society for Medical Oncology 3 as well as TRADITIONAL FACTORS FOR ADJUVANT
St. Gallens consensus statements.4 The major disadvantage of CHEMOTHERAPY DECISION MAKING
this approach is that nearly 60% of all patients with early-stage The Early Breast Cancer Trialists Collaborative Group (EBCTCG)
breast cancer receive adjuvant chemotherapy, but only 2% to meta-analyses (also called Oxford Overview) have demon-
15% of these patients derive an important benefit and others strated substantial evidence of the efficacy of adjuvant
only experience its toxic effects.5 chemotherapy in early-stage breast cancer.6,7 The last pub-
Fortunately, breast cancer was one of the first cancers with lished results in 2005 report on data from 60 randomized
advances in molecular profiling technologies, leading to a trials initiated before 1995 comparing polychemotherapy to
deeper understanding of its biology and molecular subtypes. no chemotherapy (29,000 patients of whom 10,000 died).7

From the Department of Medical Oncology, Institute of Oncology, Ljubljana, Slovenia; Breast Unit, Champalimaud Cancer Center, Champalimaud Foundation, Lisbon, Portugal.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Fatima Cardoso, MD, Champalimaud Cancer Center, Av. De Brasilia, Doca De Pedroucos, 1400-048, Lisbon, Portugal; email: fatimacardoso@fundacaochampalimaud.pt.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e31


RIBNIKAR AND CARDOSO

At 15-year follow-up, there was a substantial reduction in Generally, decisions regarding adjuvant chemotherapy are
the absolute risk of recurrence and death from breast cancer based on prognosis, that is, risk of disease recurrence, and on
for polychemotherapy compared with no chemotherapy. the likelihood of benefit from the treatment. The threshold
Among women younger than age 50, polychemotherapy of 10% risk for distant relapse is often used to make rec-
reduced the annual risk of relapse and death by 37% and ommendation for adjuvant chemotherapy.10
30%, respectively, and the absolute gain in survival was twice Clinicopathological prognostic factors for survival of early-
as great at 15 years as it was at 5 years (10% vs. 4.7%). Among stage breast cancer include tumor size, nodal status, his-
older women (age 5069), at 15-year follow-up, the annual tologic grade, and estrogen receptor, progesterone re-
risk of relapse and breast cancer mortality were reduced by ceptor, and HER2 expression as well as the presence of
19% and 12%, respectively, and translated into an absolute lymphovascular invasion. Several consensus and evidence-
gain of 4.1% and 3%, respectively.7,8 The proportional re- based guidelines3,4,11,12 provide recommendations on the
ductions in both recurrence and breast cancer mortality use of adjuvant chemotherapy in early-stage breast cancer.
were similar in node-negative and node-positive patients, The St. Gallen guidelines,4 for instance, suggest that patients
although the absolute benefit was greater in those with with early-stage breast cancer at low enough risk to avoid
node-positive disease. Furthermore, it was demonstrated adjuvant chemotherapy are characterized by node-negative
that the greatest effect of adjuvant chemotherapy occurs in disease with all of the following features: pathologic tumor
the initial few years after therapy. size of 2 cm or smaller, grade 1, estrogen receptor and pro-
Additional subgroup analyses showed that adjuvant che- gesterone receptor positive, HER2 negative, age older than
motherapy was effective in both estrogen receptornegative 35, and no vascular invasion. All other patients who do not
and estrogen receptorpositive disease, albeit the magnitude meet these criteria are considered to derive sufficient benefit
of the absolute benefit was larger in estrogen receptor from adjuvant chemotherapy because their risk of distant
negative disease. Data from multiple Cancer and Leukemia relapse is considered higher than 10%.
Group B (CALGB) trials among women with node-positive Adjuvant! Online is an independently validated, widely
breast cancer reveal greater chemotherapy benefit in es- used web-based tool for assessment of the risk of recurrence
trogen receptornegative disease compared with estrogen and mortality. It combines multiple clinical and pathologic
receptorpositive disease.7,9 Substantial reductions were factors and produces estimates for recurrence and death
observed in the absolute risk of recurrence and cancer- from early-stage breast cancer. It incorporates the effect of
specific mortality of 12.3% and 9.2% for patients younger comorbidities in the determination of prognosis and benefit
than age 50 and 8.6% and 6.1% for patients age 5069, from different therapeutic strategies.13 However, there are
respectively. data suggesting that it generally overestimates prognosis
and it lacks some crucial elements such as HER2 status.
The guidelines described above and Adjuvant! Online can-
not reveal the substantial heterogeneity that exists between
KEY POINTS patients with similar stages and grades of disease. Several
independent groups have conducted comprehensive gene
In the past, breast cancer was perceived to be one expression profiling studies using microarray technology to
disease with different anatomical extents and different develop new tools that are more effective at an individual
expressions of estrogen and progesterone receptors and level and that can assess risk of relapse to improve decision
HER2. making for adjuvant treatment.
Gene expression analysis has revealed distinct intrinsic
subtypes of breast cancer with different natural
behaviors and responses to treatment.
First-generation gene signatures have the common INTRINSIC MOLECULAR SUBTYPING
ability to further subdivide estrogen receptorpositive Over 10 years ago, Perou et al14 and Sorlie et al15 dem-
HER2-negative breast cancers into subgroups that differ onstrated that estrogen receptorpositive and estrogen
in regards to their expression of proliferation-related receptornegative breast cancers are completely distinct
genes. Additional biomarkers, particularly those with diseases on a molecular level. In addition, hierarchical
predictive value, are urgently needed. cluster analyses of genes that vary more between tumors
Classic clinicopathologic tools and new molecular than between repeated samples of the same tumor, the so-
tools should be integrated in and complementary to called intrinsic genes, revealed at least four molecular
adjuvant treatment decision making for individual subtypes of breast cancer, namely, luminal, HER2-enriched,
patients with breast cancer, in particular for cases where
basal-like, and normal breastlike.14 Multiple independent
the benefit of chemotherapy is uncertain.
Treatment of patients with low-risk tumor biology
datasets have shown the prognostic effect of intrinsic sub-
and high tumor burden is challenging since, despite typing of breast cancer with luminal A cases (high expression of
being at considerable high risk for distant relapse, the estrogen receptor and estrogen receptorregulated genes) to
expected benefit of chemotherapy might not be enough be at low risk of early recurrence. Apart from its prognostic
in terms of the low proliferative biology. implication, the intrinsic molecular subclassification seems to
predict responsiveness to chemotherapy.16 Studies evaluating

e32 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


TAILORING CHEMOTHERAPY IN EARLY-STAGE BREAST CANCER

the association between molecular subtype and path- of common clinicopathologic features traditionally used
ologic complete response (pCR) rate to preoperative for determining prognosis of early-stage breast cancer;
(neoadjuvant) chemotherapy have reported the highest important meta-analyses have shown that they com-
rate of pCR in basal-like (30%45%) and HER2-enriched plement but do not replace traditional factors since both
(33%55%) subtypes, while pCR rates for luminal B tumor size and nodal status provide independent prog-
(1%15%) and luminal A disease (0%7%) were substantially nostic information in multivariate analyses.25 In addition,
lower. the accuracy of the outcome prediction of all first-
It is important to highlight that molecular classifica- generation prognostic signatures is time-dependent,
tion is not without its inherent limitations because up with more accurate prediction during the first 5 years
to 30% of breast cancers do not fit into any of the four after diagnosis and less accurate predictions for late
molecular categories. 17 The exact number of true mo- relapses.26 Finally, there is some evidence that the
lecular subclasses of breast cancer is currently unknown, prognostic information provided by first-generation
and it is expected that the molecular classification will signatures may be comparable with that offered by
further evolve with new technological platforms and semiquantitative and centralized (high-quality) assess-
improved understanding of tumor biology. In particular, ment of estrogen and progesterone receptors, HER2, and
the immunohistochemistry-defined triple-negative sub- Ki-67.27
type is currently being subdivided into several molecularly
distinct subtypes with potential future clinical and thera-
peutic implications.18 70-Gene Profile
The 70-gene profile, also known as MammaPrint, was the
first microarray-based prognostic signature to be approved
PROGNOSTIC GENE EXPRESSION SIGNATURES by the U.S. Food and Drug Administration. The 70-gene
Concomitant to the discovery of heterogeneity of breast profile may be used for determining the prognosis of pa-
cancers, microarray-based gene expression profiling was tients with stage I and/or II, node-negative, invasive breast
used to predict the outcomes of individual patients with cancer28 as well as for nodes 1 to 3 positive disease.29 This
breast cancer and aimed to identify those patients who gene profile was first established using RNA from fresh,
could be safely omitted from adjuvant chemotherapy.19 First frozen tumor tissues; however, from 2012 onward, it is also
attempts were empirical, whereby tumors from patients evaluable in formalin-fixed, paraffin-embedded tumor
with good and poor outcomes were profiled, and collections tissue.28 The assay was based on an empirical microarray
of candidate genes that could discriminate between patients analysis of 78 patients who did not receive adjuvant systemic
with disease of good and poor prognosis were identified.19,20 therapy for their breast cancer. They were younger than age
Later, studies in which a multigene predictor was generated 55, had tumors up to 5 cm, and were node-negative. If these
on the basis of a hypothesis derived from in vivo or in vitro patients developed distant metastases within the first 5
experiments and then applied to breast cancer samples were years after primary diagnosis, they were classified as poor
conducted.21 prognosis or good prognosis in the event there were no
There are many gene-expression prognostic signatures distant metastases during this timeframe. A list of 70 genes
that were used during the past decade.22 The so-called first- that could accurately predict poor versus good prognosis for
generation signatures have several common features. De- these patients was identified by a supervised analysis of
spite the different gene sets that compose each of the 25,000 genes included in the microarrays. Subsequent studies
signatures, they characterize a unique population with a high in breast cancer cohorts that were retrospectively accrued
expression of proliferation-related genes associated with demonstrated the potential of the MammaPrint assay to
poor prognosis.23 Because the levels of expression of determine prognosis of both node-negative and node-
proliferation-related genes are usually high in estrogen positive disease29 and also for patients with HER2-positive
receptornegative disease, these signatures almost always disease.30 The prognostic subgroups, which were identified
classify estrogen receptornegative cancers as being a poor by the MammaPrint assay, correlate sensitivity to che-
prognosis disease. In estrogen receptorpositive breast can- motherapy in general: patients with high-risk signatures
cer, the strongest prognostic factor is the degree of ex- derive the greatest absolute benefit from adjuvant chemo-
pression of proliferation-related genes. Based on these therapy.31 However, it has no ability to differentiate sensi-
features, it seems that most of these signatures generally tivity to different types of chemotherapy regimens. One of
correlate with response to conventional chemotherapy main disadvantages of the MammaPrint assay is its small
regimens,24 that is, high proliferative tumors respond better discriminatory power for patients with estrogen receptor
to chemotherapy; however, they are not predictive of one negative disease (only up to 5% of patients with estrogen
type of chemotherapy agent over another and cannot be receptornegative disease are classified as having good
used to define the best chemotherapy agent/regimen for prognosis disease).32 The true clinical utility of MammaPrint is
each individual patient. being tested in a randomized prospective phase III trial EORTC
In the early years of their development, some defended 10041/BIG 3-04 MINDACT,33 the results of which will be
that these signatures would be a more objective replacement presented in April 2016.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e33


RIBNIKAR AND CARDOSO

Recurrence Score or Oncotype DX 76-Gene Signature


In parallel with the development of microarray-based prog- The 76-gene signature, also known as the Veridex signature,
nostic signatures, Paik and et al34 developed Oncotype DX, a was developed on the basis of a supervised analysis of
quantitative reverse transcription polymerase chain reaction microarray data in a set of 115 breast cancers, of which 80%
based signature that measures the expression of 21 genes were estrogen receptorpositive. Contrary to MammaPrint,
(16 cancer-related genes and five reference genes). It can estrogen receptorpositive and estrogen receptornegative
be performed with RNA extracted from formalin-fixed, cancers were analyzed separately, and this enabled the
paraffin-embedded tissue samples. Oncotype DX calcu- identification of 60 genes that could predict the develop-
lates the recurrence score (RS) to predict the risk of distant ment of distant metastases within 5 years among patients
relapse within 10 years for patients with estrogen receptor with estrogen receptorpositive disease and 16 genes that
positive, lymph nodenegative breast cancers. It was de- could predict distant dissemination in estrogen receptor
veloped on the basis of analysis of clinical samples from the negative disease.42 All 76 genes together were applied to an
National Surgical Adjuvant Breast and Bowel Project (NSABP) independent test set of 171 patients with node-negative
B-20 clinical trial. Later, it was validated with material from disease only. The 76-gene predictor was a strong prognostic
the B-14 trial. The RS is a continuous variable, ranging factor for development of metastases within the first 5 years
from 0 to 100, and is an independent prognostic factor for after diagnosis. The main disadvantages of this signature are
patients with estrogen receptorpositive, node-negative time dependency, requirement of fresh or frozen samples,
breast cancer treated with adjuvant tamoxifen. Patients prognostic power supported by level III evidence only, and,
are classified into three categories, including low risk last but not the least, that the 16-gene signature developed
(RS , 18), intermediate risk (RS 18-31), and high risk (RS . 31), for estrogen receptornegative disease might not predict
which correlate with 10-year relapse rates of 7%, 14%, the outcome of patients with triple-negative breast
and 30%, respectively. The optimal management of the cancer.23
intermediate-risk group is uncertain and is being studied
in the TailorX trial, in which patients with estrogen
Genomic Grade Index
receptorpositive, node-negative breast cancer were assessed
In 2006, Sotiriou et al developed the Genomic Grade Index
for risk of distant relapse after surgery and assigned
(GGI). It was based on the expression of 97 genes that were
to low-risk (RS , 11), intermediate-risk (RS 11-25), and
selected by comparing the gene expression of histologic
high-risk groups (RS . 25). Patients with intermediate-risk
grade 2 tumors with that of grade 1 tumors by means of a
disease were randomly assigned to either endocrine
DNA microarray.43 Importantly, this signature was able to
therapy alone or in combination with chemotherapy. In
divide estrogen receptorpositive grade 2 cancers into grade
2015, the results of the low-risk patients (RS , 11) were
1-like with a low frequency of distant relapse and grade
presented and demonstrated excellent outcome at
3-like with an aggressive clinical behavior that is similar to
5 years with endocrine therapy alone, irrespective of age,
that of histologic grade 3 tumors. Similar to MammaPrint,
tumor size, and grade. The authors concluded that these
GGI also correlates with the benefit from chemotherapy in
patients can be safely treated without chemotherapy.35
general: GGI grade 3 cancers seem to derive higher absolute
The main results of the intermediate-risk score group are
benefit from conventional chemotherapy versus GGI grade 1
eagerly awaited.
tumors.44 The main common limitations of first generation
Further analyses also demonstrated that RS generally
prognostic signatures also apply to GGI.22
correlates with benefit from chemotherapy in estrogen
receptorpositive disease.36 However, it does not provide
predictive value to differentiate between different che- PAM 50
motherapy agents/regimens. It is also not useful to dis- PAM 50, also known as Prosigna, was originally developed
criminate between subgroups of HER2-positive breast for intrinsic subtyping of breast cancer,45 but later it started
cancer because HER2 is one of the 21 genes that constitute to be used to predict recurrence.46 It was developed for
the test. patients receiving adjuvant tamoxifen. Its score, PAM 50
During the last few years, the Oncotype DX assay was ROR (PAM 50 risk of recurrence), is calculated by using the
validated among patients who had up to three positive expression profile of 50 selected genes from four intrin-
lymph nodes37 and for those who are supposed to be treated sic subtypes, a proliferation score (18-gene subset), and
with an aromatase inhibitors.38 pathologic tumor size47; it classifies patients into those at
Although the prognostic effect of the Oncotype DX assay low, intermediate, and high risk of recurrence, but it also
has been extensively validated, there is some evidence provides the score as a continuous variable. It can be used
that RS correlates with some clinicopathologic parame- with formalin-fixed, paraffin-embedded tissue samples and
ters, and, some of them, such as tumor size, nodal status, was approved by the U.S. Food and Drug Administration in
and even histologic grade, remain independent of RS.39,40 2013. An important finding was the ability of the PAM 50
Hence, a model that combines RS with traditional ana- ROR score to effectively divide patients into the three risk
tomic pathologic factors may be more prognostic than groups according to risk for recurrence between 5 and 15
RS alone.41 years after the primary diagnosis, using the samples from the

e34 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


TAILORING CHEMOTHERAPY IN EARLY-STAGE BREAST CANCER

Austrian Breast and Colorectal Cancer Study Group 8 trial DX, and wound response models. Of the five models that Fan
(ABCSG-8). It is, therefore, a more useful signature to et al analyzed in their study, only the two-gene ratio (H/I)
predict late distant relapses compared with Oncotype DX failed to identify substantial differences in outcome within
or MammaPrint. that data set.
There are two additional reports evaluating the accu-
Breast Cancer Index racy of prediction of BCI among patients with estrogen
The Breast Cancer Index (BCI) is another second-generation receptorpositive, node-negative breast cancer. One report
gene signature that was found to be a prognostic marker for by Reid et al showed that the two-gene model failed to
patients with early-stage estrogen receptorpositive breast detect differences in outcome,53 and the other study,
cancer who have or who have not received tamoxifen. It conducted by Goetz et al, showed that the H/I ratio was a
is a quantitative reverse transcription polymerase chain substantial predictor of relapse-free survival and disease-
reactionbased method that measures the expression of free survival.54 The possible explanation for these contra-
two genes, HOXB13 and IL17BR (H/I), and classifies patients dictory findings is that a model based on the analysis of only
into three recurrence risk categories (low, intermediate, and two genes is much more likely to be sensitive to technical
high).48 The BCI was found to be useful for the assessment of differences in analysis platforms than one based on many
both early and late distant recurrence, according to the genes.
Stockholm study. This study included 317 patients with Findings from multiple analyses incorporating first- and
estrogen receptorpositive node-negative disease, treated second-generation gene signatures among patients with
with adjuvant tamoxifen only, and showed that recurrence early-stage breast cancer suggest that even though there is
rates for the low-risk group based on the BCI were very low, very little gene overlap and different algorithms are used,
not only in the first 5 years after the surgery, but also in years the prediction of outcome for the majority of patients is
5 to 10. similar. One may therefore conclude that, even though
different gene sets are being used as predictors of outcome,
the signatures identify a common set of biologic charac-
EndoPredict teristics that allow for a good distinction between different
The most recently developed second-generation gene sig- subgroups of patients with breast cancer who have distinct
nature is EndoPredict. Just like the majority of first- and prognoses.
second-generation signatures, it is also used for estrogen
receptorpositive and HER2-negative breast cancers. It was
developed based on the analysis of 964 breast cancer GENE EXPRESSION SIGNATURES AND RESPONSE
samples, from which eight genes and four control genes TO CHEMOTHERAPY
were selected. The expression of these 12 genes is analyzed The predictive value of genomic signatures has been eval-
with the quantitative reverse transcription polymerase chain uated mostly in the neoadjuvant setting. Several small
reactionbased method for the classification of patients into studies suggested that the gene expression profiles of
two recurrence risk groups.49 The assay was validated in the cancers that are highly sensitive to chemotherapy differ
ABCSG-6 and ABCSG-8 trials that included patients with from those of less responsive tumors. For example, Ayers
estrogen receptorpositive, HER2-negative, lymph node et al conducted a prospectively designed study in which
positive or negative breast cancers who were treated with needle-biopsy samples from 133 patients with stage I, II, and
adjuvant tamoxifen only.50 Additionally, EndoPredictClin, III breast cancers were collected before systemic therapy.55
which combines the EndoPredict score with tumor size and Patients were then treated with preoperative chemotherapy
nodal status in a linear model, identified a subgroup of with weekly paclitaxel and a combination of fluorouracil,
patients with an excellent long-term prognosis after a doxorubicin, and cyclophosphamide. A 30-gene predictor
standard 5 years of endocrine therapy,51 thus making it was developed from the data from the first 82 patients, and
useful for evaluating risk of late relapse. it was shown to have a higher sensitivity for prediction of
pCR than a clinical predictor that included age, nuclear
grade, and estrogen receptor status (92% vs. 61%). These
CONCORDANCE AMONG GENE results need further validation and confirmation in in-
EXPRESSIONBASED PREDICTORS FOR BREAST dependent and larger studies.
CANCER Currently no multigene predictor is ready to be used in
Fan et al analyzed a single data set of 295 tumor samples and
clinical practice with the aim of predicting response to
applied five gene expressionbased models including in-
specific chemotherapy agents or regimens.
trinsic subtypes, MammaPrint assay, wound response,
Oncotype DX, and the two-gene ratio (BCI) for patients who
had been treated with tamoxifen.52 Four of these models CONCLUSION
were similar in prediction for particular risk groups. For Undoubtedly, the era of molecular oncology has brought a
instance, tumors classified as HER2-enriched, basal-like, and deeper insight into the complex biology of breast cancer
luminal Blike by intrinsic subtyping were almost all clas- and its crucial clinical and therapeutic implications. A new
sified as having a poor outcome by MammaPrint, Oncotype molecular-based classification of breast cancer exists, going

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e35


RIBNIKAR AND CARDOSO

TABLE 1. Most Common Commercially Available Prognostic Gene Signatures for Breast Cancer
MammaPrint Oncotype DX Breast Cancer Index Mapquant DX PAM 50 ROR EndoPredict
Provider Agendia Genomic Health Biotheranostics Ipsogen NanoString Sividon
Type of 70-gene assay 21-gene 2-gene ratio (H/I) Genomic grade 50-gene assay 12-gene assay
Assay recurrence score and molecular
grade index
Type of Fresh or frozen FFPE FFPE Fresh or frozen FFPE FFPE
Sample or FFPE or FFPE
Technique DNA microarray qRT-PCR qRT-PCR DNA microarray qRT-PCR qRT-PCR
or qRT-PCR or qRT-PCR
Clinical Prognosis of Prediction of Prognostic in ER+, Molecular Originally for Recurrence
Application N0, , 5 cm, recurrence risk prediction of grading for intrinsic subtyping, prediction for
stage I/II, in ER+ and N0 response to TAM ER+, histologic recurrence prediction ER+ HER2
age , 61 treated with TAM grade II disease
Results Dichotomous, Continuous variable Continuous variable Dichotomous, Continuous variable Dichotomous, low
Presentation good or poor GGI I or GGI III or high risk
prognosis
Level of II I III III I I
Evidence
FDA Approval YES NO NO NO YES NO
Abbreviations: ER+, estrogen receptorpositive; FDA, U.S. Food and Drug Administration; FFPE, formalin-fixed, paraffin-embedded; GGI, Genomic Grade Index; qRT-PCR, quantitative
reverse transcription polymerase chain reaction; TAM, tamoxifen.

beyond traditional morphologic oncology. It is, however, While we wait for the results of the large prospective ran-
indispensable to use the new molecular tools in conjunction domized trials, MINDACT, TailorX, RxPONDER, and OPTIMA,
with classic clinicopathologic parameters because the latter all major international guidelines recommend the use of
retain their independent prognostic value for early-stage genomic tools in cases for which the potential benefit of
breast cancer in multivariate models. adjuvant chemotherapy is difficult to estimate based only
All available tools have advantages and limitations, both of using on clinicopathologic factors.
which must be discussed with patients. First-generation Currently, one of the greatest challenges for the medical
prognostic gene signatures consistently demonstrate that oncologist is determining the treatment strategy for patients
about half of the patients with estrogen receptorpositive, with favorable tumor biology and high tumor burden. These
HER2-negative disease are at low risk for early distant re- patients are at substantial risk for distant relapse with en-
lapse and are likely to obtain minimal, if any, benefit from docrine therapy alone.37,56 However, they do not seem to
adjuvant chemotherapy, but do not provide information derive a substantial benefit from adjuvant chemotherapy
about late relapses. Novel second-generation multigene and are at risk for the acute and long-term toxicities of this
prognosticators have some additional advantages, such as a therapy.
better prediction of late distant relapses, a common situation In summary, adjuvant treatment decision making in
in estrogen receptorpositive, HER2-negative breast cancer. breast cancer involves an integration of the available
Combination tools, such as EndoPredictClin and Prosigna, may clinicopathologic factors and new genomic tools, when-
be even more accurate in determining short- and long-term ever appropriate, as well as a detailed and compre-
prognoses for this subgroup of breast cancer. Therefore, hensible discussion with each individual patient regarding
tumor biology and tumor burden must be complementary risk of recurrence, risk of adverse events, comorbidities,
and not mutually exclusive, so that more individualized ad- performance status, and, very importantly, patient
juvant treatment decision making can occur (Table 1). preferences.

References
1. Ravdin PM, Siminoff LA, Davis GJ, et al. Computer program to assist in 4. Jackisch C, Harbeck N, Huober J, et al. 14th St. Gallen International
making decisions about adjuvant therapy for women with early breast Breast Cancer Conference 2015: evidence, controversies, consensus -
cancer. J Clin Oncol. 2001;19:980-991. primary therapy of early breast cancer: opinions expressed by German
2. Carlson RW, Anderson BO, Burstein HJ, et al. Invasive breast cancer. experts. Breast Care (Basel). 2015;10:211-219.
J Natl Compr Canc Netw. 2007;5:246-312. 5. Early Breast Cancer Trialists Collaborative Group. Effects of chemo-
3. Senkus E, Kyriakides S, Ohno S, et al; ESMO Guidelines Committee. therapy and hormonal therapy for early breast cancer on recurrence
Primary breast cancer: ESMO Clinical Practice Guidelines for diagnosis, and 15-year survival: an overview of the randomized trials. Lancet.
treatment and follow-up. Ann Oncol. 2015;26(Suppl 5):v8-v30. 2005;365:1687-1717.

e36 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


TAILORING CHEMOTHERAPY IN EARLY-STAGE BREAST CANCER

6. Early Breast Cancer Trialists Collaborative Group. Polychemotherapy for 27. Cuzick J, Dowsett M, Pineda S, et al. Prognostic value of a combined
early breast cancer: an overview of the randomised trials. Lancet. 1998; estrogen receptor, progesterone receptor, Ki-67 and human epidermal
352:930-942. growth factor receptor 2 immunohistochemical score and comparison
7. Early Breast Cancer Trialists Collaborative Group. Effects of chemo- with the genomic health recurrence score in early breast cancer. J Clin
therapy and hormonal therapy for early breast cancer on recurrence Oncol. 2011;29:4273-4278.
and 15-year survival: an overview of the randomised trials. Lancet. 28. Sapino A, Roepman P, Linn SC, et al. MammaPrint molecular diagnostics
2005;365:1687-1717. on formalin-fixed, paraffin-embedded tissue. J Mol Diagn. 2014;16:
8. Clarke M. Meta-analyses of adjuvant therapies for women with early 190-197.
breast cancer: the Early Breast Cancer Trialists Collaborative Group 29. Mook S, Schmidt MK, Viale G, et al; TRANSBIG Consortium. The 70-gene
overview. Ann Oncol. 2006;17(Suppl 10):x59-x62. prognosis-signature predicts disease outcome in breast cancer patients
9. Berry DA, Cirrincione C, Henderson IC, et al. Estrogen-receptor status with 1-3 positive lymph nodes in an independent validation study.
and outcomes of modern chemotherapy for patients with node-positive Breast Cancer Res Treat. 2009;116:295-302.
breast cancer. JAMA. 2006;295:1658-1667. 30. Knauer M, Cardoso F, Wesseling J, et al. Identification of a low-risk
10. Kelly CM, Hortobagyi GN. Adjuvant chemotherapy in early-stage breast subgroup of HER-2-positive breast cancer by the 70-gene prognosis
cancer: what, when, and for whom? Surg Oncol Clin N Am. 2010;19: signature. Br J Cancer. 2010;103:1788-1793.
649-668. 31. Knauer M, Mook S, Rutgers EJ, et al. The predictive value of the 70-gene
11. National Comprehensive Cancer Network. NCCN Clinical Practice signature for adjuvant chemotherapy in early breast cancer. Breast
Guidelines in Oncology. Breast Cancer, V.I. 2010. http://www.nccn.org/ Cancer Res Treat. 2010;120:655-661.
professionals/physician_gls/PDF/breast.pdf. Accessed January 19, 2010. 32. Buyse M, Loi S, vant Veer L, et al; TRANSBIG Consortium. Validation and
12. National Institutes of Health Consensus Development Panel. Adjuvant clinical utility of a 70-gene prognostic signature for women with node-
therapy for breast cancer. NIH Consens Statement. 2000;17:1-35. negative breast cancer. J Natl Cancer Inst. 2006;98:1183-1192.
13. Olivotto IA, Bajdik CD, Ravdin PM, et al. Population-based validation of 33. Bogaerts J, Cardoso F, Buyse M, et al; TRANSBIG consortium. Gene
the prognostic model ADJUVANT! for early breast cancer. J Clin Oncol. signature evaluation as a prognostic tool: challenges in the design of the
2005;23:2716-2725. MINDACT trial. Nat Clin Pract Oncol. 2006;3:540-551.
14. Perou CM, Srlie T, Eisen MB, et al. Molecular portraits of human breast 34. Paik S, Shak S, Tang G, et al. A multigene assay to predict recurrence of
tumours. Nature. 2000;406:747-752. tamoxifen-treated, node-negative breast cancer. N Engl J Med. 2004;
15. Srlie T, Perou CM, Tibshirani R, et al. Gene expression patterns of 351:2817-2826.
breast carcinomas distinguish tumor subclasses with clinical implica- 35. Sparano JA, Gray RJ, Makower DF, et al. Prospective validation of a 21-
tions. Proc Natl Acad Sci USA. 2001;98:10869-10874. gene expression assay in breast cancer. N Engl J Med. 2015;373:2005-
16. Hu Z, Fan C, Oh DS, et al. The molecular portraits of breast tumors are 2014.
conserved across microarray platforms. BMC Genomics. 2006;7:96. 36. Paik S, Tang G, Shak S, et al. Gene expression and benefit of chemo-
17. Pusztai L, Mazouni C, Anderson K, et al. Molecular classification of therapy in women with node-negative, estrogen receptor-positive
breast cancer: limitations and potential. Oncologist. 2006;11:868-877. breast cancer. J Clin Oncol. 2006;24:3726-3734.
18. Curtis C, Shah PS, Chin SF, et al. The genomic and transcriptomic ar- 37. Albain KS, Barlow WE, Shak S, et al; Breast Cancer Intergroup of North
chitecture of 2,000 breast tumours reveals novel subgroups. Nature. America. Prognostic and predictive value of the 21-gene recurrence
2012;486:346-352. score assay in postmenopausal women with node-positive, oestrogen-
19. Weigelt B, Baehner FL, Reis-Filho JS. The contribution of gene ex- receptor-positive breast cancer on chemotherapy: a retrospective
pression profiling to breast cancer classification, prognostication and analysis of a randomised trial. Lancet Oncol. 2010;11:55-65.
prediction: a retrospective of the last decade. J Pathol. 2010;220: 38. Dowsett M, Cuzick J, Wale C, et al. Prediction of risk of distant re-
263-280. currence using the 21-gene recurrence score in node-negative and
20. Van;t Veer LJ, Dai H, van de Vijver MJ, et al. Gene expression profiling node-positive postmenopausal patients with breast cancer treated with
predicts clinical outcome of breast cancer. Nature. 2002;415:530- anastrozole or tamoxifen: a TransATAC study. J Clin Oncol. 2010;28:
536. 1829-1834.
21. Kim C, Paik S. Gene-expression-based prognostic assays for breast 39. Habel LA, Shak S, Jacobs MK, et al. A population-based study of tumor
cancer. Nat Rev Clin Oncol. 2010;7:340-347. gene expression and risk of breast cancer death among lymph node-
22. Sotiriou C, Pusztai L. Gene-expression signatures in breast cancer. negative patients. Breast Cancer Res. 2006;8:R25.
N Engl J Med. 2009;360:790-800. 40. Tang G, Shak S, Paik S, et al. Comparison of the prognostic and predictive
23. Wirapati P, Sotiriou C, Kunkel S, et al. Meta-analysis of gene expression utilities of the 21-gene Recurrence Score assay and Adjuvant! for
profiles in breast cancer: toward a unified understanding of breast women with node-negative, ER-positive breast cancer: results from
cancer subtyping and prognosis signatures. Breast Cancer Res. 2008;10: NSABP B-14 and NSABP B-20. Breast Cancer Res Treat. 2011;127:
R65. 133-142.
24. Iwamoto T, Bianchini G, Booser D, et al. Gene pathways associated with 41. Tang G, Cuzick J, Wale C, et al. Recurrence risk of node-negative and ER-
prognosis and chemotherapy sensitivity in molecular subtypes of breast positive early-stage breast cancer patients by combining recurrence
cancer. J Natl Cancer Inst. 2011;103:264-272. score, pathologic and clinical information: a meta-analysis approach. J
25. Desmedt C, Haibe-Kains B, Wirapati P, et al. Biological processes as- Clin Oncol. 2010;28 (suppl; abstr 509).
sociated with breast cancer clinical outcome depend on the molecular 42. Wang Y, Klijn JG, Zhang Y, et al. Gene-expression profiles to predict
subtypes. Clin Cancer Res. 2008;14:5158-5165. distant metastasis of lymph-node-negative primary breast cancer.
26. Desmedt C, Piette F, Loi S, et al; TRANSBIG Consortium. Strong time Lancet. 2005;365:671-679.
dependence of the 76-gene prognostic signature for node-negative 43. Sotiriou C, Wirapati P, Loi S, et al. Gene expression profiling in breast
breast cancer patients in the TRANSBIG multicenter independent cancer: understanding the molecular basis of histologic grade to im-
validation series. Clin Cancer Res. 2007;13:3207-3214. prove prognosis. J Natl Cancer Inst. 2006;98:262-272.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e37


RIBNIKAR AND CARDOSO

44. Liedtke C, Hatzis C, Symmans WF, et al. Genomic grade index is as- patients with endocrine-responsive early breast cancer from the
sociated with response to chemotherapy in patients with breast cancer. Austrian Breast and Colorectal Cancer Study Group. J Clin Oncol. 2012;
J Clin Oncol. 2009;27:3185-3191. 30:722-728.
45. Parker JS, Mullins M, Cheang MC, et al. Supervised risk predictor of breast 51. Dubsky P, Brase JC, Jakesz R, et al; Austrian Breast and Colorectal Cancer
cancer based on intrinsic subtypes. J Clin Oncol. 2009;27:1160-1167. Study Group (ABCSG). The EndoPredict score provides prognostic in-
46. Gnant M, Filipits M, Greil R, et al; Austrian Breast and Colorectal Cancer formation on late distant metastases in ER+/HER2- breast cancer pa-
Study Group. Predicting distant recurrence in receptor-positive breast tients. Br J Cancer. 2013;109:2959-2964.
cancer patients with limited clinicopathological risk: using the PAM50 52. Fan C, Oh DS, Wessels L, et al. Concordance among gene-expression-
Risk of Recurrence score in 1478 postmenopausal patients of the based predictors for breast cancer. N Engl J Med. 2006;355:560-569.
ABCSG-8 trial treated with adjuvant endocrine therapy alone. Ann 53. Reid JF, Lusa L, De Cecco L, et al. Limits of predictive models using
Oncol. 2014;25:339-345. microarray data for breast cancer clinical treatment outcome. J Natl
47. Filipits M, Nielsen TO, Rudas M, et al; Austrian Breast and Colorectal Cancer Inst. 2005;97:927-930.
Cancer Study Group. The PAM50 risk-of-recurrence score predicts risk 54. Goetz MP, Suman VJ, Ingle JN, et al. A two-gene expression ratio of
for late distant recurrence after endocrine therapy in postmenopausal homeobox 13 and interleukin-17B receptor for prediction of recurrence
women with endocrine-responsive early breast cancer. Clin Cancer Res. and survival in women receiving adjuvant tamoxifen. Clin Cancer Res.
2014;20:1298-1305. 2006;12:2080-2087.
48. Zhang Y, Schnabel CA, Schroeder BE, et al. Breast cancer index identifies 55. Ayers M, Symmans WF, Stec J, et al. Gene expression profiles predict
early-stage estrogen receptor-positive breast cancer patients at risk for complete pathologic response to neoadjuvant paclitaxel and fluoro-
early- and late-distant recurrence. Clin Cancer Res. 2013;19:4196-4205. uracil, doxorubicin, and cyclophosphamide chemotherapy in breast
49. Filipits M, Rudas M, Jakesz R, et al; EP Investigators. A new molecular cancer. J Clin Oncol. 2004;22:2284-2293.
predictor of distant recurrence in ER-positive, HER2-negative breast 56. Saghatchian M, Mook S, Pruneri G, et al. Combining genomic profiling
cancer adds independent information to conventional clinical risk (70-gene MammaPrint) with nodal status allows to classify patients
factors. Clin Cancer Res. 2011;17:6012-6020. with primary breast cancer and positive lymph nodes (1-9) into very
50. Dubsky PC, Jakesz R, Mlineritsch B, et al. Tamoxifen and anastrozole as a distinct prognostic subgroups that could help tailor treatment strate-
sequencing strategy: a randomized controlled trial in postmenopausal gies. Cancer Res. 2009;69:a102.

e38 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


BREAST CANCER

Targeted Therapies in Hormone


ReceptorPositive Breast Cancer

CHAIR
Hope S. Rugo, MD
University of California, San Francisco
San Francisco, CA

SPEAKERS
Cynthia Ma, MD, PhD
Washington University School of Medicine
St. Louis, MO

Dejan Juric, MD
Massachusetts General Hospital Cancer Center
Boston, MA
RUGO, VIDULA, AND MA

Improving Response to Hormone Therapy in Breast Cancer:


New Targets, New Therapeutic Options
Hope S. Rugo, MD, Neelima Vidula, MD, and Cynthia Ma, MD, PhD

OVERVIEW

The majority of breast cancer expresses the estrogen and or progesterone receptors (ER and PR). In tumors without
concomitant HER2 amplification, hormone therapy is a major treatment option for all disease stages. Resistance to
hormonal therapy is associated with disease recurrence and progression. Recent studies have identified a number of
resistance mechanisms leading to estrogen-independent growth of hormone receptorpositive (HR+) breast cancer as a
result of genetic and epigenetic alterations, which could be exploited as novel therapeutic targets. These include acquired
mutations in ER-alpha (ESR1) in response to endocrine deprivation; constitutive activation of cyclin-dependent kinases (CDK)
4 and 6; cross talk between ER and growth factor receptor signaling such as HER family members, fibroblast growth factor
receptor (FGFR) pathways, intracellular growth, and survival signals PI3K/Akt/mTOR; and epigenetic modifications by
histone deacetylase (HDAC) as well as interactions with tumor microenvironment and host immune response. Inhibitors of
these pathways are being developed to improve efficacy of hormonal therapy for treatment of both metastatic and early-
stage disease. Two agents are currently approved in the United States for the treatment of metastatic HR+ breast cancer,
including the mTOR inhibitor everolimus and the CDK4/6 inhibitor palbociclib. Management of toxicity is a critical aspect of
treatment; the primary toxicity of everolimus is stomatitis (treated with topical steroids) and of palbociclib is neutropenia
(treated with dose reduction/delay). Many agents are in clinical trials, primarily in combination with hormone therapy; novel
combinations are under active investigation.

T he estrogen-dependent nature of breast cancer has been


well established since the historical observation that
removal of the ovaries was effective in treating breast tumors.1
CDK4/6, and epigenetic and immune checkpoints as resis-
tance mechanisms and therapeutic targets in ER+ breast
cancer (Fig. 1).
A number of pharmacological agents, referred to as endocrine
therapy, are now available in the clinic. These include aro- UNDERSTANDING THE BIOLOGY OF HORMONE
matase inhibitors (AI) and gonadotropin-releasing hormone RECEPTORS AND POTENTIAL MECHANISMS OF
(GnRH) agonists that reduce estrogen biosynthesis, the RESISTANCE
selective estrogen receptor modulators (SERM) tamoxifen, Mechanisms of Action of Estrogen Receptors
and the selective ER down regulator (SERD) fulvestrant. The Estrogen receptors belong to the family of steroid hormone
clinical application of these agents has led to substantial receptors and its main mechanism of action is a transcription
improvements in survival outcomes for patients with ER+ factor (Fig. 2). There are two different forms of ER, ER-alpha
breast cancer.2 However, despite standard therapy, over and ER-beta, encoded by ESR1 and ESR2, respectively. There
20% of patients with early-stage disease experience relapse, is substantial sequence homology between ER-alpha and
and, essentially, all patients with metastatic disease suc- ER-beta, although the function of ER-beta is less known.
cumb to their illness.2,3 Although the mechanisms of en- Upon activation by estrogen, the cytosolic ER forms dimers
docrine resistance are not fully understood, significant and translocates to the nucleus where ER binds directly to
progress has been made in recent years in uncovering the estrogen response elements (ERE) or is tethered to
several key molecular pathways that promote ligand- the promoter regions of target genes via its interaction
independent activation of ER and tumor growth. In this with SP1. 4 Additional coregulators are then recruited
article, we will focus on evidence for ER-alpha (ESR1) mu- to the complex to regulate the transcription levels of
tation, growth factor receptor signaling, PI3K/Akt/mTOR, target genes, including cyclin D, to promote cell cycle

From the UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA; University of San Francisco School of Medicine, San Francisco, CA; Department of Medicine,
Washington University School of Medicine in St. Louis, St. Louis, MO.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Hope S. Rugo, MD, UCSF Helen Diller Family Comprehensive Cancer Center, 1600 Divisadero St., San Francisco, CA 94115; email: hope.rugo@ucsf.edu.

2016 by American Society of Clinical Oncology.

e40 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


IMPROVING RESPONSE TO HORMONE THERAPY

FIGURE 1. Key Pathways of Endocrine Resistance

Pink and blue color code activating and suppression signals, respectively, for pathway activation.
Abbreviations: HDAC, histone deacetylase; RTK, receptor tyrosine kinase.

progression.4,5 In addition to this classic genomic action of leading to estrogen-independent cell proliferation and resistance
ER, growth factor receptor tyrosine kinases and a variety of to endocrine therapy.6-10
proximal signaling molecules, such as G protein, Ras, Src,
PI3K, and Shc, could activate ER in the absence of estrogen,
ESR1 Mutation
Although initially discovered in the 1990s, the ESR1 mutation
was largely overlooked until recent years when recurrent,
rather than primary, breast cancers were studied. In contrast
KEY POINTS
to the extremely low incidence in treatment-naive primary
Resistance to endocrine therapy in HR+ breast cancer is breast cancers, mutations in ESR1 have been reported in
associated with diverse molecular mechanisms, 11%55% of recurrent or metastatic cancers that progressed
including acquired mutations in ESR1 in response to after long-term endocrine therapy (Fig. 3).11-14 The hot-spot
endocrine deprivation, constitutive activation of CDK4/6, mutations cluster in the ligand-binding-domain of ER, which
cross talk between the ER and growth factor receptor induce a constitutive agonist conformation. Compared with
signaling, epigenetic modifications by HDAC, and the wild-type ER, mutant ER is resistant to estrogen dep-
interactions with tumor microenvironment and host rivation and much less responsive to tamoxifen or fulves-
immune response. trant. Successful treatment of ESR1-mutant breast cancer
Increased understanding of endocrine resistance
therefore requires the development of newer generation
mechanisms has led to the development of targeted
SERMs or SERDs, or other strategies, that are effective in
agents, including two agents which are approved for
clinical use in the United States, that have improved targeting the mutated ER.
progression-free survival in combination with standard
hormone agents.
PI3K is the most commonly altered pathway in HR+ HER2 Gene Amplification
breast cancer; multiple agents that target this pathway HER family members, including HER1 (EGFR), HER2, HER3,
or related growth factor receptors are being studied. To and HER4, are tyrosine kinase receptors important in
date, markers of PI3K pathway activation have not promoting cell growth and survival.15 Cross talk between
identified a specific population most likely to benefit ER and HER family members has been well recognized in
from this therapy. preclinical and clinical studies. HER2 amplification occurs
Toxicities of targeted therapies and impact on quality of in about 10% of ER+ breast cancers and is an established
life must be taken into account and managed mechanism of endocrine resistance.16 Compared with ER+
expectantly when treating patients; common toxicities HER2- disease, ER+ HER2+ breast cancer is associated with a
are stomatitis (everolimus) and neutropenia
higher risk of relapse on adjuvant endocrine therapy17 and
(palbociclib).
Ongoing trials are evaluating inhibitors of the PI3K
incomplete cell cycle arrest to neoadjuvant endocrine
pathway, CDK4/6, and HDAC, primarily in combination therapy.18 Adjuvant trastuzumab reduces relapse in ER+
with hormone therapy in the metastatic, adjuvant, and HER2+ breast cancer and is the standard of care.19 In the
neoadjuvant settings; combination therapy targeting metastatic setting, hormonal therapy in combination with
two or more pathways is also under investigation. HER2-targeted agents provides an alternative to chemo-
therapy regimens.20-22

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e41


RUGO, VIDULA, AND MA

FIGURE 2. Estrogen Receptor Pathway and Cross Talk With Growth Factor Receptor, PI3K/AKT/mTOR, and
CDK4/6 Pathways

Estrogen-bound estrogen receptor (ER), in complex with coactivators (CoA) or corepressors (CoR), regulates target gene expression via the estrogen response element (ERE)
or the AP1 binding sites via its interaction with other transcription factors. Additionally, ER can interact with receptor tyrosine kinases (FGFR, EGFR/HER), which promote
estrogen-independent ER phosphorylation through pathways such as PI3K/Akt/mTOR and MAPK (a and b). The G1 to S phase transition is controlled by CDK4/6, which is
activated by the ER downstream target cyclin D. Constitutive activation of CDK4/6 is associated with endocrine resistance.

HER2 Mutation interaction with other HER family members. In preclinical


Somatic mutations in HER2 in otherwise HER2-negative models, many of these mutations have shown to be on-
breast cancer have attracted much attention in recent years. cogenic and are sensitive to treatment with the irreversible
Although the overall HER2 mutation rate is approximately tyrosine kinase inhibitor of HER1/HER2, neratinib.23 Results
2% in primary breast cancers,23 it reaches over 20% in in- of the breast cancer cohort of the SUMMIT study, a mul-
vasive lobular cancers.24 These mutations cluster in the ticenter, open-label, multihistology phase II basket trial of
kinase domain and the extracellular domain important for its neratinib for patients with HER2-mutant, nonamplified,

e42 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


IMPROVING RESPONSE TO HORMONE THERAPY

FIGURE 3. ESR1 Mutation as an Acquired Mechanism survival signals, and acquired endocrine resistance is often
of Endocrine Resistance accompanied by upregulation of PI3K pathway signaling.29-32
Gain of function mutations in the alpha catalytic subunit of PI3
kinase (PIK3CA) occurs at a frequency of 30%40% and is the
most frequent genetic abnormality in ER+ breast cancer.33,34
A majority of the mutations, including the three hot-spot
mutations E542K, E545K, and H1047R, are missense acti-
vating mutations that cluster in the evolutionarily conserved
accessary domain and the kinase domain.35 In preclinical
models, ER+ breast cancer carrying PIK3CA mutations was
highly dependent on p110 alpha for cell survival.31,36
Rapalogs are among the first agents introduced in the
clinic that indirectly inhibit the activity of mTOR complex 1
(mTOC1) through its interaction with FKBP12.37 Everolimus
The incidence of ESR1 mutations increases dramatically after treatment with long- is now approved for AI-resistant metastatic breast cancer.
term endocrine therapy for recurrent or metastatic ER+ disease. The hot-spot ligand-
binding domain mutations induce a constitutive agonist conformation of ER, leading
However, inhibition of mTORC1 has the potential to upre-
to estrogen independence. gulate Akt activity due to the negative feedback loop be-
Abbreviations: AF1, activation function 1; DBD, DNA-binding domain; ER, estrogen
receptor; LBD, ligand-binding domain. tween the downstream S6K and the upstream PI3K.38,39
Direct kinase inhibitors against both mTORC1 and mTORC2,
as well as inhibitors against Akt and PI3K, are in clinical trials
advanced solid tumors (NCT01953926), was recently re- for more effective pathway inhibition. Promising activity has
ported in an abstract form.25 Among the 19 evaluable pa- been observed in some studies.40-43 However, it has become
tients, objective response was observed in six (overall evident that the efficacy of pan-PI3K inhibitors is limited by
response rate 32%). As a majority of breast cancers with dose-limiting toxicities that include rash, diarrhea, and el-
HER2 mutation are ER+, the combination of fulvestrant and evated transaminases.43,44 Alpha-specific inhibitors are
neratinib is being studied among this patient population likely advantageous in improving the therapeutic window,
(NCT01670877). but resistance mechanisms through acquiring mutations in
PTEN, leading to growth dependence on PI3K-beta, have
Fibroblast Growth Factor Receptors and Ligands been reported in clinical trials of alpelisib, an alpha-specific
Amplification inhibitor.45 Preclinical modeling also indicated that effective
The fibroblast growth factor (FGF) signaling is composed inhibition of mTOR remains important for upstream in-
of 18 ligands that exert their function through four highly hibitors,46 but combined PI3K and mTOR inhibition may not
conserved transmembrane tyrosine kinase receptors be feasible due to overlapping toxicities. There has been
(FGFR1, FGFR2, FGFR3, and FGFR4) to control a wide range significant interest in developing biomarkers of treatment
of biologic functions and regulating cellular proliferation, efficacy, in particular PIK3CA mutation status. However, no
survival, migration, and differentiation.26 FGFR1 ampli- clear association has been identified even with the direct
fication has been identified in 16%27% of luminal B ER+ PI3K inhibitors.40-43,47
breast cancers and associated with increased Ki67, early
relapse, and poor survival.27 Less frequently, the pathway
could be activated by amplification of FGFR2 or different Cyclin D/CDK4/6/Rb Pathway
ligands including FGF3 and FGF4.26 In preclinical studies, The G1 to S phase transition is controlled by CDK4/6, which
FGFR1 amplification enhanced PI3K and MAPK pathway are activated upon binding to D-type cyclins,48-51 leading to
signaling and rendered cancer cells resistance to endocrine phosphorylation of retinoblastoma susceptibility (RB1) gene
therapy, which was reversed by RNAi silencing of FGFR1.27 product (Rb) and the release of the E2F transcription fac-
Dovitinib (TKI258), a first-generation oral tyrosine kinase in- tors.51 There is a strong link between the action of estrogen
hibitor of FGFR1-3, VEGFR, and PDGFR, inhibited proliferation and CDK4/6 activity.52-56 Persistent cyclin D1 expression and
of FGFR1/2-amplifiedbut not FGFRnormalbreast cancer Rb phosphorylation has been associated with resistance to
cell lines, and demonstrated promising antitumor activity for endocrine therapy in ER+ breast cancer.57 The poorer prog-
patients with FGFR1/2/3-amplified breast cancers in a phase II nosis, luminal B ER+ diseases are preferentially enriched with
trial.28 Several FGFR inhibitors are currently being in- gains of CCND1 (cyclin D1; 58% in luminal B vs. 29% in luminal
vestigated in advanced HR+ breast cancer to overcome A) and CDK4 (25% in luminal B vs. 14% in luminal A) and loss of
endocrine resistance. CDKN2A (p16) and CDKN2C (p18), which are negative regu-
lators of CDK4/6.34
In preclinical studies, the CDK4/6 inhibitor palbociclib was
PI3K/Akt/mTOR Pathway preferentially effective in inhibiting the cell proliferation of
The PI3K/Akt/mTOR pathway is a cardinal nodal point in the ER+ cancer cells, including those resistant to anti-estro-
transduction of extracellular and intracellular growth and gens.58 Synergism was observed when combining palbociclib

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e43


RUGO, VIDULA, AND MA

with various anti-estrogens,58,59 and efficacy was demon- complicated because of its functional redundancy with the
strated in patient-derived xenograft models of ER+ breast insulin receptor.75 In addition to mediating endocrine therapy
cancer carrying ESR1 mutations.59 Cells lacking Rb (and resistance, data also support the importance of IGF1R in
hence not dependent on cyclin D1/CDK4/6 for proliferation) mediating treatment resistance to inhibitors against mTOR or
were resistant to palbociclib. Therefore lack of Rb may serve Akt in endocrine-resistant ER+ breast cancer because of the
as a biomarker of resistance to this class of agents. Palbo- negative feedback loop between the downstream target S6K
ciclib in combination with letrozole is now approved as first- and the upstream IGF1R/IRSs/PI3K signaling.38,76 Preclinical
line treatment of postmenopausal women with metastatic data support the clinical investigation for the combined in-
ER+ HER2 breast cancer. Several other CDK4/6 inhibitors hibition of IGF1R/IR and Akt, along with ER targeting.76
are in various stages of clinical development.
Vascular Endothelial Growth Factor
Combining PI3K and CDK4/6 Inhibitors in PIK3CA- In addition to contributing to neoangiogenesis, VEGF has
Mutant Breast Cancer been shown to induce breast cancer cell proliferation and
A drug screen of 42 agents identified the CDK4/6 inhibitor resistance to endocrine therapy via an autocrine mecha-
ribociclib as a strong sensitizer to PI3K inhibition in three ER+ nism.77,78 VEGF and the VEGF receptor have shown to be
breast cancer cell lines with acquired resistance to PI3K overexpressed in breast cancers, and high levels of VEGF
inhibitors.60 In this study, acquired resistance to PI3K in- have been linked to early recurrence and resistance to
hibition was associated with maintained phosphorylation of hormonal therapy.79,80 These provided the preclinical ra-
S6 and Rb. Addition of ribociclib reduced Rb phosphorylation tional for investigating the addition of VEGF inhibitors to
and induced synergist antitumor effect of the PI3K inhibitor, hormonal therapy for the treatment of ER+ breast cancer.
particularly in those with PIK3CA mutation and intact Rb. The The challenge so far has been the difficulty in identifying a
synergism was also observed in the parental cell lines and those target patient population who would derive the most benefit
with de novo resistance to PI3K inhibitors. The impressive an- from these agents.
titumor activity and the nonoverlapping toxicities of these two
classes of agents has led to clinical evaluation of triplet regimens
composed of letrozole, ribociclib, and alpelisib. Microenvironment and Immune Checkpoint Pathway
The immune checkpoint pathway, PD-1, and the PD-L1 path-
Epigenetic Pathways way has been implicated in tumor immune invasion.81 In-
Preclinical studies indicated that epigenetic silencing of hibitors against PD-1 and PD-L1, which disrupt the interaction
ER and deregulation of growth factor receptorpathway between PD-1 on T cells and its ligands PD-L1, have shown
components play an important role in the development of antitumor activity in a variety of tumor types. Approximately
endocrine resistance.61-64 In addition, an array of mutations 4%20% of ER+ breast cancers, perhaps more frequently in the
have been identified in genes that regulate histone and DNA more proliferative luminal B versus luminal A subtypes, have
modification.34 An example is the MLL3 (myeloid/lymphoid been reported to overexpress PD-L1 in cancer and stroma
or mixed-lineage leukemia gene) mutation, which occurred cells.82-84 PD-1 and PD-L1 antibodies are being tested in HR+
in 8% of luminal A and 6% of luminal B breast cancers.34 breast cancer, with preliminary results available.

Insulin-Like Growth Factor/Type 1 Insulin-Like REVERSING HORMONE RESISTANCE IN THE


Growth Factor Signaling CLINIC
The insulin-like growth factor (IGF)/type 1 insulin-like With a greater understanding of the mechanisms of re-
growth factor receptor (IGF1R) signaling has a critical role sistance to hormone therapy, new therapies have emerged
in cell growth, survival, and migration and is required for that hold the potential to overcome hormone resistance and
mammary gland development.65 The IGF ligands, IGF1 and improve response, duration of response, and, hopefully,
IGF2, stimulate cell growth and survival signaling primar- survival. Targeted therapies that are being actively in-
ily via binding to IGF1R and the subsequent activation of vestigated in the clinic include mTOR inhibitors (ever-
the PI3K/Akt and RAS/MAPK pathways.65,66 Extensive bi- olimus), PI3K inhibitors (buparlisib, alpelisib, and taselisib),
directional regulation exists between ER and the IGF1R CDK4/6 inhibitors (palbociclib, ribociclib, and abemaciclib),
pathway.67 ER-alpha is a major regulator of IGF signaling as HDAC inhibitors (entinostat), FGFR inhibitors (dovitinib and
IGF1R and other IGF signaling components are its direct lucitinib), and IGFR inhibitors. Altering the host immune re-
transcription target,68-70 while IGF1R upregulates the tran- sponse with checkpoint inhibition is also being investigated in
scription level of ER-alpha and increases ER phosphorylation via advanced disease. Table 1 summarizes the major ongoing
activation of mTOR/S6K signaling.71 Hyperactivation of IGF1R phase II and III trials with the most-promising novel targeted
and downstream signaling has been associated with the de- agents. Table 2 provides a summary of these agents, their
velopment of endocrine resistance in ER+ breast cancer mechanisms of action, developmental status, and toxicity
preclinical models and patient specimens.65,72-74 However profile. This section provides a more detailed review of these
targeting IGF1R in the endocrine-resistant setting has been emerging therapies, focusing on non-HER2 amplified disease.

e44 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


IMPROVING RESPONSE TO HORMONE THERAPY

Of note, two randomized trials have demonstrated im- 0.45; 95% CI, 0.380.54). Median OS was not significantly
proved progression-free survival (PFS) with the addition of improved at 31 months with everolimus compared with
HER2-targeted agents to AIs in the treatment of metastatic, 26.6 months with placebo (HR 0.89; 95% CI, 0.731.10;
HR, and HER2+ disease.22,85 However, given the demonstrated p = .14).90 A subset analysis of BOLERO-2 demonstrated
improvement in overall survival (OS) with chemotherapy, efficacy of combination therapy among patients with visceral
trastuzumab, and pertuzumab in the first-line metastatic set- metastases and those with nonvisceral disease.91 Based on
ting, this is generally the preferred treatment approach, with the PFS data, everolimus is now being used clinically in
hormone therapy reserved for maintenance in combination combination with exemestane in the metastatic setting.
with HER2-targeted therapy following response. Toxicity is a substantial issue with everolimus, but data
from subsequent trials suggest that physician and patient
education and expectant management reduces common
Inhibition of mTOR toxicity and increases tolerability.
Several randomized trials have demonstrated that inhibition The most common toxicity in BOLERO-2 was stomatitis
of the mTOR pathway can improve duration of response to (56% all grade, aphthous-type ulcers), although the in-
hormone therapy, despite an initial rocky start. The first cidence of grade 3 stomatitis was low (8%), and no grade 4
randomized trial evaluated the efficacy of temsirolimus in events were oberved.92 Stomatitis occurs early, with 80% of
combination with the AI letrozole as first-line therapy for cases occurring in the first 6 weeks of therapy.93 A meta-
metastatic HR+ breast cancer.86 There was no difference in PFS analysis of seven phase III studies of everolimus in different
between the two arms. It may be that the dose and schedule of cancer types evaluated whether the incidence of stomatitis
temsirolimus were not optimal, as stomatitis (a marker of drug correlated with clinical efficacy.94 There was commonality of
exposure) was seen at a lower rate than expected. everolimus-induced stomatitis across solid tumors (66.9% all
Greater success leading to the first approval of a targeted grades of all patients), with 8.6% grade 3/4 stomatitis, and
therapy in combination with hormone therapy in HER2 the incidence of stomatitis correlated with at least as good if
disease was seen with everolimus. A phase II neoadjuvant not better PFS compared with the control population. This
trial was conducted to evaluate efficacy and biomarkers, suggests that stomatitis may be a marker of drug exposure
with the primary endpoint of clinical response.87 Two for everolimus and that dose reductions and delays for this
hundred-seventy postmenopausal women with HR+ early- toxicity do not impact efficacy. A phase II trial evaluated a
stage breast cancer were randomly selected to receive commercially available steroid-based mouthwash among
letrozole plus everolimus or placebo for 4 months prior to patients taking prophylactic exemestane and everolimus to
surgery. Clinical response was higher in the combination arm determine if the incidence and severity of stomatitis might
compared with placebo (68.1% vs. 59.1%; p = .062). More be reduced with this approach.95 Data will be presented at
importantly, a significant decrease in cell proliferation on the 2016 American Society of Clinical Oncology (ASCO)
repeat biopsy performed 2 weeks after treatment was noted Annual Meeting.
in the combination therapy arm compared with placebo Other commonly noted side effects with everolimus in-
(57% vs. 30% decrease; p , .01). These data encouraged clude fatigue, diarrhea, rash, hyperglycemia, pneumonitis,
further investigation of everolimus in combination with weight loss, anemia, and thrombocytopenia. The risk of
hormone therapy. pneumonitis persists over the course of therapy at a low
Two subsequent trials tested everolimus in the metastatic rate, with a 3% rate of grade 3 pneumonitis observed in
setting. The TAMRAD trial was a phase II open-label trial that BOLERO-2. Additional clinical trials with everolimus are on-
randomly selected 111 postmenopausal women whose going, summarized in Table 1. These include studies in the
disease had progressed on an AI to receive tamoxifen and neoadjuvant, adjuvant, and metastatic settings. Results from
everolimus or tamoxifen alone.88 The primary endpoint, BOLERO-6, comparing everolimus plus exemestane with
clinical benefit rate at 6 months, was improved with com- capecitabine, are expected this year.
bination therapy compared with tamoxifen alone (61% vs.
42%; p = .045). Progression-free survival in the combination
therapy arm was significantly longer at 8.6 months versus Inhibition of PI3K
4.5 months (p = .002). Two classes of PI3K inhibitors are being studied in HR+ breast
Finally, regulatory approval of everolimus in combination cancer, the pan-class I inhibitors (buparlisib [BKM120] and
with exemestane for the treatment of metastatic breast pictilisib [GDC0941]) and the alpha-specific inhibitors
cancer progressing on nonsteroidal AIs was based on data (alpelisib [BYL719] and taselisib [GDC0032]).
from the BOLERO-2 trial, a double-blind phase III trial that Results with the pan-PI3K inhibitors have been compli-
randomly selected 724 postmenopausal women in a 2:1 ratio cated by toxicity and disappointing efficacy. The FERGI trial
to receive exemestane with everolimus at 10 mg a day or was a phase II trial that randomly selected 168 women with
placebo.89 At a median follow-up of 18 months, investigator- HR+ postmenopausal metastatic breast cancer previously
assessed PFS (the primary endpoint) was more than doubled treated with an AI to receive fulvestrant with pictilisib or
with the addition of everolimus (7.8 months for everolimus placebo.44 Toxicity was increased in the pictilisib arm; 17% of
compared with 3.2 months for placebo; hazard ratio [HR] patients had at least grade 3 rash and 7% had at least grade 3

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e45


RUGO, VIDULA, AND MA

TABLE 1. Phase II/III Trials of the Novel Targeted Agents Under Development for Hormone ReceptorPositive
Breast Cancer

No.
Trial Novel Agent Primary Endpoint Patients Trial Status
Everolimus: mTOR Inhibitor
Baselga et al87 Everolimus Clinical examination response 270 Completed; response rate of 68.1% in
everolimus/letrozole arm compared with
59.1% in placebo/letrozole arm (p = .062)
GINECO88 Everolimus Clinical benefit rate at 6 months 111 Completed; clinical benefit rate of 61% in
everolimus/tamoxifen arm compared with
42% in tamoxifen arm (p = .045)
BOLERO-289 Everolimus PFS 724 Completed; median PFS of 7.8 months in
everolimus/exemestane arm compared
with 3.2 months in exemestane/placebo
(HR 0.45; 95% CI, 0.380.54)
UNIRAD127 Everolimus Disease-free survival 1,984 Ongoing
SWOG 1207128 Everolimus Invasive disease-free survival using 1,900 Ongoing, recruiting patients
a stratified log-rank test, assessed
up to 10 years
NCT02088684130 Everolimus PFS 46 Ongoing, recruiting patients
BOLERO-4143 Everolimus Percentage of patients progression- 202 Ongoing
free after completion of first-line
treatment
BOLERO-6144 Everolimus PFS 297 Ongoing, recruitment completed
BRE-43145 Everolimus Time to progression 33 Completed
LEO146 Everolimus PFS 137 Ongoing, active recruitment
147
FEVEX Everolimus PFS 745 Not yet open
PrE0102148 Everolimus PFS 130 Ongoing
NCT02236572113 Everolimus Achievement of a PEPI score of 0 66 Ongoing, recruiting patients
following neoadjuvant treatment
with everolimus and an aromatase
inhibitor at 26 weeks
NCT02123823149 BI 836845 (monoclonal antibody PFS 174 Ongoing, recruiting patients
to insulin like growth factor)
and everolimus
Taselisib (GDC0032): Alpha-Specific PI3K Inhibitor
LORELEI150 Taselisib Objective response rate and 330 Ongoing, recruiting patients
pathologic complete response
rate at 16 weeks of treatment
SANDPIPER98 Taselisib PFS 600 Ongoing, recruiting patients
Buparlisib (BKM120) Pan-Class I PI3K Inhibitor
BELLE-296 Buparlisib PFS 1147 Completed; PFS was slightly improved with
buparlisib vs. placebo (6.9 vs. 5 months; HR
0.78; 95% CI, 0.670.89)
BELLE-3151 Buparlisib PFS 420 Ongoing, recruiting patients
Pictlisib (GDC0941): Pan-Class I PI3K Inhibitor
FERGI44 Pictlisib PFS 168 PFS of 6.6 months for pictilisib/fulvestrant
arm compared with 5.1 months in
fulvestrant/placebo arm (HR 0.7; 95% CI,
0.521.06)
Palbociclib: CDK4/6 Inhibitor
PALOMA-1/TRIO- Palbociclib PFS 165 Completed; PFS for palbociclib/letrozole was
1899 20.2 months compared with 10.2 months
in letrozole arm (HR 0.49; 95% CI,
0.3190.748)
PALOMA-2100 Palbociclib PFS 650 Ongoing, closed to accrual
Continued

e46 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


IMPROVING RESPONSE TO HORMONE THERAPY

TABLE 1. Phase II/III Trials of the Novel Targeted Agents Under Development for Hormone ReceptorPositive
Breast Cancer (Contd)
No.
Trial Novel Agent Primary Endpoint Patients Trial Status
PALOMA-3101 Palbociclib PFS 417 Completed; PFS of 9.2 months in palbociclib/
fulvestrant arm vs. 3.8 months in fulves-
trant/placebo arm (HR 0.42; 95% CI,
0.320.56)
PEARL104 Palbociclib PFS 348 Ongoing, recruiting patients
PENELOPEB103 Palbociclib Invasive disease-free survival 800 Ongoing, recruiting patients
PALOMA-4152 Palbociclib PFS 330 Ongoing, recruiting patients
PALOMA-2139 Palbociclib Treatment discontinuation rate 160 Ongoing, not recruiting patients
PALLET153 Palbociclib Change in proliferation rate (Ki67) 306 Ongoing, recruiting patients
at 2 weeks
FLIPPER154 Palbociclib PFS 190 Ongoing, recruiting participants
PARSIFAL155 Palbociclib PFS 304 Ongoing, currently recruiting patients
NCT02384239156 Palbociclib Tumor Progression as measured by 70 Ongoing, currently recruiting patients
RECIST 1.1
NCT02592746157 Palbociclib PFS 122 Ongoing, not yet recruiting patients
Abemaciclib: CDK4/6 Inhibitor
neoMONARCH134 Abemaciclib Change in proliferation rate (Ki67) 220 Ongoing, recruiting patients
at 2 weeks
monarcHER158 Abemaciclib PFS 225 Ongoing, not yet recruiting patients
MONARCH-1110 Abemaciclib Objective response rate 128 Closed to accrual
MONARCH-2112 Abemaciclib PFS 550 Ongoing
MONARCH-3113 Abemaciclib PFS 450 Ongoing
NCT02308020114 Abemaciclib Percentage of participants achieving 247 Recruiting
complete response or partial
response: objective intracranial
response rate
Ribociclib: CDK4/6 Inhibitor
MONALEESA-2141 Ribociclib PFS 667 Ongoing
159
MONALEESA-3 Ribociclib PFS 660 Ongoing, recruiting participants
Entinostat: HDAC Inhibitor
Yardley et al117 Entinostat PFS 130 Completed; PFS was 4.3 months in
exemestane/entinostat arm compared
with PFS of 2.3 months with exemestane
alone arm (p = .055); overall survival was
28.1 months in exemestane/entinostat
arm compared with 19.8 months in
exemestane alone arm (HR 0.59; 95% CI,
0.360.97)
E2112118 Entinostat PFS 600 Ongoing
Azacitidine: DNA Methylation Inhibitor
NCT01349959120 Azacitidine and entinostat Confirmed response rate 40 Ongoing
NCT02374099119 Azacitidine Time period of PFS 92 Ongoing, recruiting patients
Abbreviations: HR, hazard ratio; PFS, progression-free survival.

diarrhea, and 18% of patients discontinued due to adverse randomly selected to fulvestrant with either buparlisib or
events. Progression-free survival was similar between the placebo, with a primary endpoint of PFS in the full pop-
two treatment arms at 6.6 months for pictilisib and ulation and in those with PI3K pathwayactivated tumors. In
5.1 months for placebo (HR 0.7; 95% CI, 0.521.06; p = .096), the 1,147 women who received study therapy, PFS was
and among patients with PI3KCA-mutant tumors. In an slightly improved with buparlisib compared with placebo
exploratory analysis, there was a suggestion of benefit for (6.9 vs. 5 months; HR 0.78; 95% CI, 0.670.89; p , .001).
patients with PR+ disease. Progression-free survival was not significantly improved for
The pan-PI3K inhibitor buparlisib was evaluated in the patients with PI3K activation (6.8 vs. 4 months). In an ex-
phase III BELLE-2 trial.96 Patients with prior AI therapy were ploratory analysis of PIK3CA mutations in circulating tumor

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e47


RUGO, VIDULA, AND MA

TABLE 2. Summary of the Most-Promising Targeted Agents Under Development in the Treatment of Hormone
ReceptorPositive Breast Cancer

Agent Mechanism of Action Developmental Status Toxicity Profile


Everolimus mTOR inhibition FDA approval for everolimus plus exemestane as at Main toxicities include stomatitis, hyperglycemia,
least second-line therapy for HR+ advanced fatigue, pneumonitis
breast cancer
Palbociclib, Ribociclib, CDK4/6 inhibition FDA approval for palbociclib plus letrozole as first- Main toxicities include neutropenia without
Abemaciclib line therapy, and palbociclib plus fulvestrant as an increase in febrile neutropenia, QTc
at least second-line therapy for advanced breast prolongation, and fatigue; diarrhea most
cancer; ongoing trials with all 3 agents in the common with abemaciclib
metastatic, adjuvant, and neoadjuvant settings
Alpha-Specific
Alpelisib, Taselisib PI3K inhibition Ongoing trials Main toxicities include hyperglycemia, rash, and
liver dysfunction
Pan-PI3K
Buparlisib, Pictilisib PI3K inhibition Ongoing trials Main toxicities include hyperglycemia, rash, and
liver dysfunction
Entinostat HDAC inhibition FDA breakthrough status; ongoing phase III trial Main toxicities include fatigue, myelosuppression,
with exemestane diarrhea, nausea, and vomiting
Dovitinib, Lucitinib FGFR inhibition Pre-clinical and preliminary clinical efficacy with Main toxicities include diarrhea, nausea, and
FGFR/VEGF inhibition dovitinib and lucitinib hypertension, proteinuria, and liver dysfunction
(lucitinib)
Abbreviation: FDA, U.S. Food and Drug Administration.

DNA (ctDNA; 87 patients for buparlisib and 113 for placebo), cancer. Palbociclib and ribociclib are given on a 3-week-on,
PFS was improved for those with PIK3CA mutations receiving 1-week-off schedule, and abemaciclib is given continuously.
buparlisib compared with placebo (7.0 vs. 3.2 months; HR A number of studies are evaluating CDK4/6 inhibitors in the
0.56), with no difference in the wild-type population. neoadjuvant, adjuvant, and metastatic settings, as sum-
Toxicity was increased in the buparlisib arm, with grade marized in Table 1. The results of pivotal trials and key
3/4 transaminitis (. 20%; approximately 0% grade 4), hy- planned trials are summarized here.
perglycemia (15.4%), rash (7.9%), anxiety (5.4%), and de- PALOMA-1 was a phase II trial that randomly selected 165
pression (4.4%); 13% of patients discontinued treatment due postmenopausal women with untreated HR metastatic
to adverse events. breast cancer to receive letrozole with or without palbociclib.99
The data from BELLE-2, although disappointing in the Progression-free survival with combination therapy was
overall population, provide the first data suggesting that 20.2 months vs. 10.2 months in the control arm (HR 0.49;
ctDNA could be a potential marker to select patients who 95% CI 0.3190.748; p = .0004). The trial was not powered
might benefit from specific targeted therapies. Ongoing to evaluate OS, which was similar between the two arms.
trials are evaluating the use of PIK3CA mutations in ctDNA as
The primary toxicity was at least grade 3 neutropenia,
predictive markers for response to alpha-specific PI3K in-
occurring in 54% of patients, with no cases of febrile
hibitors, such as alpelisib and taselisib.
neutropenia; 13% of patients in the study arm dis-
The alpha-specific PI3K inhibitors alpelisib and taselisib
continued therapy due to adverse events.
have demonstrated encouraging efficacy in early-phase
Based on this data, in 2015, the U.S. Food and Drug Ad-
trials, with modest toxicity. Two ongoing placebo-controlled
ministration (FDA) granted accelerated approval for pal-
phase III trials are evaluating the potential improvement in
bociclib and letrozole for the first-line treatment of
PFS with the addition of alpelisib (SOLAR-197) or taselisib
(SANDPIPER98) to fulvestrant among patients with HR+ postmenopausal patients with HR+ metastatic breast can-
metastatic breast cancer, and neoadjuvant trials in combi- cer. PALOMA-2 is a phase III trial designed to validate these
nation with AIs are also ongoing (the buparlisib arm of the findings; data are expected to be presented at the 2016
NeoORB study was recently closed based on results from ASCO Annual Meeting.100
BELLE-2). More details regarding ongoing studies are pro- PALOMA-3, a phase III trial, randomly selected 521 women
vided in Table 1. Results are eagerly awaited. with advanced pretreated breast cancer 2:1 to treatment
with palbociclib and fulvestrant versus fulvestrant and
placebo.101 Progression-free survival was significantly im-
Inhibition of CDK4/6 proved in the combination arm versus placebo (9.2 months vs.
Three CDK4/6 inhibitors (palbociclib, ribociclib, and abe- 3.8 months; HR 0.42; 95% CI, 0.320.56; p , .001). Pre-
maciclib) are in advanced clinical testing, and palbociclib has menopausal women with ovarian suppression and pa-
regulatory approval for treatment of metastatic breast tients with one line of prior chemotherapy appeared to

e48 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


IMPROVING RESPONSE TO HORMONE THERAPY

have similar benefit. Toxicity was similar to PALOMA-1, al- toxicities; 11% of patients discontinued due to adverse
though only 2.6% of patients discontinued treatment due to events. A phase III trial, E2112, with a similar design is
adverse events. Fulvestrant and palbociclib were approved ongoing (Table 1).118
by the FDA in early 2016. Azacitidine, a DNA methylation inhibitor, is also being
An international adjuvant trial of 4,600 patients is compar- investigated in breast cancer with hormonal therapy119 and
ing 2 years of treatment with palbociclib versus placebo entinostat.120
(PALLAS),102 and PENELOPE-B103 is studying 1 year of palbo-
ciclib as postneoadjuvant therapy for high-risk patients. Nu- Inhibition of IGFR
merous other trials are evaluating palbociclib as neoadjuvant A number of IGFR inhibitors have been studied in the clinic, with
therapy compared with capecitabine in metastatic disease generally disappointing results to date. A randomized phase II
(PEARL)104 and following chemotherapy, among others. trial evaluated the benefit of adding ganitumab to either ful-
Ribociclib is a similar CDK4/6 inhibitor that is being studied vestrant or exemestane as second-line therapy for 156 patients
in the metastatic and neoadjuvant settings in the ongoing with metastatic HR+ breast cancer.121 There was no difference
MONALEESA trials.105,106 In addition, ongoing studies are in PFS (3.9 months for the ganitumab arm vs. 5.7 months for
evaluating ribociclib combined with exemestane and ever- placebo). New agents that do not cause hyperglycemia may
olimus,107 and letrozole and the PI3K inhibitor alpelisib.108 offer advantages; the monoclonal antibody BI 836845 is being
Abemaciclib is a potent inhibitor of CDK4, more than CDK6, studied in combination with everolimus and exemestane in a
and is the only agent in this class that can be given con- randomized phase II trial based on promising phase I results.107
tinuously. A phase IB trial including 47 patients demon-
strated single-agent activity with a clinical benefit rate of
61.1%,109 leading to the single agent MONARCH-1 trial of Inhibition of Angiogenesis
patients with heavily pretreated HR+ metastatic breast Although not currently in the clinical arena, a discussion of
cancer; data are expected to be presented at the 2016 ASCO this approach is important. Two randomized phase III trials
Annual Meeting.110 An expansion cohort of the phase IB trial evaluated the potential efficacy from adding bevacizumab,
evaluated abemaciclib plus fulvestrant; the clinical benefit the VEGF antibody, to first-line hormone therapy with
rate was 72.2%.111 The most common toxicity from abe- letrozole or fulvestrant.122,123 Both trials demonstrated im-
maciclib is diarrhea, with a lower incidence of neutropenia. provement in PFS comparing letrozole or fulvestrant plus
Additional ongoing MONARCH trials are evaluating abe- bevacizumab with hormone therapy alone, although only the
maciclib in the neoadjuvant setting,105 in metastatic disease CALGB trial found this difference to be statistically significant
in combination with fulvestrant,112 or nonsteroidal AIs113 in (LEA trial: 19.3 vs. 14.4 months; HR 0.83; 95% CI, 0.651.06;
brain metastases114 and in HER2+ disease.112 p = .126; CALGB 40503: 20 vs. 16 months; HR 0.75; 95% CI,
0.590.96; p = .016); no difference was seen in OS. Adverse
events included at least grade 3 hypertension and proteinuria.
Inhibition of FGFR Further combined biomarkers analysis is ongoing.
Dovitinib, an oral tyrosine kinase inhibitor of FGFR, has been
shown to have both preclinical and clinical efficacy.28 In a
Immune Checkpoint Inhibition
study of 81 patients, five patients with tumor FGFR ampli-
Based on the expression of PD-L1 in HR+ breast cancer, there
fication experienced clinical benefit at 6 months. These
has been interest in evaluating the efficacy of this approach
results prompted further trials with dovitinib and hormonal
in the advanced disease setting. Pembrolizumab, a PD-1
therapy combinations, although toxicity is a limiting fac-
antibody, was tested in a number of solid malignancies in the
tor.115 Lucitanib is a FGFR/VEGFR tyrosine kinase inhibitor
phase IB KEYNOTE-28 trial.124 Out of 248 patients who had
with encouraging phase IB response data under evaluation
tissue available for screening, 48 (19.4%) had at least 1%
in a phase II single-agent trial.116 Toxicity is again limiting,
staining for PD-L1. The overall response rate among 25
including hypertension, proteinuria, and transaminitis.
treated patients was 12%, with a clinical benefit rate of 20%.
The median time to response was 8 weeks, and the median
Inhibition of HDAC duration of response had not yet been reached.
A randomized phase II trial evaluated the addition of enti- The phase IB Javelin trial evaluated the efficacy of the anti-
nostat or placebo to exemestane for 130 women with ad- PD-L1 antibody avelumab for patients with metastatic solid
vanced breast cancer.117 Progression-free survival was tumors, including breast cancer.125 Out of the 72 patients
slightly improved in those receiving entinostat compared with HR+ disease, an objective response was seen in only
with placebo (4.3 vs. 2.3 months; p = .055), but the ex- 2.8%, but 54% were found to have PD-L1 expression. This
ploratory endpoint of OS was significantly improved (28.1 vs. brings into question the antibodies used for PD-L1 testing as
19.8 months; HR 0.59; 95% CI, 0.360.97; p = .036), leading well as potential differences in efficacy between different
to breakthrough drug status by the FDA. Interestingly, checkpoint inhibitors.
protein lysine hyperacetylation in the entinostat biomarker Overall, therapy was well tolerated in both trials, with
subset was associated with prolonged PFS. Fatigue and the primary toxicity reversible and immune related (e.g.,
neutropenia were the most common grade 3 or worse thyroid disorders, hepatitis). Further studies will evaluate

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e49


RUGO, VIDULA, AND MA

combination therapy with agents that could increase The development of ESR1 mutation following endocrine
expression of neo-antigens (such as HDAC inhibitors), and therapy provides further evidence that ER remains relevant
therefore enhance immunogenicity. in resistant tumors. Combination strategies that target both
ER and resistant pathways are therefore rational approaches
CONCLUSION to overcome endocrine resistance. Toxicity and cost are
Progress in the understanding of endocrine resistance increased with the addition of targeted agents. Awareness of
mechanisms has led to the development of a number of both expected toxicity and appropriate management is
agentssome approved and many in clinical trialsto important. There is a critical need to develop predictive
target growth factor receptor signaling, PI3K/Akt/mTOR, biomarkers; recent data using ctDNA are encouraging.
CDK4/6, HDAC, and immune checkpoints, among others. Further studies are ongoing.

References
1. Beatson GT. On the treatment of inoperable carcinoma of the 17. Ejlertsen B, Aldridge J, Nielsen KV, et al. Prognostic and predictive role
mamma: suggestions for a new method of treatment with illustrative of ESR1 status for postmenopausal patients with endocrine-
cases. Lancet. 1896;2:104-107. responsive early breast cancer in the Danish cohort of the BIG 1-98
2. Davies C, Godwin J, Gray R, et al; Early Breast Cancer Trialists Col- trial. Ann Oncol. 2011;23:1138-1144.
laborative Group (EBCTCG). Relevance of breast cancer hormone re- 18. Ellis MJ, Tao Y, Young O, et al. Estrogen-independent proliferation is
ceptors and other factors to the efficacy of adjuvant tamoxifen: patient- present in estrogen-receptor HER2-positive primary breast cancer
level meta-analysis of randomised trials. Lancet. 2011;378:771-784. after neoadjuvant letrozole. J Clin Oncol. 2006;24:3019-3025.
3. Mauri D, Pavlidis N, Polyzos NP, et al. Survival with aromatase in- 19. Perez EA, Romond EH, Suman VJ, et al. Four-year follow-up of tras-
hibitors and inactivators versus standard hormonal therapy in ad- tuzumab plus adjuvant chemotherapy for operable human epidermal
vanced breast cancer: meta-analysis. J Natl Cancer Inst. 2006;98: growth factor receptor 2-positive breast cancer: joint analysis of data
1285-1291. from NCCTG N9831 and NSABP B-31. J Clin Oncol. 2011;29:3366-3373.
4. Osborne CK, Schiff R. Estrogen-receptor biology: continuing progress 20. Schwartzberg LS, Franco SX, Florance A, et al. Lapatinib plus letrozole
and therapeutic implications. J Clin Oncol. 2005;23:1616-1622. as first-line therapy for HER-2+ hormone receptor-positive metastatic
5. Egeland NG, Lunde S, Jonsdottir K, et al. The role of MicroRNAs as breast cancer. Oncologist. 2010;15:122-129.
predictors of response to tamoxifen treatment in breast cancer pa- 21. Marcom PK, Isaacs C, Harris L, et al. The combination of letrozole and
tients. Int J Mol Sci. 2015;16:24243-24275. trastuzumab as first or second-line biological therapy produces du-
6. Losel R, Wehling M. Nongenomic actions of steroid hormones. Nat Rev rable responses in a subset of HER2 positive and ER positive advanced
Mol Cell Biol. 2003;4:46-56. breast cancers. Breast Cancer Res Treat. 2007;102:43-49.
7. Levin ER. Integration of the extranuclear and nuclear actions of es- 22. Kaufman B, Mackey JR, Clemens MR, et al. Trastuzumab plus anas-
trogen. Mol Endocrinol. 2005;19:1951-1959. trozole versus anastrozole alone for the treatment of postmenopausal
8. Bjornstrom L, Sjoberg M. Mechanisms of estrogen receptor signaling: women with human epidermal growth factor receptor 2-positive,
convergence of genomic and nongenomic actions on target genes. hormone receptor-positive metastatic breast cancer: results from the
Mol Endocrinol. 2005;19:833-842. randomized phase III TAnDEM study. J Clin Oncol. 2009;27:5529-5537.
9. Campbell RA, Bhat-Nakshatri P, Patel NM, et al. Phosphatidylinositol 23. Bose R, Kavuri SM, Searleman AC, et al. Activating HER2 mutations in
3-kinase/AKT-mediated activation of estrogen receptor alpha: a new HER2 gene amplification negative breast cancer. Cancer Discov. 2013;
model for anti-estrogen resistance. J Biol Chem. 2001;276:9817-9824. 3:224-237.
10. Kato S, Endoh H, Masuhiro Y, et al. Activation of the estrogen receptor 24. Ross JS, Wang K, Sheehan CE, et al. Relapsed classic E-cadherin (CDH1)-
through phosphorylation by mitogen-activated protein kinase. Sci- mutated invasive lobular breast cancer shows a high frequency of HER2
ence. 1995;270:1491-1494. (ERBB2) gene mutations. Clin Cancer Res. 2013;19:2668-2676.
11. Li S, Shen D, Shao J, et al. Endocrine-therapy-resistant ESR1 variants 25. Hyman DM, Piha-Paul SA, Rodon J, et al. Neratinib for ERBB2 mutant,
revealed by genomic characterization of breast-cancer-derived xe- HER2 non-amplified, metastatic breast cancer: preliminary analysis
nografts. Cell Reports. 2013;4:1116-1130. from a multicenter, open-label, multi-histology phase II basket trial.
12. Robinson DR, Wu YM, Vats P, et al. Activating ESR1 mutations in Cancer Res. 2016;76 (suppl 4; abstr PD5-05).
hormone-resistant metastatic breast cancer. Nat Genet. 2013;45: 26. Turner N, Grose R. Fibroblast growth factor signalling: from devel-
1446-1451. opment to cancer. Nat Rev Cancer. 2010;10:116-129.
13. Toy W, Shen Y, Won H, et al. ESR1 ligand-binding domain mutations in 27. Turner N, Pearson A, Sharpe R, et al. FGFR1 amplification drives
hormone-resistant breast cancer. Nat Genet. 2013;45:1439-1445. endocrine therapy resistance and is a therapeutic target in breast
14. Roodi N, Bailey LR, Kao WY, et al. Estrogen receptor gene analysis in cancer. Cancer Res. 2010;70:2085-2094.
estrogen receptor-positive and receptor-negative primary breast 28. Andre F, Bachelot T, Campone M, et al. Targeting FGFR with dovitinib
cancer. J Natl Cancer Inst. 1995;87:446-451. (TKI258): preclinical and clinical data in breast cancer. Clin Cancer Res.
15. Saxena R, Dwivedi A. ErbB family receptor inhibitors as therapeutic 2013;19:3693-3702.
agents in breast cancer: Current status and future clinical perspective. 29. deGraffenried LA, Friedrichs WE, Russell DH, et al. Inhibition of mTOR
Med Res Rev. 2012;32:166-215. activity restores tamoxifen response in breast cancer cells with ab-
16. Dowsett M. Overexpression of HER-2 as a resistance mechanism to errant Akt Activity. Clin Cancer Res. 2004;10:8059-8067.
hormonal therapy for breast cancer. Endocr Relat Cancer. 2001;8: 30. Miller TW, Hennessy BT, Gonzalez-Angulo AM, et al. Hyperactivation
191-195. of phosphatidylinositol-3 kinase promotes escape from hormone

e50 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


IMPROVING RESPONSE TO HORMONE THERAPY

dependence in estrogen receptor-positive human breast cancer. J Clin 48. Sherr CJ. Cancer cell cycles. Science. 1996;274:1672-1677.
Invest. 2010;120:2406-2413. 49. Sherr CJ, Roberts JM. CDK inhibitors: positive and negative regulators
31. Sanchez CG, Ma CX, Crowder RJ, et al. Preclinical modeling of com- of G1-phase progression. Genes Dev. 1999;13:1501-1512.
bined phosphatidylinositol-3-kinase inhibition with endocrine therapy 50. van den Heuvel S, Harlow E. Distinct roles for cyclin-dependent kinases
for estrogen receptor-positive breast cancer. Breast Cancer Res. 2011; in cell cycle control. Science. 1993;262:2050-2054.
13:R21. 51. Weinberg RA. The retinoblastoma protein and cell cycle control. Cell.
32. Vivanco I, Sawyers CL. The phosphatidylinositol 3-Kinase AKT pathway 1995;81:323-330.
in human cancer. Nat Rev Cancer. 2002;2:489-501. 52. Butt AJ, McNeil CM, Musgrove EA, et al. Downstream targets of
33. Ellis MJ, Ding L, Shen D, et al. Whole-genome analysis informs breast growth factor and oestrogen signalling and endocrine resistance: the
cancer response to aromatase inhibition. Nature. 2012;486:353-360. potential roles of c-Myc, cyclin D1 and cyclin E. Endocr Relat Cancer.
34. Cancer Genome Atlas Network.; Cancer Genome Atlas Network. 2005;12:S47-S59.
Comprehensive molecular portraits of human breast tumours. Na- 53. Altucci L, Addeo R, Cicatiello L, et al. Estrogen induces early and timed
ture. 2012;490:61-70. activation of cyclin-dependent kinases 4, 5, and 6 and increases cyclin
35. Saal LH, Holm K, Maurer M, et al. PIK3CA mutations correlate with messenger ribonucleic acid expression in rat uterus. Endocrinology.
hormone receptors, node metastasis, and ERBB2, and are mutually 1997;138:978-984.
exclusive with PTEN loss in human breast carcinoma. Cancer Res. 54. Geum D, Sun W, Paik SK, et al. Estrogen-induced cyclin D1 and D3 gene
2005;65:2554-2559. expressions during mouse uterine cell proliferation in vivo: differential
36. Crowder RJ, Phommaly C, Tao Y, et al. PIK3CA and PIK3CB inhibition induction mechanism of cyclin D1 and D3. Mol Reprod Dev. 1997;46:
produce synthetic lethality when combined with estrogen deprivation 450-458.
in estrogen receptor-positive breast cancer. Cancer Res. 2009;69: 55. Said TK, Conneely OM, Medina D, et al. Progesterone, in addition to
3955-3962. estrogen, induces cyclin D1 expression in the murine mammary ep-
37. Guertin DA, Sabatini DM. The pharmacology of mTOR inhibition. Sci ithelial cell, in vivo. Endocrinology. 1997;138:3933-3939.
Signal. 2009;2:pe24. 56. Tong W, Pollard JW. Progesterone inhibits estrogen-induced cyclin D1
38. OReilly KE, Rojo F, She QB, et al. mTOR inhibition induces upstream and cdk4 nuclear translocation, cyclin E- and cyclin A-cdk2 kinase
receptor tyrosine kinase signaling and activates Akt. Cancer Res. 2006; activation, and cell proliferation in uterine epithelial cells in mice. Mol
66:1500-1508. Cell Biol. 1999;19:2251-2264.
39. Wan X, Harkavy B, Shen N, et al. Rapamycin induces feedback acti- 57. Thangavel C, Dean JL, Ertel A, et al. Therapeutically activating RB:
vation of Akt signaling through an IGF-1R-dependent mechanism. reestablishing cell cycle control in endocrine therapy-resistant breast
Oncogene. 2007;26:1932-1940. cancer. Endocr Relat Cancer. 2011;18:333-345.
40. Mayer IA, Abramson VG, Isakoff SJ, et al. Stand up to cancer phase Ib 58. Finn RS, Dering J, Conklin D, et al. PD 0332991, a selective cyclin D
study of pan-phosphoinositide-3-kinase inhibitor buparlisib with kinase 4/6 inhibitor, preferentially inhibits proliferation of luminal
letrozole in estrogen receptor-positive/human epidermal growth estrogen receptor-positive human breast cancer cell lines in vitro.
factor receptor 2-negative metastatic breast cancer. J Clin Oncol. Breast Cancer Res. 2009;11:R77.
2014;32:1202-1209. 59. Wardell SE, Ellis MJ, Alley HM, et al. Efficacy of SERD/SERM Hybrid-
41. Ma CX, Sanchez C, Gao F, et al. A phase I study of the AKT inhibitor MK- CDK4/6 inhibitor combinations in models of endocrine therapy re-
2206 in combination with hormonal therapy in postmenopausal sistant breast cancer. Clin Cancer Res. 2015;21:5121-5130.
women with estrogen receptor positive metastatic breast cancer. Clin 60. Vora SR, Juric D, Kim N, et al. CDK 4/6 inhibitors sensitize PIK3CA
Cancer Res. Epub 2016 Jan 18. mutant breast cancer to PI3K inhibitors. Cancer Cell. 2014;26:136-149.
42. Ma CX, Luo J, Naughton M, et al. A phase 1 trial of BKM120 (buparlisib) 61. Pathiraja TN, Stearns V, Oesterreich S. Epigenetic regulation in es-
in combination with fulvestrant in postmenopausal women with trogen receptor positive breast cancerrole in treatment response.
estrogen receptor positive metastatic breast cancer. Clin Cancer Res. J Mammary Gland Biol Neoplasia. 2010;15:35-47.
2016;22:1583-1591. 62. Yi X, Wei W, Wang SY, et al. Histone deacetylase inhibitor SAHA in-
43. Baselga J, Im S-A, Iwata H, et al. PIK3CA status in circulating tumor DNA duces ERalpha degradation in breast cancer MCF-7 cells by CHIP-
(ctDNA) predicts efficacy of buparlisib (BUP) plus fulvestrant (FULV) in mediated ubiquitin pathway and inhibits survival signaling. Biochem
postmenopausal women with endocrine-resistant HR+/HER2 ad- Pharmacol. 2008;75:1697-1705.
vanced breast cancer (BC): First results from the randomized, phase III 63. Thomas S, Thurn KT, Biaku E, et al. Addition of a histone deacetylase
BELLE-2 trial. Presented at: San Antonio Breast Cancer Symposium; inhibitor redirects tamoxifen-treated breast cancer cells into apo-
2015; San Antonio, Texas. ptosis, which is opposed by the induction of autophagy. Breast Cancer
44. Krop I, Johnston S, Mayer I, et al. The FERGI phase II study of the PI3K Res Treat. 2011;130:437-447.
inhibitor pictilisib (GDC-0941) plus fulvestrant vs fulvestrant plus 64. Biaku E, Marchion DC, Schmitt ML, et al. Selective inhibition of
placebo in patients with ER+, aromatase inhibitor (AI)-resistant ad- histone deacetylase 2 silences progesterone receptor-mediated sig-
vanced or metastatic breast cancer Part I results. Cancer Res. 2015; naling. Cancer Res. 2008;68:1513-1519.
75:S2-02. 65. Farabaugh SM, Boone DN, Lee AV. Role of IGF1R in breast cancer
45. Juric D, Castel P, Griffith M, et al. Convergent loss of PTEN leads to subtypes, stemness, and lineage differentiation. Front Endocrinol
clinical resistance to a PI(3)Ka inhibitor. Nature. 2015;518:240-244. (Lausanne). 2015;6:59.
46. Elkabets M, Vora S, Juric D, et al. mTORC1 inhibition is required for 66. Shelton JG, Steelman LS, White ER, et al. Synergy between PI3K/Akt
sensitivity to PI3K p110a inhibitors in PIK3CA-mutant breast cancer. and Raf/MEK/ERK pathways in IGF-1R mediated cell cycle progression
Sci Transl Med. 2013;5:196ra99. and prevention of apoptosis in hematopoietic cells. Cell Cycle. 2004;3:
47. Hortobagyi GN, Piccart-Gebhart MJ, Rugo HS, et al. Correlation of 372-379.
molecular alterations with efficacy of everolimus in hormone receptor 67. Fagan DH, Yee D. Crosstalk between IGF1R and estrogen receptor
positive, HER2-negative advanced breast cancer: Results from BOLERO-2. signaling in breast cancer. J Mammary Gland Biol Neoplasia. 2008;13:
J Clin Oncol. 2013 (suppl; abstr LBA509). 423-429.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e51


RUGO, VIDULA, AND MA

68. Lee AV, Jackson JG, Gooch JL, et al. Enhancement of insulin-like growth postmenopausal hormone receptor-positive metastatic breast can-
factor signaling in human breast cancer: estrogen regulation of insulin cer. J Clin Oncol. 2009;27:5538-5546.
receptor substrate-1 expression in vitro and in vivo. Mol Endocrinol. 86. Wolff AC, Lazar AA, Bondarenko I, et al. Randomized phase III placebo-
1999;13:787-796. controlled trial of letrozole plus oral temsirolimus as first-line en-
69. Maor S, Mayer D, Yarden RI, et al. Estrogen receptor regulates insulin- docrine therapy in postmenopausal women with locally advanced or
like growth factor-I receptor gene expression in breast tumor cells: metastatic breast cancer. J Clin Oncol. 2013;31:195-202.
involvement of transcription factor Sp1. J Endocrinol. 2006;191: 87. Baselga J, Semiglazov V, van Dam P, et al. Phase II randomized study of
605-612. neoadjuvant everolimus plus letrozole compared with placebo plus
70. Mauro L, Salerno M, Panno ML, et al. Estradiol increases IRS-1 gene letrozole in patients with estrogen receptor-positive breast cancer.
expression and insulin signaling in breast cancer cells. Biochem Bio- J Clin Oncol. 2009;27:2630-2637.
phys Res Commun. 2001;288:685-689. 88. Bachelot T, Bourgier C, Cropet C, et al. Randomized phase II trial of
71. Becker MA, Ibrahim YH, Cui X, et al. The IGF pathway regulates ERa everolimus in combination with tamoxifen in patients with hormone
through a S6K1-dependent mechanism in breast cancer cells. Mol receptor-positive, human epidermal growth factor receptor 2-nega-
Endocrinol. 2011;25:516-528. tive metastatic breast cancer with prior exposure to aromatase in-
72. Knowlden JM, Hutcheson IR, Barrow D, et al. Insulin-like growth hibitors: a GINECO study. J Clin Oncol. 2012;30:2718-2724.
factor-I receptor signaling in tamoxifen-resistant breast cancer: 89. Yardley DA, Noguchi S, Pritchard KI, et al. Everolimus plus exemestane
a supporting role to the epidermal growth factor receptor. Endocri- in postmenopausal patients with HR(+) breast cancer: BOLERO-2 final
nology. 2005;146:4609-4618. progression-free survival analysis. Adv Ther. 2013;30:870-884.
73. Arnedos M, Drury S, Afentakis M, et al. Biomarker changes associated 90. Piccart M, Hortobagyi GN, Campone M, et al. Everolimus plus
with the development of resistance to aromatase inhibitors (AIs) in exemestane for hormone-receptor-positive, human epidermal
estrogen receptor-positive breast cancer. Ann Oncol. 2014;25: growth factor receptor-2-negative advanced breast cancer: overall
605-610. survival results from BOLERO-2. Ann Oncol. 2014;25:2357-2362.
74. Fox EM, Miller TW, Balko JM, et al. A kinome-wide screen identifies the 91. Campone M, Bachelot T, Gnant M, et al. Effect of visceral metastases
insulin/IGF-I receptor pathway as a mechanism of escape from on the efficacy and safety of everolimus in postmenopausal women
hormone dependence in breast cancer. Cancer Res. 2011;71: with advanced breast cancer: subgroup analysis from the BOLERO-2
6773-6784. study. Eur J Cancer. 2013;49:2621-2632.
75. Fagan DH, Uselman RR, Sachdev D, et al. Acquired resistance to ta- 92. Baselga J, Campone M, Piccart M, et al. Everolimus in postmenopausal
moxifen is associated with loss of the type I insulin-like growth factor hormone-receptor-positive advanced breast cancer. N Engl J Med.
receptor: implications for breast cancer treatment. Cancer Res. 2012; 2012;366:520-529.
72:3372-3380. 93. Rugo HS, Pritchard KI, Gnant M, et al. Incidence and time course of
76. Fox EM, Kuba MG, Miller TW, et al. Autocrine IGF-I/insulin receptor everolimus-related adverse events in postmenopausal women with
axis compensates for inhibition of AKT in ER-positive breast cancer hormone receptor-positive advanced breast cancer: insights from
cells with resistance to estrogen deprivation. Breast Cancer Res. 2013; BOLERO-2. Ann Oncol. 2014;25:808-815.
15:R55. 94. Dean JL, Thangavel C, McClendon AK, et al. Therapeutic CDK4/6 in-
77. Liang Y, Hyder SM. Proliferation of endothelial and tumor epithelial hibition in breast cancer: key mechanisms of response and failure.
cells by progestin-induced vascular endothelial growth factor from Oncogene. 2010;29:4018-4032.
human breast cancer cells: paracrine and autocrine effects. Endo- 95. NCT02069093. Phase II study of stomatitis prevention with a steroid-
crinology. 2005;146:3632-3641. based mouthwash in post-menopausal women with ER+, HER2-
78. Schoeffner DJ, Matheny SL, Akahane T, et al. VEGF contributes to metastatic or locally advanced breast cancer. https://clinicaltrials.
mammary tumor growth in transgenic mice through paracrine and gov/ct2/show/NCT02069093. Accessed January 30,2016.
autocrine mechanisms. Lab Invest. 2005;85:608-623. 96. Baselga J, Im SA, Iwata H, et al. PIK3CA status in circulating tumor DNA
79. Foekens JA, Peters HA, Grebenchtchikov N, et al. High tumor levels of (ctDNA) predicts efficacy of buparlisib (BUP) plus fulvestrant (FULV) in
vascular endothelial growth factor predict poor response to systemic postmenopausal women with endocrine-resistant HR+/HER2 ad-
therapy in advanced breast cancer. Cancer Res. 2001;61:5407-5414. vanced breast cancer (BC): First results from the randomized, phase III
80. Manders P, Beex LV, Tjan-Heijnen VC, et al. Vascular endothelial BELLE-2 trial. Cancer Res. 2015;76 (4 suppl; abstr S6-01).
growth factor is associated with the efficacy of endocrine therapy in 97. NCT02437318. Study assessing the efficacy and safety of alpelisib
patients with advanced breast carcinoma. Cancer. 2003;98: plus fulvestrant in men and postmenopausal women with advanced
2125-2132. breast cancer which progressed on or after aromatase inhibitor
81. Pardoll DM. The blockade of immune checkpoints in cancer immu- treatment. (SOLAR-1). https://clinicaltrials.gov/ct2/show/NCT02437318?
notherapy. Nat Rev Cancer. 2012;12:252-264. term=alpelisib&rank=1. Accessed March 3, 2016.
82. Ghebeh H, Mohammed S, Al-Omair A, et al. The B7-H1 (PD-L1) 98. NCT02340221. SANDPIPER Study: A study of taselisib + fulvestrant
T lymphocyte-inhibitory molecule is expressed in breast cancer pa- versus placebo + fulvestrant in patients with advanced or metastatic
tients with infiltrating ductal carcinoma: correlation with important breast cancer who have disease recurrence or progression during or
high-risk prognostic factors. Neoplasia. 2006;8:190-198. after aromatase inhibitor therapy. https://clinicaltrials.gov/ct2/
83. Muenst S, Schaerli AR, Gao F, et al. Expression of programmed death show/NCT02340221. Accessed March 8, 2016.
ligand 1 (PD-L1) is associated with poor prognosis in human breast 99. Finn RS, Crown JP, Lang I, et al. The cyclin-dependent kinase 4/6
cancer. Breast Cancer Res Treat. 2014;146:15-24. inhibitor palbociclib in combination with letrozole versus letrozole
84. Sabatier R, Finetti P, Mamessier E, et al. Prognostic and predictive alone as first-line treatment of oestrogen receptor-positive, HER2-
value of PDL1 expression in breast cancer. Oncotarget. 2015;6: negative, advanced breast cancer (PALOMA-1/TRIO-18): a rando-
5449-5464. mised phase 2 study. Lancet Oncol. 2015;16:25-35.
85. Johnston S, Pippen J Jr, Pivot X, et al. Lapatinib combined with 100. NCT01740427. A study of palbociclib (PD-0332991) + letrozole vs. letrozole
letrozole versus letrozole and placebo as first-line therapy for for 1st line treatment of postmenopausal women with ER+/HER2- advanced

e52 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


IMPROVING RESPONSE TO HORMONE THERAPY

breast cancer (PALOMA-2). http://www.cancer.gov/clinicaltrials/search/ 116. NCT01723774. PD 0332991 and anastrozole for stage 2 or 3 estrogen
view?cdrid=743730&version=HealthProfessional&protocolsearchid= receptor positive and HER2 negative breast cancer. http://www.
6378902. Accessed May 16, 2014. cancer.gov/clinicaltrials/search/view?cdrid=742660&version=
101. Turner NC, Ro J, Andre F, et al. Palbociclib in hormone-receptor- HealthProfessional&protocolsearchid=12773356. Accessed May
positive advanced breast cancer. N Engl J Med. 2015;373:209-219. 27, 2015.
102. NCT02513394. PALbociclib CoLlaborative Adjuvant Study: a randomized 117. Yardley DA, Ismail-Khan RR, Melichar B, et al. Randomized phase II,
phase III trial of palbociclib with standard adjuvant endocrine ther- double-blind, placebo-controlled study of exemestane with or
apy versus standard adjuvant endocrine therapy alone for hormone without entinostat in postmenopausal women with locally recurrent
receptor positive (HR+) / human epidermal growth factor receptor 2 or metastatic estrogen receptor-positive breast cancer progressing on
(HER2)-negative early breast cancer (PALLAS). https://clinicaltrials. treatment with a nonsteroidal aromatase inhibitor. J Clin Oncol. 2013;
gov/show/NCT02513394undefined. Accessed March 3, 2016. 31:2128-2135.
103. NCT01864746. A study of palbociclib in addition to standard endo- 118. Connolly RM, Zhao F, Miller K, et al. E2112: a randomized phase III trial
crine treatment in hormone receptor positive Her2 normal patients of endocrine therapy plus entinostat/placebo in patients with hor-
with residual disease after neoadjuvant chemotherapy and surgery mone receptor-positive advanced breast cancer. J Clin Oncol. 2015;33
(PENELOPE-B). http://clinicaltrials.gov/show/NCT01864746. Accessed (suppl; abstr TPS636).
May 16, 2014. 119. NCT02374099. Study to assess the efficacy and safety of the epige-
104. NCT02028507. Phase III study of palbociclib (PD-0332991) in com- netic modifying effects of CC-486 (oral azacitidine) in combination
bination with exemestane versus chemotherapy (capecitabine) in with Fulvestrant. https://clinicaltrials.gov/ct2/show/NCT02374099?
hormonal receptor (HR) positive/HER2 negative metastatic breast term=vidaza+and+breast+cancer&rank=5. Accessed January 31, 2016.
cancer (MBC) patients with resistance to non-steroidal aromatase 120. NCT01349959. Azacitidine and entinostat in treating patients with
inhibitors (PEARL). http://clinicaltrials.gov/ct2/show/NCT02028507. advanced breast cancer. https://clinicaltrials.gov/ct2/show/NCT01349959?
Accessed May 16, 2014. term=vidaza+and+breast+cancer&rank=1. Accessed January 31, 2016.
105. NCT01919229. A pharmacodynamics pre-surgical study of LEE011 in 121. Robertson JF, Ferrero JM, Bourgeois H, et al. Ganitumab with either
early breast cancer patients (MONALEESA-1). http://clinicaltrials.gov/ exemestane or fulvestrant for postmenopausal women with ad-
show/NCT01919229. Accessed May 27, 2014. vanced, hormone-receptor-positive breast cancer: a randomised,
106. NCT01958021. Study of efficacy and safety of LEE011 in post- controlled, double-blind, phase 2 trial. Lancet Oncol. 2013;14:
menopausal women with advanced breast cancer: (MONALEESA-2). 228-235.
http://clinicaltrials.gov/show/NCT01958021. Accessed May 27, 2014. 122. Dickler MN, Barry WT, Cirrincione CT, et al. Phase III trial evaluating
107. Bardia A, Modi S, Chavez-Mac M, et al. Phase Ib/II study of LEE011, the addition of bevacizumab to letrozole as first-line endocrine
everolimus, and exemestane in postmenopausal women with ER therapy for treatment of hormone-receptor positive advanced breast
+/HER2- metastatic breast cancer. J Clin Oncol. 2014;32 (suppl; cancer: CALGB 40503 (Alliance). J Clin Oncol. 2015;33 (suppl; abstr
abstr 535). 501).
108. Juric D, Ismail-Khan R, Campone M, et al. Phase Ib/II study of ribociclib 123. Martn M, Loibl S, von Minckwitz G, et al. Phase III trial evaluating the
and alpelisib and letrozole in ER+, HER2 breast cancer: Safety, addition of bevacizumab to endocrine therapy as first-line treatment
preliminary efficacy and molecular analysis. Cancer Res. 2016;76 for advanced breast cancer: the letrozole/fulvestrant and avastin
(4 suppl; abstr P3-14-01). (LEA) study. J Clin Oncol. 2015;33:1045-1052.
109. Patnaik A, Rosen LS, Tolaney SM, et al. Clinical activity of 124. Rugo HS, Delord JP, Im SA, et al. Preliminary efficacy and safety of
LY2835219, a novel cell cycle inhibitor selective for CDK4 and pembrolizumab (MK-3475) in patients with PD-L1positive, estrogen
CDK6, in patients with metastatic breast cancer. Cancer Res. 2014; receptor-positive (ER+)/HER2-negative advanced breast cancer en-
74 (suppl; abstr CT232). rolled in KEYNOTE-028. Cancer Res. 2016;76 (4 suppl; abstr S5-07).
110. NCT02102490. A study of abemaciclib (LY2835219) in participants 125. Dirix LY, Takacs I, Nikolinakos P, et al. Avelumab (MSB0010718C), an
with previously treated breast cancer that has spread (MONARCH 1). anti-PD-L1 antibody, in patients with locally advanced or metastatic
https://clinicaltrials.gov/ct2/show/NCT02102490?term=ABEMACICLIB& breast cancer: a phase Ib JAVELIN solid tumor trial. Cancer Res. 2016;
rank=16. Accessed March 3, 2016. 76 (suppl 4; abstr S1-04).
111. Patnaik A, Rosen LS, Tolaney SM, et al. LY2835219, a novel cell cycle 126. Munster PN, Hamilton EP, Franklin C, et al. Phase Ib study of LEE011
inhibitor selective for CDK 4/6, in combination with fulvestrant for and BYL719 in combination with letrozole in estrogen receptor-
patients with hormone receptor positive (HR+) metastatic breast positive, HER2-negative breast cancer (ER+, HER2- BC). J Clin Oncol.
cancer. J Clin Oncol. 2014;32 (suppl; abstr 534). 2014;32:5s (suppl; abstr 533).
112. NCT02107703. A study of abemaciclib (LY2835219) combined with 127. Andre F. UNIRAD. Randomised, double blind, multicentre phase III
fulvestrant in women with hormone receptor positive HER2 negative trial evaluating the safety and benefit of adding everolimus to ad-
breast cancer (MONARCH 2). https://clinicaltrials.gov/ct2/show/ juvant hormone therapy in women with high risk of relapse, ER+ and
NCT02107703. Accessed March 26, 2016. HER2- primary breast cancer who remain free of disease after re-
113. NCT02236572. Neoadj ph 2 AI plus everolimus in postmenopausal ceiving at least 1 year of adjuvant hormone therapy. http://www.icr.
women w/ ER pos/HER2 neg, low risk score. https://clinicaltrials.gov/ ac.uk/our-research/our-research-centres/clinical-trials-and-
ct2/show/NCT02236572. Accessed March 26, 2016. statistics-unit/clinical-trials/unirad. Accessed January 30, 2016.
114. NCT02308020. A study of abemaciclib (LY2835219) in participants with 128. NCT01674140. SWOG 1207. Phase III randomized, placebo-controlled
breast cancer, non-small cell lung cancer, or melanoma that has spread to clinical trial evaluating the use of adjuvant endocrine therapy +/2 one
the brain. https://clinicaltrials.gov/ct2/show/NCT02308020?term= year of everolimus in patients with high-risk, hormone receptor-
abemaciclib+brain&rank=1. Accessed March 3, 2016. positive and HER2/neu negative breast cancer. e3 breast cancer
115. NCT01484041. Dovitinib plus an aromatase inhibitor for metastatic study- evaluating everolimus with endocrine therapy. http://swog.org/
breast cancer. https://clinicaltrials.gov/show/NCT01484041. Accessed Visitors/ViewProtocolDetails.asp?ProtocolID=2248. Accessed January 30,
January 31, 2016. 2016.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e53


RUGO, VIDULA, AND MA

129. NCT02308020. A study of abemaciclib (LY2835219) in participants breast cancer. (BOLERO-6). https://clinicaltrials.gov/ct2/show/
with breast cancer that has spread to the brain. https://clinicaltrials. NCT01783444. Accessed March 26, 2016.
gov/ct2/show/record/NCT02308020?term=abemaciclib&rank=6. 145. NCT00570921. Study of combined fulvestrant and everolimus in
Accessed January 30, 2016. advanced/metastatic breast cancer after aromatase inhibitor failure
130. NCT02088684. Study of LEE011 with fulvestrant and BYL719 or (BRE-43). https://clinicaltrials.gov/ct2/show/NCT00570921. Accessed
BKM120 in advanced breast cancer. http://www.cancer.gov/ March 26, 2016.
clinicaltrials/search/view?cdrid=759365&version=HealthProfessional& 146. NCT02344550. Study of GnRH plus leterozole +/2 everolimus for
protocolsearchid=12773476. Accessed May 27, 2014. premenopausal women with metastatic breast cancer (LEO). https://
131. NCT01872260. Study of LEE011. BYL719 and letrozole in advanced ER+ clinicaltrials.gov/ct2/show/NCT02344550. Accessed March 26, 2016.
breast cancer. http://www.cancer.gov/clinicaltrials/search/view?cdrid= 147. NCT02404051. Fulvestrant and EVerolimus Plus EXemestane in
750319&version=HealthProfessional&protocolsearchid= metastatic breast cancer (FEVEX). https://clinicaltrials.gov/ct2/show/
12773476. Accessed May 27, 2014. NCT02404051. Accessed March 26, 2016.
132. NCT02057133. A study of LY2835219 in combination with therapies 148. NCT01797120. Study of fulvestrant +/2 everolimus in post-menopausal,
for breast cancer that has spread. https://clinicaltrials.gov/ct2/show/ hormone-receptor 1 metastatic breast ca resistant to AI (PrE0102). https://
record/NCT02057133. Accessed May 27, 2014. clinicaltrials.gov/ct2/show/NCT01797120. Accessed March 26, 2016.
133. NCT02117648. A study of LY2835219 in participants with cancer. 149. NCT02123823. A phase Ib/II randomized study of BI 836845 in com-
https://clinicaltrials.gov/ct2/show/NCT02117648. Accessed March bination with exemestane and everolimus versus exemestane and
26, 2016. everolimus alone in women with locally advanced or metastatic breast
134. NCT02441946. A neoadjuvant study of abemaciclib (LY2835219) in cancer. https://clinicaltrials.gov/ct2/show/record/NCT02123823?term5
postmenopausal women with hormone receptor positive, HER2 BI1836845&rank53. Accessed March 26, 2016.
negative breast cancer (neoMONARCH). https://clinicaltrials.gov/ct2/ 150. NCT02273973. A study of neoadjuvant letrozole 1 GDC-0032 versus
show/NCT02441946?term=abemaciclib&rank=3. Accessed May 15, letrozole 1 placebo in post-menopausal women with breast cancer
2015. (LORELEI). https://clinicaltrials.gov/ct2/show/NCT02273973. Accessed
135. Caldon CE, Sergio CM, Kang J, et al. Cyclin E2 overexpression is as- March 26, 2016.
sociated with endocrine resistance but not insensitivity to CDK2 in- 151. NCT01633060. A phase III study of BKM120 with fulvestrant in
hibition in human breast cancer cells. Mol Cancer Ther. 2012;11: patients with HR1, HER2-, AI treated, locally advanced or meta-
1488-1499. static breast cancer who progressed on or after mTORi (BELLE-3).
136. Casimiro MC, Velasco-Velazquez M, Aguirre-Alvarado C, et al. Over- https://clinicaltrials.gov/ct2/show/NCT01633060. Accessed March
view of cyclins D1 function in cancer and the CDK inhibitor landscape: 26, 2016.
past and present. Expert Opin Investig Drugs. 2014;23:295-304. 152. NCT02297438. A study of palbociclib (PD-0332991) 1 letrozole vs.
137. Finn RS, Crown JP, Lang I, et al. Final results of a randomized phase II placebo1 letrozole for 1st line treatment of Asian postmeno-
study of PD 0332991, a cyclin-dependent kinase (CDK-4/6 inhibitor), in pausal women with ER1/HER2- advanced breast cancer [PALOMA-4].
combination with letrozole vs letrozole alone for first-line treatment https://clinicaltrials.gov/ct2/show/NCT02297438. Accessed March
of ER+/HER2- advanced breast cancer (PALOMA-1; TRIO-18). Cancer 26, 2016.
Res. 2014;74 (suppl; abstr CT101). 153. NCT02296801. A phase II randomized study evaluating the biological
138. NCT01320592. PD0332991 and paclitaxel in treating patients with and clinical effects of the combination of palbociclib with letrozole
metastatic breast cancer. http://www.cancer.gov/clinicaltrials/ as neoadjuvant therapy in post-menopausal women with estrogen-
search/view?cdrid=697909&version=HealthProfessional&protocolsearchid= receptor positive primary breast cancer (PALLET). https://clinicaltrials.
12773356. Accessed May 27, 2014. gov/ct2/show/NCT02296801. Accessed March 26, 2016.
139. NCT02040857. Palbociclib and endocrine therapy in treating patients 154. NCT02690480. Comparison of efficacy and tolerability of fulvestrant1
with hormone receptor-positive stage II-III breast cancer. http:// placebo vs fulvestrant1palbociclib as first line therapy for post-
www.cancer.gov/clinicaltrials/search/view?cdrid=757030&version= menopausal women with HR1 metastatic BC treated with 5 years of
HealthProfessional&protocolsearchid=12773356. Accessed May 27, hormonal therapy remaining disease free more than 12 months after
2014. completion or have de novo metastatic disease (FLIPPER). https://
140. Byrd JC, Peterson BL, Gabrilove J, et al; Cancer and Leukemia Group B. clinicaltrials.gov/ct2/show/NCT02690480. Accessed March 26, 2016.
Treatment of relapsed chronic lymphocytic leukemia by 72-hour 155. NCT02491983. Study to evaluate the efficacy and safety of palbociclib
continuous infusion or 1-hour bolus infusion of flavopiridol: results in combination with fulvestrant or letrozole in patients with ER1,
from Cancer and Leukemia Group B study 19805. Clin Cancer Res. HER2- locally advanced or metastatic breast cancer (PARSIFAL). https://
2005;11:4176-4181. clinicaltrials.gov/ct2/show/NCT02491983. Accessed March 26, 2016.
141. NCT01709370. Letrozole and CDK 4/6 inhibitor for ER positive, HER2 156. NCT02384239. A study of palbociclib in combination with fulvestrant
negative breast cancer in postmenopausal women. https://clinicaltrials. or tamoxifen as treatment for metastatic breast cancer. https://
gov/ct2/show/NCT01709370. Accessed March 26, 2016. clinicaltrials.gov/ct2/show/NCT02384239. Accessed March 26, 2016.
142. NCT02102490. A study of abemaciclib (LY2835219) in participants 157. NCT02592746. A study of palbociclib with exemestane plus goserelin
with previously treated breast cancer that has spread (MONARCH 1). versus capecitabine in premenopausal women with HR1 MBC. https://
https://clinicaltrials.gov/ct2/show/NCT02102490. Accessed March clinicaltrials.gov/ct2/show/NCT02592746. Accessed March 26, 2016.
26, 2016. 158. NCT02675231. A study of abemaciclib (LY2835219) in women with
143. NCT01698918. Open-label, phase II, study of everolimus plus letrozole HR1, HER21 locally advanced or metastatic breast cancer (monarcHER).
in postmenopausal women with ER1, HER2- metastatic or locally https://clinicaltrials.gov/ct2/show/NCT02675231. Accessed March
advanced breast cancer (Bolero-4). https://clinicaltrials.gov/ct2/ 26, 2016.
show/NCT01698918. Accessed March 26, 2016. 159. NCT02422615. Study of efficacy and safety of LEE011 in postmenopausal
144. NCT01783444. A phase II study of everolimus in combination with women with advanced breast cancer (MONALEESA-3). https://clinicaltrials.
exemestane versus everolimus alone versus capecitabine in advance gov/ct2/show/NCT02422615. Accessed March 26, 2016.

e54 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


BREAST CANCER

Treatment of Premenopausal
Women With Endocrine-Sensitive
Breast Cancer

CHAIR
Nancy E. Davidson, MD
University of Pittsburgh Cancer Institute
Pittsburgh, PA

SPEAKERS
Hatem A. Azim Jr., MD, PhD
Institut Jules Bordet
Brussels, Belgium

Kathryn J. Ruddy, MD, MPH


Mayo Clinic
Rochester, MN
TREATING PREMENOPAUSAL ENDOCRINE-SENSITIVE BREAST CANCER

Challenges in Treating Premenopausal Women with


Endocrine-Sensitive Breast Cancer
Hatem A. Azim Jr., MD, PhD, Nancy E. Davidson, MD, and Kathryn J. Ruddy, MD, MPH

OVERVIEW

For the hundreds of thousands of premenopausal women who are diagnosed annually with endocrine-sensitive breast
cancer, treatment strategies are complex. For many, chemotherapy may not be necessary, and endocrine therapy decision
making is paramount. Options for adjuvant endocrine regimens include tamoxifen for 5 years, tamoxifen for 10 years,
ovarian function suppression (OFS) plus tamoxifen for 5 years, and OFS plus an aromatase inhibitor for 5 years. There are
modest differences in efficacy between these regimens, with a benefit from OFS most obvious among patients with higher-risk
disease; therefore, choosing which should be used for a given patient requires consideration of expected toxicities and
patient preferences. An aromatase inhibitor cannot be safely prescribed without OFS in this setting. Additional research is
needed to determine whether genomic tests such as Prosigna and Endopredict can help with decision making about
optimal duration of endocrine therapy for premenopausal patients. Endocrine therapy side effects can include hot flashes,
sexual dysfunction, osteoporosis, and infertility, all of which may impair quality of life and can encourage nonadherence
with treatment. Ovarian function suppression worsens menopausal side effects. Hot flashes tend to be worse with
tamoxifen/OFS, whereas sexual dysfunction and osteoporosis tend to be worse with aromatase inhibitors/OFS. Preg-
nancy is safe after endocrine therapy, and some survivors can conceive naturally. Still, embryo or oocyte cryopreservation
should be considered at the time of diagnosis for patients with endocrine-sensitive disease who desire future childbearing,
particularly if they will undergo chemotherapy.

I n the United States, approximately 20% of patients with


breast cancer are diagnosed before age 50.1 This translates
into almost 50,000 new patients per year, which well ex-
have unique challenges, most notably fertility-related issues,
poorer perceived quality of life, and difficulties adhering
to prescribed endocrine therapy. Therefore, treating pre-
ceeds the total number of women diagnosed with acute menopausal women with early breast cancer, particularly
leukemia and brain and stomach cancers in all ages com- those with endocrine-sensitive disease, is rather complex.
bined.1 A higher proportion of patients with breast cancer
are diagnosed at a younger age in other parts of the world,
such as Africa and the Middle East, where the average age of HOW TO CHOOSE ENDOCRINE THERAPY FOR
developing breast cancer hardly exceeds 50.2 PREMENOPAUSAL PATIENTS?
Over the past decade, several studies have indicated In recent years, several trials have evaluated different
that diagnosis at a younger age is associated with poorer strategies of adjuvant endocrine therapy for premenopausal
prognosis,3-5 especially for women with endocrine-sensitive patients. These include tamoxifen for 5 years, tamoxifen for
disease.3,5 Compared with patients with breast cancer who 10 years, OFS plus tamoxifen for 5 years, and OFS plus an
are postmenopausal, younger patients are more commonly aromatase inhibitor for 5 years. As reviewed below, modest
diagnosed with highly proliferative estrogen receptor differences between these options in terms of long-term
positive tumors (i.e., luminal-B breast cancer).6 Further- outcome have been reported. These approaches have different
more, these tumors are enriched for molecular aberrations toxicity profiles. Deciding which approach should be used for
that could render them more aggressive, including high a given patient requires consideration of risk of recurrence,
expression of stem cell markers and high prevalence of fertility considerations, and patient preferences regarding
GATA3 mutations,3,7 the latter of which has been implicated expected toxicities.8 Unfortunately, premenopausal patients
in endocrine resistance. In addition, younger patients often with breast cancer have been found to have poor persistence

From the Department of Medicine, Institut Jules Bordet, Universite Libre de Bruxelles, Brussels, Belgium; University of Pittsburgh Cancer Institute and UPMC Cancer Center, Pittsburgh,
PA; Department of Oncology, Mayo Clinic, Rochester, MN.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Hatem A. Azim Jr., MD, PhD, Institut Jules Bordet, Boulevard de Waterloo, 121, 1000 Brussels, Belgium; email: hatemazim@icloud.com.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 23


AZIM, DAVIDSON, AND RUDDY

with endocrine therapy.9,10 Some studies have shown that only study did not accrue as planned, and it closed early with only
50% of women complete the 5 years of scheduled tamoxifen.9 345 patients. Notably, 85% of patients had T1 disease and,
Therefore, it is very important for clinicians to engage young per protocol, none were treated with adjuvant chemo-
patients in therapeutic decision making and to discuss toxicities therapy. This underpowered study showed no difference in
and expectations. Salient points pertaining to the use of the 10-year disease-free survival (DFS; 85.9% vs. 87.1%; p = .62)
different endocrine therapy options are discussed in this article. or overall survival (OS; 92.5% vs. 92.4%; p = .67) between
Figure 1 summarizes a schema to help optimize treatment both arms. The Suppression of Ovarian Function Trial (SOFT)
decisions in daily practice. was a larger, properly powered trial that included 2,033
patients who were randomly assigned to either tamoxifen
with OFS or tamoxifen alone.14 In this trial, the main analysis
Value of Ovarian Function Syndrome indicated no significant difference in 5-year DFS between the
Induction of amenorrhea for premenopausal patients with OFS plus tamoxifen and tamoxifen alone arms (84.7% vs.
endocrine-sensitive breast cancer has consistently been 86.6%; p = .10). Subgroup analyses suggested a benefit with
shown to reduce risk of recurrence.11,12 This is true for OFS the addition of OFS in women who had received chemo-
by luteinizing hormone-releasing hormones (LHRH) agonists therapy (who were more likely to have node-positive dis-
or surgical or radiation ablation. However, because adjuvant ease) and those younger than age 35 at diagnosis. Of those
tamoxifen was not routinely administered to patients who diagnosed with breast cancer younger than age 35 (94% of
were premenopausal until the early 2000s, uncertainty whom received chemotherapy), the proportion who were
remained regarding the benefit of amenorrhea induction in free of breast cancer for at least 5 years was 67.7% with
the presence of tamoxifen. Within the last several years, two tamoxifen alone and 78.9% with tamoxifen with OFS, sug-
studies have provided important insights on this issue. The gesting that OFS cut the risk of recurrence by approximately
first is the E-3193, INT-0142 trial, which randomly assigned one-third in this group.
node-negative, estrogen receptorpositive patients to ta- These results underscore several important points. First,
moxifen or OFS plus tamoxifen, both given for 5 years.13 This endocrine therapy alone, without chemotherapy, is a very
effective option for selected premenopausal women with
early-stage breast cancer. Second, in older premenopausal
women with low-risk early-stage breast cancer, OFS adds
KEY POINTS little if any benefit to 5 years of tamoxifen. Finally, induction
of amenorrhea is therapeutic for younger premenopausal
Several adjuvant endocrine therapy options are
currently available for premenopausal patients with
patients with higher-risk disease.
endocrine-responsive early-stage breast cancer, and it
can be challenging to decide which is best for a given
patient. Tamoxifen Versus Aromatase Inhibitors
Recent evidence from randomized trials indicates that a In the postmenopausal setting, aromatase inhibitors have
substantial proportion of premenopausal women with largely replaced tamoxifen as the standard endocrine adju-
early-stage breast cancer can achieve excellent vant therapy.15 Among premenopausal patients, the final
outcomes with endocrine therapy only, in the absence results of the ABCSG-12 trial did not show a difference in DFS
of chemotherapy, challenging the notion that adjuvant between tamoxifen and anastrozole when administered for
chemotherapy should be considered in all young 3 years in combination with LHRH agonists, but OS was inferior
patients with breast cancer.
in the aromatase inhibitor arm.16 This raised concerns about
Careful attention must be paid to the toxicities of
endocrine therapies including hot flushes, sexual
the use of aromatase inhibitors with OFS among pre-
dysfunction, osteoporosis, and infertility, which could all menopausal patients. Recently, the results of a joint analysis
reduce adherence to endocrine therapy and of the long-awaited SOFT and TEXT trials were published.17 In
detrimentally impact patients quality of life. total, 4,690 women were randomly assigned to receive either
Pregnancy following endocrine-responsive breast tamoxifen or exemestane in addition to OFS for 5 years.
cancer is safe, although challenges exist in the era of Approximately 40% of patients had node-positive disease and
extended adjuvant therapy. A prospective trial currently nearly 50% received adjuvant chemotherapy. The results
is addressing the feasibility of temporary interruption of demonstrated a 28% reduction in DFS hazard ratio (HR) at
endocrine treatment to allow pregnancy. 5 years with exemestane compared to tamoxifen (91.1% vs.
Embryo or oocyte cryopreservation prior to 87.3%, p , .001), with no difference in OS.
chemotherapy is the best option to preserve fertility for
Although in line with the hypothesis and results observed
premenopausal patients with breast cancer, even those
who have endocrine-sensitive disease. Evolving data
in the postmenopausal setting, the results of this pooled
suggest that LHRH agonists could reduce risk of analysis contrasted with those observed in the ABCSG-12
chemotherapy-induced amenorrhea and, thus, may trial. However, it is important to note that the ABCSG-12 trial
serve as a valid alternative to embryo or oocyte was smaller in size and administered therapy for only
cryopreservation if these are not feasible. 3 years, which does not reflect todays standard practice. An
unplanned analysis of ABCSG-12 suggested that the worse

24 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


TREATING PREMENOPAUSAL ENDOCRINE-SENSITIVE BREAST CANCER

FIGURE 1. Schema to Guide Decision Making of Primary Systemic Therapy of Premenopausal Women With
Endocrine ReceptorPositive (HER-Negative) Breast Cancer

Abbreviations: AI, aromatase inhibitor; ER, estrogen receptor; LHRH, luteinizing hormone-releasing hormone; OFS, ovarian function suppression; PR, progesterone receptor; y, years.
*No evidence on extended adjuvant therapy in women treated with adjuvant OFS/AI during first 5 years.
**Clinical utility of genomic tests to guide extended endocrine therapy is yet to be proven.

outcome observed in the aromatase inhibitor arm was re- Thus, although the combination of OFS and exemestane
stricted to patients who were overweight or obese18; these may be the most effective 5-year endocrine therapy regi-
patients constituted nearly one-third of the study population. men, adequate suppression of estradiol levels appears to be
This was hypothesized to reflect incomplete estrogen level more challenging for patients who are overweight or obese,
suppression by aromatase inhibitor for these women, which in whom ABCSG-12 suggests that OFS with tamoxifen may
has been seen among postmenopausal patients who were produce equal if not better outcomes. Yet it is unclear
obese,19 or incomplete ovarian suppression by LHRH agonists whether measurement of estradiol levels during LHRH ag-
in larger patients. Research is limited regarding the impact of onists therapy will be clinically useful in guiding the choice of
body weight on the efficacy of OFS, but it is possible that adjuvant endocrine therapy in daily practice.24
surgical or radiation ablation would be more effective than One final point that should be underscored is the very
LHRH agonists in women who are overweight and obese.20 favorable outcome of the 50% of women in the joint analysis
Alternatively, cross talk between estrogen signaling and in- who did not receive adjuvant chemotherapy. Although these
sulin growth factor signaling may play a role in the impact of patients were mostly low risk by virtue of stage, their 5-year
weight on the efficacy of OFS and AI.21 A randomized pro- DFS was greater than 93%. Because the benefit of adjuvant
spective study from Japan in the neoadjuvant setting showed chemotherapy is largely a reduction in risk of recurrence
superiority of aromatase inhibitor plus OFS over tamoxifen during the first few years after diagnosis, it is likely that
plus OFS with a near doubling of clinical response rate on chemotherapy would not have been beneficial in the ma-
MRI.22 However, in this study, only 17% of patients were jority of these patients. These results strongly challenge the
overweight or obese. Correlation between body mass index notion that young premenopausal women should be offered
(BMI) and outcome in the joint analysis of SOFT and TEXT is yet more chemotherapy, and they indicate that endocrine
to be published, but high BMI was shown to be associated therapy alone remains a very effective option for a large
with high on-treatment estradiol levels in SOFT patients fraction of young patients with endocrine-sensitive breast
randomly assigned to exemestane and OFS.23 Although most cancer.
SOFT patients maintained a low estradiol level on treatment,
34% of patients on OFS and aromatase inhibitor had high
estradiol levels at some point during the 12 months after 10-Year Versus 5-Year Schedule
study initiation. No correlation with long-term outcome was Unlike estrogen receptornegative tumors, endocrine-
seen, perhaps because of the small number of patients in this sensitive cancers have a propensity for late relapse. 25
analysis (86 patients on LHRH agonist plus exemestane). Therefore, extending treatment beyond 5 years seems

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 25


AZIM, DAVIDSON, AND RUDDY

logical to improve the outcomes of these patients. In the report, and thus validation of the role of genomic tests to
ATLAS and aTTom trials,26,27 taking tamoxifen for 10 years inform on the benefit of this approach is warranted. Until then,
was shown to be superior to the standard 5-year schedule, no solid statements could be made regarding their role in
and benefit was observed independent of menopausal guiding the use of extended endocrine therapy.
status. Given the established role of aromatase inhibitors in
treating postmenopausal women during the first 5 years,
these results appear to be most relevant for the treatment
TOXICITIES OF ENDOCRINE THERAPY FOR
of premenopausal women for whom tamoxifen is still
PREMENOPAUSAL WOMEN
Premenopausal women with breast cancer who require
considered a mainstay treatment. Another study, MA17,
endocrine therapy are vulnerable to a variety of treatment
which previously showed the added benefit of extended
toxicities. Although excess tamoxifen-associated uterine
therapy with letrozole after 5 years of tamoxifen, demon-
cancer diagnoses and thromboembolic events have not been
strated that this benefit is mostly observed in women who
observed among patients younger than age 50, some side
were premenopausal at the time they initiated tamoxifen
effects of endocrine therapy are more severe for young
but became postmenopausal during its administration.28
patients, particularly if OFS is used. These side effects are
These results in aggregate underscore the value of con-
discussed below.
sidering extending endocrine therapy to 10 years for pre-
menopausal women. However, the question remains as to
whether all women need this approach. This question is Hot Flashes
relevant as prolonged treatment increases toxicity and could Hot flashes are problematic for the majority of young re-
impair quality of life. Acknowledging the adherence issues cipients of adjuvant endocrine therapy,33 especially with the
highlighted earlier, it is important to define tools that can addition of OFS,34 and perhaps more substantially with
identify the subsets of patients who will benefit the most tamoxifen than aromatase inhibitors. These hot flashes
from this approach and spare others the burden of long-term often are accompanied by night sweats, which can interrupt
treatment. sleep and impair quality of life. In 345 young women enrolled
Recently, several genomic tests, particularly Prosigna and in the E-3193, INT-0142 phase III trial, grade 3 hot flashes
Endopredict, have been evaluated for their ability to predict occurred in 4.7% of those receiving tamoxifen monotherapy
long-term recurrence in women who were treated with compared with 16.1% of those receiving tamoxifen with
5 years of adjuvant endocrine therapy in the context of OFS.13 In SOFT, grade 3 or 4 hot flashes occurred in 7.6% of
randomized phase III trials.29-31 Notably, these trials were patients assigned to receive tamoxifen, compared with
conducted among postmenopausal women and genomic 13.2% of those assigned to OFS and tamoxifen.14 However,
testing was not part of the initial study plan. In the ABCSG-8 the use of an aromatase inhibitor instead of tamoxifen with
trial, which randomly assigned postmenopausal patients to OFS was associated with less sweating (reported by 54.5%
receive either tamoxifen or anastrozole, approximately vs. 59%, respectively).17
1,300 patients were free of relapse at completion of 5 years
of endocrine therapy and were analyzed by Prosigna test-
Sexual Dysfunction
ing.29 None of these patients received adjuvant chemo-
In contrast, sexual dysfunction is most severe for patients
therapy and 30% were node-positive. Thirty-seven percent
who received an aromatase inhibitor with OFS.17 In the TEXT
of patients were classified as low risk by the genomic test, of
trial, 52.4% of 2,318 women who received exemestane with
whom only 2.4% developed distant recurrence. In contrast,
OFS and 47.4% of 2,325 women who received tamoxifen
17.5% of the 30% of patients classified as high risk developed
with OFS had vaginal dryness. Forty-five percent of the
distant recurrence between years 5 and 15. Similar results
exemestane/OFS group and 40.9% of the tamoxifen/OFS
were observed in a combined analysis of the ABCSG-8 and
group had decreased libido, although dyspareunia was re-
ATAC datasets.30 Endopredict showed comparable findings
ported in 30.5% and 25.8%, respectively. Of note, ovarian
when tested on the ABCSG-6 and ABCSG-8 trial. In 1,702
dysfunction secondary to chemotherapy can cause similar
patients free of relapse after 5 years of endocrine therapy,
vaginal symptoms and sexual dysfunction.35 Tamoxifen
Endopredict identified 64% as low risk, with an absolute risk
alone is less problematic,13 and it may even improve vaginal
of recurrence of 1.8% at 10 years.31
dryness in some survivors by causing vaginal discharge.34
Thus, it appears that extending endocrine therapy beyond
Sexual dysfunction in survivors of breast cancer can be
5 years for women in the low-risk group category is very
compounded by body image issues associated with local
unlikely to add any benefit because their risk of recurrence is
therapies and the weight gain that many experience during
negligible. On the other hand, extended therapy could be more
systemic chemotherapy.
useful for patients classified as high risk. Of note, none of these
studies included premenopausal patients, yet data with other
genomic tests such as Oncotype Dx32 and an in silico analysis3 Depression and Cognitive Dysfunction
indicated that the prognostic performance of genomic tests SOFT results showed a nonstatistically significant trend
is largely comparable irrespective of age. Currently, several toward more mood impairment in those who received ta-
studies investigating extended endocrine therapy have yet to moxifen with OFS than in those who received tamoxifen

26 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


TREATING PREMENOPAUSAL ENDOCRINE-SENSITIVE BREAST CANCER

monotherapy, with depressive symptoms identified in received OFS with tamoxifen compared with those who
51.9% and 46.6%, respectively.14 An aromatase inhibitor received tamoxifen monotherapy,14 identified in 23.3%
does not appear to cause more depression than tamoxifen.17 versus 17.2% of patients, respectively. But among TEXT
Treatment of depression can be complex for patients re- and SOFT participants who received OFS, hypertension
ceiving tamoxifen because of concerns that certain anti- was equally likely with tamoxifen versus exemestane.17
depressants (e.g., fluoxetine, paroxetine, and sertraline) Thrombosis or embolism occurred in 2.2% (95% CI, 1.62.8)
may reduce the efficacy of tamoxifen by inhibiting of patients who received OFS plus tamoxifen and 1.0% (95%
CYP2D6.36,37 One study found that survivors of breast cancer CI, 0.71.5) of those who received OFS plus aromatase
who took paroxetine with tamoxifen had increased breast inhibitors.17
cancer mortality, although another study identified no im-
pact of CYP2D6 inhibitor use on recurrence risk for patients
taking tamoxifen. Possibly, the CYP2D6 genotype is partic- PREGNANCY AFTER ESTROGEN
ularly relevant for tamoxifen efficacy for premenopausal RECEPTORPOSITIVE BREAST CANCER: SAFETY
patients,38 but definitive studies of this issue are needed. AND FEASIBILITY IN THE ERA OF EXTENDED
Cognitive dysfunction related to endocrine therapy is ADJUVANT THERAPY
understudied among premenopausal patients. However, in Infertility remains a main concern for young patients with
the ZIPP trial, patient self-evaluation of memory and con- breast cancer. A recent prospective study including more
centration was similar for patients with breast cancer who than 600 patients demonstrated that up to 50% of young
received chemotherapy with or without goserelin, goserelin patients with breast cancer are concerned about infertility,
plus tamoxifen, or tamoxifen alone.34 In addition, pre- and, in a substantial fraction, this has an impact on treat-
liminary results at 1 year from 86 of a planned 321 par- ment decisions.44
ticipants in the cognitive function substudy of SOFT The safety of subsequent pregnancy in women with a
demonstrated no worse objective cognitive function in history of estrogen receptorpositive breast cancer con-
those who received tamoxifen plus OFS compared with tinues to be an area of concern. In a recent survey, ap-
tamoxifen monotherapy.39 proximately 40% of oncologists believed that hormonal
changes secondary to pregnancy could increase risk of re-
currence particularly during the first 2 years after cancer
Bone Health
diagnosis.45 Although this question has been addressed in
Tamoxifen is known to improve bone mineral density for
several studies over the past 3 to 4 decades,46-48 definitive
postmenopausal women, but it can thin bones in pre-
answers were complicated by lack of information on es-
menopausal women.40 This is likely because of its mixed
trogen receptor status in many of these studies and
agonist/antagonist effect on estrogen receptor. Therefore,
methodological concerns about patient selection. In 2013, a
bone loss is a concern in most young patients with breast
large multicenter study was published in which the long-
cancer, not just in those who experience chemotherapy-
term outcome of 333 women who became pregnant after
related premature menopause or receive OFS. A recent
breast cancer diagnosis was compared with 874 survivors of
study of survivors of breast cancer who were premenopausal
breast cancer who did not become pregnant.49 Both groups
at the time of breast cancer diagnoses and had no known
were matched according to relevant tumor characteristics
history of osteoporosis revealed a 12% fracture risk during a
and use of adjuvant systemic therapy. The primary endpoint
median 3.1-year follow-up after the completion of 56 years
was 5-year DFS of the estrogen receptorpositive cohort. In
of endocrine therapy. This was similar to the 15% fracture
this cohort, which comprised 194 pregnant and 492 non-
risk this study identified in survivors of postmenopausal
pregnant patients, no difference in 5-year DFS was observed
breast cancer.41 However, the fractures in survivors of
by pregnancy status (HR 0.91; 95% CI, 0.671.24; p = .55).
premenopausal breast cancer were more commonly toe/
Secondary endpoints, which included DFS in estrogen
finger fractures, although those in postmenopausal women
receptornegative patients and OS, also did not differ be-
were more commonly hip fractures. The median time to
tween the pregnant and nonpregnant groups. These results
first fracture was shorter in premenopausal than in post-
underscore the safety of pregnancy following breast cancer
menopausal women (1.4 vs. 2.4 years; p = .01). Zoledronic
irrespective of the estrogen receptor status of the primary
acid has been proven an effective preventative agent against
tumor. Similar to other studies,47 approximately 30% of
bone loss in this population.42,43 The lifetime fracture risk of
patients underwent abortion in this study. A preplanned
young recipients of endocrine therapy deserves additional
subgroup analysis showed that abortion did not have any
study in the new era of increased use of OFS and prolonged
impact on patient prognosis, indicating that abortion should
duration of treatment.
not be recommended for therapeutic purposes.
However, with the increasing use of long-term endocrine
Cardiovascular Toxicity therapy up to 10 years, the feasibility of becoming pregnant
Although it occurs at low frequency in young patients, after completion of 510 years of endocrine therapy is a
cardiovascular toxicity also must be considered. Hyperten- major challenge. This raises the question of whether tem-
sion developed more frequently in SOFT participants who porary interruption of endocrine therapy to allow pregnancy

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 27


AZIM, DAVIDSON, AND RUDDY

is safe. This approach is sometimes considered in selected Potential Detrimental Impact on Breast Cancer
cases, particularly for patients with low risk of recurrence or Outcome Particularly in Patients With Estrogen
advanced age at the time of diagnosis. Presently, it is not ReceptorPositive Disease
known whether this approach is detrimental because it was This is perhaps the greatest concern when considering assisted
not investigated in any of the reported studies. As such, the reproduction in young patients with breast cancer, as this
first international prospective trial (POSITIVE) was recently procedure is associated with significant rise in serum estradiol
launched to address the safety of temporary interruption of levels. Despite the fact that estradiol peaks only last for a few
endocrine therapy to allow pregnancy in young women days, defining protocols that would reduce estradiol peaks
with estrogen receptorpositive breast cancer who wish to without majorly compromising oocyte yield is desirable. Oktay
become pregnant.50 This trial allows patients to receive et al have performed a series of studies investigating the
1830 months of endocrine therapy. Patients with adequate addition of letrozole to standard ovarian stimulation protocols
ovarian function can temporarily interrupt endocrine therapy to reduce estradiol peaks.60,61 They showed that this strategy
for up to 2 years to allow pregnancy with or without breast- was associated with a comparable oocyte yield and significantly
feeding. Afterward, they are encouraged to resume endocrine lower estradiol peaks. In one study, the mean estradiol level at
therapy for a total course of 510 years according to local human chorionic gonadotropin (known as hCG) triggering was
practice. POSITIVE is sponsored by the International Breast 483.4 pg/mL with letrozole compared with 1,464 pg/mL for
Cancer Study Group and is performed in collaboration with the standard protocol (p , .001), with similar numbers of
ALLIANCE in the United States. This study is expected to recruit mature oocytes (8.7 vs. 9.7; p = .43) and fertilization rates
approximately 500 patients, which would provide solid evi- (74.1% vs. 73.2%; p = .71). In addition, there was no difference
dence for the safety and feasibility of endocrine therapy in- in days needed for stimulation, which was approximately
terruption to allow pregnancy. 12 days.61 Other studies have confirmed these results.62,63 In
terms of fertility and pregnancy outcome, recent results from
ASSISTED REPRODUCTION TO BECOME 131 patients with breast cancer who underwent ovarian
PREGNANT AFTER BREAST CANCER: TIMING stimulation with letrozole before initiating adjuvant chemo-
AND IMPACT ON OUTCOME therapy showed that 40 patients attempted to transfer
Given the progressive decline in ovarian function with age51 embryos back at an average time of 5.2 years following initial
and the detrimental impact of chemotherapy on ovarian embryo cryopreservation. The overall live birth rate was 45%,
function,52 fertility preservation must be considered and which is comparable to figures observed in the general pop-
offered soon after breast cancer diagnosis to improve the ulation of women of the same age. Importantly, no congenital
chances of future conception. Ovarian stimulation for oocyte malformations were observed in 25 children after a mean
or embryo cryopreservation is a standard procedure. It is follow-up of 40 months.64
currently endorsed by several guidelines for patients who To provide more robust evidence regarding the safety of
are newly diagnosed with breast cancer and are also con- this approach, a prospective study was initiated to eval-
sidering future pregnancy.53-55 However, several concerns uate the association between using ovarian stimulation
hinder the wide application of this approach. with letrozole and long-term breast cancer outcome.62 In
this study, 337 young patients with breast cancer (median
Potential Delay of Adjuvant Systemic Therapy age 35) were included, of whom 120 underwent ovarian
Optimally, ovarian stimulation should be performed before stimulation with letrozole. At a median follow-up of
the initiation of adjuvant therapy to precede the de- 5 years, there was no difference in DFS between patients
struction of oocytes by cytotoxic agents. Standard stimu- who underwent stimulation and those who did not.65 No-
lation protocols are classicaly applied during the first days tably, in this study, patients who underwent stimulation
of the menstrual cycle, which can result in a delay in ini- initiated adjuvant chemotherapy later compared with the
tiation of chemotherapy as some woman can have to wait control group (45 days vs. 33 days; p , .01). This is not
up to 3 weeks for stimulation. Such a delay could be po- unexpected because this study did not offer random stimu-
tentially detrimental to patient outcome because the in- lation protocols. A subsequent analysis investigating patient
terval between surgery and initiation of chemotherapy is outcome according to the number of ovarian stimulation
crucial, particularly in treating young patients with breast cycles (one vs. two cycles) showed no difference in patient
cancer.56 Over the past 5 years, several studies were prognosis with better fertility preservation cycle outcome in
published on random stimulation protocols, in which ovarian those undergoing two cycles in terms of number of mature
stimulation is performed at any time during the menstrual oocytes (10.3 vs. 6.2; p = .004), fertilized oocytes (7.4 vs. 4.4;
cycle.57-59 The number of days required for ovarian stimu- p = .04) and embryos (6.4 vs. 3.7; p =.019).66
lation is slightly longer using the random protocol, on av- Although more data on larger series of patients with
erage 1 to 2 days more, yet the oocyte yield is almost breast cancer are still needed to establish the safety and
identical. This approach increases the feasibility of per- effectiveness of using ovarian stimulation with letrozole
forming ovarian stimulation prior to starting systemic in newly diagnosed patients, this approach is currently
therapy without substantial delay. preferred.

28 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


TREATING PREMENOPAUSAL ENDOCRINE-SENSITIVE BREAST CANCER

Feasibility and Safety of Ovarian Stimulation for p = .04), although absolute numbers remain low (33 vs. 19
Patients Already Treated With Chemotherapy pregnancies) in the combined results from five different
The postchemotherapy setting is suboptimal for ovarian trials.72
stimulation because of the detrimental effects of adjuvant Although these data suggest that giving LHRH agonists
systemic therapy on ovarian function.52 Nevertheless, often before and during chemotherapy could improve chances of
we are confronted by situations in which a patient did not future fertility for young patients with breast cancer,
freeze embryos or oocytes before chemotherapy initiation several important considerations should be taken into
and wants to become pregnant but requires assisted re- account. First, these trials evaluated chemotherapy-
production to conceive. In the one study reported in this induced amenorrhea, which is an imperfect surrogate for
setting, 25 women underwent ovarian stimulation some adequate ovarian function. Previous studies have shown
years after primary chemotherapy to conceive. Their that even women who resume menses after chemo-
outcome was compared with more than 170 women therapy have reduced ovarian reserve73 and will most
who had spontaneous pregnancy after breast cancer likely develop early menopause.74 Chemotherapy has
diagnosis.67 Approximately 50% of patients had endocrine- been shown to induce structural changes in the ovary
sensitive disease. At a median follow-up of 4.5 years after including hyalinization of cortical vessels, collagen de-
conception, no difference in breast cancer outcome was position, and cortical fibrosis, even in women who
observed between the two groups. This small study sug- resume menses after chemotherapy.75 Whether the co-
gests that this approach may be considered in selected administration of LHRH agonists avoids any of these phe-
patients who did not have fertility preservation at di- nomena remains unclear. Second, long-term data are scant.
agnosis, completed their adjuvant therapy, and cannot The PROMISE trial did report that the 5-year cumulative
conceive spontaneously. incidence estimate of resumption of menses was 72.6% in
the LHRH agonist group versus 64% in the control group
USE OF CONCOMITANT ADMINISTRATION OF (age-adjusted HR 1.48; 95% CI, 1.121.95; p = .006).76
LHRH AGONISTS WITH CHEMOTHERAPY TO Unfortunately, none of the published studies has ade-
PRESERVE FERTILITY quate evaluation of ovarian function parameters over
Several randomized studies have addressed the role of LHRH time.
agonists as a mean of preserving fertility with conflicting An individual-patient meta-analysis including all ran-
results.68-71 Table 1 summarizes results from the main trials, domized trials of LHRH agonist in this setting currently is
the largest of which, PROMISE and POEMS, showed reduced underway. This will allow subgroup analyses according to
rates of chemotherapy-induced amenorrhea with the con- age and estrogen receptor status. Yet based on current data,
current administration of LHRH agonists.70,71 A recent meta- LHRH agonists can be considered during chemotherapy to
analysis of published data showed higher odds of achieving potentially preserve the fertility of women in whom ovarian
pregnancy as well (odds ratio 1.83; 95% CI, 1.023.28; stimulation was not possible.

TABLE 1. Main Randomized Trials That Investigated the Role of LHRH Agonist to Reduce Chemotherapy-Induced
Amenorrhea

PROMISE71,76 GBG 37 ZORO68 NCT0009084469 POEMS70


Country Italy Germany United States International
Number of Patients 281 61 49 135
Median Age 39 36 39 38
Chemotherapy Any chemotherapy AC-T AC-T or FEC Any chemotherapy
LHRH Agonists/28d Triptorelin Goserelin Triptorelin Goserelin
ER Status Any Negative Any Negative
Median Follow-up 7.3 years NR 1.5 years 4.1 years
Primary Endpoint (POF defined as) Amenorrhea + postmenopausal Amenorrhea at 6 Amenorrhea at Amenorrhea + postmenopausal
FSH and E2 at 12 months months 12 months FSH at 24 months
Absence of POF (%) LHRH: 91% LHRH: 56% LHRH: 88% LHRH: 92%
No LHRH: 74% No LHRH: 70% No LHRH: 90% No LHRH: 78%
Positive Negative Negative Positive
Pregnancies Achieved (number) LHRH: 8 LHRH: 1 LHRH: 0 LHRH: 22
No LHRH: 3 No LHRH: 1 No LHRH: 2 No LHRH: 12
Abbreviations: AC, doxorubicin, cyclophosphamide; E2, estradiol; ER, estrogen receptor; FEC, 5-fluorouracil, epirubicin, cyclophosphamide; FSH, follicle-stimulating hormone; LHRH,
luteinizing hormone-releasing hormone; POF, premature ovarian failure; T, taxane.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 29


AZIM, DAVIDSON, AND RUDDY

References
1. DeSantis C, Ma J, Bryan L, et al. Breast cancer statistics, 2013. CA Cancer 19. Ligibel JA, Winer EP. Aromatase inhibition in obese women: how much
J Clin. 2014;64:52-62. is enough? J Clin Oncol. 2012;30:2940-2942.
2. DeSantis CE, Bray F, Ferlay J, et al. International variation in female 20. Dowsett M, Lnning PE, Davidson NE. Incomplete estrogen suppression
breast cancer incidence and mortality rates. Cancer Epidemiol Bio- with gonadotropin-releasing hormone agonists may reduce clinical
markers Prev. 2015;24:1495-1506. efficacy in premenopausal women with early breast cancer. J Clin Oncol.
3. Azim HA, Jr, Michiels S, Bedard PL, et al. Elucidating prognosis and Epub 2016 Jan 4.
biology of breast cancer arising in young women using gene expression 21. Hamelers IH, Steenbergh PH. Interactions between estrogen and
profiling. Clin Cancer Res. 2012;18:1341-1351. insulin-like growth factor signaling pathways in human breast tumor
4. Kim EK, Noh WC, Han W, et al. Prognostic significance of young age (,35 cells. Endocr Relat Cancer. 2003;10:331-345.
years) by subtype based on ER, PR, and HER2 status in breast cancer: 22. Masuda N, Sagara Y, Kinoshita T, et al. Neoadjuvant anastrozole versus
a nationwide registry-based study. World J Surg. 2011;35:1244-1253. tamoxifen in patients receiving goserelin for premenopausal breast
5. Cancello G, Maisonneuve P, Rotmensz N, et al. Prognosis and adjuvant cancer (STAGE): a double-blind, randomised phase 3 trial. Lancet Oncol.
treatment effects in selected breast cancer subtypes of very young 2012;13:345-352.
women (,35 years) with operable breast cancer. Ann Oncol. 2010;21: 23. Bellet M, Gray KP, Francis PA, et al. Twelve-month estrogen levels in
1974-1981. premenopausal women with hormone receptor-positive breast cancer
6. Azim HA, Jr, Partridge AH. Biology of breast cancer in young women. receiving adjuvant triptorelin plus exemestane or tamoxifen in the
Breast Cancer Res. 2014;16:427. Suppression of Ovarian Function Trial (SOFT): the SOFT-EST substudy.
7. Azim HA, Jr, Nguyen B, Brohee S, et al. Genomic aberrations in young J Clin Oncol. Epub 2016 Jan 4.
and elderly breast cancer patients. BMC Med. 2015;13:266. 24. Burstein HJ, Lacchetti C, Anderson H, et al. Adjuvant endocrine therapy
8. Mathew A, Davidson NE. Adjuvant endocrine therapy for pre- for women With hormone receptor-positive breast cancer: American
menopausal women with hormone-responsive breast cancer. Breast. Society of Clinical Oncology clinical practice guideline update on ovarian
2015;24(Suppl 2):S120-S125. suppression. J Clin Oncol. Epub 2016 Feb 16.
9. Huiart L, Bouhnik AD, Rey D, et al. Early discontinuation of tamoxifen 25. Copson E, Eccles B, Maishman T, et al; POSH Study Steering Group.
intake in younger women with breast cancer: is it time to rethink the Prospective observational study of breast cancer treatment outcomes
way it is prescribed? Eur J Cancer. 2012;48:1939-1946. for UK women aged 18-40 years at diagnosis: the POSH study. J Natl
10. Ruddy K, Mayer E, Partridge A. Patient adherence and persistence with Cancer Inst. 2013;105:978-988.
oral anticancer treatment. CA Cancer J Clin. 2009;59:56-66. 26. Davies C, Pan H, Godwin J, et al; Adjuvant Tamoxifen: Longer Against
11. Cuzick J, Ambroisine L, Davidson N, et al; LHRH-agonists in Early Breast Shorter (ATLAS) Collaborative Group. Long-term effects of continuing
Cancer Overview group. Use of luteinising-hormone-releasing hormone adjuvant tamoxifen to 10 years versus stopping at 5 years after di-
agonists as adjuvant treatment in premenopausal patients with hormone- agnosis of oestrogen receptor-positive breast cancer: ATLAS, a rand-
receptor-positive breast cancer: a meta-analysis of individual patient data omised trial. Lancet. 2013;381:805-816.
from randomised adjuvant trials. Lancet. 2007;369:1711-1723. 27. Gray RG, Rea D, Handley K, et al. aTTom: Long-term effects of continuing
12. Swain SM, Jeong JH, Geyer CE, Jr, et al. Longer therapy, iatrogenic adjuvant tamoxifen to 10 years versus stopping at 5 years in 6,953
amenorrhea, and survival in early breast cancer. N Engl J Med. 2010; women with early breast cancer. J Clin Oncol. 2013;31s (suppl; abstr 5).
362:2053-2065. 28. Goss PE, Ingle JN, Martino S, et al. Impact of premenopausal status at breast
13. Tevaarwerk AJ, Wang M, Zhao F, et al. Phase III comparison of tamoxifen cancer diagnosis in women entered on the placebo-controlled NCIC CTG
versus tamoxifen plus ovarian function suppression in premenopausal MA17 trial of extended adjuvant letrozole. Ann Oncol. 2013;24:355-361.
women with node-negative, hormone receptor-positive breast cancer 29. Filipits M, Nielsen TO, Rudas M, et al. The PAM50 risk-of-recurrence
(E-3193, INT-0142): a trial of the Eastern Cooperative Oncology Group. score predicts risk for late distant recurrence after endocrine therapy in
J Clin Oncol. 2014;32:3948-3958. postmenopausal women with endocrine-responsive early breast can-
14. Francis PA, Regan MM, Fleming GF, et al; SOFT Investigators; In- cer. Clin Cancer Res. 2014;20:1298-1305.
ternational Breast Cancer Study Group. Adjuvant ovarian suppression in 30. Sestak I, Cuzick J, Dowsett M, et al. Prediction of late distant recurrence
premenopausal breast cancer. N Engl J Med. 2015;372:436-446. after 5 years of endocrine treatment: a combined analysis of patients
15. Dowsett M, Cuzick J, Ingle J, et al. Meta-analysis of breast cancer from the Austrian breast and colorectal cancer study group 8 and
outcomes in adjuvant trials of aromatase inhibitors versus tamoxifen. arimidex, tamoxifen alone or in combination randomized trials using the
J Clin Oncol. 2010;28:509-518. PAM50 risk of recurrence score. J Clin Oncol. 2015;33:916-922.
16. Gnant M, Mlineritsch B, Stoeger H, et al. Zoledronic acid combined with 31. Dubsky P, Brase JC, Jakesz R, et al; Austrian Breast and Colorectal Cancer
adjuvant endocrine therapy of tamoxifen versus anastrozol plus ovarian Study Group (ABCSG). The EndoPredict score provides prognostic in-
function suppression in premenopausal early breast cancer: final formation on late distant metastases in ER+/HER2- breast cancer pa-
analysis of the Austrian Breast and Colorectal Cancer Study Group Trial tients. Br J Cancer. 2013;109:2959-2964.
12. Annal Oncol. 2015;26:313-320. 32. Paik S, Shak S, Tang G, et al. A multigene assay to predict recurrence of
17. Pagani O, Regan MM, Walley BA, et al; TEXT and SOFT Investigators; tamoxifen-treated, node-negative breast cancer. N Engl J Med. 2004;
International Breast Cancer Study Group. Adjuvant exemestane with 351:2817-2826.
ovarian suppression in premenopausal breast cancer. N Engl J Med. 33. Ruddy KJ, Gelber S, Ginsburg ES, et al. Menopausal symptoms and fertility
2014;371:107-118. concerns in premenopausal breast cancer survivors: a comparison to age-
18. Pfeiler G, Konigsberg R, Fesl C, et al. Impact of body mass index on the and gravidity-matched controls. Menopause. 2011;18:105-108.
efficacy of endocrine therapy in premenopausal patients with breast 34. Nystedt M, Berglund G, Bolund C, et al. Side effects of adjuvant en-
cancer: an analysis of the prospective ABCSG-12 trial. J Clin Oncol. 2011; docrine treatment in premenopausal breast cancer patients: a pro-
29:2653-2659. spective randomized study. J Clin Oncol. 2003;21:1836-1844.

30 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


TREATING PREMENOPAUSAL ENDOCRINE-SENSITIVE BREAST CANCER

35. Azim HA Jr, de Azambuja E, Colozza M, et al. Long-term toxic effects of 54. Lambertini M, Del Mastro L, Pescio MC, et al. Cancer and fertility
adjuvant chemotherapy in breast cancer. Ann Oncol. 2011;22: preservation: international recommendations from an expert meeting.
1939-1947. BMC Med. 2016;14:1.
36. Jin Y, Desta Z, Stearns V, et al. CYP2D6 genotype, antidepressant use, 55. Peccatori FA, Azim HA, Jr, Orecchia R, et al; ESMO Guidelines Working
and tamoxifen metabolism during adjuvant breast cancer treatment. Group. Cancer, pregnancy and fertility: ESMO clinical practice guide-
J Natl Cancer Inst. 2005;97:30-39. lines for diagnosis, treatment and follow-up. Ann Oncol. 2013;24(suppl
37. Binkhorst L, Mathijssen RH, van Herk-Sukel MP, et al. Unjustified 6):vi160-vi170.
prescribing of CYP2D6 inhibiting SSRIs in women treated with ta- 56. Balduzzi A, Leonardi MC, Cardillo A, et al. Timing of adjuvant systemic
moxifen. Breast Cancer Res Treat. 2013;139:923-929. therapy and radiotherapy after breast-conserving surgery and mas-
38. Margolin S, Lindh JD, Thoren L, et al. CYP2D6 and adjuvant tamoxifen: tectomy. Cancer Treat Rev. 2010;36:443-450.
possible differences of outcome in pre- and post-menopausal patients. 57. Buendgen NK, Schultze-Mosgau A, Cordes T, et al. Initiation of ovarian
Pharmacogenomics. 2013;14:613-622. stimulation independent of the menstrual cycle: a case-control study.
39. Phillips KA, Feng Y, Ribi K, et al. Co-SOFT: the cognitive function sub-study of Arch Gynecol Obstet. 2013;288:901-904.
the suppression of ovarian function trial (SOFT). Paper presented at: 37th 58. Cakmak H, Katz A, Cedars MI, et al. Effective method for emergency
Annual CTRC-AACR San Antonio Breast Cancer Symposium; December fertility preservation: random-start controlled ovarian stimulation.
2014; San Antonio, TX. Fertil Steril. 2013;100:1673-1680.
40. Powles TJ, Hickish T, Kanis JA, et al. Effect of tamoxifen on bone mineral 59. Cakmak H, Rosen MP. Ovarian stimulation in cancer patients. Fertil
density measured by dual-energy x-ray absorptiometry in healthy Steril. 2013;99:1476-1484.
premenopausal and postmenopausal women. J Clin Oncol. 1996;14: 60. Oktay K, Buyuk E, Libertella N, et al. Fertility preservation in breast
78-84. cancer patients: a prospective controlled comparison of ovarian
41. Koopal C, Janssen-Heijnen ML, van de Wouw AJ, et al. Fracture stimulation with tamoxifen and letrozole for embryo cryopreservation.
incidence in pre- and postmenopausal women after completion of J Clin Oncol. 2005;23:4347-4353.
adjuvant hormonal therapy for breast cancer. Breast. 2015;24: 61. Oktay K, Hourvitz A, Sahin G, et al. Letrozole reduces estrogen and
153-158. gonadotropin exposure in women with breast cancer undergoing
42. Hershman DL, McMahon DJ, Crew KD, et al. Zoledronic acid prevents ovarian stimulation before chemotherapy. J Clin Endocrinol Metab.
bone loss in premenopausal women undergoing adjuvant chemo- 2006;91:3885-3890.
therapy for early-stage breast cancer. J Clin Oncol. 2008;26:4739-4745. 62. Azim AA, Costantini-Ferrando M, Oktay K. Safety of fertility preservation
43. Gnant M, Mlineritsch B, Luschin-Ebengreuth G, et al; Austrian Breast by ovarian stimulation with letrozole and gonadotropins in patients
and Colorectal Cancer Study Group (ABCSG). Adjuvant endocrine with breast cancer: a prospective controlled study. J Clin Oncol. 2008;
therapy plus zoledronic acid in premenopausal women with early-stage 26:2630-2635.
breast cancer: 5-year follow-up of the ABCSG-12 bone-mineral density 63. Goldrat O, Gervy C, Englert Y, et al. Progesterone levels in letrozole
substudy. Lancet Oncol. 2008;9:840-849. associated controlled ovarian stimulation for fertility preservation in
44. Ruddy KJ, Gelber SI, Tamimi RM, et al. Prospective study of fertility breast cancer patients. Hum Reprod. 2015;30:2184-2189.
concerns and preservation strategies in young women with breast 64. Oktay K, Turan V, Bedoschi G, et al. Fertility preservation success
cancer. J Clin Oncol. 2014;32:1151-1156. subsequent to concurrent aromatase inhibitor treatment and ovarian
45. Biglia N, Torrisi R, DAlonzo M, et al. Attitudes on fertility issues in breast stimulation in women with breast cancer. J Clin Oncol. 2015;33:
cancer patients: an Italian survey. Gynecol Endocrinol. 2015;31: 2424-2429.
458-464. 65. Kim J, Turan V, Oktay K. Long-term safety of letrozone and gonadotropin
46. Kroman N, Jensen MB, Wohlfahrt J, et al; Danish Breast Cancer Co- stimulation for fertility preservation in women with breast cancer. J Clin
operative Group. Pregnancy after treatment of breast cancera Endocrinol Metab. 2016;101:1364-1371.
population-based study on behalf of Danish Breast Cancer Cooperative 66. Turan V, Bedoschi G, Moy F, et al. Safety and feasibility of performing
Group. Acta Oncol. 2008;47:545-549. two consecutive ovarian stimulation cycles with the use of letrozole-
47. Ives A, Saunders C, Bulsara M, et al. Pregnancy after breast cancer: gonadotropin protocol for fertility preservation in breast cancer pa-
population based study. BMJ. 2007;334:194. tients. Fertil Steril. 2013;100:1681-1685.e1.
48. Azim HA, Jr, Santoro L, Pavlidis N, et al. Safety of pregnancy following 67. Goldrat O, Kroman N, Peccatori FA, et al. Pregnancy following breast
breast cancer diagnosis: a meta-analysis of 14 studies. Eur J Cancer. cancer using assisted reproduction and its effect on long-term outcome.
2011;47:74-83. Eur J Cancer. 2015;51:1490-1496.
49. Azim HA, Jr, Kroman N, Paesmans M, et al. Prognostic impact of 68. Gerber B, von Minckwitz G, Stehle H, et al; German Breast Group In-
pregnancy after breast cancer according to estrogen receptor status: vestigators. Effect of luteinizing hormone-releasing hormone agonist on
a multicenter retrospective study. J Clin Oncol. 2013;31:73-79. ovarian function after modern adjuvant breast cancer chemotherapy:
50. NCT02308085. Pregnancy Outcome and Safety of Interrupting Therapy the GBG 37 ZORO study. J Clin Oncol. 2011;29:2334-2341.
for Women with Endocrine Responsive Breast Cancer (POSITVE). http:// 69. Munster PN, Moore AP, Ismail-Khan R, et al. Randomized trial using
clinicaltrials.gov/ct2/show/NCT02308085. Accessed March 16, 2016. gonadotropin-releasing hormone agonist triptorelin for the preserva-
51. Lobo RA. Potential options for preservation of fertility in women. N Engl tion of ovarian function during (neo)adjuvant chemotherapy for breast
J Med. 2005;353:64-73. cancer. J Clin Oncol. 2012;30:533-538.
52. Torino F, Barnabei A, De Vecchis L, et al. Chemotherapy-induced ovarian 70. Moore HC, Unger JM, Phillips KA, et al; POEMS/S0230 Investigators.
toxicity in patients affected by endocrine-responsive early breast Goserelin for ovarian protection during breast-cancer adjuvant che-
cancer. Crit Rev Oncol Hematol. 2014;89:27-42. motherapy. N Engl J Med. 2015;372:923-932.
53. Loren AW, Mangu PB, Beck LN, et al; American Society of Clinical 71. Del Mastro L, Boni L, Michelotti A, et al. Effect of the gonadotropin-
Oncology. Fertility preservation for patients with cancer: American releasing hormone analogue triptorelin on the occurrence of
Society of Clinical Oncology clinical practice guideline update. J Clin chemotherapy-induced early menopause in premenopausal women
Oncol. 2013;31:2500-2510. with breast cancer: a randomized trial. JAMA. 2011;306:269-276.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 31


AZIM, DAVIDSON, AND RUDDY

72. Lambertini M, Ceppi M, Poggio F, et al. Ovarian suppression using breast cancer: long-term results from International Breast Cancer Study
luteinizing hormone-releasing hormone agonists during chemotherapy Group Trials V and VI. Eur J Cancer. 2007;43:1646-1653.
to preserve ovarian function and fertility of breast cancer patients: 75. Meirow D, Dor J, Kaufman B, et al. Cortical fibrosis and blood-vessels
a meta-analysis of randomized studies. Ann Oncol. 2015;26:2408-2419. damage in human ovaries exposed to chemotherapy. Potential
73. Partridge AH, Ruddy KJ, Gelber S, et al. Ovarian reserve in women who mechanisms of ovarian injury. Hum Reprod. 2007;22:1626-1633.
remain premenopausal after chemotherapy for early stage breast 76. Lambertini M, Boni L, Michelotti A, et al; GIM Study Group. Ovarian
cancer. Fertil Steril. 2010;94:638-644. suppression with triptorelin during adjuvant breast cancer chemo-
74. Partridge A, Gelber S, Gelber RD, et al. Age of menopause among therapy and long-term ovarian function, pregnancies, and disease-free
women who remain premenopausal following treatment for early survival: a randomized clinical trial. JAMA. 2015;314:2632-2640.

32 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


BREAST CANCER

Triple-Negative Breast Cancer: Is


Change on the Horizon?

CHAIR
Carey K. Anders, MD
The University of North Carolina at Chapel Hill
Chapel Hill, NC

SPEAKERS
Rebecca Dent, MD
National Cancer Center Singapore
Singapore

Vandana Abramson, MD
Vanderbilt University Medical Center
Nashville, TN
ANDERS ET AL

The Evolution of Triple-Negative Breast Cancer: From Biology


to Novel Therapeutics
Carey K. Anders, MD, Vandana Abramson, MD, Tira Tan, MBBS, and Rebecca Dent, MD

OVERVIEW

Triple-negative breast cancer (TNBC) is clinically defined as lacking expression of the estrogen receptor (ER), progesterone receptor
(ER), and HER2. Historically, TNBC has been characterized by an aggressive natural history and worse disease-specific outcomes
compared with other breast cancer subtypes. The advent of next-generation sequencing (NGS) has allowed for the dissection of
TNBC into molecular subtypes (i.e., basal-like, claudin-low). Within TNBC, several subtypes have emerged as immune-activated,
consistently illustrating better disease outcome. In addition, NGS has revealed a host of molecular features characteristic
of TNBC, including high rates of TP53 mutations, PI3K and MEK pathway activation, and genetic similarities to serous ovarian
cancers, including inactivation of the BRCA pathway. Identified genetic vulnerabilities of TNBC have led to promising therapeutic
approaches, including DNA-damaging agents (i.e., platinum salts and PARP inhibitors), as well as immunotherapy. Platinum salts are
routinely incorporated into the treatment of metastatic TNBC; however, best outcomes are observed among those with deficiencies
in the BRCA pathway. Although the incorporation of platinum in the neoadjuvant care of patients with TNBC yields higher pathologic
complete response (pCR) rates, the impact on longer-term outcome is less clear. The presence of immune infiltrate in TNBC has
shown both a predictive and prognostic role. Checkpoint inhibitors, including PD-1 and PD-L1 inhibitors, are under investigation in
the setting of metastatic TNBC and have shown responses in initial clinical trials. Finally, matching emerging therapeutic strategies to
optimal subtype of TNBC is of utmost importance as we design future research strategies to improve patient outcome.

T riple-negative breast cancer is a unique subset of breast


cancer that accounts for approximately 15%20% of all
breast cancer diagnoses.1 Clinically defined as lacking ex-
commonly targeted biomarkers in the treatment of breast
cancer: ER, PR, and HER2. The definition of TNBC has varied
throughout the decades, namely around the definition of
pression of ER/PR and HER2, TNBC is characterized by an ER and/or PR positivity (i.e., 1%10% vs. $ 1% by immu-
aggressive natural history and worse disease-specific out- nohistochemistry [IHC]). More recently, ER/PR positivity
comes compared with other breast cancer subtypes.2 Despite has been more strictly defined as greater than or equal to 1% by
initial responses to chemotherapy, early and higher rates of the American Society of Clinical Oncology guidelines.7 Endo-
distant, typically visceral, recurrences are observed for TNBC.3 crine therapy is generally recommended for breast cancers
Anthracycline and taxane-based chemotherapy traditionally exhibiting greater than or equal to 1% ER and/or PR positivity
has been the mainstay of therapy for TNBC, but with the across all stages of breast cancer. HER2-postivity also has been
advent of NGS, molecular dissection of TNBC is revealing novel
strictly defined as protein expression of 3+, and/or HER2/neu
targets currently under investigation.4-6 Herein, we will review
gene amplification greater than or equal to 2.0 by fluorescence
the natural history of TNBC, our current understanding of the
in situ hybridization (FISH).8 Thus, the pathologic definition of
molecular characteristics of this unique subset of breast
TNBC is currently defined as ER and/or PR IHC expression of
cancer, and emerging clinical strategies in the modern era,
with a highlight on immunotherapy and DNA damaging agents. 0 and HER2 negativity defined as either IHC expression of 01+
or lack of gene amplification (FISH , 2.0). This universal
definition is being incorporated into the design of TNBC clinical
DEFINITION OF TRIPLE-NEGATIVE BREAST trials. Such an approach will help interpret results of individual
CANCER clinical trials in the context of others because the biology of
The term triple-negative breast cancer is a pathologic breast cancers expressing hormone receptors at 1%10% is
definition based on the protein expression of the three most likely distinct from those without any expression.

From the Department of Medicine, Vanderbilt University, Vanderbilt-Ingram Cancer Center, Nashville, TN; Department of Medicine, National Cancer Center Singapore, Singapore;
Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC; UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Carey K. Anders, MD, Division of Hematology Oncology, University of North Carolina at Chapel Hill, 710 Oxfordshire Ln., Chapel Hill, NC 27517; email:
carey_anders@medunc.edu.

2016 by American Society of Clinical Oncology.

34 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


TRIPLE-NEGATIVE BREAST CANCER: EVOLUTION OF BIOLOGY AND NOVEL THERAPEUTICS

NATURAL HISTORY AND CLINICAL BEHAVIOR OF to a pCR with novel chemotherapy combinations (platinums)
TRIPLE-NEGATIVE BREAST CANCER and novel targeted therapies (inhibitors of PARP inhibitors)
In addition to its distinct pathologic definition, TNBC is have shown superior pCR rates on a trial-level basis.13-15 It is
characterized by a unique clinical history as compared with unclear, however, if incremental increases in pCR rates ul-
other subsets of breast cancer. Smid and colleagues reported timately will translate into improvement in recurrence-free
among 344 primary lymph nodenegative breast cancers survival.16
that the site of distant recurrence differed by breast cancer Triple-negative breast cancer is characterized by recurrence
subtype.9 In this analysis, visceral relapses, including lung patterns and natural history that are distinct from other
(p = .01) and brain metastases (p = .0035), more commonly were subtypes of breast cancer. Dent et al compared the natural
observed among patients with basal-like tumors. In contrast, history and clinical behavior of TNBC to non-TNBC subtypes
bone relapse was more commonly seen among the luminal among 1,601 patients diagnosed with early-stage breast
subtype (p = .0031), and those with HER2-enriched breast cancer in Toronto between 1987 and 1997.2 At a median
cancer more commonly recurred in the liver (p = .17; Fig. 1). follow-up of 8 years, this analysis illustrated that women with
Moreover, Harrell et al evaluated tumor gene expression TNBC were more likely to experience a distant recurrence
using a dataset of approximately 1,000 human breast tumors (hazard ratio [HR] 2.6; p , .0001); survival for patients with
and confirmed that HER2-enriched tumors more commonly TNBC was inferior compared with others (HR 3.2; p , .001)
spread to the liver, whereas basal-like and claudin-low breast within 5 years of diagnosis. Interestingly, and compared with
tumors, both commonly triple-negative, spread to the brain patients with non-TNBC breast cancer, the risk of recurrence
and lung.10 Among patients with advanced TNBC, brain re- peaked at the 3-year mark and declined thereafter. This
lapse has been observed in approximately 50% of patients.11 observation is in contrast to non-TNBC, in which the risk of
Response to chemotherapy also differs by breast cancer recurrence is lower during the initial 3-year follow-up period,
subtype, with the high-grade TNBC subset typically more but it remains constant over time. These important dis-
responsive to preoperative chemotherapy (i.e., higher rates tinctions between the clinical behaviors of TNBC versus
of pCR) but with higher overall rates of relapse.3,12 This non-TNBC must be considered when designing clinical
phenomenon has been coined the triple-negative para- interventions to prevent recurrence for this subset of
dox.3 The higher rate of distant recurrence for patients with aggressive breast cancer.
TNBC following neoadjuvant chemotherapy is likely attrib-
uted to residual disease (i.e., inability to achieve pCR).
Multiple studies have shown superior survival for patients TRIPLE-NEGATIVE BREAST CANCER SUBTYPES
who experience pCR compared with those who do not Breast cancer is a heterogeneous disease characterized by
experience pCR in this setting. Efforts to convert patients distinct intrinsic subtypes, traditionally defined as luminal A
with residual disease following neoadjuvant chemotherapy and B, normal-like, HER2-enriched, and basal-like, each with
corresponding unique clinical behavior.1,17,18 Over the past
decade, TNBC also has been recognized as a similarly het-
erogeneous disease. Several groups have sought to dissect
KEY POINTS the biology of TNBC using IHC, gene expression, and se-
quencing tools. Perhaps the most well-known classification
Triple-negative breast cancer (TNBC) is a pathologic of TNBC by gene expression, when compared with all breast
classification defined as lack of estrogen and cancers, is the basal-like subtype. The basal-like subtype,
progesterone receptors (0% by IHC) expression and
which represents 15%20% of all breast cancers, originally
HER2 negativity (01+ by IHC or lack of HER2/neu gene
amplification).
was classified by a basal epithelial cell gene expression
Although TNBC comprises only 15% to 20% of all breast cluster including keratin 5, keratin 17, integrin-b4, and
cancer diagnoses, it is over-represented among laminin.1 Basal-like breast cancer is approximately 80%
advanced disease as recurrences are early and typically concordant with the IHC classification of TNBC.19 A more
visceral (e.g., lung and brain). rare subtype of breast cancer, termed the claudin-low
Next-generation sequencing has provided a powerful subtype, is also commonly triple-negative.20 The claudin-
platform to dissect the biology of TNBC to provide novel low subtype is characterized by low to absent expression of
therapeutic targets under clinical investigation. luminal differentiation markers, high enrichment for
Checkpoint inhibitors, including PD-1 and PD-L1, are epithelial-to-mesenchymal transition (EMT) markers, im-
showing responses in TNBC in initial clinical trials, and mune response genes, and cancer stem celllike features.
several studies are underway to confirm activity and
This breast cancer subtype, with high frequency of meta-
understand biomarkers of response.
Platinum-based chemotherapy, with a DNA-damaging
plastic and medullary differentiation, illustrates a response
mechanism of action, is active in TNBC and is routinely to therapy that is intermediate between the basal-like and
incorporated into the neoadjuvant and metastatic luminal breast cancer subtypes.
settings. Investigation of platinums in the adjuvant A different, yet complementary, approach has been taken
setting is ongoing. by other investigators in which only TNBCs have been
subtyped using gene expression and sequencing tools to

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 35


ANDERS ET AL

FIGURE 1. Subtype-Specific Patterns of Distant Recurrence From 344 Primary Breast Cancers

Copyright pending as of February 1, 2016.

reveal distinct biologic subtypes with unique therapeutic MOLECULAR DISSECTION OF TRIPLE-NEGATIVE
implications.5,6 Lehmann and colleagues analyzed gene BREAST CANCER
expression from over 500 TNBC tumors and identified six As with other breast cancer subtypes, the biology of triple-
unique subtypes: basal-like (two subtypes, BL1 and BL2), negative, specifically basal-like, breast cancer was a focus of
immunomodulatory (IM), mesenchymal (M), mesenchymal The Cancer Genome Atlas (TCGA) project.4 In this large-scale
stemlike (MSL), and luminal androgen receptor (LAR).5 Cell- analysis of primary human breast cancers, genomic DNA
line models representative of each subtype were identified copy number arrays, DNA methylation, exome sequencing,
and therapeutically targeted. Results indicated that cell lines messenger RNA arrays, microRNA sequencing, and reverse-
of the BL1 and BL2 subtypes, with higher expression of cell phase protein arrays were evaluated. Among the 81 basal-
cycle and DNA damage response genes, were sensitive to like breast cancers, of which 65 were both triple-negative
cisplatin. Mesenchymal and MSL subtype cell lines, which
and basal-like, several hallmark findings were reported: (1) a
were enriched for EMT and growth factor pathways, illus-
high frequency of TP53 mutations (80%); (2) loss of RB1 and
trated sensitivity to dual PI3K/mTOR inhibition and Src in-
BRCA1; (3) high activation of the PI3K pathway (either
hibition. Finally, LAR subtype cell lines were sensitive to an
through PIK3CA gene mutation of approximately 9% or loss of
androgen receptor (AR) antagonist, a strategy that has shown
early success in the clinical arena.21 In parallel, Burnstein et al the negative regulators INPP4B and/or PTEN); (4) an enhanced
performed DNA and RNA sequencing on approximately 200 proliferation signature with hyperactivated FOXM1 as a tran-
TNBC tumors with confirmation in independent datasets and scriptional driver of this signature; and (5) genetic similarity
identified four subtypes: LAR, mesenchymal (MES), basal-like between basal-like breast cancer and serous ovarian cancers.
immunosuppressed (BLIS), and basal-like immune-activated These similarities included high frequencies of ATM and TP53
(BLIA).6 The BLIS subtype conferred the worst prognosis and mutations, high expression of AKT3 and MYC, inactivation of
BLIA the best prognosis, for both disease-free and disease- BRCA1 and BRCA2, RB1 loss, and cyclin E1 amplification. From a
specific survival. In this analysis, subtype-specific targets were therapeutic targeting perspective, approximately 20% of basal-
identified (such as AR in the LAR and STAT signal transduction like tumors harbored a germline or somatic BRCA mutation,
molecules and cytokines in the BLIA) that are worthy of which may confer sensitivity to platinum and/or PARP in-
continued exploration. hibitors that will be discussed below. Finally, copy number

36 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


TRIPLE-NEGATIVE BREAST CANCER: EVOLUTION OF BIOLOGY AND NOVEL THERAPEUTICS

analysis revealed several amplifications or deletions that may prone. BRCA1 and BRCA2 are well-described tumor sup-
be targetable in the clinical arena. Amplifications included pressor genes that participate directly in homologous
PIK3CA (49%), KRAS (32%), BRAF (30%), and EGFR (23%). Other recombinationmediated repair of double-stranded breaks.27
less common amplifications were seen in FGFR1, FGFR2, IGFR1, In humans, mutation in one copy of either of these genes in the
KIT, MET, and PDGFRA. As cited above, deletions were seen in germline results in hereditary breast and ovarian cancer syn-
PTEN and INPP4B, which confer activation of the PI3K pathway. drome; tumors that subsequently develop are defective in
Results of this valuable analysis are summarized in Sidebar 1 homologous recombinationmediated DNA repair.27 Recent
and provide a wealth of knowledge pertaining to our un- reports have expanded the range of genes implicated in familial
derstanding of the biology of TNBC and ways to target this breast cancers, many of which are involved in the homologous
disease more effectively. recombination pathway.23,28-30 In addition, defective homol-
ogous recombination also can be found in sporadic breast
cancers as shown by comparative genome hybridization
TRIPLE-NEGATIVE BREAST CANCER: IS CHANGE studies.31 Finally, impairment of homologous recombination
ON THE HORIZON? deficiency can occur through epigenetic mechanisms, such as
In addition to our evolving understanding of the biology of methylation of BRCA1/2, which is also part of the BRCAness
TNBC over the decades, therapeutic strategies to treat TNBC spectrum.22
across different stages are maturing. Although many mo-
lecularly targeted strategies under investigation exist, for
the purposes of this review, we will focus on the use of Incorporation of Platinums in the Neoadjuvant
platinums and the incorporation of immunotherapy into the Treatment of Triple-Negative Breast Cancer
treatment of TNBC. Over the past few decades, there has been considerable
interest in the use of platinum salts in the treatment of TNBC
Optimal Use of Platinum in Triple-Negative on the basis that dysfunction of homologous recombination
Breast Cancer DNA repair sensitizes tumor cells to these agents and in-
A subset of TNBC has been postulated to be phenotypically duces cell death. As a proof of concept, a small neoadjuvant
and molecularly similar to familial BRCA1-associated breast study reported a strikingly high pCR of 90% to four cycles of
cancers, so called BRCAness.22 It is estimated that more neoadjuvant cisplatin in a group of 10 patients from Poland
than 75% of tumors arising in BRCA1 mutation carriers are with BRCA1 mutation, the majority of whom were the TNBC
triple-negative and/or have a basal-like phenotype.23 These phenotype.32 Larger phase II studies involving patients with
tumors are characterized by their heightened sensitivity to predominantly sporadic TNBC have described conflicting
damage by DNA cross-linking.24-26 Double-stranded DNA results. Alba and colleagues reported a randomized phase II
breaks caused by agents such as platinum salts are con- study of standard neoadjuvant epirubicin plus cyclophos-
sidered the most incisive form of DNA damage, and they are phamide chemotherapy followed by docetaxel with or
repaired by homologous recombination, an error-free without carboplatin AUC 6 given every 21 days.33 The
process, and nonhomologous end-joining, which is error prespecified primary endpoint of improvement in pCR rates
(defined as pCR in breast) was not met, with pCR rates in
breast and axilla of 30% of patients in both treatment
arms.33 In contrast, the German Breast Cancer Group ad-
SIDEBAR 1. The Cancer Genome Atlas Molecular ministered neoadjuvant paclitaxel, liposomal doxorubicin,
Characteristics of Basal-like Breast Cancer and bevacizumab with or without weekly carboplatin AUC 1.5-2
Characteristics in the GeparSixto trial. Of the 315 patients with TNBC, 53.2%
High concordance with TNBC (approximately 80%) of those treated with the addition of weekly carboplatin, as
High rates of TP53 pathway alterations (TP53 mutations in compared with 36.9% treated without the addition of carbo-
84%; gain of MDM2 14%) platin, achieved a pCR (p = .005).14 Similarly, CALGB 40603, a
High rates of PI3K pathway alterations (PIK3CA mutation
two-by-two factorial randomized phase II trial evaluating
7%; PTEN mutation or loss 35%; INPP4B loss 30%) weekly paclitaxel with or without carboplatin (AUC 6) and/or
High rates of RB pathway alterations (RB1 mutation or loss
bevacizumab followed by dose dense doxorubicin plus cyclo-
20%; cyclin E1 amplification 9%; high expression of
phosphamide showed an increase in pCR rate (defined as
CDKN2A; low expression of RB1)
High genomic instability
ypT0/is) from 46% to 60% (odds ratio 1.76; p = .0018).15
Hypomethylation compared with other breast cancer
Furthermore, an important clinical question revolves
subtypes around these data: does the purported improvement in pCR
Similarities with serous ovarian carcinomas, including rates translate to improved survival outcomes? Early survival
approximately 20% rate of germline or somatic BRCA analysis of GeparSixto and CALGB 40603 simultaneously
mutations were reported at the San Antonio Breast Cancer Symposium
in December 2015. In the GeparSixto study, the improved
Abbreviation: TNBC, triple-negative breast cancer. pCR rate translated into a significant increase in 3-year
disease-free survival (DFS) from 76.1% to 85.8% (HR 0.56;

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 37


ANDERS ET AL

95% CI, 0.330.96; p = .035).34 This contrasts with the CALGB outcome. The addition of carboplatin enhances acute tox-
40603 study, which reported a numerical improvement, but icities associated with chemotherapy with higher discon-
no statistical difference in 3-year event-free survival nor tinuation rates, more dose reductions, and higher grade 3/4
overall survival (OS), with the caveat that this trial was adverse events, which could potentially compromise the
underpowered to determine a survival benefit.13 Of note, current standard of care in an already high-risk population
GeparSixto used a nonstandard, more intensive regimen of of patients. It may be reasonable to consider off-trial use of
concurrent anthracycline-taxane chemotherapy with the carboplatin in locally advanced TNBC for rapid control of
incorporation of bevacizumab in all arms. Pharmacologic locoregional disease and in young, fit patients with a high
synergy, as well as potentially the weekly schedule of risk of relapse as we await definitive results from ongoing
carboplatin, may have contributed to the better overall phase III trials powered for survival outcomesPEARLY
prognosis in the GeparSixto trial. (NCT02441933), Gerpar-Octo (NCT021253144), NRG-
BR003 (NCT02488967), and TPPC (NCT02455141).
Incorporation of Platinums in the Metastatic
Treatment of Triple-Negative Breast Cancer
In the metastatic setting, retrospective studies have been
BIOMARKER DISCOVERY IN TRIPLE-NEGATIVE
conflicting and largely focused on comparing TNBC with
BREAST CANCER
Triple-negative breast cancer is a heterogeneous disease.
other subtypes.35-38 In the largest randomized phase III trial,
Given the trade-off of greater toxicities in the early setting
the Triple Negative Breast Cancer Trial (TNT), 376 patients in
and mixed results in the metastatic setting, it would be ideal
U.K. centers were randomly assigned to receive either
to be better able to identify patients most likely to benefit
carboplatin or docetaxel monotherapy as first-line treat-
from this treatment strategy. TBCRC009, a phase II bio-
ment with cross-over on progression.39 In the unselected
marker discovery trial, has reported an improved response
TNBC population, there was no difference in the primary
rate in patients who harbored deleterious germline BRCA
outcome of objective response rates in both treatment arms
mutation(s) without durability of response nor improve-
when treated up front (31.4% vs. 35.6%; p = .44) or following
ment in PFS or OS.42 The TNT trial lends support to BRCA
cross-over (22.8% vs. 25.6%; p = .73).39 Similarly, the sec-
genotyping and gene expression profiling to improved
ondary endpoint of progression-free survival (PFS) did not
therapy selection in metastatic TNBC.39 Patients with BRCA1
significantly differ in both arms.39 To add to the burgeoning
or BRCA2 mutations treated with carboplatin fared better
evidence in favor of a role for platinum-based chemother-
with a greater response rate (68% vs. 33.3%; p = .03) and PFS
apy, two studies from China have been reported: (1) a
(6.8 vs. 3.1 months; p = .03) as compared with docetaxel,
randomized phase II trial comparing docetaxel and cisplatin
whereas nonbasal-like tumors did better with docetaxel
versus docetaxel and capecitabine and (2) a multicenter
(73.7% vs. 16.7%; p # .01).39 Lastly, DNA-based assays have
randomized phase III trial of gemcitabine and cisplatin versus
been developed based on whole genome tumor loss of
gemcitabine and paclitaxel.40,41 In the phase II study, 53
heterozygosity score (LOH),43 telomeric allelic imbalance
patients with metastatic TNBC were randomly selected to
score (TAI),44 and large-scale state transitions score (LST).45
receive docetaxel with cisplatin or capecitabine in the first-
These assays measure patterns of genomic instability derived
line setting. Both overall response rates (63.0% vs. 15.4%;
through comparison of BRCA1/2 mutated and nonmutated
p = .001) and PFS (10.9 vs. 4.8 months; p # .001) were
tumors. The unweighted sum of LOH, TAI, and LST together
significantly better in the cisplatin group.40 CBCSG006 was a
form the homologous recombination deficiency score. This
phase III study in which 240 patients were randomly assigned
has been evaluated in the translational component of the
to receive a maximum of eight cycles of gemcitabine and
TBCRC009 and TNT trials. Again, results are conflicting. In
either cisplatin or paclitaxel dosed every 21 days.41 The trial
TBCRC009, tumors from patients with platinum-responsive
used a hybrid design and was powered for noninferiority by
disease exhibited higher values for LST and LOH assays.42 This
allowing for a prespecified superiority hypothesis to be
was not so in TNT, in which the homologous recombination
tested. Gemcitabine plus cisplatin was both noninferior to and
deficiency score did not select for sensitivity to carboplatin
superior to gemcitabine plus paclitaxel (PFS: HR 0.692; 95% CI,
over docetaxel.39 Therefore, further development of an assay
0.5230.915; p , .0001 [noninferiority] and p , .009
to identify homologous recombination defect in non-BRCA1/2
[superiority]).41 Therefore, in both studies, the cisplatin-
tumors is needed. Finally, the group from Memorial Sloan
containing regimen outperformed the comparator.
Kettering Cancer Center reported a comparison of homologous
recombination deficiency scoring methodologies and con-
Challenges in Incorporating Platinums Into Standard cluded that homologous recombination DNA repair gene
Practice: Toxicities and Dosing Strategies sequencing demonstrated good sensitivity and specificity,
Given this encouraging clinical data on the role of platinum providing a potential biomarker for clinical trials of homologous
agents in TNBC, important clinical questions still remain on recombination deficiencytargeted therapies.46
how best to choose the dose and/or schedule of the plat- In summary, for the practicing oncologist, what would be
inum agent. No trials to date have been powered adequately the current standard of care with respect to the use of
to demonstrate an improvement in long-term survival platinum salts? In the neoadjuvant setting, platinum is a

38 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


TRIPLE-NEGATIVE BREAST CANCER: EVOLUTION OF BIOLOGY AND NOVEL THERAPEUTICS

reasonable choice for selected patients, especially if those substantial infiltration with TILs.55-58 Higher levels of TILs
who are BRCA positive. There are no data to support platinum generally are associated with poor-prognostic clinicopath-
salts in the adjuvant setting; clinical trials are ongoing. In the ologic features, including ER negativity, higher grade, higher
metastatic setting, where the goal of therapy is palliative, proliferative rate, and lymph node positivity.55,59-62 How-
platinum is one of the standard chemotherapies oncologists ever, despite worse clinical features, higher levels of TILs are
include in their armamentarium of chemotherapies, although associated with improved DFS and OS, independent of
with the caveat that the most convincing data are in the front- systemic therapy.59,63,64 This apparent paradox highlights
line setting for BRCA-germline mutation carriers. the role that the immune system may play in a subset of
TNBCs and suggests that TILs may be a surrogate for an
adaptive immune response in these cancers.
IMMUNOTHERAPY AND IMMUNOCONJUGATES
FOR TRIPLE-NEGATIVE BREAST CANCER:
PROMISE AND POTENTIAL ASSOCIATION OF TUMOR INFILITRATIVE
Modern immunotherapy, in the form of monoclonal anti- LYMPHOCYTES AND RESPONSE OF TRIPLE-
bodies that act as checkpoint inhibitors, has revolutionized NEGATIVE BREAST CANCER TO CHEMOTHERAPY
the landscape of the treatment of metastatic melanoma in a Generally, TILs are divided into intratumoral TILs that have
relatively short period of time. Antibodies to cytotoxic direct contact with tumor cells, and stromal TILs, which are
T-lymphocyte antigen 4 (CTLA-4), PD-1, and PD-L1, all of which found between tumor cells within the tumor stroma, but
increase the immune response against the tumor by blocking they do not have direct contact with tumor cells. A study of
immune regulating proteins that downregulate the immune patients with breast cancer receiving neoadjuvant chemo-
system, have increased response rates and OS.47-49 Perhaps therapy with an anthracycline/taxane combination showed
more interestingly, other solid tumor malignancies, such as lung an independent association between percentage of intra-
cancer, which traditionally are not considered immunogenic, tumoral TILs and pCR.64 Those with lymphocyte-predominant
have shown clinical benefit to checkpoint inhibitors.50 The role breast cancer, defined as more than 60% of stromal or
of immunotherapy in breast cancer has yet to be defined, but tumoral infiltration, had a particularly high rate of pCR (41.7%)
increasing evidence points to TNBCs as possibly having unique compared with only a 2% pCR rate in those tumors without any
characteristics that may make them more responsive to tumoral lymphocyte infiltration. In the GeparSixto study that
checkpoint inhibition. Given the lack of targeted treatment evaluated the addition of carboplatin to chemotherapy in TNBC
options for TNBC, immunotherapy poses an exciting opportu- or HER2-positive cancers, the presence of stromal TILs was
nity for the treatment of this aggressive disease. predictive of response to the addition of carboplatin, although
this correlation reached statistical significance only in HER2-
positive tumors.65 Regardless, this association between re-
OVERVIEW OF TUMOR IMMUNE SURVEILLANCE sponse to chemotherapy based on the presence of TILs suggests
IN THE SETTING OF BREAST CANCER that TILs not only are predictive markers of response in TNBC,
In a process called tumor immune surveillance, tumor immune
but that cell death induced by platinum agents may prime or
cells, including CD8+ T cells, recognize tumor-associated an-
generate neoantigens to stimulate nearby lymphocytes.
tigens presented by tumor cells and attack the tumor cells. The
PD-1 receptor pathway plays an important role in modulating
immune responses and provides an escape mechanism to PROGNOSTIC IMPLICATIONS OF IMMUNE GENE
tumor immune surveillance. PD-1 is an inhibitory immune SIGNATURES IN TRIPLE-NEGATIVE BREAST
checkpoint receptor expressed on activated T cells, B cells, CANCER
natural killer cells, and other lymphocytes that can remain Several other studies promote the exploration of the PD-1
active in inflammatory states such as cancer.51,52 PD-L1 is a pathway and immunotherapy in TNBC. Data from TCGA4
ligand of PD-1 and acts to suppress antitumor immunity by have confirmed higher PD-L1 mRNA expression in TNBC
binding PD-1. Ligation of PD-L1 with PD-1 inhibits T-cell pro- versus non-TNBC samples.66 This and other studies have
liferation, cytokine production, and cytolytic activity, which shown that PD-L1 is not detected in normal breast tissue but
leads to the functional inactivation or exhaustion of T cells.53 is expressed in about half of all breast cancers, including
Through upregulation of PD-L1 and other adaptive immune approximately 20% to 30% of TNBCs.67,68 PD-L1 expression
resistance mechanisms, tumors are able to use this pathway to is associated with TILs,69 correlates with higher histologic
evade the antitumor immune response. grade, greater tumor size, and higher expression of the
The balance of cells in the tumor microenvironment, which proliferation marker Ki-67.70
includes tumor cells, stromal cells (i.e., fibroblasts and mi- Further supporting the link between immunotherapy and
crovasculature), and immune cells (i.e., lymphocytes), ap- TNBC are gene expression profiling studies that demonstrate
pears to influence breast cancer outcome.54 An association an association between expression of IM genes and better
between greater tumor infiltrative lymphocytes (TILs) and clinical outcomes in TNBC.71 Desmedt et al were among the
better prognosis in breast cancer has been recognized for first to create gene modules and correlate them with out-
some time, but newer studies have shown the specific come in different subtypes of breast cancer. Of the seven
relevance in TNBC, which has been shown to have gene expression modules they described (tumor invasion,

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 39


ANDERS ET AL

immune response, angiogenesis, apoptosis, proliferation, FUTURE DIRECTIONS FOR IMMUNOTHERAPY IN


and ER and HER2 signaling), only the immune response TRIPLE-NEGATIVE BREAST CANCER
module correlated with prognosis in the ER-/HER2- sub- A decade ago, very little was known about the make-up of
group. Since that time, several groups have described the TNBC, and the only promising treatments on the horizon
prognostic value of immune gene signatures and TNBC.72-74 involved cytotoxic chemotherapy. Now, with a better
More recently, analysis of gene expression profiles of 587 understanding of the heterogeneity of TNBC and the
TNBC samples identified six distinct subtypes, including an understanding that immune targets are relevant in this
IM subtype characterized by high expression of immune- disease, we are in a new era in which we are exploring
related genes.5 This subtype is composed of immune- checkpoint inhibitors, vaccines, and immune antagonists.
activated and associated signaling components contributed Two large phase III randomized studies of atezolizumab
from both the tumor and the infiltrating lymphocytes, and and pembrolizumab are now ongoing in metastatic TNBC;
it has been associated with improved relapse-free survival the first is nab-paclitaxel with or without atezolizumab
compared with other subtypes.5 RNA sequencing showed and the other is pembrolizumab versus single agent
this subtype to have substantially higher expression of PD-L1, chemotherapy. Studies with both agents are also planned
PD-1, and CTLA-4. These and other data provide evidence that in the neoadjuvant setting. Finally, two studies evaluating
there may be a subset of TNBC in which checkpoint inhibitors carboplatin with atezolizumab are to begin accrual
may have particular efficacy. through the TBCRC in upcoming months to capitalize on
data with platinums in TNBC. Tissue samples from these
CLINICAL EXPERIENCE AND OUTCOMES WITH studies will further clarify the relationship of response and
IMMUNOTHERAPY IN TRIPLE-NEGATIVE BREAST resistance to PD-1 and PD-L1 expression, and to the
CANCER presence of TILs. The reported studies to date that show
The first reported study of an immune checkpoint inhibitor in responses in heavily pretreated patients with TNBC are
TNBC was a single-arm phase IB study of single-agent notable, but more impressive is the duration of response
pembrolizumab, a PD-1 antibody (KEYNOTE-012).75 This study seen in a disease that usually has a PFS of 2 to 3 months
showed that pembrolizumab 10 mg/kg given every 2 weeks was after the first or second line of treatment of metastatic
not only well-tolerated; it also showed activity in heavily pre- disease. Immunotherapy rapidly has become a great
treated patients with metastatic PD-L1positive TNBC. In- contender in the treatment of TNBC. Our next challenges
terestingly, of all patients with TNBC who were screened, 58% will be to identify how to use it best, whether it is with
tested positive for PD-L1 in greater than 1% of tumor stroma or chemotherapy or in combination with other immune
tumor cells. Over 45% of patients had received more than three modulators, and to identify biomarkers of response.
prior treatments in the metastatic setting, and 21.9% had
received five or more treatments. Of the 27 participants
with centrally confirmed measurable disease (32 patients CONCLUSION
total enrolled), one participant (3.7%) had a complete Triple-negative breast cancer is an established subset of
response (CR), four participants (14.8%) had a confirmed breast cancer with characteristic clinical behavior and
partial response (PR), 25.9% had stable disease, and 44.4% natural history. Until recently, the mainstay of therapy
had progressive disease by central review. The median against TNBC has been cytotoxic chemotherapy. The
time to response was 18 weeks (range, 732 weeks), and advent of NGS has opened our eyes to the possibility of
the median duration of response had not been reached targeted therapy in a subset of breast cancer that lacks the
(range, 15more than 40 weeks). classic biomarkers (ER, PR, and HER2) in breast cancer.
The PD-L1 antagonist atezolizumab (MPDL3280A) also is Despite this challenge, investigators spanning the re-
being evaluated in TNBC. An ongoing phase I study of atezo- search spectrum from bench-to-bedside are tackling TNBC
lizumab has an expansion cohort in heavily pretreated patients head on. We are seeing an explosion of activity in basic,
with PD-L1positive and negative TNBC.76 Initial data show translational, and clinical research that includes plati-
the overall response rate for evaluable patients with metastatic nums, immunotherapy, and others. Coordinated research
TNBC is 19%, including 9.5% CR and 9.5% PR, with 75% of efforts such as this are the way forward to improve the
responses ongoing (range, 18 to more than 56 weeks). outcome for our thousands of patients with TNBC.

References
1. Perou CM, Srlie T, Eisen MB, et al. Molecular portraits of human breast 3. Carey LA, Dees EC, Sawyer L, et al. The triple negative paradox: primary
tumours. Nature. 2000;406:747-752. tumor chemosensitivity of breast cancer subtypes. Clin Cancer Res.
2. Dent R, Trudeau M, Pritchard KI, et al. Triple-negative breast cancer: 2007;13:2329-2334.
clinical features and patterns of recurrence. Clin Cancer Res. 2007; 4. The Cancer Genome Atlas Network. Comprehensive molecular portraits
13:4429-4434. of human breast tumours. Nature. 2012;490:61-70.

40 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


TRIPLE-NEGATIVE BREAST CANCER: EVOLUTION OF BIOLOGY AND NOVEL THERAPEUTICS

5. Lehmann BD, Bauer JA, Chen X, et al. Identification of human triple- 23. Foulkes WD, Smith IE, Reis-Filho JS. Triple-negative breast cancer.
negative breast cancer subtypes and preclinical models for selection of N Engl J Med. 2010;363:1938-1948.
targeted therapies. J Clin Invest. 2011;121:2750-2767. 24. Bhattacharyya A, Ear US, Koller BH, et al. The breast cancer susceptibility
6. Burstein MD, Tsimelzon A, Poage GM, et al. Comprehensive genomic gene BRCA1 is required for subnuclear assembly of Rad51 and survival
analysis identifies novel subtypes and targets of triple-negative breast following treatment with the DNA cross-linking agent cisplatin. J Biol
cancer. Clin Cancer Res. 2015;21:1688-1698. Chem. 2000;275:23899-23903.
7. Hammond ME, Hayes DF, Dowsett M, et al. American Society of Clinical 25. Moynahan ME, Cui TY, Jasin M. Homology-directed DNA repair,
Oncology/College of American Pathologists guideline recommenda- mitomycin-c resistance, and chromosome stability is restored with
tions for immunohistochemical testing of estrogen and progesterone correction of a Brca1 mutation. Cancer Res. 2001;61:4842-4850.
receptors in breast cancer. J Clin Oncol. 2010;28:2784-2795. 26. Hastak K, Alli E, Ford JM. Synergistic chemosensitivity of triple-negative
8. Wolff AC, Hammond ME, Hicks DG, et al; American Society of Clinical breast cancer cell lines to poly(ADP-ribose) polymerase inhibition,
Oncology; College of American Pathologists. Recommendations for gemcitabine, and cisplatin. Cancer Res. 2010;70:7970-7980.
human epidermal growth factor receptor 2 testing in breast cancer: 27. Roy R, Chun J, Powell SN. BRCA1 and BRCA2: different roles in a common
American Society of Clinical Oncology/College of American Pathologists pathway of genome protection. Nat Rev Cancer. 2011;12:68-78.
clinical practice guideline update. J Clin Oncol. 2013;31:3997-4013. 28. Kurian AW, Hare EE, Mills MA, et al. Clinical evaluation of a multiple-
9. Smid M, Wang Y, Zhang Y, et al. Subtypes of breast cancer show gene sequencing panel for hereditary cancer risk assessment. J Clin
preferential site of relapse. Cancer Res. 2008;68:3108-3114. Oncol. 2014;32:2001-2009.
10. Harrell JC, Prat A, Parker JS, et al. Genomic analysis identifies unique 29. Castera L, Krieger S, Rousselin A, et al. Next-generation sequencing for
signatures predictive of brain, lung, and liver relapse. Breast Cancer Res the diagnosis of hereditary breast and ovarian cancer using genomic
Treat. 2012;132:523-535. capture targeting multiple candidate genes. Eur J Hum Genet. 2014;22:
11. Lin NU, Claus E, Sohl J, et al. Sites of distant recurrence and clinical 1305-1313.
outcomes in patients with metastatic triple-negative breast cancer: high 30. Couch FJ, Hart SN, Sharma P, et al. Inherited mutations in 17 breast
incidence of central nervous system metastases. Cancer. 2008;113: cancer susceptibility genes among a large triple-negative breast cancer
2638-2645. cohort unselected for family history of breast cancer. J Clin Oncol. 2015;
12. Liedtke C, Mazouni C, Hess KR, et al. Response to neoadjuvant therapy 33:304-311.
and long-term survival in patients with triple-negative breast cancer. 31. Stefansson OA, Jonasson JG, Johannsson OT, et al. Genomic profiling of
J Clin Oncol. 2008;26:1275-1281. breast tumours in relation to BRCA abnormalities and phenotypes.
13. Sikov WM, Berry DA, Peou CM, et al. Event-free and overall survival Breast Cancer Res. 2009;11:R47.
following neoadjuvant weekly paclitaxel and dose-dense AC +/2 car- 32. Byrski T, Huzarski T, Dent R, et al. Response to neoadjuvant therapy with
boplatin and/or bevacizumab in triple-negative breast cancer: out- cisplatin in BRCA1-positive breast cancer patients. Breast Cancer Res
comes from CALGB 40603 (Alliance). 2015, San Antonio, TX: 2016 San Treat. 2009;115:359-363.
Antonio Breast Cancer Symposium. 33. Alba E, Chacon JI, Lluch A, et al. A randomized phase II trial of platinum
14. von Minckwitz G, Schneeweiss A, Loibl S, et al. Neoadjuvant carboplatin salts in basal-like breast cancer patients in the neoadjuvant setting.
in patients with triple-negative and HER2-positive early breast cancer Results from the GEICAM/2006-03, multicenter study. Breast Cancer
(GeparSixto; GBG 66): a randomised phase 2 trial. Lancet Oncol. 2014; Res Treat. 2012;136:487-493.
15:747-756. 34. von Minckwitz G, Loibl S, Schneeweiss A, et al. Early survival analysis of
15. Sikov WM, Berry DA, Perou CM, et al. Impact of the addition of car- the randomized phase II trial investigating the addition of carboplatin to
boplatin and/or bevacizumab to neoadjuvant once-per-week paclitaxel neoadjuvant therapy for triple-negative and HER2-positive early breast
followed by dose-dense doxorubicin and cyclophosphamide on path- cancer (GeparSixto). 2015, San Antonio, TX: 2015 San Antonio Breast
ologic complete response rates in stage II to III triple-negative breast Cancer Symposium.
cancer: CALGB 40603 (Alliance). J Clin Oncol. 2015;33:13-21. 35. Staudacher L, Cottu PH, Dieras V, et al. Platinum-based chemotherapy in
16. Cortazar P, Zhang L, Untch M, et al. Pathological complete response and metastatic triple-negative breast cancer: the Institut Curie experience.
long-term clinical benefit in breast cancer: the CTNeoBC pooled Ann Oncol. 2011;22:848-856.
analysis. Lancet. 2014;384:164-172. 36. Khalaf D, Hilton JF, Clemons M, et al. Investigating the discernible and
17. Srlie T, Perou CM, Tibshirani R, et al. Gene expression patterns of distinct effects of platinum-based chemotherapy regimens for meta-
breast carcinomas distinguish tumor subclasses with clinical implica- static triple-negative breast cancer on time to progression. Oncol Lett.
tions. Proc Natl Acad Sci USA. 2001;98:10869-10874. 2014;7:866-870.
18. Sorlie T, Tibshirani R, Parker J, et al. Repeated observation of breast 37. Uhm JE, Park YH, Yi SY, et al. Treatment outcomes and clinicopathologic
tumor subtypes in independent gene expression data sets. Proc Natl characteristics of triple-negative breast cancer patients who received
Acad Sci USA. 2003;100:8418-8423. platinum-containing chemotherapy. Int J Cancer. 2009;124:1457-1462.
19. Livasy CA, Karaca G, Nanda R, et al. Phenotypic evaluation of the basal-like 38. Villarreal-Garza C, Khalaf D, Bouganim N, et al. Platinum-based che-
subtype of invasive breast carcinoma. Mod Pathol. 2006;19:264-271. motherapy in triple-negative advanced breast cancer. Breast Cancer Res
20. Prat A, Parker JS, Karginova O, et al. Phenotypic and molecular char- Treat. 2014;146:567-572.
acterization of the claudin-low intrinsic subtype of breast cancer. Breast 39. Tutt A, Ellis P, Kilburn L, et al. The TNT trial: A randomized phase III trial
Cancer Res. 2010;12:R68. of carboplatin (C) compared with docetaxel (D) for patients with
21. Gucalp A, Tolaney S, Isakoff SJ, et al; Translational Breast Cancer metastatic or recurrent locally advanced triple negative or BRCA1/2
Research Consortium (TBCRC 011). Phase II trial of bicalutamide in breast cancer (CRUK/07/012). 2014, San Antonio, TX: 2014 San Antonio
patients with androgen receptor-positive, estrogen receptor-negative Breast Cancer Symposium.
metastatic breast cancer. Clin Cancer Res. 2013;19:5505-5512. 40. Fan Y, Xu BH, Yuan P, et al. Docetaxel-cisplatin might be superior to
22. Turner N, Tutt A, Ashworth A. Hallmarks of BRCAness in sporadic docetaxel-capecitabine in the first-line treatment of metastatic triple-
cancers. Nat Rev Cancer. 2004;4:814-819. negative breast cancer. Ann Oncol. 2013;24:1219-1225.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 41


ANDERS ET AL

41. Hu XC, Zhang J, Xu BH, et al. Cisplatin plus gemcitabine versus paclitaxel 60. Issa-Nummer Y, Darb-Esfahani S, Loibl S, et al. Prospective validation of
plus gemcitabine as first-line therapy for metastatic triple-negative immunological infiltrate for prediction of response to neoadjuvant
breast cancer (CBCSG006): a randomised, open-label, multicentre, chemotherapy in HER2-negative breast cancera substudy of the
phase 3 trial. Lancet Oncol. 2015;16:436-446. neoadjuvant GeparQuinto trial. PLoS One. 2013;8:e79775.
42. Isakoff SJ, Mayer EL, He L, et al. TBCRC009: a multicenter phase II clinical 61. Ladoire S, Arnould L, Apetoh L, et al. Pathologic complete response to
trial of platinum monotherapy with biomarker assessment in metastatic neoadjuvant chemotherapy of breast carcinoma is associated with the
triple-negative breast cancer. J Clin Oncol. 2015;33:1902-1909. disappearance of tumor-infiltrating Foxp3+ regulatory T cells. Clin
43. Abkevich V, Timms KM, Hennessy BT, et al. Patterns of genomic loss of Cancer Res. 2008;14:2413-2420.
heterozygosity predict homologous recombination repair defects in 62. Loi S, Michiels S, Salgado R, et al. Tumor infiltrating lymphocytes are
epithelial ovarian cancer. Br J Cancer. 2012;107:1776-1782. prognostic in triple negative breast cancer and predictive for trastu-
44. Birkbak NJ, Wang ZC, Kim JY, et al. Telomeric allelic imbalance indicates zumab benefit in early breast cancer: results from the FinHER trial. Ann
defective DNA repair and sensitivity to DNA-damaging agents. Cancer Oncol. 2014;25:1544-1550.
Discov. 2012;2:366-375. 63. Mahmoud SM, Paish EC, Powe DG, et al. Tumor-infiltrating CD8
45. Popova T, Manie E, Rieunier G, et al. Ploidy and large-scale genomic + lymphocytes predict clinical outcome in breast cancer. J Clin Oncol.
instability consistently identify basal-like breast carcinomas with 2011;29:1949-1955.
BRCA1/2 inactivation. Cancer Res. 2012;72:5454-5462. 64. Denkert C, Loibl S, Noske A, et al. Tumor-associated lymphocytes as an
46. Powell SN, Riaz N, Mutter RW, et al. A functional assay for homologous independent predictor of response to neoadjuvant chemotherapy in
recombination (HR) DNA repair and whole exome sequencing reveal breast cancer. J Clin Oncol. 2010;28:105-113.
that HR-defective sporadic breast cancers are enriched for genetic 65. Denkert C, von Minckwitz G, Brase JC, et al. Tumor-infiltrating lympho-
alterations in DNA repair genes. 2015, San Antonio, TX: 2015 San cytes and response to neoadjuvant chemotherapy with or without
Antonio Breast Cancer Symposium. carboplatin in human epidermal growth factor receptor 2-positive
47. Hodi FS, ODay SJ, McDermott DF, et al. Improved survival with ipilimumab and triple-negative primary breast cancers. J Clin Oncol. 2015;33:
in patients with metastatic melanoma. N Engl J Med. 2010;363:711-723. 983-991.
48. Robert C, Long GV, Brady B, et al. Nivolumab in previously untreated 66. Mittendorf EA, Philips AV, Meric-Bernstam F, et al. PD-L1 expression in
melanoma without BRAF mutation. N Engl J Med. 2015;372:320-330. triple-negative breast cancer. Cancer Immunol Res. 2014;2:361-370.
49. Hamid O, Robert C, Daud A, et al. Safety and tumor responses with 67. Ghebeh H, Mohammed S, Al-Omair A, et al. The B7-H1 (PD-L1)
lambrolizumab (anti-PD-1) in melanoma. N Engl J Med. 2013;369:134-144. T lymphocyte-inhibitory molecule is expressed in breast cancer pa-
50. Garon EB, Rizvi NA, Hui R, et al; KEYNOTE-001 Investigators. Pem- tients with infiltrating ductal carcinoma: correlation with important
brolizumab for the treatment of non-small-cell lung cancer. N Engl J high-risk prognostic factors. Neoplasia. 2006;8:190-198.
Med. 2015;372:2018-2028. 68. Emens LA, Braiteh FS, Cassier P, et al. Inhibition of PD-L1 by MPDL3280A
51. Blank C, Gajewski TF, Mackensen A. Interaction of PD-L1 on tumor cells leads to clinical activity in patients with metastatic triple-negative
with PD-1 on tumor-specific T cells as a mechanism of immune evasion: breast cancer. 2014, San Antonio, TX: 2014 San Antonio Breast Cancer
implications for tumor immunotherapy. Cancer Immunol Immunother. Sympsoium.
2005;54:307-314. 69. Ali HR, Glont SE, Blows FM, et al. PD-L1 protein expression in breast
52. Keir ME, Butte MJ, Freeman GJ, et al. PD-1 and its ligands in tolerance cancer is rare, enriched in basal-like tumours and associated with
and immunity. Annu Rev Immunol. 2008;26:677-704. infiltrating lymphocytes. Ann Oncol. 2015;26:1488-1493.
53. Butte MJ, Keir ME, Phamduy TB, et al. Programmed death-1 ligand 1 70. Muenst S, Schaerli AR, Gao F, et al. Expression of programmed death
interacts specifically with the B7-1 costimulatory molecule to inhibit ligand 1 (PD-L1) is associated with poor prognosis in human breast
T cell responses. Immunity. 2007;27:111-122. cancer. Breast Cancer Res Treat. 2014;146:15-24.
54. Dunn GP, Bruce AT, Ikeda H, et al. Cancer immunoediting: from 71. Desmedt C, Haibe-Kains B, Wirapati P, et al. Biological processes as-
immunosurveillance to tumor escape. Nat Immunol. 2002;3:991-998. sociated with breast cancer clinical outcome depend on the molecular
55. Loi S, Sirtaine N, Piette F, et al. Prognostic and predictive value of tumor- subtypes. Clin Cancer Res. 2008;14:5158-5165.
infiltrating lymphocytes in a phase III randomized adjuvant breast 72. Nagalla S, Chou JW, Willingham MC, et al. Interactions between im-
cancer trial in node-positive breast cancer comparing the addition of munity, proliferation and molecular subtype in breast cancer prognosis.
docetaxel to doxorubicin with doxorubicin-based chemotherapy: BIG Genome Biol. 2013;14:R34.
02-98. J Clin Oncol. 2013;31:860-867. 73. Calabro` A, Beissbarth T, Kuner R, et al. Effects of infiltrating lymphocytes
56. Liu S, Lachapelle J, Leung S, et al. CD8+ lymphocyte infiltration is an and estrogen receptor on gene expression and prognosis in breast
independent favorable prognostic indicator in basal-like breast cancer. cancer. Breast Cancer Res Treat. 2009;116:69-77.
Breast Cancer Res. 2012;14:R48. 74. Rody A, Holtrich U, Pusztai L, et al. T-cell metagene predicts a favorable
57. Liyanage UK, Moore TT, Joo HG, et al. Prevalence of regulatory T cells is prognosis in estrogen receptor-negative and HER2-positive breast
increased in peripheral blood and tumor microenvironment of patients with cancers. Breast Cancer Res. 2009;11:R15.
pancreas or breast adenocarcinoma. J Immunol. 2002;169:2756-2761. 75. Nanda R, Chow LQ, Dees EC, et al. A phase Ib study of pembrolizumab
58. Bates GJ, Fox SB, Han C, et al. Quantification of regulatory T cells enables (MK-3475) in patients with advanced triple-negative breast cancer.
the identification of high-risk breast cancer patients and those at risk of 2014, San Antonio, TX: 2014 San Antonio Breast Cancer Symposium.
late relapse. J Clin Oncol. 2006;24:5373-5380. 76. Emens LA, Braiteh FS, Cassier P, et al. Inhibition of PD-L1 by MPDL3280A
59. Ono M, Tsuda H, Shimizu C, et al. Tumor-infiltrating lymphocytes are leads to clinical activity in patients with metastatic triple-negative
correlated with response to neoadjuvant chemotherapy in triple- breast cancer (TNBC) 2015, Philadelphia, PA: American Association
negative breast cancer. Breast Cancer Res Treat. 2012;132:793-805. for Cancer Research 106th Annual Meeting.

42 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


BREAST CANCER

Updates and Controversies in


HER2-Positive Breast Cancer

CHAIR
Sunil Verma, MD, MSEd, FRCPC
Cumming School of Medicine, University of Calgary
Calgary, AB, Canada

SPEAKERS
Kimberly L. Blackwell, MD
Duke University Medical Center
Durham, NC

Speaker
Miguel Martin, MD, PhD
~
Hospital General Universitario Gregorio Maranon
Madrid, Spain
NIXON AND VERMA

A Value-Based Approach to Treatment of HER2-Positive


Breast Cancer: Examining the Evidence
Nancy Nixon, MD, FRCPC, and Sunil Verma, MD, MSEd, FRCPC

OVERVIEW

Over the past decade, treatment of HER2-positive breast cancer has been revolutionized with the introduction of targeted
therapies. Survival in both early and advanced HER2-positive breast cancer has improved significantly. With evidence for
major clinical benefit, it is imperative that health systems evaluate new treatments to maximize the value of health
expenditures. Physicians, funding agencies, and policy makers are tasked with analyzing available evidence to ensure that
each individual patient receives the optimal treatment in a resource-challenged environment.

A pproximately 15% to 20% of breast cancer patients


overexpress HER2 protein. 1 Over the past decade,
treatment of HER2-positive breast cancer has been revo-
the EGFR family. The rationale for the addition of pertu-
zumab to trastuzumab is to overcome resistance to tras-
tuzumab caused specifically by formation of HER2:HER3
lutionized. Although previously a poor prognostic marker, heterodimers. It was evaluated for metastatic disease in the
HER2-positive breast cancer now gives patients an equiva- CLEOPATRA trial,6 which studied patients with HER2-positive
lent, if not superior, survival to those patients that are HER2- disease with no prior treatment of metastatic disease and
negative.2 In metastatic disease, the median overall survival randomly assigned them 1:1 to trastuzumab and docetaxel,
(OS) has dramatically increased over the course of 15 years, plus either pertuzumab or placebo. Ninety percent of the
from 20 months, 50 months, and beyond.3 For metastatic patients in this trial had not been on prior trastuzumab
patients, American Society of Clinical Oncology (ASCO) therapy in the adjuvant setting, meaning the majority of the
guidelines4 now recommend HER2-targeted therapies in all patients were naive to anti-HER2 therapies. The results of
lines of treatment of metastatic disease. this trial established a new first-line standard of metastatic
HER2-positive breast cancer, with an increase in progression-
TREATMENT OF METASTATIC HER2-POSITIVE free survival (PFS) of 6 months (18.6 vs. 12.4 months; p , .001)
BREAST CANCER and OS (56.5 vs. 40.8 months; p = .002; Table 1).7
First Line
Trastuzumab, a human monoclonal IgG antibody that se- Second Line
lectively targets HER2, was the first targeted drug to be For patients progressing on an anti-HER2 therapy combined
approved. It has since has been shown to be effective in with a cytotoxic or endocrine agent, additional anti-HER2
combination with multiple chemotherapy drugs including therapy should be offered based on evidence showing it is
docetaxel, paclitaxel, vinorelbine, and capecitabine. In the beneficial to continue suppression of the HER2 pathway.8-12
landmark trial by Slamon et al,5 patients were randomly Laptinib is an oral tyrosine kinase inhibitor that functions
assigned 1:1 to standard chemotherapy alone versus downstream of HER2, inhibiting both EGFR and HER2 re-
standard chemotherapy plus trastuzumab. In this study, ceptors. It was shown to be an effective option for second-
there was a substantial improvement in time to pro- line treatment in 2006 by Geyer et al13 for patients who
gression (TTP, 7.4 vs. 4.6 months; p , .001). Median OS had locally advanced or metastatic HER2-positive breast
increased from 20.3 to 25.1 months (p = .046), establishing cancer previously treated with regimens that included an
trastuzumab as a standard of care in combination with anthracycline, a taxane, and trastuzumab. The addition of
taxanes (Table 1). lapatinib resulted in a significant improvement in median
Pertuzumab, a recombinant humanized monoclonal an- TTP of 8.4 versus 4.4 months at interim analysis, meeting
tibody, binds to a separate domain on the HER2 receptor, specified criteria for early reporting (hazard ratio [HR] 0.49;
preventing the dimerization of HER2 with other members of p , .001). There was no noticeable improvement in OS

From the University of Calgary, Calgary, Alberta, Canada.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Sunil Verma, MD, MSEd, FRCPC, University of Calgary, 1331 29 St. NW, Calgary, Alberta, Canada; email: drsunil.verma@albertahealthservices.ca.

2016 by American Society of Clinical Oncology.

e56 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


HER2-POSITIVE BREAST CANCER: A VALUE-ORIENTED APPROACH TO TREATMENT

TABLE 1. Clinical Benefit of Targeted Therapies for Early HER2-Positive Breast Cancer
Indication Regimen Overall Survival Disease-Free Survival
NSABP-B31 and Adjuvant early breast AC T vs. AC TH HR 0.63; 75.2% vs. 84.0% at HR 0.60; p , .001; 62.3% vs.
NCCTG N983120 cancer 8.4 years follow-up 73.7% at 8.4 years follow-up
10-Year Follow-up Adjuvant early breast AC T vs. AC TH vs. TCH HR 0.63 (AC-TH p , .0001; 85.9% HR 0.72 (AC-TH p , .001; 74.6%
BCIRG 00621 cancer vs. 78.7%) vs. 67.9%)
HR 0.76 (TCH p = .0075; 83.8% vs. HR 0.77 (TCH p = .0011; 73.0%
78.7%) vs. 67.9%)
10-Year Follow-up Adjuvant early breast Standard chemotherapy vs. standard HR 0.74; p , .001; 79.4% vs. HR 0.75; p , .001; 69.3% vs.
HERA cancer chemotherapy + 1-y trastuzumab 72.9% at 10 years follow-up 62.5% at 10 years follow-up
Abbreviations: T, taxane; H, trastuzumab, C, cyclophosphamide, c, carboplatin; AC, doxorubicin and cyclophosphamide; HR, hazard ratio.

observed in this trial, likely in part because of early crossover Beyond Second Line
(Table 1). After progression on trasuzumab, pertuzumab, and treat-
In 2012, the EMILIA study14 was published, establishing a ment with T-DM1, there remain multiple options of treat-
new standard of care for second-line treatment. This trial ment, still including anti-HER2targeted treatments. These
looked at T-DM1, an antibody-drug conjugate of trastuzumab include capecitabine plus lapatinib, trastuzumab plus
and the cytotoxic agent emtansine (DM1). The antibody lapatinib,15-17 or trastuzumab in combination with another
trastuzumab binds to the HER2 on tumor cell surfaces and cytotoxic agent. There is insufficient evidence to recommend
upon internalization the emtansine moiety is released, one regimen over another. Alternatively, patients may be
binding to tubules and disrupting microtubule dynamics. considered for enrollment on clinical trial.
EMILIA compared T-DM1 versus lapatinib plus capectiabine In 2015, the THERESA trial was presented at the San
in patients with locally advanced or metastatic HER2-positive Antonio Breast Cancer Symposium. Showing OS benefit for
breast cancer who had previously received zero to more than T-DM1 for patients who have had prior therapy with taxane,
three prior regimens. All the patients on this trial had prior trastuzumab, and lapatinib. This confirms the efficacy for
exposure to trastuzumab and a taxane. T-DM1 showed a T-DM1 in later lines of therapy for patients who may not
significant improvement in median PFS (9.6 vs. 6.4 months, HR have had received T-DM1 in the second-line setting.
0.65; p , .001) and median OS (30.9 vs. 25.1 months; HR 0.68;
p , .001; Table 1). In addition, rates of grade 3 or higher TREATMENT OF EARLY HER2-POSITIVE BREAST
adverse events were higher in the lapatinib plus capecitabine CANCER
group. This established T-DM1 as the new standard for second- Adjuvant
line treatment. With the success of anti-HER2 therapy in the metastatic
setting, trastuzumab was evaluated in the adjuvant setting
in a series of pivotal trials, first reported in 2005. In the HERA
trial,18 patients were randomly assigned 1:1:1 to observa-
tion, versus 1 or 2 years of trastuzumab given every 3 weeks.
When compared with observation, trastuzumab given after
KEY POINTS primary therapy reduced the rate of recurrence, in particular
Treatment of HER2-positive breast cancer, and its
distant recurrence, by approximately 50% after interim ef-
prognosis, has been revolutionized since the ficacy analysis with median follow-up of 12 months. This
introduction of targeted treatment. benefit was confirmed in a 2012 meta-analysis of eight trials
Significant improvements in overall survival have been of chemotherapy plus trastuzumab versus trastuzumab
observed in both metastatic and adjuvant settings over alone.19 The HR for recurrence was 0.60, also with an im-
the past decade. provement in OS and HR for death of 0.66. The benefit in OS
Despite significant clinical benefit with introduction of was most strongly associated with concurrent administra-
targeted treatments, there is a risk that cost on a tion of trastuzumab with chemotherapy, as opposed to with
population level may become prohibitive for sequential treatment. In 2014, Perez et al published the
widespread use. results of a planned joint analysis on survival from NSABP-31
Physicians, funding agencies, and policy makers are
and NCCTG N9831, where patients were randomly assigned
tasked with finding a means to ensure that treatments
are cost-effective in an environment of limited
to doxorubicin plus cyclophosphamide followed by pacli-
resources. taxel plus trastuzumab for 1 year versus paclitaxel alone.20
Careful evaluation of the evidence and consideration of Adding trastuzumab led to 37% relative improvement in OS
clinical benefit alongside the cost-effectiveness of novel (HR 0.63; p , .001). An improvement in disease-free survival
therapies will help to guide clinical practice. (DFS) of 40% was observed (HR 0.60; p , .001) with increase
in 10-year DFS rate from 62.2% to 73.7% (Table 2).

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e57


NIXON AND VERMA

TABLE 2. Clinical Benefit of Targeted Therapies for Early and Advanced HER2-Positive Breast Cancer
Indication Regimen Overall Survival Progression-Free Survival
Slamon et al, First-line advanced breast Standard chemotherapy vs. standard che- 25.1 vs. 20.3 months (p = .046) 7.4 vs. 4.6 months
20015 cancer motherapy + H (p , .001)
CLEOPATRA6 First-line advanced breast TH + placebo vs. THP 56.5 months vs. 40.8 months 7.7 vs. 6. 3 months
cancer (HR 0.68; p , .001) (HR 0.68; p , .001)
EMILIA14 Second-line advanced T-DM1 vs. capecitbine + lapatinib 20.9 vs. 25.1 months (HR 0.68; 9.6 vs. 6.4 months
breast cancer p , .001) (HR 0.65; p , .001)
Geyer et al, Second-line advanced Lapatinib + capecitabine vs. capecitabine TTP 8.4 vs. 4.4 months
200613 breast cancer (HR 0.49; p , .001)
Abbreviations: T, taxane; H, trastuzumab; P, pertuzumab; DM1, emtansine; HR, hazard ratio; TTP, time to progression.

The benefit of adjuvant trastuzumab has been maintained Several studies have been done to look at lapatinib in
with longer follow-up. An updated analysis of the HERA trial neoadjuvant treatment, both in combination with or in place
showed continued significant DFS and OS benefit of tras- of trastuzumab. Lapatinib has been consistently inferior to
tuzumab despite substantial crossover (Table 2). Ten-year trastuzumab when compared head to head.25-27 The com-
follow-up from the BCIRG 006 trial, which randomly assigned bination was looked at in the NeoALLTO study, which en-
patients to three arms (doxorubicin plus cyclophosphamide rolled women with HER2-positive, early breast cancer and
followed by taxane plus or minus trastuzumab, vs. docetaxel randomly assigned them 1:1:1 to lapatinib, trastuzumab, or
plus carboplatin plus trastuzumab) was presented in 2015 trastuzumab plus laptinib for 6 weeks followed by the anti-
at the San Antonio Breast Cancer Symposium (Table 2). HER2 treatment combined with paclitaxel for 12 weeks.
With 10.3 years of follow-up, there was significant benefit After surgery, women received three cycles of FEC followed
in OS for both trastuzumab-containing arms compared by 34 weeks of the assigned anti-HER2 neoadjuvant therapy.
with chemotherapy alone (p , .001 for doxorubicin plus Although patients in the combination group had improve-
cyclophosphamide followed by docetaxel plus trastuzu- ment in pCR, this did not translate to improved DFS or OS in
mab, and p = .0018 for docetaxel plus carboplatin plus the confirmatory phase III adjuvant ALTTO study.
trastuzumab).
Near Future Directions
There continues to be new data that will add to our
Neoadjuvant
knowledge of how to optimize treatment of early disease.
There is greater interest in evaluating HER2 therapies in the
The APHINITY trial evaluating adjuvant pertuzumab plus
neoadjuvant setting, given the correlation between path-
trastuzumab versus trastuzumab after surgery is yet to be
ologic complete response (pCR) and long-term outcomes
reported but may provide evidence for adjuvant utilization
including event-free survival (EFS).22 The U.S. Food and Drug
of pertuzumab. Similarly, the KATHERINE trial, which recently
Administration granted approval to the addition of neo-
completed accrual, is evaluating the addition of T-DM1 as ad-
adjuvant pertuzumb to trastuzumab based on two random-
juvant treatment of patients treated with neoadjuvant che-
ized phase II studies, in which higher pCR rates were seen
motherapy and trastuzumab who do not achieve pCR. T-DM1 is
compared with trastuzumab arms. NeoSphere compared four
also being evaluated in direct comparison to paclitaxel and
regimens preoperatively (docetaxel plus trastuzumab vs.
trastuzumab in the adjuvant setting for patients with low-risk
docetaxel plus trastuzumab plus pertuzumab vs. trastuzumab
HER2-positive disease (ATEMPT trial).
plus pertuzumab. vs. docetaxel plus pertuzumab).23 After
surgery, patients in the docetaxel arms received three cycles
of 5-fluorouracil, epirubicin, and cyclophosphamide (FEC) VALUE-BASED APPROACH
and completed a year of trastuzumab, whereas patients Over the past decade, there has been rapid growth in the
who had received only antibody treatment received both cost of cancer care as a whole. In the United States, it is
docetaxel and FEC followed by trastuzumab for a full year. expected to increase from $125 billion in 2010 to $158 billion
pCR rates were 29%, 46%, 17%, and 24% in each group, in 2020.28 Although drug treatment costs account for as
respectively. In the TRYPHA-ENA study, women were ran- much as 20% of total cost, it is imperative that health
domly assigned to trastuzumab plus pertuzumab and con- systems evaluate new treatments strategically to maximize
current FEC followed by concurrent docetaxel, FEC alone value of health expenditures. In HER2 breast cancer, the
followed by concurrent docetaxel, or concurrent docetaxel benefits gained by new treatments have been undeniably
and carboplatin.24 After surgery, patients completed 1 year important clinically. Physicians, policy makers, and funding
of trastuzumab. The pCR rate was equivalent across arms. In agencies are tasked with identifying economic trade-offs and
both studies, patients with ER-negative disease were more ensuring patients have access to treatments with mean-
likely to achieve pCR. ingful health benefits in a resource-limited environment.

e58 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


HER2-POSITIVE BREAST CANCER: A VALUE-ORIENTED APPROACH TO TREATMENT

Clinical Benefit Value Assessment


The definition of clinical benefit is variable. Ideally, it would Economists use a variety of methods to assess value.
include all patient-important outcomes. In practice, the evi- Commonly used in evaluations of medical intervention are
dence used to create treatment guidelines is the highest quality quality-adjusted life-years (QALYs) and incremental cost
available, such as the randomized controlled trials described. In effectiveness ratios (ICERs; Table 3). QALY incorporates both
ASCOs 2015 value framework, clinical endpoints used to assess quality and quantity of life achieved with treatment. A
benefit (OS, PFS, DFS) were selected based on their repre- treatment with a low cost per QALY is more cost effective
sentation of data collected in clinical trials.29 Based on this than one with high cost per QALY. An ICER is simply the ratio
framework, Table 1 summarizes the clinical benefit observed in between the difference in cost and the difference in benefit
landmark trials in early breast cancer. Table 2 summarizes of two interventions. In cost-effectiveness analyses, ICER are
clinical benefit in advanced disease. commonly expressed as incremental cost per QALY. Defining

TABLE 3. Summary of Key North American Cost-Effectiveness Analyses on Targeted HER2 Therapies in Various
Lines of Treatment
Study Country HER2 Test and Treatment ICER/QALY
33
Neoadjuvant Pertuzumab Attard et al Canada HER2 test: not included in analysis Neosphere: $25,388 per QALY
NeoSphere: neoadjuvant TRYPHAENA: $46,196 per
docetaxel + trastuzumab 6 QALY
pertuzumab surgery
fluorouracil + epirubicin +
cyclophosphamide (FEC)
TRYPHAENA: docetaxel + carbo-
platin + trastuzumab +
pertuzumab
Adjuvant Trastuzumab Kurian et al32 United States HER2 test: not included in analysis Anthracycline based: $39,98
per QALY
Treatment: anthracycline-based Nonanthracycline is more ex-
regimens in NSABP B-31 and pensive and less effective,
NCCTGN9831 trials; nonan- therefore both with and
thracycline regimen from without trastuzumab ex-
BCIRG 006 + trastuzumab for 52 ceed $100,000 per QALY
weeks vs. chemotherapy only compared with anthracy-
cline regimens
Garrison et al46 United States HER2 test: not included in analysis $26,417 per QALY over
a lifetime
Treatment: anyhtacycline-based
regimens from NSABP B-31 and
NCCTG N9831 trials plus
trastuzumab (at actual dose/
duration received in trial) vs.
chemotherapy alone
Metastatic trastuzumab Elkin et al34 United States HER2 test: IHC +3; IHC with FISH Initial IHC with FISH confir-
for IHC 2+ and 3+; IHC with FISH mation: $125,0000 per
for IHC 2+; FISH only QALY
Treatment: trastuzumab + Initial FISH: $145,000 per
paclitaxel vs. paclitaxel only QALY
Other strategies for HER2
testing less cost effective
First-line pertuzumab Durkee et al36 United States HER2 testing: not included in $713,219 per QALY
analysis
Treatment: docetaxel + trastuzu-
mab + pertuzumab vs. doce-
taxel and trastuzumab alone
Metastatic T-DM1 PCODR Canada HER2 testing: not included in $162,839 per QALY
analysis
Treatment: T-DM1 vs. lapatinib
capecitabine
Metastatic capecitabine + lapatinib Le et al35 United States HER2 testing: not included in $166,113 per QALY
analysis
Treatment: capecitabine + lapati-
nib vs. capecitabine alone
Abbreviations: ICER, incremental cost-effectiveness ratio; QALY, quality-adjusted life-years; IHC, immunohistochemistry; FISH, fluorescence in situ hybridization; DM1, emtansine.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e59


NIXON AND VERMA

the threshold for cost effectiveness is a focus for funding focus on treatment according to best evidence, while finding
sources worldwide and is variable depending on the funding ways to minimize costs and avoid overtreatment.
party and the basic structure of the health care system. In
most analysis from the United States using USD, a threshold Should dual anti-HER2 maintenance be continued after
level of $100,000 per QALY is considered cost-effective maximal response? There is limited evidence to answer
(range 50,000 to three times the United States per capita whether maintenance with dual anti-HER2 blockade should
gross domestic product, or $160,000 per QALY).30 This be continued once maximal response is achieved. There is,
method of assessment is fraught with variability that is however, evidence of continued efficacy of trastuzumab
highly subjective to the approach used to estimate OS in the after progression on a trastuzumab-based regimen. A trial
control group when crossover is allowed in the trial, in- by Von Minckwitz et al looked at the combination of
clusion of treatment past progression, and the cost of HER2 capecitabine plus trastuzumab versus trastuzumab alone
testing as demonstrated in a systemic review by Parkinson as a second-line treatment.10 This showed a significant im-
et al.31 provement in TTP (8.2 vs. 5.6 months; p = .03) and a non-
In the adjuvant setting, trastuzumab was shown to be cost- significant trend in OS (25.5 vs. 20.4 months; p = .257). In a
effective by Kurian et al, who estimated an ICER of $39,982 separate study, the combination of trastuzumab plus laptinib
USD per QALY.32 Cost-effectiveness of pertuzumab in the versus lapatinib alone after progression on a trastuzumab-
neoadjuvant setting was investigated by Attard et al based based regimen was investigated.16 This showed a significant
on pCR data from both NeoSphere and TRYPHAENA.33 improvement for PFS in the combination arm (12 weeks vs.
Published EFS and OS data for patients achieving and not 8.1 weeks; p = .008) and OS (14 vs. 9.5 months).
achieving pCR were used in combination with percentage Currently, no trials have been done to address con-
achieving pCR in trials to estimate survival. The incremental tinuation of targeted therapy after maximal response is
cost per QALY ranged from $25,388 CAD (NeoSphere) to achieved. In most trials, HER2-targeted therapy was ad-
$46,196 CAD (TRYPHAENA analysis), making the addition of ministered until disease progression or until significant
pertuzumab cost-effective with a threshold of $100,000. toxicity leading to discontinuation of therapy. Current
For treatment of advanced disease, the cost of even a recommendations on duration from ASCO and the cost-
single targeted agent increases significantly per QALY. For effectiveness analysis mentioned before are based on the
HER2 testing and treatment with trastuzumab in the met- approach used in relevant clinical trials. Potential gains in
astatic setting, Elkin et al determined an ICER of $125,000 both quality of life and cost-effectiveness may be achieved
USD per QALY.34 A similar method was used to determine if we consider treatment breaks in patients who have
the ICER for lapatinib pus capecitabine to be $166,113 USD sustained a response with no evidence of progression. This
per QALY, not including testing for HER2 positivity.35 The approach needs to evaluated and studied in the metastatic
pan-Canadian Oncology Drug Review (pCODR) calculated a setting and could be guided by utilization of functional
cost of $162,839 CAD for T-DM1 based on EMILIA trial re- imaging/circulating tumor cells.
sults. The addition of pertuzumab to docetaxel and tras-
tuzumab according to the CLEOPATRA trial, was found to be, Do all patients need dual anti-HER2 blockade up front? If
again, incrementally higher in a recent publication, with an we are able to identify a certain subgroup within the HER2
estimated ICER of $713,219 USD per QALY gained.36 Despite population that derives the greatest benefit from dual-
the high cost, most funding agencies have adopted these antibody treatment, it allows us to be selective in who re-
therapies into clinical practice. This brings to question what ceives the treatment, sparing others both toxicities and cost.
practical considerations can be made to maximize access to Baselga et al looked to answer this question by assessing a
highly effective, costly treatments in an environment of protocol-prespecified panel of biomarkers in tumor and
limited resources. serum samples from patients on the CLEOPATRA trial.17
Biomarkers were identified based on previous under-
PRACTICAL CONSIDERATIONS standing of HER2-signaling pathways, or HER2-targeted
Metastatic Disease therapy resistance. Pertuzumab was found to have a con-
There are several limitations to QALY as a means of eval- sistent benefit for PFS, independent of biomarker sub-
uation for treatment of advanced breast cancer patients. In groups. This indicates that to our current knowledge, HER2 is
the metastatic setting, successful treatments are victims of the only marker that should be used for patient selection for
their own success, because longer PFS extrapolates to more the combination of trastuzumab plus pertuzumab. There is a
time accruing costs for expensive therapies. Additionally, need for further studies that may help to elucidate tumor, or
QALY does not capture an individual patients priorities. For patient-related factors that may ultimately help us de-
patients with metastatic disease, the balance between termine which patients are best candidates for dual anti-
quality versus quantity of life is a much more personal bodies up front.
determination than for early breast cancer, where the goal One of the key questions that must be asked when ap-
is to cure. It also does not take into account the benefit of plying evidence to practice is whether the patient population
time-off treatment or symptom improvement. Based on in the study matches the patient being treated in the real-
these limitations, we pose questions for discussion that still world setting. In the CLEOPATRA trial, 90% of patients were

e60 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


HER2-POSITIVE BREAST CANCER: A VALUE-ORIENTED APPROACH TO TREATMENT

nave to any HER2-targeted therapy. In practice, many of the patients, 47% had been given adjuvant chemotherapy and
patients we treat in the first line for metastatic disease are trastuzumab. Disease-free survival at 40 months was 93% for
being treated for recurrence, and have already received patients who had not received chemotherapy versus 99% for
antibody treatment in the adjuvant setting. Swain et al those who had. In multivariate analysis, tumor size was not
recently published the results that suggest incremental associated with clinical outcomes, leading the authors to
improvement for patients who relapsed postadjuvant state adjuvant treatment should be discussed with all HER2-
trastuzumab; however, although the HR was similar, the positive T1a-b tumors. Based on the available data, one has
prognosis and overall results for this population were in- to weigh the potential benefit:risk of toxicity and cost im-
ferior compared with de novo patients.37 Forty-one patients plications specifically for patients with T1a/T1b tumors.
in the control group and 47 patients in the treatment arm
were enrolled after 1 year of adjuvant trastuzumab. The OS Does everyone need neoadjuvant pertuzumab? In the
was 46.6 months and 53.8 months, respectively (HR 0.8; adjuvant setting similarly to advanced disease, there is
95% CI, 0.441.47), with a large confidence interval owing question of whether patients who are positive for both HER2
to relatively small numbers. The study authors point out, and hormone receptors should be treated the same. In the
however, that since the introduction of trastuzumab in the NeoSphere study, patients were stratified according to
adjuvant setting the proportion of patients who had re- hormone receptor status at the time of randomization. They
current disease had significantly decreased, as shown in the subsequently compared pCR results across these groups.
SystHERs trial.38 This means that the results of the CLEO- Patients that were estrogen receptor (ER) and progesterone
PATRA trial may reflect what is seen in practice today, with receptor (PgR)negative showed a pCR rate of 63% when
increased proportion of patients being treated with HER2- pertuzumab was added to docetaxel and trastuzumab, com-
positive de novo presentation. pared with 36.8% with trastuzumab and docetaxel alone. In
patients who were also ER-positive, PgR-positive, or both, the
three-drug regimen had a pCR of 26%, compared with 20% with
Early Breast Cancer docetraxel and trastuzumab. The updated EFS data show a
Treatment of early breast cancer is distinct from advanced nonsignificant trend for improvement in EFS. One requires
breast cancer because the treatment goals are more clearly confirmatory adjuvant data to truly assess the benefit of adding
defined and uniform across patients. Treatment with the pertuzumab to standard trastuzumab-based therapy.42
goal of cure makes decreasing treatment intensity an un-
common target. However, because outcomes of HER2- Do patients need 1 year of trastuzumab? Duration of
positive breast cancer exceed survival rates of 95%, it is trastuzumab has been looked at in multiple studies. In the
important to consider whether a subset of patients exists in original HERA study, trastuzumab for 1 year plus chemo-
whom we can de-escalate therapy. therapy was compared with chemotherapy alone, and in a
third arm trastuzumab for 2 years was given. At a median
Is there a population who do not need HER2-targeted follow-up of 8 years, there was no difference between 1 and
treatment? For patients who are HER2-negative, chemo- 2 years with respect to DFS (HR 0.99; p = .86) or OS (HR 1.05;
therapy is generally not recommended unless they are p = .63). The other pivotal trials discussed before used the
lymph nodepositive, or high-risk lymph nodenegative. In duration of 1 year somewhat empirically. Shorter duration of
the HER2+ population, however, the threshold for treatment adjuvant trastuzumab was studied in the FinHER trial.43 This
is lower based on our knowledge that even earlier stage study tested nine cycles of weekly trastuzumab with adju-
tumors are more likely to recur. OSullivan et al performed a vant docetaxel or vinorelbine followed by FEC in a subset of
meta-analysis of five trastuzumab randomized controlled early breast cancer patients with HER2-positive, high-risk
trials in early breast cancer and tumors 2 cm or smaller.39 disease (node-positive or node-negative with tumors at least
They concluded that patients with HER2-positive and tumors 2 cm). The primary endpoint of recurrence-free survival
2 cm or smaller had major DFS and OS benefits from tras- favored the trastuzumab arm (HR 0.41; p = .01), as did OS (HR
tuzumab as part of the treatment regimen. 0.41; p = .07). Documented clinical improvement was similar
For patients with T1a or T1b tumors that are node- to standard of 1-year therapy seen in previous trials.
negative, prognosis is less well-defined, even in the set- In a larger phase III study, PHARE randomly assigned 1,693
ting of HER2 positivity. Ferenbacher et al did a retrospective patients with HER2-positive high-risk (node-positive) disease
analysis of 234 patients with HER2-positive, lymph node to chemotherapy plus 6 months versus 1 year of trastuzu-
negative, and T1a or T1b tumors.40 Distant invasive re- mab.44 This study failed to achieve its primary outcome of
currence was the primary endpoint. Five-year distant noninferiority at a median follow-up of 3.5 years. Addi-
relapse-free survival was 98.2% for T1a, 89.4% for T1b, and tionally, more patients in the 6 months group had a distant
84.5% for 1-cm tumors. A majority (74%) of these patients recurrence as the first DFS event.
had no adjuvant treatment, raising the possibility of not There are a number of ongoing studies to evaluate
requiring therapy for certain T1a patients. In contrast, a shorter treatments of adjuvant trastuzumab comparing 6 to
study by Rodriguez et al41 looked at 276 patients with 12 months (e.g., the Hellenic group trial, the Persephone
node-negative, T1a-b HER2-positive breast cancer. Of these study) and 3 to 12 months (e.g., SOLD study, Short-HER

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e61


NIXON AND VERMA

study). These studies will provide insight into possibly reducing There are important questions raised that must be con-
the duration of adjuvant treatment and, subsequently, costs. sidered despite the attraction of reduced cost. These include
While awaiting these results, 1 year continues to be the questions on what patient population was included to es-
standard of care. tablish comparable clinical efficacy. To date, studies have
been in metastatic patients, which is a varied population
Biosimilars compared with patients with early breast cancer. Primary
The most intuitive way to receive the benefit of targeted endpoints have also been questioned, with the overall re-
treatments without the cost is to find a cheaper, equally sponse rate being most commonly chosen. Another concern
effective alternative. For this purpose, there has been great is that, up to now, biosimilar trials have looked at trastu-
interest in the production of biosimilar agents. Biosimilars zumab as a single agent, but the new standard of care in first-
are distinct from generic formulations in that they are not line advanced disease is a combination treatment with
biochemically identical to the original. Biologic agents are pertuzumab. Ongoing efforts are underway to evaluate
complex, with larger size and more intricate structure, such biosimilars in a neoadjuvant setting and with more clinically
that exact replication is impossible.45 Biosimilars, therefore, relevant endpoints to guide us about its use in early-stage
must be tested to ensure noninferior biologic activity, safety, and advanced breast cancer.
and efficacy.46 At least 10 trastuzumab biosimilar mono- Overall, we have made major strides in the management
clonal antibodies are in active development. The furthest of HER2-positive early-stage and advanced breast cancer.
along in testing is CT-P6, produced by Celltrion. CT-P6 has These impressive gains are based on inclusion of newer
been through phase III testing and has demonstrated an HER2-targeted therapies and the related financial impact of
equivalent overall response rate (primary endpoint) to integrating these drugs. The future in HER2-positive breast
trastuzumab in patients with metastatic disease. Based on cancer clinical research needs to focus on how best to
this trial, CT-P6 has already been approved as a biosimilar in optimize therapy that provides us with a more individualized
South Korea. As this becomes available, the cost of tras- approach based on inclusion of biomarkers, imaging, and
tuzumab may drop by as much as 40%. patient factors.

References
1. Slamon DJ, Clark GM, Wong SG, et al. Human breast cancer: correlation advanced breast cancer that has progressed on trastuzumab: updated
of relapse and survival with amplification of the HER-2/neu oncogene. efficacy and biomarker analyses. Breast Cancer Res Treat. 2008;112:
Science. 1987;235:177-182. 533-543.
2. Zurawksa U, Baribeau DA, Giilck S, et al. Outcomes of HER 2-positive 10. von Minckwitz G, du Bois A, Schmidt M, et al. Trastuzumab beyond
early-stage breast cancer in the trastuzumab era: a population based progression in human epidermal growth factor receptor 2-positive
study of Canadian patients. Curr Oncol. 2013;20:6. advanced breast cancer: a german breast group 26/breast international
3. Verma S, Joy AA, Rayson D, et al. HER story: the next chapter in HER-2- group 03-05 study. J Clin Oncol. 2009;27:1999-2006.
directed therapy for advanced breast cancer. Oncologist. 2013;18: 11. Von Minckwitz G, Vogel P, Schmidt M, et al. Trastuzumab treatment
1153-1166. beyond progression in patients with HER-2 positive metastatic breast
4. Giordano SH, Temin S, Kirshner JJ, et al; American Society of Clinical cancer: the TBP study (GBG 26/BIG 3-05). Presented at: 30th San
Oncology. Systemic therapy for patients with advanced human epi- Antonio Breast Cancer Symposium; December 2007; San Antonio, TX.
dermal growth factor receptor 2-positive breast cancer: American Abstract 4056. (abstr 4056).
Society of Clinical Oncology clinical practice guideline. J Clin Oncol. 12. Von Minckwitz G, Zielinski C, Maarteense P, et al. Capecitabine vs.
2014;32:2078-2099. capecitabine + trastuzumab in patients with HER2-positive metastatic
5. Slamon DJ, Leyland-Jones B, Shak S, et al. Use of chemotherapy plus a breast cancer progressing durgin trastuzumab treatment: the TBP
monoclonal antibody against HER2 for metastatic breast cancer that phase III study (GBG 26/BIG 3-05). 2008. J Clin Oncol. 26:47s (suppl;
overexpresses HER2. N Engl J Med. 2001;344:783-792. abstr 1025).
6. Swain SM, Baselga J, Kim SB, et al; CLEOPATRA Study Group. Pertu- 13. Geyer CE, Forster J, Lindquist D, et al. Lapatinib plus capecitabine for
zumab, trastuzumab, and docetaxel in HER2-positive metastatic breast HER2-positive advanced breast cancer. N Engl J Med. 2006;355:
cancer. N Engl J Med. 2015;372:724-734. 2733-2743.
7. Swain SM, Kim SB, et al. Confirmatory overall survival (OS) analysis of 14. Verma S, Miles D, Gianni L, et al; EMILIA Study Group. Trastuzumab
CLEOPATRA: A randomized, double-blind, placebo controlled Phase III emtansine for HER2-positive advanced breast cancer. N Engl J Med.
study with peruzumab (P), trastuzumab (T), and docetaxel (D) in pa- 2012;367:1783-1791.
tients with HER2-postiive first-line metastatic breast cancer (MBC). 15. Blackwell KL, Burstein HJ, Storniolo AM, et al. Randomized study of
Cancer Res. 2012;72:3s (suppl; abstr P5-18-26). Lapatinib alone or in combination with trastuzumab in women with
8. Cameron D, Casey M, Oliva C, et al. Lapatinib plus capecitabine in ErbB2-positive, trastuzumab-refractory metastatic breast cancer. J Clin
women with HER-2-positive advanced breast cancer: final survival Oncol. 2010;28:1124-1130.
analysis of a phase III randomized trial. Oncologist. 2010;15:924-934. 16. Blackwell KL, Burstein HJ, Storniolo AM, et al. Overall survival benefit
9. Cameron D, Casey M, Press M, et al. A phase III randomized comparison with lapatinib in combination with trastuzumab for patients with hu-
of lapatinib plus capecitabine versus capecitabine alone in women with man epidermal growth factor receptor 2-positive metastatic breast

e62 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


HER2-POSITIVE BREAST CANCER: A VALUE-ORIENTED APPROACH TO TREATMENT

cancer: final results from the EGF104900 Study. J Clin Oncol. 2012;30: 30. Ubel PA, Loewenstein G, Jepson C. Whose quality of life? A commentary
2585-2592. exploring discrepancies between health state evaluations of patients
17. Baselga J, Corte s J, S-A Im, et al. Biomarker analyses in CLEOPATRA: and the general public. Qual Life Res. 2003;12:599-607.
a phase III, placebo-controlled study of pertuzumab in human epi- 31. Parkinson B, Pearson SA, Viney R. Economic evaluations of trastuzumab
dermal growth factor receptor 2positive, first-line metastatic breast in HER2-positive metastatic breast cancer: a systematic review and
cancer. J Clin Oncol. 2014;32:3753-3761. critique. Eur J Health Econ. 2014;15:93-112.
18. Piccart-Gebhart MJ, Procter M, Leyland-Jones B, et al; Herceptin Ad- 32. Kurian AW, Thompson RN, Gaw AF, et al. A cost-effectiveness analysis of
juvant (HERA) Trial Study Team. Trastuzumab after adjuvant chemo- adjuvant trastuzumab regimens in early HER2/neu-positive breast
therapy in HER2-positive breast cancer. N Engl J Med. 2005;353: cancer. J Clin Oncol. 2007;25:634-641.
1659-1672. 33. Attard CL, Pepper AN, Brown ST, et al. Cost-effectiveness analysis of
19. Moja L, Tagliabue L, Balduzzi S, et al. Trastuzumab containing regi- neoadjuvant pertuzumab and trastuzumab therapy for locally ad-
mens for early breast cancer. Cochrane Database Syst Rev. 2012;4: vanced, inflammatory, or early HER2-positive breast cancer in Canada.
CD006243. J Med Econ. 2015;18:173-188.
20. Perez EA, Romond EH, Suman VJ, et al. Trastuzumab plus adjuvant 34. Elkin EB, Weinstein MC, Winer EP, et al. HER-2 testing and trastuzumab
chemotherapy for human epidermal growth factor receptor 2-positive therapy for metastatic breast cancer: a cost-effectiveness analysis. J Clin
breast cancer: planned joint analysis of overall survival from NSABP B-31 Oncol. 2004;22:854-863.
and NCCTG N9831. J Clin Oncol. 2014;32:3744-3752. 35. Le QA, Hay JW. Cost-effectiveness analysis of lapatinib in HER-2-positive
21. Slamon DJ, Eiermann W, Robert NJ, et al. Ten year follow up of the advanced breast cancer. Cancer. 2009;115:489-498.
BCIRG-006 Trial comparing doxorubicin plus cyclophosphamide 36. Durkee BY, Qian Y, Pollom EL, et al. Cost-effectiveness of pertuzumab in
followed by docetaxel (ACT) with doxorubicin plus cyclosphosphamide human epidermal growth factor receptor 2-positive metastatic breast
followed by docetaxel and trastuzumab (ACTH) with docetaxel, carbo- cancer. J Clin Oncol. 2015;63:1-12.
platin, and trastuzumab (TCH) in HER2+ early breast cancer. Presented at: 37. Swain S, Baselga, J. Treatment of HER2-positive metastatic breast
San Antonio Breast Cancer Symposium; December 2015; San Antonio, cancer. N Engl J Med. Correspondence. 2015;372:1964-1965.
TX. Abstract S5-S04. 38. Tripathy D, Brufsky A, Cobleigh M, et al. Increasing proportion of de
22. Cortazar P, Zhang L, Untch M, et al. Pathological complete response and novo compared with recurrent HER2-positive metastatic breast cancer:
long-term clinical benefit in breast cancer: the CTNeoBC pooled early results from the systemic therapies for HER2-positive metastatic
analysis. Lancet. 2014;384:164-172. breast cancer registry study. Presented at the 37th Annual San Antonio
23. Gianni L, Pienkowski T, Im YH, et al. Efficacy and safety of neoadjuvant Breast Cancer Symposium; December 2014; San Antonio, TX.
pertuzumab and trastuzumab in women with locally advanced, in- 39. OSullivan CC, Bradbury I, Campbell C, et al. Efficacy of adjuvant
flammatory, or early HER2-positive breast cancer (NeoSphere): trastuzumab for patients with human epidermal growth factor receptor
a randomised multicentre, open-label, phase 2 trial. Lancet Oncol. 2012; 2-positive early breast cancer and tumors #2cm: A meta-analysis of the
13:25-32. randomized trastuzumab trials. J Clin Oncol. 2015;33:2600-2608.
24. Schneeweiss A, Chia S, Hickish T, et al. Pertuzumab plus trastuzumab in 40. Fehrenbacher L, Capra AM, Quesenberry CP Jr, et al. Distant invasive
combination with standard neoadjuvant anthracycline-containing and breast cancer recurrence risk in human epidermal growth factor re-
anthracycline-free chemotherapy regimens in patients with HER2- ceptor 2-positive T1a and T1b node-negative localized breast cancer
positive early breast cancer: a randomized phase II cardiac safety diagnosed from 2000 to 2006: a cohort from an integrated health care
study (TRYPHAENA). Ann Oncol. 2013;24:2278-2284. delivery system. J Clin Oncol. 2006;32:2151-2158.
25. Baselga J, Bradbury I, Eidtmann H, et al; NeoALTTO Study Team. 41. Rodrigues JM, Wassermann J, Albiges L, et al. Trastuzumab treatment
Lapatinib with trastuzumab for HER2-positive early breast cancer in T1ab, node-negative human epidermal growth factor receptor
(NeoALTTO): a randomised, open-label, multicentre, phase 3 trial. 2-overexpressing breast carcinomas. J Clin Oncol. 2010;28:e541-e542.
Lancet. 2012;379:633-640. 42. Gianni L, Pienkowski T, Im Y, et al. Five-year analysis of the phase II
26. Guarneri V, Frassoldati A, Bottini A, et al. Preoperative chemo- NeoSphere trial evaluating four cycles of neoadjuvant docetaxel (D)
therapy plus trastuzumab, lapatinib, or both in human epidermal and/or trastuzumab (T) and/or pertuzumab (P). J Clin Oncol. 2015;33:
growth factor receptor 2-positive operable breast cancer: results of 15s (suppl; abstr 505).
the randomized phase II CHER-LOB study. J Clin Oncol. 2012;30: 43. Joensuu H, Bono P, Kataja V, et al. Fluorouracil, epirubicin, and cy-
1989-1995. clophosphamide with either docetaxel or vinorelbine, with or without
27. Robidoux A, Tang G, Rastogi P, et al. Lapatinib as a component of trastuzumab, as adjuvant treatments of breast cancer: final results of
neoadjuvant therapy for HER2-positive operable breast cancer (NSABP the FINHER trial. J Clin Oncol. 2009;27:5684-5692.
protocol B-41): an open-label, randomised phase 3 trial. Lancet Oncol. 44. Pivot X, Romieu G, Debled M, et al. 6 months versus 12 months of
2013;14:1183-1192. adjuvant trastuzumab for patients with HER2-positive early breast cancer
28. Mariotto AB, Yabroff KR, Shao Y, et al. Projections of the cost of cancer PHARE: a randomized phase 3 trial. Lancet Oncol. 2013;14:741-748.
care in the United States: 2010-2020. J Natl Cancer Inst. 2011;103: 45. Thill M. New frontiers in oncology: biosimilar monoclonal antibodies for
117-128. the treatment of breast cancer. Expert Rev Anticancer Ther. 2015;15:
29. Schnipper LE, Davidson NE, Wollins DS, et al; American Society of 331-338.
Clinical Oncology. American Society of Clinical Oncology Statement: A 46. Garrison LP Jr, Lubeck D, Lalla D, et al. Cost-effectiveness analysis of
Conceptual Framework to Assess the Value of Cancer Treatment Op- trastuzumab in the adjuvant setting for treatment of HER2-positive
tions. J Clin Oncol. 2015;33:2563-2577. breast cancer. Cancer. 2007;110:489-498.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e63



MARTIN AND LOPEZ-TARRUELLA

Emerging Therapeutic Options for HER2-Positive Breast


Cancer
Miguel Martin, MD, PhD, and Sara Lopez-Tarruella,
MD, PhD

OVERVIEW

The natural history of HER2-positive breast cancer has progressively improved since the introduction of the first anti-HER2
directed therapy (trastuzumab). Trastuzumab has significantly increased survival of patients with HER2-positive metastatic
breast cancer and, after the standardization of the use of this drug in the adjuvant setting in 2005, has also avoided many
disease recurrences and, consequently, saved many lives. Later on, the introduction of lapatinib offered new choices for
patients with advanced HER2-positive breast cancer, although the drug has failed to show a clear efficacy in the adjuvant
setting. New promising drugs have been approved to broaden the horizon of HER2-positive breast cancer such as per-
tuzumab or T-DM1, but we need new options to further improve the management of these diseases. In this review, we cover
new strategies that are currently under evaluation for the treatment of patients with HER2-positive breast cancer, including
new tyrosine kinase inhibitors (neratinib, ONT-380), new antibody-drug conjugates targeting HER2 (MM-302), and new
indications of already approved drugs (T-DM1), as well as the potential dual combinations of anti-HER2 therapy with
phosphoinositide 3-kinase/mTOR or cell cycle inhibitors (palbociclib, abemaciclib). Last but not least, we briefly review a new
paradigm of emerging approaches that involve the host immune response, HER2 breast cancer vaccines, and other immune
strategies, including immune checkpoint inhibition.

A dvances in the treatment of patients with HER2-positive


breast cancer have been made over the past few de-
cades, both in the metastatic setting and in the neo/adjuvant
ORAL, SMALL-MOLECULE INHIBITORS
Neratinib
Neratinib is an oral, small-molecule, anti-HER receptor ty-
treatment of earlier stages. In the neo/adjuvant setting, the rosine kinase inhibitor with a complex history. It was designed
combination of chemotherapy, trastuzumab, and perhaps in the early 1990s at Lederle Laboratories (Pearl River, NY) and
pertuzumab (for high-risk patients) has significantly im- was later was developed by Wyeth-Ayerst (Quebec, Canada),
proved patient outcomes. In the metastatic setting, the Pfizer (Quebec, Canada), and, finally, Puma Biotechnology
outcome of patients with HER2-positive tumors has also (Los Angeles, CA), the current owner of the drug.3,4 Neratinib
significantly improved, owing to the introduction of effective is a covalent drug that, differently from lapatinib, binds ir-
anti-HER therapies, including trastuzumab, pertuzumab, reversibly to the ATP active site of the tyrosine kinase domain
lapatinib, and T-DM1. The reported median survival time of HER2. A phase I study of neratinib among patients with
using these modern combination therapies is now approx- solid tumors showed that the maximum tolerated dose and
imately 5 years compared with approximately 1.5 years in the recommended dose for phase II trials was 320 mg and
the pretrastuzumab era.1,2 240 mg once daily, respectively. Grade 3 diarrhea was the
Despite this, 15%20% of patients with HER2-positive local- dose-limiting toxicity.5 The drug has good oral bioavailability
regional breast cancer still relapse after receiving the currently and, in the presence of food, the half-life elimination on day
available neo/adjuvant therapies. In addition, metastatic HER2- 1 after a single 240-mg administration is 14 hours, supporting
positive breast cancer essentially remains an incurable disease, a once-daily dosing regimen.
despite the improvement in survival seen after the introduction Neratinib was initially tested in a phase II trial of patients
of the new anti-HER2 therapies. Newer therapies and newer with breast cancer with HER2-positive metastatic tumors (66
approaches are therefore necessary to further improve the patients with and 70 without prior trastuzumab therapy,
current results. Here, we review the most promising emerging respectively) at a dose of 240 mg once daily. The study
therapeutic options currently in development for the treat- showed that single-agent neratinib has relevant antitumor
ment of HER2-positive breast cancer (Table 1). activity among patients with HER2-positive breast cancer

From the Instituto de Investigacion ~on,


Sanitaria Gregorio Maran Universidad Complutense, Madrid, Spain.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Miguel Martin, MD, PhD, Servicio de Oncologa Medica,


Instituto de Investigacion ~on,
Sanitaria Gregorio Maran Universidad Complutense, Dr. Esquerdo 46,
28007 Madrid, Spain; email: mmartin@geicam.org.

2016 by American Society of Clinical Oncology.

e64 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


EMERGING THERAPEUTIC OPTIONS FOR HER2 BREAST CANCER

who were pretreated. Objective response rates (ORRs) were TABLE 1. Emerging Therapies for HER2-Positive Breast
24% for patients who received prior trastuzumab treatment Cancer
and 56% for trastuzumab-naive patients. Responses were of
long duration (9 months for patients pretreated with tras- Class Drug Mechanism of Action
tuzumab and 12 months for trastuzumab-naive patients). Oral, Anti-HER2, Neratinib Tyrosine kinase inhibitor (HER2
Median progression-free survival (PFS) was approximately Small and HER1)
Molecules
5 months for patients pretreated with trastuzumab and ONT-380 Tyrosine kinase inhibitor (HER2)
approximately 9 months for trastuzumab-naive patients. Anti-HER2 Anti- T-DM1 Trastuzumab-like effect
The downside of the treatment was the high incidence of Body-Drug
Intracellular release of emtan-
Conjugates
adverse events, particularly diarrhea and, to a much lesser sine-cytotoxic toxicity in HER2-
overexpressing cells
extent, nausea, vomiting, and fatigue. Diarrhea was reported
MM-302 Intracellular release of pegylated
for 85% of patients pretreated with trastuzumab (grade 3 to liposomal doxorubicin in
4 for 30% of patients). The incidence of diarrhea was greater HER2-overexpressing cells
in the first week of therapy (85%) but significantly declined mTOR/PI3K/Akt Everolimus mTOR inhibition
with loperamide therapy from that time onward (60% at Inhibitors
Buparlisib Pan-class I PI3K inhibitor
weeks 2 to 4, less than 40% at month 2, and approximately
Pictilisib Pan-class I PI3K inhibitor
15% at month 3).6
Taselisib Alpha-specific PI3K inhibitor
Based on these appealing phase II data, single-agent
Alpelisib Alpha-specific PI3K inhibitor
neratinib was compared with the combination of lapatinib
and capecitabine in a phase II noninferiority study. The CDK4/6 Palbociclib CDK4/6 inhibition, cell cycle arrest
Inhibitors
comparator was the standard therapy for trastuzumab- Abemaciclib CDK4/6 inhibition, cell cycle arrest
pretreated HER2-positive metastatic breast cancer at the Vaccines E75 Peptide-based vaccine
time the study was designed (capecitabine plus lapatinib). Of GP2 Peptide-based vaccine
the study patients, 117 were allocated to receive 240 mg Immune Pembrolizumab AntiPD-1
of neratinib daily and 116 patients received 1,250 mg of Checkpoint
lapatinib daily plus 2,000 mg/m2 of capecitabine daily on Inhibitors
days 1 to 14 of each 21-day cycle. The study failed to show Abbreviation: PI3K, phosphoinositide 3-kinase.

noninferiority of neratinib compared with lapatinib plus


capecitabine. However, neratib showed indisputable single-agent
activity in the trial, with a response rate of 29% versus 41%
with the combination of lapatinib and capecitabine (p = .067).
KEY POINTS Median PFS and overall survival (OS) times were 4.5 and
19.7 months (neratinib) versus 6.8 and 23.6 months for the
The currently available anti-HER2 therapies have combination, respectively. Eighty-five percent of patients taking
changed the natural history of HER2-positive breast neratinib experienced diarrhea (grade 3 or greater for 28% of
cancer, but new therapeutic options are necessary patients). Diarrhea was more frequent in the first cycle of therapy
because the disease is essentially incurable in the and was properly managed with loperamide or dose modifica-
metastatic setting and a relevant proportion of patients tions for most patients.7
with early-stage disease still relapse in spite of the use of In view of the lack of demonstration of noninferiority of
currently available therapies.
neratinib versus the standard lapatinib plus capecitabine and to
Neratinib is one of the most promising new drugs for
HER2-positive breast cancer, although the management
prepare the registration trial of neratinib for trastuzumab-
of its main side effect, diarrhea, should be addressed pretreated HER2-positive metastatic breast cancer, a phase
and resolved to allow safe use of the drug. I/II study combining neratinib and capecitabine was con-
Other drugs, such as ONT-380 (an oral, small-molecule, ducted.8 The study was carried out in two parts. The first part
HER2-selective inhibitor) and MM-302 (an anti-HER2 was a dose-escalation trial among 33 patients with advanced
antibody-drug conjugate carrying pegylated liposomal solid tumors; they received oral neratinib once a day contin-
doxorubicin) have shown initial promising activity and uously plus capecitabine twice a day (days 1 to 14 every
good tolerance among patients with HER2-positive 3 weeks) at predefined dose levels, to define the recom-
breast cancer. mended dose/schedule for phase II/III trials. In part two, 72
Interest in the inhibition of downstream signaling of the patients with trastuzumab-pretreated HER2-positive meta-
HER2 pathway with mTOR/phosphoinositide 3-kinase/
static breast cancer received the combination at the recom-
Akt and CDK4/6 inhibitors is currently under clinical
evaluation.
mended dose for phase II trials (240 mg of neratinib per day
Some immunologic approaches (vaccines, alone or in continuously plus 1,500 mg/m2 of capecitabine for 2 weeks
combination with trastuzumab, inhibitors of every 21 days).
immunocheckpoints, and others) are also being tested In part two, the ORR was 64% for patients with no prior
in HER2-positive breast cancer. lapatinib exposure and 57% for patients previously treated with
lapatinib. Median PFS was approximately 9 and 8 months,

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e65



MARTIN AND LOPEZ-TARRUELLA

respectively. The most common side effects of the combination that, in combination with standard therapy, meet a high
were diarrhea (88%) and palmar-plantar erythrodysesthesia Bayesian predictive probability of statistical significance in a
syndrome (48%). This study showed that the combination of future phase III neoadjuvant trial performed among the same
neratinib and capecitabine possesses significant antitumor population. Patients with HER2-positive breast cancer were
activity in trastuzumab-pretreated metastatic HER2-positive randomly assigned to receive 12 weekly administrations of
breast cancer. A randomized registration trial (NALA; paclitaxel plus trastuzumab or paclitaxel plus neratinib, fol-
NCT01808573) comparing neratinib plus capecitabine with lowed by four cycles of doxorubicin hydrochloride plus cy-
lapatinib plus capecitabine is underway. clophosphamide (AC) before surgery. Neratinib met the
A randomized phase II trial (NEFERTT; NCT00915018), predictive probability criterion in the population with HR-
which included 479 patients with HER2-positive breast negative/HER2-positive tumors. A phase III confirmatory trial
cancer without any therapy for metastatic disease, com- with neratinib is planned for patients with HER2-positive breast
pared paclitaxel plus trastuzumab with paclitaxel plus ner- cancer (I-SPY 3, a phase III registration trial).
atinib. No statistically significant differences in PFS (the Neratinib has been evaluated in an adjuvant phase III trial,
primary endpoint) or ORR (one of the trials secondary the ExteNET study. Eligible patients had operable-stage
endpoints) were seen.9 Grade 3 diarrhea was reported for HER2-positive breast cancer with no relapse after surgery,
approximately 30% of patients receiving neratinib plus chemotherapy, and up to 2 years after the completion of
paclitaxel compared with approximately 4% of patients in 1 year of trastuzumab. Patients were randomly assigned to
the paclitaxel plus trastuzumab arm. Central nervous system receive neratinib or placebo for 1 year (together with
(CNS) progression occurred in 8.3% of patients in the ner- hormonal therapy for patients with HR-positive tumors). In
atinib arm versus 17.3% in the trastuzumab arm (RR 0.48; the initial design, the trial had a 90% power to detect a
p = .002). However, the proportion of patients with CNS hazard ratio of 0.7 for invasive disease-free survival (iDFS),
involvement at study initiation was superior in the paclitaxel at a two-sided 5% significance level. The study design suf-
arm (12 patients; 5.1%) compared with the neratinib arm fered from some modifications over time because the drug
(6 patients; 2.5%). owner changed and three different sponsors assumed re-
The significant activity of neratinib observed in metastatic sponsibility. In the initial design, patients who were T$1c
HER2-positive breast cancer has prompted the evaluation of node negative were eligible, a sample size of 3,850 patients
the drug in earlier stages. A phase II trial conducted by the was established, and iDFS was the main endpoint. In view of
NSABP group (NSABP FG-7 Trial) compared weekly paclitaxel the good outcome of node-negative patients with standard
plus neratinib or trastuzumab or the combination of ner- trastuzumab-containing chemotherapy later reported in
atinib and trastuzumab followed by standard doxorubicin other adjuvant trials (particularly the BCIRG 006 study), an
and cyclophosphamide as neoadjuvant therapy among initial amendment restricted recruitment to patients with
126 patients with HER2-positive locally advanced, operable node-positive disease with trastuzumab completion 1 year
breast cancer.10 The proportions of patients with inflam- or earlier before randomization. In October 2011, the new
matory breast cancer (a bad prognostic feature) were 16.7% sponsor introduced two key changes: cessation of enroll-
(neratinib arm) and 9.5% (in both the trastuzumab and the ment (only 2,842 patients of the initially planned of 3,850
trastuzumab plus neratinib arms). The pCR rates (breast plus had been enrolled at that time) and shortening of follow-up
axillary lymph nodes) were 38.1% (trastuzumab arm), 33.3% to 2 years from randomization. In January 2014, a new
(neratinib arm), and 50% (neratinib plus trastuzumab arm). amendment re-established the extension of data collection
The apparent superiority of the combination arm was for disease events and deaths to 5 years postrandomization
mainly attributable to the effect on patients with hormone for consenting patients and the primary endpoint of iDFS at
receptor (HR)negative disease (pCR rates of 57.1%, 46.2%, 2 years of follow-up was resumed. Treatment allocation
and 73.7%, in the trastuzumab, neratinib, and combination remained blinded to the sponsor prior to the first analysis
arms, respectively). For patients with HR-positive disease, and an independent statistical evaluation by a reputed
the pCR rates were not significantly different among arms biostatistician who had access to all data was performed.
(29.6%, 27.6%, and 30.4%, respectively). Grade 3 diarrhea The detailed analyses showed that the results remained
was the most relevant side effect of neratinib (31% in both consistent throughout the trial and were trustable. The
neratinib arms vs. 0% in the trastuzumab arm). Intensive primary analysis was first reported in 201512 and published
primary prophylaxis with loperamide in neratinib-treated in detail in 2016.13 At 2 years from randomization, the hazard
patients implemented at European sites apparently reduced ratio for iDFS in the neratinib group compared with the
the incidence of grade 3 diarrhea (17% in the neratinib placebo group was 0.67 (p = .009), with 2-year iDFS rates of
arm and 24% in the neratinib plus trastuzumab arm across 93.9% and 91.6%, respectively. A preplanned subgroup
all cycles). analysis according to HR status showed that patients
Neratinib has also been evaluated in the I-SPY 2 program, with HR-positive tumors (the majority of whom were re-
a multicenter neoadjuvant trial among women with oper- ceiving concurrent hormonal therapy) obtained the greatest
able breast cancer using adaptive randomization within benefit with neratinib (hazard ratio for iDFS of 0.51, p = .001),
biomarker subtypes to evaluate novel antitumor drugs added whereas patients with HR-negative tumors had little or
to conventional therapy.11 I-SPY 2 intends to identify new drugs no benefit (hazard ratio for iDFS, 0.93; p = .735; interaction

e66 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


EMERGING THERAPEUTIC OPTIONS FOR HER2 BREAST CANCER

p = .054). Another preplanned analysis of iDFS for 1,463 the MARIANNE trial, in which T-DM1 plus pertuzumab was not
patients with centrally confirmed HER2-positive disease superior to taxane plus trastuzumab as a first-line treatment for
showed a hazard ratio of 0.51 (95% CI, 0.330.77; p = .002). patients in the metastatic setting.
The 3-year update of the ExteNET trial was recently reported The ADAPT trial19 is a phase II trial that enrolled 375 patients
and confirmed the earlier findings.14 OS data were not with HR-positive, operable, HER2-positive tumors and ran-
mature but will continue to be monitored. domly assigned them to receive neoadjuvant T-DM1, T-DM1
Diarrhea was the most frequent side effect of neratinib in plus endocrine therapy, or trastuzumab plus endocrine therapy
the ExteNET trial and occurred among most patients (grade 2 for 12 weeks prior to surgery. The pathologic complete re-
in 32.5%, grade 3 in 39.8%, and grade 4 in 0.1%). Ustaris sponse rates (breast plus axilla) were 41%, 41.5%, and 15.1%,
et al15 recently reviewed the characteristics and treatment respectively.
of neratinib-induced diarrhea. This side effect usually starts The phase III KATHERINE trial (NCT01772472) compares
during the first month of therapy and resolves or decreases adjuvant trastuzumab versus adjuvant T-DM1 for patients with
later among most patients either spontaneously or with HER2-positive tumors in which the tumor persisted in the
loperamide or dose reductions. Diarrhea is a cause of dose breast or axilla after neoadjuvant chemotherapy plus trastu-
reduction for 10%15% of patients. Permanent discontin- zumab. The KRISTINE trial (TRIO-021; NCT02131064) is an
uation of neratinib because of diarrhea occurs in a minority ongoing phase III trial in which patients are randomly assigned
of patients (0%14% of patients in the reviewed trials) and to neoadjuvant standard docetaxel, carboplatin, trastuzumab,
seems to be reduced by the introduction of early, intense and pertuzumab or to T-DM1 plus pertuzumab, with pCR as the
loperamide prophylaxis. A prospective clinical trial is eval- main endpoint.
uating the efficacy of prophylactic loperamide among pa-
tients with HER2-positive operable breast cancer receiving
extended adjuvant neratinib (NCT02400476). Loperamide is MM-302
administered at a dose of 4 mg three times a day for the first MM-302 is an ADC composed of a HER2-directed antibody
2 weeks of neratinib and 4 mg twice a day until week 8. (lacking anti-HER2 activity) linked to pegylated liposomal
doxorubicin. The antibody acts as a carrier delivering pegylated
liposomal doxorubicin into HER2-positive breast cancer cells.
ONT-380 The drug has been tested in a phase I trial with 69 patients
ONT-380 is an oral, small-molecule, HER2-selective inhibitor in with HER2-positive metastatic breast cancer after a median of
clinical development. It is speculated that the lack of inhibition four prior regimens for metastatic disease. Patients were
of HER1 could translate into a lower frequency of skin and treated with either MM-302 alone, MM-302 plus trastuzumab,
gastrointestinal toxicity with respect to HER2/HER1 dual in- or MM-plus trastuzumab and cyclophosphamide. The most
hibitors (i.e., lapatinib, or neratinib). In fact, a phase I trial with frequent adverse events (. 20% of patients) were con-
ONT-380 as a single agent showed no treatment-related grade stipation, cough, decreased appetite, diarrhea, dyspnea, fa-
3 diarrhea and only minimal skin toxicity. In a phase Ib trial of tigue, nausea, neutropenia, stomatitis, and vomiting. As a
ONT-380 in combination with T-DM1 among patients who single agent, a maximum tolerated dose was not reached at
received prior taxane and trastuzumab treatment, a 41% ORR 50 mg/m2. Cardiotoxicity was seen only among patients with
was reported.16 A second study reported the CNS activity of extensive prior exposure to regular anthracyclines. Antitumor
ONT-380 in combination with either T-DM1 or trastuzumab or activity was observed for 49 patients treated with 30 mg/m2 of
capecitabine. Patients with brain metastases assessable for MM-302 or greater, alone or in combination with trastuzumab,
response were included in the combined analysis. Responses with a response rate of 12% and median PFS of 7.6 months.
and clinical benefit in the CNS were reported with the three Responses were seen for patients pretreated with trastuzu-
combinations tested, supporting future development of the mab, T-DM1, and pertuzumab. The median PFS among the 13
drug for this particular indication.17 patients receiving MM-302 plus trastuzumab and cyclophos-
phamide was 10.6 months.20 In view of these data, a ran-
ANTIBODY-DRUG CONJUGATES domized phase II study (the HERMIONE trial; NCT02213744) is
T-DM1 ongoing. The study includes patients with metastatic HER2-
T-DM1 is the first antibody-drug conjugate (ADC) approved for positive breast cancer who received prior trastuzumab, T-DM1,
metastatic HER2-positive breast cancer for the indication of and pertuzumab treatment (but not anthracyclines in any
patients pretreated with taxanes and trastuzumab.18 The role setting) and randomly assigns patients to receive MM-302 plus
of T-DM1 in earlier stages of the disease is currently under trastuzumab or trastuzumab plus a single chemotherapy agent
evaluation. The KAITLIN trial (NCT01966471) is an adjuvant at the physicians choice.
study in operable HER2-positive breast cancer. After surgery,
patients are randomly assigned to receive four cycles of INHIBITORS OF DOWNSTREAM SIGNALING
anthracycline-containing chemotherapy followed by combi- PATHWAYS
nation treatment with taxane, trastuzumab, and pertuzumab mTOR/Phosphoinositide 3-Kinase/Akt Inhibitors
or with T-DM1 and pertuzumab. The study enrollment has The mTOR/phosphoinositide 3-kinase (PI3K)/Akt pathway
stopped and the protocol was amended in view of the results of plays an important role in the downstream signaling of

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e67



MARTIN AND LOPEZ-TARRUELLA

the HER2 pathway and is a potential target for combined study is exploring the combination of palbociclib and T-DM1
therapies.21 The BOLERO 3 trial studied adding the mTOR in metastatic HER2-positive breast cancer after prior tras-
inhibitor everolimus to conventional chemotherapy tuzumab or other HER2-directed therapies (NCT01976169).
(vinorelbine-trastuzumab) among patients with metastatic The PATRICIA trial (NCT02448420) is a phase II study of
HER2-positive breast cancer who were pretreated with palbociclib and trastuzumab with or without letrozole among
trastuzumab and taxanes. The addition of everolimus sig- patients with HER2-positive metastatic breast cancer who
nificantly improved PFS compared with vinorelbine- have received chemotherapy and treatment with trastu-
trastuzumab alone.22 The BOLERO 3 trial provided the zumab for their metastatic disease.
proof of principle that the blockade of mTOR can play a role Abemaciclib, another CDK4/6, has been safely combined
in the management of HER2-positive breast cancer; how- with trastuzumab in a phase Ib study (NCT02057133). Di-
ever, the significant increase in toxicity associated with arrhea was a dose-limiting toxicity. The recommended dose
everolimus administration makes this combination un- for further studies is 150 mg/12 hours.28
attractive. The BOLERO 1 trial, a first-line study comparing
paclitaxel plus trastuzumab with or without everolimus, VACCINES AND OTHER FORMS OF
failed to show any significant advantage in PFS with ever- IMMUNOTHERAPY
olimus in the overall group of 719 patients. However, the HR- Tumor Vaccines
negative subpopulation obtained an apparent benefit with This strategy aims to amplify the patients immune response
the addition of everolimus (median PFS of 20.27 months vs. against HER2-positive breast cancer. The majority of breast
13.08 months with placebo, p = .0049), although the pre- cancer vaccine trials were initially performed in advanced
planned protocol-specified significance threshold (p = .0044) stages of the disease and the results were somewhat dis-
was not crossed.23 Slamon et al24 used next-generation appointing.29 The high genomic instability and the loss of
sequencing (NGS) and immunohistochemistry (IHC) to es- antigens,30 together with defects in the antitumor immune
tablish the status of the PI3K pathway in tumor samples response (T-cell activation/function and regulation) found in
(mainly primary tumors) from patients from the BOLERO metastatic breast cancer, can explain these negative results.
1 and 3 trials. Exons of 282 cancer-related genes were an- Afterward, the cornerstones of clinical development of
alyzed by NGS, whereas PTEN (phosphatase and tensin cancer vaccines were redefined,31 emphasizing the need for
homolog) levels were determined by IHC. Patients with low selecting an appropriate clinical context; herein, early or
PTEN or known PIK3CA or AKT1 E17K mutations were minimal residual disease stages were proposed as more
considered to have PI3K pathway activation. Patients with suitable scenarios to be explored. Several vaccine formu-
PI3K pathway hyperactivity showed a significantly better PFS lations have been tested among patients with HER2-positive
with exemestane, but the remaining patients did not.24 The breast cancer.32
role of everolimus in the routine therapy of metastatic HER2- Peptide-based vaccines use antigen epitopes of cancer
positive breast cancer is therefore currently undefined. cells combined with different adjuvants to elicit immune
Clinical trials of PI3K pathway inhibitors (both pan-class responses. The main advantages of this approach include
I and alpha-specific inhibitors) for the treatment of meta- their easy manufacture at low cost, storage stability, safety,
static HER2-positive breast cancer are underway. In a phase and measurable antigen-specific immune response in the
Ib trial of buparlisib plus trastuzumab for patients with HER2- blood (direct monitoring of T-cell response).33
positive metastatic breast cancer that progressed while they E75 is an immunogenic peptide derived from the extra-
were taking trastuzumab, the combination was well toler- cellular domain of HER2. Small-size phase I trials tested E75
ated and showed some activity; at the recommended dose with different immune-adjuvants in metastatic breast and
for phase II trials, there were two partial responses (17%) ovarian cancer, demonstrating that the peptide was able to
and seven disease stabilizations of 6 weeks or longer induce specific cytotoxic T-lymphocyte responses with a
(58%).25 Several studies are testing the activity of other favorable safety profile. However, no meaningful antitumor
PI3K inhibitors, including alpelisib (NCT02038010), taselisib activity was seen. Because the metastatic studies showed
(NCT02390427), and pictilisib (NCT00960960). that this active immunization could last several years after
the vaccination process, the vaccine was later tested in the
adjuvant setting. E75, in combination with granulocyte-
CDK4/6 Inhibitors macrophage colony-stimulating factor (GM-CSF), was eval-
CDK4/6 controls a key pathway downstream of HER2. The uated in a small randomized trial of patients with high-risk
activated cyclin DCDK4/6 heterodimer promotes cell cycle breast cancer with any level of HER2 and HLA-A2+, dem-
progression. Deregulation of cell cycle control is not un- onstrating its safety and effectiveness in eliciting immune
common in estrogen receptorpositive and HER2-positive response in vivo in node-positive breast cancer. Recurrence
breast cancer; therefore, inhibition of CDK4/6 could be a rates were lower in the vaccination arm (21% in the ob-
way to augment the effectiveness of standard anti-HER2 servation group vs. 8% in the vaccine group at a median
therapies. 22 months of follow-up).34 The final results of the phase I/II
Palbociclib, a CDK4/6 inhibitor, is synergistic with trastuzumab clinical trial of E75 among 195 patients with early breast
and T-DM1 in HER2-positive preclinical models.26,27 A phase Ib cancer were recently reported with a 60-month follow-up

e68 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


EMERGING THERAPEUTIC OPTIONS FOR HER2 BREAST CANCER

time. A nonstatistically significant difference in 5-year autologous or from cell-line cultures), and dendritic cell
disease-free survival (DFS) in favor of the vaccine group was vaccines.36,37
reported (89.7% vs. 80.2%, p = .08). A phase III clinical trial The synergy of trastuzumab and vaccines was explored in a
with E75 is currently ongoing (NCT01479244). phase I/II trial obtaining prolonged and robust T-cell re-
GP2 is a peptide from the transmembrane HER2 protein sponses with very low toxicity.38 Ongoing trials are even
domain found to be able to stimulate CD8 lymphocytes to considering the addition of chemotherapy to vaccine/
destroy HER2-overexpressing cells. The preliminary results antibody combinations (NCT00266110). A further step in
of a phase II trial among patients with HLA-A2+ with any level the HER2 vaccination field is the generation of HER2 vaccine-
of expression of HER2 were recently reported.35 The com- primed autologous T cells for therapeutic infusion reported
bination of GP2 plus GM-CSF was compared with GM-CSF to be feasible and well tolerated in early phase I trials.39
alone in patients with breast cancer whose tumors had
diverse expression of HER2. The treatment was well toler-
ated. In the subset of patients with HER2-overexpressing Immune Checkpoint Inhibitors
tumors treated with trastuzumab, the combination of GP2 The clinical testing of immune checkpoint inhibitors in breast
vaccine plus GM-CSF was associated with a numerically cancer, initially focused on triple-negative breast cancer, has
superior DFS (94% vs. 89% in the GM-CSF group, p = .86). recently been extended to the HER2-positive subtype.
Other immunogenic approaches are also under clinical PANACEA (NCT02129556) is a phase IB/II trial evaluating the
testing, including peptides derived from the HER2 intracellular efficacy of pembrolizumab and trastuzumab in trastuzumab-
domain, DNA-based and whole tumor cell vaccines (either resistant metastatic breast cancer.

References
1. Swain SM, Baselga J, Kim SB, et al; CLEOPATRA Study Group. Pertu- 11. Park JW, Liu MC, Yee D, et al. Neratinib plus standard neoadjuvant
zumab, trastuzumab, and docetaxel in HER2-positive metastatic breast therapy for high-risk breast cancer: efficacy results from the I-SPY 2
cancer. N Engl J Med. 2015;372:724-734. TRIAL. Cancer Res. 2014;74 (abstr CT227).
2. Slamon DJ, Leyland-Jones B, Shak S, et al. Use of chemotherapy plus a 12. Chan A, Delaloge S, Holmes FA, et al. Neratinib after adjuvant che-
monoclonal antibody against HER2 for metastatic breast cancer that motherapy and trastuzumab in HER2-positive early breast cancer:
overexpresses HER2. N Engl J Med. 2001;344:783-792. primary analysis at 2 years of a phase 3 randomized, placebo-controlled
3. Tsou HR, Overbeek-Klumpers EG, Hallett WA, et al. Optimization of 6,7- trial (ExteNET). J Clin Oncol. 2015;33 (suppl; abstr 508).
disubstituted-4-(arylamino)quinoline-3-carbonitriles as orally active, 13. Chan A, Delaloge S, Holmes FA, et al. Neratinib after trastuzumab-based
irreversible inhibitors of human epidermal growth factor receptor-2 adjuvant therapy in HER2-positive breast cancer (ExteNET): a ran-
kinase activity. J Med Chem. 2005;48:1107-1131. domized, double-blind, phase 3 trial. Lancet Oncol. 2016;17:367-377.
4. Lopez-Tarruella
S, Jerez Y, Marquez-Rodas I, et al. Neratinib (HKI-272) in 14. Chan A, Delaloge S, Holmes FA, et al. Neratinib after trastuzumab-based
the treatment of breast cancer. Future Oncol. 2012;8:671-681. adjuvant therapy in early-stage HER2+ breast cancer: 3-year analysis
5. Wong KK, Fracasso PM, Bukowski RM, et al. A phase I study with from a phase 3 randomized, placebo-controlled double-blind trial
neratinib (HKI-272), an irreversible pan ErbB receptor tyrosine kinase (ExteNET). Presented at: San Antonio Breast Cancer Symposium. San
inhibitor, in patients with solid tumors. Clin Cancer Res. 2009;15: Antonio, TX; 2015. Abstract S5-02.
2552-2558. 15. Ustaris F, Saura C, Di Palma J, et al. Effective management and pre-
6. Burstein HJ, Sun Y, Dirix LY, et al. Neratinib, an irreversible ErbB receptor vention of neratinib-induced diarrhea. Am J Hematol Oncol. 2015;11:
tyrosine kinase inhibitor, in patients with advanced ErbB2-positive 13-22.
breast cancer. J Clin Oncol. 2010;28:1301-1307. 16. Ferrario C, Hamilton E, Aucoin N, et al. A phase 1b study of ONT 380, an
7. Martin M, Bonneterre J, Geyer CE Jr, et al. A phase two randomised oral HER2-specific inhibitor, combined with ado trastuzumab emtansine
trial of neratinib monotherapy versus lapatinib plus capecitabine (T-DM1), in HER2+ metastatic breast cancer (MBC). Presented at: San
combination therapy in patients with HER2+ advanced breast cancer. Antonio Breast Cancer Symposium. San Antonio, TX; 2015. Abstract P4-
Eur J Cancer. 2013;49:3763-3772. 14-20.
8. Saura C, Garcia-Saenz JA, Xu B, et al. Safety and efficacy of neratinib in 17. Murthy RK, Hamilton E, Borges VF, et al. ONT-380 in the treatment of
combination with capecitabine in patients with metastatic human HER2+ breast cancer central nervous system (CNS) metastases (mets).
epidermal growth factor receptor 2-positive breast cancer. J Clin Oncol. Presented at: San Antonio Breast Cancer Symposium. San Antonio, TX;
2014;32:3626-3633. 2015. Abstract P4-14-19.
9. Awada A, Colomer R, Bondarenko I, et al. Efficacy and CNS progression 18. Krop IE, Kim SB, Gonza lez-Martn A, et al; TH3RESA Study Collab-
analysis from the randomized phase 2 trial of neratinib + paclitaxel vs orators. Trastuzumab emtansine versus treatment of physicians
trastuzumab + paclitaxel as first-line treatment for HER2+ MBC choice for pretreated HER2-positive advanced breast cancer
(NEfERTT). J Clin Oncol. 2015;33 (suppl; abstr 610). (TH3RESA): a randomised, open-label, phase 3 trial. Lancet Oncol.
10. Jacobs SA, Robidoux A, Garcia JMP, et al. NSABP FB-7: a phase II 2014;15:689-699.
randomized trial evaluating neoadjuvant therapy with weekly paclitaxel 19. Harbeck N, Gluz O, Christgen M, et al. Final analysis of WSG-ADAPT HER2
(P) plus neratinib (N) or trastuzumab (T) or neratinib and trastuzumab +/HR+ phase II trial: Efficacy, safety, and predictive markers for 12-
(N+T) followed by doxorubicin and cyclophosphamide (AC) with weeks of neoadjuvant TDM1 with or without endocrine therapy versus
postoperative T in women with locally advanced HER2-positive breast trastuzumab+endocrine therapy in HER2-positive hormone-receptor-
cancer. Presented at: San Antonio Breast Cancer Symposium. San positive early breast cancer. Presented at: San Antonio Breast Cancer
Antonio, TX; 2015. Abstract PD5-04. Symposium. San Antonio, TX; 2015. Abstract S5-03.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e69



MARTIN AND LOPEZ-TARRUELLA

20. LoRusso P, Krop I, Miller K, et al. A phase I study of MM-302, a HER2- 29. Rosenberg SA, Yang JC, Restifo NP. Cancer immunotherapy: moving
targeted PEGylated liposomal doxorubicin, in patients with HER2+ beyond current vaccines. Nat Med. 2004;10:909-915.
metastatic breast cancer. Cancer Res. 2015;75 (abstr CT234). 30. Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: integrating
21. Rexer BN, Arteaga CL. Optimal targeting of HER2-PI3K signaling in breast immunitys roles in cancer suppression and promotion. Science. 2011;
cancer: mechanistic insights and clinical implications. Cancer Res. 2013; 331:1565-1570.
73:3817-3820. 31. Hoos A, Parmiani G, Hege K, et al; Cancer Vaccine Clinical Trial Working
22. Andre F, ORegan R, Ozguroglu M, et al. Everolimus for women with Group. A clinical development paradigm for cancer vaccines and related
trastuzumab-resistant, HER2-positive, advanced breast cancer biologics. J Immunother. 2007;30:1-15.
(BOLERO-3): a randomised, double-blind, placebo-controlled phase 3 32. Milani A, Sangiolo D, Montemurro F, et al. Active immunotherapy in
trial. Lancet Oncol. 2014;15:580-591. HER2 overexpressing breast cancer: current status and future per-
23. Hurvitz SA, Andre F, Jiang Z, et al. Combination of everolimus with spectives. Ann Oncol. 2013;24:1740-1748.
trastuzumab plus paclitaxel as first-line treatment for patients with 33. Clifton GT, Mittendorf EA, Peoples GE. Adjuvant HER2/neu peptide
HER2-positive advanced breast cancer (BOLERO-1): a phase 3, rando- cancer vaccines in breast cancer. Immunotherapy. 2015;7:1159-1168.
mised, double-blind, multicentre trial. Lancet Oncol. 2015;16:816-829. 34. Peoples GE, Gurney JM, Hueman MT, et al. Clinical trial results of a
24. Slamon DJ, Hurvitz SA, Chen D, et al. Predictive biomarkers of ever- HER2/neu (E75) vaccine to prevent recurrence in high-risk breast cancer
olimus efficacy in HER2+ advanced breast cancer: combined exploratory patients. J Clin Oncol. 2005;23:7536-7545.
analysis from BOLERO-1 and BOLERO-3. J Clin Oncol. 2015;33 (suppl; 35. Schneble EJ, Perez SA, Murray JL, et al. Primary analysis of the pro-
abstr 512). spective, randomized, phase II trial of GP2+GMCSF vaccine versus GM-
25. Saura C, Bendell J, Jerusalem G, et al. Phase Ib study of buparlisib plus CSF alone administered in the adjuvant setting to high-risk breast
trastuzumab in patients with HER2-positive advanced or metastatic cancer patients. J Clin Oncol. 2014;32 (suppl 26; abstr 134).
breast cancer that has progressed on tsrastuzumab-based therapy. Clin 36. Disis ML, Schiffman K, Guthrie K, et al. Effect of dose on immune re-
Cancer Res. 2014;20:1935-1945. sponse in patients vaccinated with an HER-2/neu intracellular domain
26. Cadoo KA, Gucalp A, Traina TA. Palbociclib: an evidence-based review of proteinbased vaccine. J Clin Oncol. 2004;22:1916-1925.
its potential in the treatment of breast cancer. Breast Cancer (Dove Med 37. Morse MA, Hobeika A, Osada T, et al. Long term disease-free survival
Press). 2014;6:123-122. and T cell and antibody responses in women with high-risk Her2+ breast
27. Knudsen E, Cox D, Franco J, et al. Targeting CDK4/6 in HER2 positive cancer following vaccination against Her2. J Transl Med. 2007;5:42.
breast cancer: therapeutic effect, markers, and combination strategies. 38. Disis ML, Wallace DR, Gooley TA, et al. Concurrent trastuzumab and
Ann Oncol. 2014;25(suppl 1):i21-i22. HER2/neu-specific vaccination in patients with metastatic breast can-
28. Goetz MP, Beeram M, Beck T, et al. Abemaciclib, an inhibitor of CDK4 cer. J Clin Oncol. 2009;27:4685-4692.
and CDK6, combined with endocrine and HER2-targeted therapies for 39. Disis ML, Dang Y, Coveler AL, et al. HER-2/neu vaccine-primed autol-
women with metastatic breast cancer. Presented at: San Antonio Breast ogous T-cell infusions for the treatment of advanced stage HER-2/neu
Cancer Symposium. San Antonio, TX; 2015. Abstract P4-13-25. expressing cancers. Cancer Immunol Immunother. 2014;63:101-109.

e70 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


CANCER PREVENTION, HEREDITARY GENETICS,
AND EPIDEMIOLOGY

Controversies in Genetic
Evaluation

CHAIR
Noah D. Kauff, MD
Memorial Sloan Kettering Cancer Center
New York, NY

SPEAKERS
Claudine Isaacs, MD, FRCPC
Georgetown Lombardi Comprehensive Cancer Center
Washington, DC

Victoria Raymond, MS, CGC


University of Michigan Medical School
Ann Arbor, MI
LYNCE AND ISAACS

How Far Do We Go With Genetic Evaluation? Gene, Panel, and


Tumor Testing
Filipa Lynce, MD, and Claudine Isaacs, MD, FRCPC

OVERVIEW

The traditional model by which an individual was identified as harboring a hereditary susceptibility to cancer was to test for a mutation
in a single gene or a finite number of genes associated with a particular syndrome (e.g., BRCA1 and BRCA2 for hereditary breast and
ovarian cancer or mismatch repair genes for Lynch syndrome). The decision regarding which gene or genes to test for was based on
a review of the patients personal medical history and their family history. With advances in next-generation DNA sequencing
technology, offering simultaneous testing for multiple genes associated with a hereditary susceptibility to cancer is now possible.
These panels typically include high-penetrance genes, but they also often include moderate- and low-penetrance genes. A number of
the genes included in these panels have not been fully characterized either in terms of their cancer risks or their management options.
Another way some patients are unexpectedly identified as carrying a germline mutation in a cancer susceptibility gene is at the time
they undergo molecular profiling of their tumor, which typically has been carried out to guide treatment choices for their cancer. This
article first focuses on the issues that need to be considered when deciding between recommending more targeted testing of a single
or a small number of genes associated with a particular syndrome (single/limited gene testing) versus performing a multigene panel.
This article also reviews the issues regarding germline risk that occur within the setting of ordering molecular profiling of tumors.

T hroughout the past several decades, we have witnessed


tremendous advances in our knowledge of evaluating
and treating patients with germline mutations in hereditary
supported by the recent American Society of Clinical Oncology
(ASCO) policy statement on testing for genetic and genomic
cancer susceptibility,5 a number of criteria must be considered
cancer syndromes, with studies clearly demonstrating the when evaluating existing or emerging genetic tests. These cri-
feasibility and clinical utility of genetic testing. Perhaps most teria include analytical and clinical validity, clinical utility, and
importantly, studies have provided convincing evidence the associated ethical, legal, and social issues. In the context of
that implementing prevention strategies in some instances genetic testing, analytic validity refers to the accuracy and re-
prolongs the survival of mutation carriers. For example, for producibility by which the assay detects the presence or absence
unaffected women who carry a BRCA1 or BRCA2 mutation, of a mutation. Clinical validity focuses on whether the test ac-
risk-reducing salpingo-oophorectomy results in a significant curately and reproducibly predicts the clinically defined disorder.
reduction in all-cause mortality (3% vs. 10%; hazard ratio Clinical utility can be defined as the evidence that a genetic test
[HR] 0.40; 95% CI, 0.260.6), breast cancer-specific mor- results in improved health outcomes typically based on early
tality (2% vs. 6%; HR 0.44; 95% CI, 0.260.76) and ovarian detection or prevention strategies, and the tests usefulness
cancerspecific mortality (0.4 vs. 3%; HR 0.21; 95% CI, and added value to patient management decision making. For
0.060.8) when compared with carriers who chose not to genetic testing, particularly for moderate-penetrance genes,
clinical utility remains the fundamental issue.5 The EGAPP
undergo this procedure.1 Additionally, Markov modeling sug-
framework is key when evaluating the utility of genetic testing
gests that a 30-year old healthy BRCA1 mutation carrier
for hereditary cancer syndromes. Failure to meet some of
would gain 0.2 to 1.8 years in life expectancy with risk-
these criteria forms the basis for many concerns regarding the
reducing salpingo-oophorectomy and 0.6 to 2.1 years from
current clinical actionability of multigene panel testing.
risk-reducing mastectomies.2,3 Given these findings, genetic
testing for hereditary cancer syndromes has now become part
of standard practice. SINGLE/LIMITED GENE TESTING
As set forth in the Evaluation of Genomic Applications in For well over a century, it has been recognized that some
Practice and Prevention (known as EGAPP) initiative4 and further families harbor a hereditary predisposition to a variety of

From the Lombardi Comprehensive Cancer Center, MedStar Georgetown University Hospital, Washington, DC; Lombardi Comprehensive Cancer Center, Georgetown University,
Washington, DC.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Claudine Isaacs, MD, Lombardi Comprehensive Cancer Center, 3800 Reservoir Rd. NW, Washington, DC 20057; email: isaacsc@georgetown.edu.

2016 by American Society of Clinical Oncology.

e72 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


SINGLE GENE VERSUS MULTIGENE PANEL TESTING FOR HEREDITARY CANCER

malignancies. In 1913, Warthin described a kindred known with rare high-penetrance syndromes or a more moderate
as Family G, in which he noted an aggregation of endometrial penetrance were identified.
carcinoma along with gastric and colorectal cancer.6 This Based on these findings, the general paradigm of testing
family, among others, formed the basis of the initial evolved whereby the more common genes such as BRCA1
descriptions of hereditary nonpolyposis colorectal cancer and BRCA2 were tested first, and, if negative, sequential
syndrome, now more commonly known as Lynch syn- testing for additional gene(s) was performed if the patient
drome. Similarly, astute clinicians recognized other hered- met criteria for testing for other syndromes. This process
itary cancer syndromes such as Li-Fraumeni and Cowden had both advantages and disadvantages. In terms of ad-
syndromes and hereditary breast and ovarian cancer based vantages, the genes tested in this setting typically have
on the cancer phenotype of the family.7-12 By the mid-1990s, well-described cancer risks and often have established
linkage analyses and other studies resulted in the abil- management guidelines. Additionally, through the pretest
ity to pinpoint individual genes associated with some of counseling process, patients undergoing this testing have
these hereditary cancer syndromes.13,14 These included the had the opportunity to fully consider the benefits, risks, and
identification of BRCA1 and BRCA2 associated with hered- limitations of testing in their particular situation. In terms
itary breast and ovarian cancer; MLH1, MSH2, MSH6, PMS2, of disadvantages, such testing is less comprehensive than
and more recently EPCAM associated with Lynch syndrome; multigene testing, and, if performed, sequential testing is
FAP with familial adenomatous polyposis; and TP53 with quite time-consuming and costly.
Li-Fraumeni syndrome.
However, it became apparent that many families with
striking histories consistent with either a hereditary colo- MULTIGENE PANELS
rectal or breast/ovarian cancer syndrome are not found to There has been a dramatic shift in the genetic testing
carry a mutation in one of the mismatch repair genes as- landscape over the past several years in large part because of
sociated with Lynch syndrome or in BRCA1/2. For example, two major factors. The first is the development of next-
studies indicate that a mutation in a mismatch repair gene is generation sequencing, a high-throughput approach to DNA
found in approximately 40% to 80% of families that meet sequencing that allows for massively parallel sequencing of
the Amsterdam I criteria and only about 5% to 50% of multiple genes more efficiently and at a lower cost than the
families meeting the Amsterdam II criteria.15 Similarly, only traditional Sanger sequencing methods. The second is the
about 5% to 10% of unselected patients with breast cancer16 Supreme Court decision in 2013 for Association for Mol-
and 20% to 25% of patients with hereditary breast cancer17 ecular Pathology v. Myriad Genetics, which invalidated many
are found to carry a deleterious BRCA1 or BRCA2 mutation. patents restricting BRCA1/2 testing. Very shortly after the
Additionally, a number of other genes associated either ruling, many companies and some academic institutions
announced they would offer BRCA testing in addition to the
existing genes on their multigene panels.18,19 As a result of
these two factors, offering relatively rapid turnaround times
for multigene testing in a reasonably affordable manner
KEY POINTS became feasible.
The panels differ from company to company. They may
Advances in technology have resulted in the ability to be comprehensive, tumor-specific, and focus only on highly
test for multiple genes associated with a hereditary penetrant genes, or be customizable (Table 1). The price of
predisposition to cancer.
testing also has dropped significantly. It now can range from
Multigene panel testing can allow for a more
comprehensive and efficient approach to testing, but
$249 to $6,040, with most costing $1,500 to $6,040. The cost
many of the genes included in multigene panels have varies among laboratories and differs based on the number
not been fully characterized either in terms of their of genes included. In most cases, the cost of multigene panel
cancer risks or management strategies. In many cases, testing does not significantly differ from the cost of more
single/limited gene testing remains a very appropriate single/limited gene testing. Furthermore, the cost of testing
testing option. likely will continue to diminish over time. Adding to the
The decision to pursue single or limited gene testing is complexity of testing choices is that insurance companies
complex and referral to clinicians with expertise in have different policies and may cover some but not all
cancer genetics is critical. Testing must be carried out choices.
with pre- and post-test genetic counseling. A number of studies have evaluated the utility and impact
In the tumor molecular profiling setting, secondary
of multigene testing in a variety of settings. The key ques-
incidental germline mutations may be detected. ASCO
recommends that the possibility of identifying
tions that must be addressed revolve around the clinical
secondary incidental germline information and the utility or actionability of the findings from such testing,
clinical relevance, benefits, risks, and limitations of such namely (1) the numbers of patients who are found to have a
findings be discussed with all patients before they deleterious mutation in a gene for which cancer risks are
undergo tumor sequencing. known and management strategies exist, (2) patients who
are found to have a mutation with uncertain cancer risks

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e73


LYNCE AND ISAACS

TABLE 1. Examples of Genes Included on Some Next-Generation Sequencing Cancer Panels*


Number
Company Test of Genes Genes Included
20
Comprehensive Ambry Genetics CancerNext 32 APC, ATM, BARD1, BRCA1, BRCA2, BRIP1, BMPR1A, CDH1, CDK4,
Panels CDKN2A, CHEK2, EPCAM, GREM1, MLH1, MRE11A, MSH2, MSH6,
MUTYH, NBN, NF1, PALB2, PMS2, POLD1, POLE, PTEN, RAD50,
RAD51C, RAD51D, SMAD4, SMARCA4, STK11, TP53
GeneDx OncoGene Dx Compre- 32 APC, ATM, AXIN2, BARD1, BMPR1A, BRCA1, BRCA2, BRIP1, CDH1,
hensive Cancer Panel21 CDK4, CDKN2A, CHEK2, EPCAM, FANCC, MLH1, MSH2, MSH6,
MUTYH, NBN, PALB2, PMS2, POLD1, POLE, PTEN, RAD51C, RAD51D,
SCG5/GREM1, SMAD4, STK11, TP53, VHL, XRCC2
Myriad Genetics MyRisk22 25 BRCA1, BRCA2, MLH1, MSH2, MSH6, PMS2, EPCAM, APC, MUTYH,
CDKN2A, CDK4, TP53, PTEN, STK11, CDH1, BMPR1A, SMAD4, PALB2,
CHEK2, ATM, NBM, BARD1, BRIP1, RAD51C, RAD51D
Invitae Invitae Multi-Cancer 79 ALK, APC, ATM, AXIN2, BAP1, BARD1, BLM, BMPR1A, BRCA1, BRCA2,
Panel23 BRIP1, CASR,CDC73, CDH1, CDK4, CDKN1B, CDKN1C, CDKN2A,
CEBPA, CHEK2, DICER1, DIS3L2, EGFR, EPCAM, FH, FLCN, GATA2,
GPC3, GREM1, HOXB13, HRAS, KIT, MAX, MEN1, MET, MITF, MLH1,
MSH2, MSH6, MUTYH, NBN, NF1, NF2, PALB2,PDGFRA, PHOX2B,
PMS2, POLD1, POLE, PRKAR1A, PTCH1, PTEN, RAD50,RAD51C,
RAD51D, RB1, RECQL4, RET, RUNX1, SDHA, SDHAF2, SDHB, SDHC,
SDHD, SMAD4, SMARCA4, SMARCB1, SMARCE1, STK11, SUFU, TERC,
TERT, TMEM127, TP53, TSC1, TSC2, VHL, WRN, WT1
Breast/Ovarian Ambry Genetics BRCAplus24 6 BRCA1, BRCA2, CDH1, PALB2, PTEN, TP53
Panels
BreastNext25 17 ATM, BARD1, BRCA1, BRCA2, BRIP1, CDH1, CHEK2, MRE11A, MUTYH,
NBN, NF1, PALB2, PTEN, RAD50, RAD51C, RAD51D, TP53
OvaNext26 23 ATM, BARD1, BRCA1, BRCA2, BRIP1, CDH1, CHEK2, EPCAM, MLH1,
MRE11A, MSH2, MSH6, MUTYH, NBN, NF1, PALB2, PMS2, PTEN,
RAD50, RAD51C, RAD51D, SMARCA4, STK11, TP53
Invitae Breast and Gynecologic 14 ATM, BRCA1, BRCA2, CDH1, CHEK2, EPCAM, MLH1, MSH2, MSH6,
Cancers Guidelines PALB2, PMS2,PTEN, STK11, TP53
Based Panel27
Breast Cancer Guidelines 9 ATM, BRCA1, BRCA2, CDH1, CHEK2, PALB2, PTEN, STK11, TP53
Based Panel28
Color Color 19 ATM, BARD1, BRCA1, BRCA2, BRIP1, CDH1, CHEK2, EPCAM, MHL1,
Genomics29 MSH2, MSH6, NBM, PALB2, PMS2, PTEN, RAD51C, RAD51D, STK11,
TP53
GeneDx Breast Cancer High/Mod- 9 ATM, BRCA1, BRCA2, CDH1, CHEK2, PALB2, PTEN, STK11, TP53
erate Risk Panel21
Breast/Ovarian Cancer 21 ATM, BARD1, BRCA1, BRCA2, BRIP1, CDH1, CHEK2, EPCAM, FANCC,
Panel21 MLH1, MSH2, MSH6, NBN, PALB2, PMS2, PTEN, RAD51C, RAD51D,
STK11, TP53, XRCC2
Gastrointestinal Ambry Genetics ColoNext30 17 APC, BMPR1A, CDH1, CHEK2, EPCAM, GREM1, MLH1, MSH2, MSH6,
Panels MUTYH, PMS2, POLD1, POLE, PTEN, SMAD4, STK11, TP53
Invitae Colorectal Cancer Guide- 12 APC, BMPR1A, EPCAM, MLH1, MSH2, MSH6, MUTYH, PMS2, PTEN,
lines Based Panel31 SMAD4, STK11, TP53
Myriad Genetics COLARIS32 6 MLH1, MSH2, EPCAM, MSH6, PMS2, MUTYH
COLARIS AP33 APC, MUTYH
GeneDx Colorectal Cancer Panel21 19 APC, ATM, AXIN2, BMPR1A, CDH1, CHEK2, EPCAM, MLH1, MSH2,
MSH6, MUTYH, PMS2, POLD1, POLE, PTEN, SCG5/GREM1, SMAD4,
STK11, TP53
Lynch/Colorectal High Risk 7 APC, EPCAM, MLH1, MSH2, MSH6, MUTYH, PMS2
Panel21
Abbreviation: VUS, variants of uncertain significance.
*Current as of February 2, 2016.

and/or no evidence-based recommendations for manage- future because of the rapid accumulation of data from
ment, and (3) the rate detection of variants of uncertain multigene panel testing.
significance (VUS). As summarized in Tables 2 and 3, the rate Given the high rate of VUS and the detection of genes for
of VUS varies between 3.3% and 42%, and many patients which the cancer risks are not well-defined, several regis-
were reported to have two or more VUS. The VUS rate is still tries have been created to catalog and curate these vari-
high in some reports, but it is expected to fall in the near ants with the goal of advancing our knowledge about their

e74 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


SINGLE GENE VERSUS MULTIGENE PANEL TESTING FOR HEREDITARY CANCER

TABLE 2. Rate of Deleterious Mutations and VUS Reported in Multigene Testing Associated with Hereditary Breast
Cancer

BRCA Mutations Non-BRCA Mutations


Publication No. of Participants Panel Tested Detected (%) Detected (%) VUS Rate (%)
38
Kapoor et al 337 Ambry Genetics; different 3.6% 3.9%; gene mutations more often 3.3% BRCA1/2 and
panel types and genes detected: PALB2, CHEK2, ATM 13.4% non-BRCA1/2
tested
Slavin et al39 348 Not provided 3.4% 16.4% 42%
40
Tung et al 2,158 Cohort 1: Referred for 9.3% 4.2% 41.7%
commercial BRCA1/2
gene testing and
Cohort 2: Previously 3.7% 41.6%
tested negative for
BRCA1/2 mutations
Chong et al41 3,000 BRCAPlus: 6-gene panel 4.6% 1% 7.6%
(BRCA1, BRCA2, TP53,
CDH1, PTEN, SKT11)
Kurian et al42 198 (57 BRCA1/2, Invitae 11.4% 2.1% VUS/average per
141 BRCA1/2 neg) participant
LaDuca et al43 2,079 (874 had Ambry Genetics All previously underwent 7.4% gene mutations detected: 19.8%
breast panel) BRCA sequencing and CHEK2 (19), ATM (18), PALB2
BART testing and were (15), TP53 (4), PTEN (3), RAD50
negative (3), RAD51C (2), BRIP1 (1),
MRE11A (1), NBN (1)
Abbreviation: VUS, variants of uncertain significance.

clinical utility. The Prospective Registry of Multiplex Testing syndrome versus multigene panel testing. These include
(PROMPT)34 is a multi-institutional online registry that en- (1) the characteristics of the probands personal and family
courages patients to self-enter information about their history, (2) an individuals preferences and tolerance re-
genetic testing results and to complete questionnaires about garding the possibility of ambiguous results, (3) insurance-
their personal medical and family histories. Others involved related issues, and (4) the rapidity with which results are
in reclassifying variants include ENIGMA (Evidence-based needed. A publication in the New England Journal of Medicine
Network for the Interpretation of Germline Mutant Alleles) in 2015, authored by experts from the United States, United
Consortium35 and ClinVar, a peer-reviewed database funded Kingdom, the Netherlands, Germany, Australia, and Canada,
by the National Institutes of Health, which is a freely available thoughtfully reviewed the issues that must be addressed
archive of reports of relationships among medically important when considering multigene panels.45 Additionally, ASCO
variants and phenotypes.36 Professional societies such as the released a policy statement on genetic and genomic testing
American Medical Association also have adopted positions in for cancer susceptibility to reflect the impact of advances in
favor of data sharing.37 this field.5
Single/limited gene testing remains an excellent option
when the clinical features, such as the patients personal and
SINGLE/LIMITED GENE VERSUS MULTIGENE family history, are strongly indicative of a particular syn-
PANEL TESTING drome associated with a single or finite set of genes. This
Several factors guide the decision to pursue testing of a single approach allows for a focused and comprehensive pretest
gene or a finite set of genes associated with a particular evaluation in which individuals have an opportunity to more

TABLE 3. Rate of Deleterious Mutations and VUS Reported in Multigene Testing Associated with Colorectal Cancer

LS Genes (MLH1, MSH2, MSH6,


PMS2, EPCAM) Mutations De- Non-LS Genes Mutations
Publication No. Patients Panel Tested tected (%) Detected (%) VUS Rate (%)
LaDuca et al43 557 Ambry Genetics 4.5%: MSH (7), MLH1 (7), 4.7% (26): APC (6), CHEK2 (6), MUTYH 15.1% (84)
PMS2 (6), MSH (5) biallelic (6), SMAD4 (4), PTEN (3),
CDH1 (1), STK11 (1), TP53 (1)
Yurgelun et al44 1,260 Myriad MyRisk Hereditary 9.5% 3.8% (48): BRCA1/2 (15), APC biallelic 44%
Cancer MUTYH, PTEN, STK11, ATM,
BARD1, BRIP1, CHEK2, NBN, PALB2,
and RAD51C
Abbreviations: LS, Lynch syndrome; VUS, variants of uncertain significance.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e75


LYNCE AND ISAACS

fully consider the impact of testing for a particular gene or critical to guide the choice of which test to order and from
set of genes. Additionally, such testing minimizes the like- which laboratory. These issues further underscore the im-
lihood of detecting a VUS or a deleterious mutation in a gene portance of ensuring that patients undergo pre- and post-test
with limited clinical information. genetic counseling by well-trained professionals, as endorsed
Multigene panel testing is an appropriate option when by ASCO and the National Comprehensive Cancer Network.46
the family phenotype is not suggestive of a single specific
mutation and one or more hereditary cancer syndromes
are in the differential. Additionally, panel testing is often GERMLINE FINDINGS ON MOLECULAR
considered if more focused initial testing is negative (e.g., PROFILING OF TUMORS
BRCA1/2 testing followed by multigene breast/ovarian An important challenge when performing vast-scale se-
panel). Multigene panel testing has a number of advan- quencing is the potential for detecting incidental findings.
tages and potential disadvantages (Table 4). The advan- Incidental findings are defined as unexpected positive
tages include gains in efficiency both in terms of cost and findings. In this context, they refer to the detection of
time. Such testing also would result in a more compre- deleterious or likely deleterious alterations in genes that
hensive assessment of the genes that could account for have clinical significance and are unrelated to the in-
the cancer phenotype in the family. Finally, pragmatically, dication for obtaining the sequencing test. Typically, these
in this era of multigene panel testing, it is unclear if an are germline mutations. As such, the only way to truly
individual could obtain insurance coverage for repeat determine if an identified sequence variant is somatic or
testing if the initial, more limited testing results were inherited is to simultaneously analyze tumor and normal
negative. In terms of disadvantages, as described pre- DNA. This analysis allows for a determination on which
viously, multigene panel testing has a higher rate of de- variants are unique to the cancer (i.e., somatic) and which
tection of VUS. Individuals undergoing testing must be are germline. Determining whether, which, and how in-
fully informed of this possibility before testing and coun- cidental findings are returned to the patient is becoming
seled on the interpretation of such a result. Furthermore, increasingly important and controversial. The American
it is important that an individual undergoing panel test- College of Medical Genetics and Genomics published a
ing understands it is possible that a high-penetrance mutation policy statement on clinical sequencing that included
in an uncommon or rare gene may be identified, even in the exome and genome sequencing.47 The policy state-
absence of a classic presentation of the associated syndrome. ment recommended that constitutional mutations from a
Consequently, aggressive interventions may be recommended, panel of 56 disease-associated genes be reported to the
such as consideration of prophylactic gastrectomy if a CDH1 ordering clinician, regardless of the indication for which
mutation was found, even in the absence of gastric cancer in the clinical sequencing was ordered (Table 5). These
the family. At this moment, it is unclear if the cancer risks for genes were chosen because they result in disorders for
patients identified through panel testing without features of which preventive strategies and/or treatments are
the associated syndrome are the same as quoted in the lit- available. About half are associated with syndromes that
erature because of ascertainment bias. Moreover, laboratories increase the risk of cancer; the others are primarily as-
have varying methods by which they assure the analytic and sociated with various cardiac diseases. The American
clinical validity and the clinical utility of the variants they re- College of Medical Genetics and Genomics also states
port. Expertise in this area is required to ensure accurate in- that the ordering clinician/team is responsible for pro-
terpretation of the clinical significance of the findings reported. viding the patient with comprehensive pre- and post-test
Given the panoply of testing options, this expertise is also counseling.

TABLE 4. Pros and Cons of Single/Limited Gene Testing and Multigene Panels

Single/Limited Gene Testing Multigene Panels


Advantages Phenotype-directed testing More cost effective (less expensive per gene cost)
Cancer risks and management More time efficient
options often more established Decrease in testing fatigue for patients and providers
Lower likelihood of detecting VUS Efficient use of single specimen
More rapid turnaround time Higher mutation detection rate, genes individually rare but
collectively significant
Disadvantages Higher risk of loss to follow-up during sequential testing multiple Increased prevalence of VUS
single genes (test fatigue) Cancer risks and management options often not well-defined,
Less comprehensive particularly for some moderate- and low-penetrance genes
Unexpected findings such as off-phenotypic-target gene
mutation
Longer turnaround time
Panels may include genes that patients dont wish to test for
Abbreviation: VUS, variants of uncertain significance.

e76 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


SINGLE GENE VERSUS MULTIGENE PANEL TESTING FOR HEREDITARY CANCER

TABLE 5. Conditions and Genes Recommended by the American College of Medical Genetics and Genomics for
Return of Incidental Findings in Clinical Sequencing47

Phenotype Gene
Hereditary breast and ovarian cancer BRCA1, BRCA2
Li-Fraumeni syndrome TP53
Peutz-Jeghers syndrome STK11
Lynch syndrome MLH1, MSH2, MSH6, PMS2
Familial adenomatous polyposis APC
MYH-associated polyposis; adenomas; multiple colorectal cancers; familial amyloid MUTYH
polyneuropathy type 2; colorectal adenomatous polyposis, autosomal recessive,
with pilomatricomas
Von Hippel-Lindau syndrome VHL
Multiple endocrine neoplasia type 1 MEN1
Multiple endocrine neoplasia type 2 RET
Familial medullary thyroid cancer RET
PTEN hamartoma tumor syndrome PTEN
Retinoblastoma RB1
Hereditary paraganglioma-pheochromocytoma syndrome SDHD, SDHAF2, SDHC, SDHB
Tuberous sclerosis complex TSC1, TSC2
WT1-related Wilm syndrome WT1
Neurofibromatosis type 2 NF2
Ehlers-Danlos syndrome (vascular type) COL3A1
Marfan syndrome, Loeys-Dietz syndrome, familial thoracic aortic aneurysms and FBN1, TGFBR1, TGFBR2, SMAD3, ACTA2, MYLK, MYH11
dissections
Hypertrophic cardiomyopathy, dilated cardiomyopathy MYBPC3, MYH7, TNNT2, TNNI3, TPM1, MYL3, ACTC1, PRKAG2,
GLA, MYL2, LMNA
Catecholaminergic polymorphic ventricular tachycardia RYR2
Arrhythmogenic right-ventricular cardiomyopathy PKP2, DSP, DSC2, TMEM43, DSG2
Romano-Ward Long QT syndrome types 1, 2, and 3; Brugada syndrome KCNQ1, KCNH2, SCN5A
Familial hypercholesterolemia LDLR, APOB, PCSK9
Malignant hyperthermia susceptibility RYR1, CACNA1S

Several challenges are inherent in this process. They range Although multigene panel testing provides a more com-
from analytic issues (e.g., determining if the variant is prehensive and efficient approach to testing an individual
germline and, if so, if it is deleterious/likely deleterious, for a hereditary susceptibility to cancer, the informa-
a VUS, or a benign polymorphism) to practical issues related tion obtained can be challenging to interpret. Further-
to obtaining informed consent and delivering traditional more, many of the genes included in multigene panels
pre- and post-testing genetic counseling. ASCO recently have not been fully characterized either in terms of
published a policy statement on genetic and genomic testing their cancer risks or management strategies. In many
for cancer susceptibility that includes germline implications of cases, single/limited gene testing remains a very ap-
somatic mutation profiling.5 The policy statement recognizes propriate testing option. Presently, we live in an era in
that standard pre- and post-test counseling may not be which our technical capabilities have outstripped our
feasible in this setting for all patients. It recommends that the medical knowledge. A strong and continuous part-
possibility of identifying secondary incidental germline in- nership among clinicians, individuals with genetics
formation as well as the clinical relevance, benefits, risks, and expertise, and laboratory geneticists is critical to bridge
limitations of such incidental findings be discussed with all this gap.
patients before they undergo tumor sequencing. ASCO also As to the detection of incidental findings on tumor se-
endorses that providers should honor patients decisions if quencing, more research is clearly necessary to better clarify
they elect not to receive information about such incidental how to approach this complex area. Until such time, as
findings. stated by ASCO, it is critical that individuals undergoing
tumor sequencing be fully apprised of the possibility,
CONCLUSION benefits, risks, and limitations that such testing could
Next-generation sequencing has introduced substantial uncover unanticipated mutations in cancer susceptibility
complexity and promise in the field of cancer risk assessment. genes.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e77


LYNCE AND ISAACS

References

1. Domchek SM, Friebel TM, Singer CF, et al. Association of risk-reducing 26. Ambry Genetics. OvaNext. http://www.ambrygen.com/tests/ovanext.
surgery in BRCA1 or BRCA2 mutation carriers with cancer risk and Accessed February 2, 2016.
mortality. JAMA. 2010;304:967-975. 27. Invitae. Breast and Gyn Cancers Guidelines-Based Panel. https://www.
2. Schrag D, Kuntz KM, Garber JE, et al. Benefit of prophylactic mastectomy for invitae.com/en/physician/tests/01204/. Accessed February 2 ,2016.
women with BRCA1 or BRCA2 mutations. JAMA. 2000;283:3070-3072. 28. Invitae Breast Cancers Guidelines-Based Panel. https://www.invitae.
3. Grann VR, Jacobson JS, Thomason D, et al. Effect of prevention strategies on com/en/physician/tests/01206/. Accessed February 2, 2016.
survival and quality-adjusted survival of women with BRCA1/2 mutations: 29. Color Genomics. Get Color. https://getcolor.com/learn/the-science#gene-panel.
an updated decision analysis. J Clin Oncol. 2002;20:2520-2529. Accessed February 2, 2016.
4. Teutsch SM, Bradley LA, Palomaki GE, et al; EGAPP Working Group. The 30. Ambry Genetics. ColoNext. http://www.ambrygen.com/tests/colonext.
Evaluation of Genomic Applications in Practice and Prevention (EGAPP) Accessed February 2, 2016.
initiative: methods of the EGAPP working group. Genet Med. 2009;11:3-14. 31. Invitae. Colorectal Cancer Guidelines Based Panel. https://www.invitae.
5. Robson ME, Bradbury AR, Arun B, et al. American Society of Clinical com/en/physician/tests/01252/. Accessed February 2, 2016.
Oncology policy statement update: genetic and genomic testing for 32. Myriad Pro. COLARIS FAQ. https://www.myriadpro.com/hereditary-cancer-
cancer susceptibility. J Clin Oncol. 2015;33:3660-3667. testing/hereditary-colon-and-gynecologic-cancers/lynch-syndrome-hnpcc/
6. Warthin AS. Heredity with reference to carcinoma. Arch Intern Med colaris-faq/. Accessed February 2, 2016.
(Chic). 1913;12:546-555. 33. Myriad Pro. COLARIS AP FAQ. https://www.myriadpro.com/hereditary-
7. Li FP, Fraumeni JF Jr. Soft-tissue sarcomas, breast cancer, and other cancer-testing/hereditary-colon-and-gynecologic-cancers/polyposis-
neoplasms. A familial syndrome? Ann Intern Med. 1969;71:747-752. syndromes-fap-afap-map/colaris-ap-faq/. Accessed February 2, 2016.
8. Li FP, Fraumeni JF Jr. Rhabdomyosarcoma in children: epidemiologic 34. PROMPT. Prospective Registry of Multiplex Testing. http://www.
study and identification of a familial cancer syndrome. J Natl Cancer promptstudy.org. Accessed February 22, 2016.
Inst. 1969;43:1365-1373. 35. ENIGMA. Evidence-based Network for the Interpretation of Germline
9. Lloyd KM II, Dennis M. Cowdens disease. A possible new symptom Mutant Alleles. http://www.enigmaconsortium.org. Accessed February
complex with multiple system involvement. Ann Intern Med. 1963;58: 22, 2016.
136-142. 36. Landrum MJ, Lee JM, Riley GR, et al. ClinVar: public archive of re-
10. Le Dran H. Memoire avec un precis de plusieurs observations sur le lationships among sequence variation and human phenotype. Nucleic
cancer. Mem Acad Chir (Paris). 1757;3:1-54. Acids Res. 2014;42:D980-D985.
11. Smithers DW. Family histories of 459 patients with cancer of the breast. 37. American Medical Association. Support of Public Access to Genetic
Br J Cancer. 1948;2:163-167. Data. http://www.ama-assn.org/ama/pub/news/news/2013/2013-06-
12. Broca PP. Traite des tumeurs. Paris: P. Asselin; 1866. 18-new-ama-policies-annual-meeting.page. Accessed February 2, 2016.
13. Hall JM, Lee MK, Newman B, et al. Linkage of early-onset familial breast 38. Kapoor NS, Curcio LD, Blakemore CA, et al. Multigene panel testing detects
cancer to chromosome 17q21. Science. 1990;250:1684-1689. equal rates of pathogenic BRCA1/2 mutations and has a higher diagnostic
14. Nakamura Y, Lathrop M, Leppert M, et al. Localization of the genetic yield compared to limited BRCA1/2 analysis alone in patients at risk for
defect in familial adenomatous polyposis within a small region of chro- hereditary breast cancer. Ann Surg Oncol. 2015;22:3282-3288.
mosome 5. Am J Hum Genet. 1988;43:638-644. 39. Slavin TP, Niell-Swiller M, Solomon I, et al. Clinical application of multigene
15. Lynch HT, de la Chapelle A. Hereditary colorectal cancer. N Engl J Med. panels: challenges of next-generation counseling and cancer risk man-
2003;348:919-932. agement. Front Oncol. 2015;5:208.
16. Antoniou A, Pharoah PDP, Narod S, et al. Average risks of breast and 40. Tung N, Battelli C, Allen B, et al. Frequency of mutations in individuals
ovarian cancer associated with BRCA1 or BRCA2 mutations detected in with breast cancer referred for BRCA1 and BRCA2 testing using next-
case series unselected for family history: a combined analysis of 22 generation sequencing with a 25-gene panel. Cancer. 2015;121:25-33.
studies. Am J Hum Genet. 2003;72:1117-1130. 41. Chong HK, Wang T, Lu HM, et al. The validation and clinical imple-
17. Nathanson KL, Wooster R, Weber BL. Breast cancer genetics: what we mentation of BRCAplus: a comprehensive high-risk breast cancer di-
know and what we need. Nat Med. 2001;7:552-556. agnostic assay. PLoS One. 2014;9:e97408.
18. Azvolinsky A. Supreme Court ruling broadens BRCA testing options. 42. Kurian AW, Hare EE, Mills MA, et al. Clinical evaluation of a multiple-
J Natl Cancer Inst. 2013;105:1671-1672. gene sequencing panel for hereditary cancer risk assessment. J Clin
19. Cook-Deegan R, Niehaus A. After Myriad: genetic testing in the wake of Oncol. 2014;32:2001-2009.
recent Supreme Court decisions about gene panels. Curr Genet Med 43. LaDuca H, Stuenkel AJ, Dolinsky JS, et al. Utilization of multigene panels
Rep. 2014;2:223-241. in hereditary cancer predisposition testing: analysis of more than 2,000
20. Ambry Genetics. CancerNext. http://www.ambrygen.com/tests/cancernext. patients. Genet Med. 2014;16:830-837.
Accessed February 2, 2016. 44. Yurgelun MB, Allen B, Kaldate R, et al. Multigene panel testing in
21. Ambry Genetics. GeneDx Oncology Genetics. http://www.genedx.com/ patients suspected to have Lynch syndrome. J Clin Oncol. 2014;32:5s
oncology-genetics/. Accessed February 2, 2016. (suppl; abstr 1509).
22. Myriad Pro. MyRisk FAQ. https://www.myriadpro.com/myrisk/frequently- 45. Easton DF, Pharoah PDP, Antoniou AC, et al. Gene-panel sequencing and
asked-questions/. Accessed February 2, 2016. the prediction of breast-cancer risk. N Engl J Med. 2015;372:2243-2257.
23. Invitae Multi-Cancer Panel. https://www.invitae.com/en/physician/tests/ 46. Hall MJ, Forman AD, Pilarski R, et al. Gene panel testing for inherited
01101/. Accessed February 2, 2016. cancer risk. J Natl Compr Canc Netw. 2014;12:1339-1346.
24. Ambry Genetics. BRCAPlus. http://www.ambrygen.com/tests/brcaplus. 47. Green RC, Berg JS, Grody WW, et al; American College of Medical
Accessed February 2, 2016. Genetics and Genomics. ACMG recommendations for reporting of in-
25. Ambry Genetics. BRCANext. http://www.ambrygen.com/tests/breastnext. cidental findings in clinical exome and genome sequencing. Genet Med.
Accessed February 2, 2016. 2013;15:565-574.

e78 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


CANCER PREVENTION, HEREDITARY GENETICS,
AND EPIDEMIOLOGY

Evolving Recommendations on
Prostate Cancer Screening

CHAIR
Otis W. Brawley, MD, FASCO
American Cancer Society
Atlanta, GA

SPEAKERS
Henrik Gronberg, MD, PhD
Karolinska Institutet
Stockholm, Sweden

Ian M. Thompson Jr., MD


The University of Texas Health Science Center at San Antonio
San Antonio, TX

BRAWLEY, THOMPSON, AND GRONBERG

Evolving Recommendations on Prostate Cancer Screening


Otis W. Brawley, MD, FASCO, Ian M. Thompson Jr., MD, and Henrik Gronberg, MD, PhD

OVERVIEW

Results of a number of studies demonstrate that the serum prostate-specific antigen (PSA) in and of itself is an inadequate
screening test. Today, one of the most pressing questions in prostate cancer medicine is how can screening be honed to
identify those who have life-threatening disease and need aggressive treatment. A number of efforts are underway. One
such effort is the assessment of men in the landmark Prostate Cancer Prevention Trial that has led to a prostate cancer
risk calculator (PCPTRC), which is available online. PCPTRC version 2.0 predicts the probability of the diagnosis of no
cancer, low-grade cancer, or high-grade cancer when variables such as PSA, age, race, family history, and physical findings
are input. Modern biomarker development promises to provide tests with fewer false positives and improved ability to
find high-grade cancers. Stockholm III (STHLM3) is a prospective, population-based, paired, screen-positive, prostate
cancer diagnostic study assessing a combination of plasma protein biomarkers along with age, family history, previous
biopsy, and prostate examination for prediction of prostate cancer. Multiparametric MRI incorporates anatomic and
functional imaging to better characterize and predict future behavior of tumors within the prostate. After diagnosis of
cancer, several genomic tests promise to better distinguish the cancers that need treatment versus those that need
observation. Although the new technologies are promising, there is an urgent need for evaluation of these new tests in
high-quality, large population-based studies. Until these technologies are proven, most professional organizations have
evolved to a recommendation of informed or shared decision making in which there is a discussion between the doctor
and patient.

T he U.S. Food and Drug Administration (FDA) first ap-


proved sale and use of a serum PSA test in 1986. It was
approved for assessing response to prostate cancer therapy
They were studies of hormones and radiation therapy for
locally advanced disease.4,5 A prospective randomized study
demonstrating a positive effect for radical prostatectomy
and monitoring for relapse. Large-scale prostate cancer was first published in 2002.6,7
screening began in the United States in 1991 after publica- Two large relatively well-designed randomized trials
tions showing serum PSA useful in finding localized prostate assessing prostate cancer screening were published in
cancer. Use increased dramatically after the American Cancer 2009.8,9 Although relatively well designed, both have some
Society recommended it in 1992.1 The FDA approved it as a flaws. One suggests screening is ineffective, and one sug-
diagnostic test in 1994. gests a regimented screening program does reduce risk of
In the United States, use of the PSA test was rampant prostate cancer death. Both studies documented the harms
throughout the 1990s, despite the lack of conclusive data to of prostate cancer screening, especially overdiagnosis and
show that screening and aggressive treatment of localized resultant overtreatment.
prostate cancer saved lives. Screening spread outside of the The fact that there is considerable overdiagnosis associ-
physician-patient relationship. There was mass screening at ated with prostate cancer screening was confirmed in as-
shopping malls, community centers, and even on the floor of sessment of the placebo arm of the large multi-institutional
national political conventions. The effect was that prostate Prostate Cancer Prevention Trial (PCPT).10 This trial of men at
cancer incidence rates soared, and a large number of men average risk of prostate cancer demonstrated that annual
were treated for localized prostate cancer with radical pros- screening could diagnose prostate cancer in upward of 14%
tatectomy, radiation therapy, or cryotherapy.2 Screening has of men in their fifties, sixties, and seventies. Men underwent
not been as popular in Europe, and prostate cancer incidence biopsy for diagnosis when their PSA was 4 ng/mL or greater
rates have not risen as much.3 or when their PSA changed by 1 ng/mL in 1 years time.
It is of note that the first definitive studies to show that Planned biopsies for men who screened normal every year
localized therapy reduces mortality were published in 1997. for 7 years (median age 69) found 15% of these men had

From the American Cancer Society, Emory University, Atlanta, GA; The University of Texas Health Science Center at San Antonio, San Antonio, TX; Karolinska Institute, Stockholm, Sweden.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Otis W. Brawley, MD, American Cancer Society, 250 Williams St., Ste. 600, Atlanta, GA 30303-1002; email: otis.brawley@cancer.org.

2016 by American Society of Clinical Oncology.

e80 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


EVOLVING RECOMMENDATIONS ON PROSTATE CANCER SCREENING

prostate cancer. This demonstrates that PSA screening missed USPSTF recommended against PSA-based prostate cancer
as much cancer as it found. screening for men age 75 and older.17 USPSTF recom-
Radical prostatectomy was compared with conservative mended against screening for all men in 2012.18 During this
therapy in the Prostate Intervention versus Observation Trial period, other organizations moved toward informed de-
(PIVOT).11 Mortality reductions were only found among cision making in which the decision to screen or not to screen
patients treated for intermediate- and high-risk disease. This is made within the physician-patient relationship, with the
again demonstrates that a considerable portion of men with patient taking into account the potential harms and po-
screened diagnosed prostate cancer have disease that does tential benefits (informed decision making) or the physician
not need treatment. and patient taking into account potential harms and po-
By 2010, the data became very clear that PSA testing iden- tential benefits (shared decision making).19
tifies a lot of cancer, and a lot of that cancer is of no risk to According to the 2008 National Health Interview Survey,
the patients lives. Today some even go so far as to ask 40.6% of American men age 50 and older reported having
whether low-grade disease (Gleason 6) should be consid- PSA screening within the previous year. By 2013, it was
ered cancer.12 Gleason score 6 and lower disease constitutes 20.8%. From 2010 to 2013 the prostate cancer incidence
about 50% of screen-detected prostate cancer in the United declined by 18% in the United States. Longer follow-up is
States. needed to see whether these decreases are associated with
It took a number of years for the screening activity to trends in mortality.
subside. It is estimated that more than a million American
men were diagnosed and received treatment because of PSA
screening.13 Although a significant proportion of screen- NEXT-GENERATION PROSTATE CANCER
diagnosed prostate cancers are of no clinical significance, SCREENING AND TREATMENT
prostate cancer is a significant cause of death. The historical approach to prostate cancer screening was
The U.S. prostate cancer mortality rate has declined by based in part on the historically poor outcomes of treat-
53% since 1991.14 Although some of this decline may be due ment of metastatic prostate cancer and the realization that
to screening and aggressive treatment, some is certainly not PSA-detected tumors were often cured with radiation or
due to screening and aggressive treatment. The U.S. death surgery. As noted in Fig. 1, all patients over a certain age
rate began declining the same year that widespread PSA (often age 50) were recommended to undergo annual
screening was introduced. One would expect screening to screening. If a PSA was found to be greater than 4.0 ng/mL,
reduce mortality 8 or more years after introduction. There a biopsy was recommended. If biopsy found cancer,
have been declines in prostate cancer mortality in more than treatment with radiation or surgery was generally rec-
two dozen countries. PSA screening is uncommon in most of ommended. This approach led to a remarkable spike
these countries.3 There is also speculation that some of the in prostate cancer detection, treatment, and the con-
decline is an artifact.15 The decline in mortality also corre- duct of the PLCO and ERSPC studies.8,20 Recognizing the
sponded with changes in coding causes of death and the use questionable impact of such an approach when com-
of PSA for assessing progression of prostate cancer. In re- paring the potential benefit and the known risks of
ality, the U.S. decline may be due to all of the above, treatment, the USPSTF recommended against PSA test-
including a screening benefit.15,16 ing in 2012.20
The U.S. Preventive Services Task Force (USPSTF) is a Twenty years of experience with this initial approach to
health advisory group for the U.S. government. In 2008, screening began to change with several sets of observations.
First, it was recognized that many tumors detected via PSA
screening were small and low-grade; the possible extent of
overdetection was illustrated in the end-of-study biopsies of
KEY POINTS the PCPT in which all men with a PSA , 4.0 ng/mL were
recommended to undergo biopsy. In this group of men, 15%
Prostate cancer screening has clear documented harms were found to have cancer, a stunning risk of cancer when it
and some possible benefits. is recognized that the lifetime risk of cancer death in all men
Men with low-grade or Gleason 6 disease generally have is between 2% and 3%.21 Concurrent with this observation,
long-term survival with observation. the first reports of the outcomes of active surveillance for
The benefit-risk ratio of prostate cancer screening low-risk prostate cancer detected through PSA testing began
improves if screening finds high-grade disease. to appear.9 With the maturation of these series, it became
The Prostate Cancer Prevention Trial risk calculator is
clear that low-grade (Gleason 3 + 3) and low-volume
useful in predicting whether a man of a specific PSA has
no cancer, low-grade cancer, or high-grade cancer and
intermediate-grade (Gleason 3 + 4) prostate cancers will
reduces the number of unnecessary prostate biopsies. achieve high prostate cancerspecific survival at periods up
The Stockholm III model is a promising predictor of low- to 15 years.22 In a cohort of 993 men with such tumors, of
grade versus high-grade disease that may further whom 13% had Gleason 3 + 4 tumors, combined cause-
decrease the number of unnecessary prostate biopsies. specific survival at 10 years was 98.1% and at 15 years was
94.3%.23

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e81



BRAWLEY, THOMPSON, AND GRONBERG

FIGURE 1. Early Approach to Prostate Cancer low-grade cancer, the benefit-risk ratio would more likely
Screening and Treatment argue against a biopsy. The first tool developed for this
was the PCPTRC (www.prostate-cancer-risk-calculator.
com).8
Two clinical scenarios illustrate how such a tool can be
used in next-generation screening.24
Case 1: a healthy 55-year-old white man who has a
normal examination. His current PSA is 3.0 (previous
year was 1.5). He has no family history of prostate
cancer and no prior biopsy.
Case 2: a 72-year-old black man who runs 5K races each
weekend, winning his age group times (i.e., in good
health). His father had prostate cancer, and on ex-
amination, he has a prostate nodule. He has heard
that PSA doesnt matter, but, because of his family
history, asked his physician to have one drawn. It is
3.0 ng/mL.
The risk calculator displays outcomes as green smiley faces
(no cancer), yellow uncertain faces (low-grade cancer), and
red frowny faces (high-grade cancer). As can be seen in these
Abbreviation: PSA, prostate-specific antigen. two examples, whereas the patient in case 1 (Fig. 2A) has a
3% risk of a high-grade cancer (potential benefit), he also
has a 15% risk of a low-grade cancer (unlikely benefit, likely
By studying the biopsy outcomes of the European Ran- harm) and 82% likelihood of a negative biopsy (unnecessary
domized Study of Prostate Cancer Screening and the PCPT, with risk of pain, bleeding, and infection). In contrast, the
it also has become apparent that using a single biomarker patient in case 2 (Fig. 2B), with precisely the same PSA
(i.e., PSA) to determine a mans risk of prostate cancer was value, has an eightfold greater risk of a high-grade cancer
naive; other measures of risk had a profound impact on (24%). In clinical practice, when provided these data with the
prostate cancer risk.8 Also phase III randomized clinical accompanying information regarding what happens to you if
trials have demonstrated very little (if any) benefit of you have any of these three outcomes, most patients simi-
treatment of low-grade tumors but mortality benefit for lar to that in case 1 opt to not undergo biopsy, whereas
high-grade tumors, the clinical community recognized most patients similar to that in case 2 opt for a biopsy. It is
that screening should seek to identify the man with high- also important to note that, again in clinical practice, two
grade cancer in whom a biopsy may have net potential patients with precisely the same risks will often reach
benefit from detection (and, presumably, treatment). On two different decisions, on the basis of their own personal
the other hand, if biopsy would preferentially detect a preferences and values that they place on the different

FIGURE 2. Prostate Cancer Prevention Trial Risk Calculator Patient Displays for (A) Case 1 and (B) Case 2

e82 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


EVOLVING RECOMMENDATIONS ON PROSTATE CANCER SCREENING

outcomes of each possible finding. As can be seen at the FIGURE 3. Next-Generation Approach to Prostate
PCPTRC website, other biomarkers (e.g., percent free PSA) or Cancer Screening and Treatment
biomeasures (e.g., body mass index) can be incorporated to
better estimate risk.
It has also been recognized that annual PSA testing is
unnecessary for many men. In most population-based studies,
the median PSA is about 1.0 ng/mL. We have found, in a large
community-dwelling cohort, if a mans PSA is 1.0 ng/mL or
lower, his risk of a prostate cancer diagnosis within 10 years
is 3.4% and 90% of the cancers are low-grade. Thus, con-
ceivably, half of the general population could have their PSA
performed concurrently with their colonoscopy, eliminating
an enormous burden on society.
Finally, information is now available to better help patients
decide on treatment options. As up to two-thirds of patients
will qualify for active surveillance, it is important that men
with low-risk cancers are aware of this option.18 Multiple
series now attest to the safety of this approach and the Abbreviations: HG, high grade; LG, low grade; PSA, prostate-specific antigen.

low risk of prostate cancer mortality. Studies are ongoing


to examine how to reduce the current burden of surveil- unscreened population from eight countries with different
lance. By comparison, phase III clinical trials have been screening and treatment strategies. After 13 years of follow-
completed that demonstrate the importance of multi- up, a significant 21% reduction (rate ratio 0.79; 95% CI,
modal therapy (e.g., androgen deprivation plus radiation 0.690.91) in death from prostate cancer is found in a
or adjuvant radiotherapy after prostatectomy) for high- predefined subgroup of men age 5569. After adjustment
risk tumors to reduce cancer recurrence and to reduce for nonparticipation, the rate ratio of prostate cancer
mortality.18,25 To help patients with decision making, con- mortality among men screened was 0.73 (95% CI, 0.610.88).
siderable data are also available regarding the impact of After 13 years of follow-up, there was no evidence of a
these treatments on genitourinary, sexual, and gastro- mortality benefit for organized annual PSA screening in
intestinal functions. the PLCO trial compared with opportunistic screening
It is unequivocal that using the current body of knowl- used in normal care. Men randomly assigned to the annual
edge regarding whom to screen, how frequently to screen screening group had a higher incidence of prostate cancer
them, what their risk is for different biopsy outcomes, in- (relative rate [RR] 1.12; 95% CI, 1.071.17) but no reduction
corporating active surveillance, and, as appropriate, multi- in prostate cancer mortality (RR 1.09; 95% CI, 0.871.36).
modal therapies for high-risk cancers, that the harms of Differences in follow-up protocols and screening intervals
screening and treatment can be dramatically reduced. It exist between the two trials. The major differences can
remains to be determined whether such an intelligent mostly be explained by the high screening rates (52% after
approach, summarized only briefly in Fig. 3, to screening 6 years) in the control arm and low compliance to biopsies
and treatment reduces prostate cancer mortality and in the PLCO trial.
morbidity. An interesting observation from cancer registries shows
that in a majority of countries in northern Europe and North
America, the age-adjusted mortality rates for prostate
A NEW VIEW ON PROSTATE CANCER SCREENING cancer have been decreasing substantially in the last 20
Prostate cancer mortality and morbidity is substantial in years. However, this trend is not global, as several countries
many parts of the world, and it is the most common cancer in Asia and Africa record increasing mortality during the
among males in most parts of Europe, North and South same period (Fig. 4). Lithuania is an outlier with a very rapid
America, Australia, and sub-Saharan Africa.26 The over- increase in mortality in the last 20 years.26 The drop in
arching goal of prostate cancer screening is to decrease the mortality in some countries can be explained by PSA testing,
morbidity and suffering from advanced prostate cancer better treatment, or changes in how prostate cancer deaths
without causing harm to men who do not require treatment are recorded. On the basis of SEER data modeling, 50% of the
for low-risk prostate cancer. observed decrease is estimated due to earlier detection with
The first question to ask is, is there evidence that PSA PSA, and 50% can be accounted to better treatment.28 More
screening decreases prostate cancer mortality? The highest detailed analysis from the Swedish Cancer Registries during
scientific weight is carried by randomized trials, and two 1990 to 2009 shows a lower prostate cancer mortality in
large randomized PSA screening studies have been pub- counties with high PSA testing as compared with less-intense
lished, ERSPC (Europe) and PLCO (United States), un- opportunistic PSA testing areas.29 Data from cancer regis-
fortunately with contradictory results.21,27 The European tries also show that the incidence of advanced and meta-
ERSPC trial examined the role of PSA screening in a largely static prostate cancer is going down.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e83



BRAWLEY, THOMPSON, AND GRONBERG

FIGURE 4. Trends in Prostate Cancer Mortality Rates So what is the way forward? Continuing to argue over the
in 10 Selected Countries (Age Adjusted to World use of the PSA test would result in polarized discussion
Standard) rather than promote development of new ideas. We suggest
an alternative route forward, getting away from the PSA test
into a more modern diagnostic pipeline using the rapidly
advancing technology in biomarkers, imaging, and geno-
mics. The goal of such a new diagnostic pipeline is to sub-
stantially reduce the harm of current PSA testing while
maintaining or increasing the sensitivity for diagnosing high-
risk cancers.

Blood-Based Biomarkers and Risk Calculators


The currently used biomarker PSA has low specificity
(only 20%30%), which leads to many men without can-
cer undergoing prostate biopsies. Biopsies are unpleas-
ant, and 2%5% of men will get septicemia after the
biopsies.
Use of PCPTRC has been previously discussed. These risk
calculators are important tools, but unfortunately not used
as much as they should be.
With the inefficient use of PSA in Stockholm,33 we initiated
In a recent review of early detection of prostate cancer, 16 of the STHLM3 study in 2011 with the goal of developing a
26 (62%) key opinion leaders agree that PSA screening does better prostate cancer test than PSA (Fig. 5). This new pros-
reduce death from prostate cancer; others thought that the tate cancer test should at least have the same sensitivity to
present evidence is not sufficiently conclusive.30 The consensus identify men with high-risk prostate cancer but with much
was that the magnitude of the effect was uncertain and that better specificity. This improved specificity should lead
substantial overdiagnosis and overtreatment exists, which to fewer unnecessary biopsies and fewer men diagnosed
needs to be reduced before recommendations for PSA with low-risk cancer. The STHLM3 study is a prospective,
screening in the general population can be made. population-based, paired, screen-positive, diagnostic study
With available data, guideline committees have come to of men without prostate cancer aged 50 to 69 randomly
very different conclusions, ranging from not recommending invited by date of birth from the Swedish Population Reg-
PSA testing (USPTF) to recommending population-based, ister. The predefined STHLM3 model included a combination
organized PSA testing (Lithuania).31 Most professional or- of six plasma protein biomarkers (PSA, free PSA, intact PSA,
ganizations are somewhere in between, recommending PSA hK2, MSMB, MIC1), genetic polymorphisms (232 SNPs) as-
testing for men age 45 or age 50 to 70 years but in a context sociated with prostate cancer susceptibility, and clinical
of shared decision making.32 variables (age, family, history, previous prostate biopsy,

FIGURE 5. Scheme for Prostate Cancer Screening, Diagnostics, and Treatment: Today and Tomorrow

Abbreviations: PSA, prostate-specific antigen; STKHLM3, Stockholm3.

e84 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


EVOLVING RECOMMENDATIONS ON PROSTATE CANCER SCREENING

FIGURE 6. Variables Used in the Stockholm III Model sensitivity as the PSA test using a cutoff of at least 3 ng/mL
to diagnose high-risk prostate cancer, use of the STHLM3
model could reduce the overall number of biopsies by 32%
(95% CI, 24%39%) and small Gleason score 6 cancers by
17% (95% CI, 7%26%) and could avoid 44% (95% CI,
35%54%) of biopsies for men without cancer. The STHLM3
model also identifies a significant number of men with high-
risk cancers in the PSA range of 1-3 ng/mL (Fig. 7).
We see the STHLM3 test as a first step in reducing the
harms of current ongoing PSA screening. The Institute of
Health Economy in Lund, Sweden, has conducted a detailed
health-economic evaluation of the STHLM3 test. Compared
with current clinical practice in Stockholm today, the new
test can save money for the health care provider and at the
same time increase the quality of life in the population.
Abbreviations: DRE, digital rectal examination; PSA, prostate-specific antigen;
STHLM3, Stockholm III
Targeted Prostate Biopsies Using MRI With Fusion
Technology
and prostate examination) with the endpoint of Gleason Men at increased risk of prostate cancer are traditionally
score 7+ cancers. assessed using systematic prostate biopsies. The procedure
The STHLM3 model was predefined in a test cohort of is performed under local anesthesia using antibiotic pro-
11,130 men and then prospectively validated in an in- phylaxis and includes 10 to 14 cores taken from predefined
dependent cohort of 47,688 men. All variables used in the areas of the peripheral zone of the gland as visualized by
STHLM3 model were significantly associated with prostate endorectal ultrasound. While the biopsies systematically
cancers with a Gleason score 7+ (p , .05) in a multiple cover the prostatic gland rather than targeting a lesion, and
logistic regression model (Fig. 6). At the same level of as low-risk cancers are common, the risk of overdiagnosis

FIGURE 7. Comparison of Number of Intermediate- and High-Grade Prostate Cancers Diagnosed with Serum PSA
Screening Versus Stockholm III

Abbreviation: PSA, prostate-specific antigen

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e85



BRAWLEY, THOMPSON, AND GRONBERG

(i.e., detection of nonsignificant tumors) is high. The risk The field of MRI in prostate cancer diagnostics is rapidly
of nonrepresentative biopsy findings results in under- evolving, and, in many centers, MRI is regarded as standard
estimation of Gleason grade compared with subsequent of care, despite solid scientific evidence to support such.
prostatectomy in up to 40% of men undergoing surgery. There is a lack of large randomized studies comparing these
Multiparametric MRI incorporating anatomic and func- two biopsy techniques. In Stockholm, we are initiating a
tional imaging has now been validated as a means of de- randomized study (STHLM3-MRI/fusion study) during 2016,
tecting and characterizing prostate tumors. In 2012, the including a population-based sample of 8,000 men age 50
European Society of Urogenital Radiology established the to 69.
Prostate Imaging Reporting and Data System (PI-RADS)
guidelines aimed at standardizing the acquisition, inter-
Genomic Profiling of Prostate Biopsies
pretation, and reporting of prostate multiparametric MRI.
The knowledge of cancer genomics has exploded with the
Consensus on an updated version (PI-RADS v2) has recently
introduction of modern sequencing technology. New
been published, outlining aspects of both interpretation and
technologies make it possible to conduct both RNA- and
technical execution.34
DNA-based sequencing on small amounts of formalin-fixed
Targeted biopsies of the prostate consist of imaging (MRI)
tissue. Several commercially available RNA-based gene
detecting a lesion and a biopsy procedure where biopsies are
panels with some external validation are now available.
targeted to that area using various devices for guidance.
Today, the commercially available tests are Prolaris,
Cognitive fusion is based on a skilled physician interpreting
Oncotype DX Genomic Prostate Score, and Decipher, which
the MRI images and directing needles solely on the basis of
can be used to estimate disease outcome in addition to
the ultrasound images. The disadvantages of cognitive fu-
clinical parameters. ConfirmMDx is a DNA-based epigenetic
sion lie in the potential for human error when attempting to
test used to predict the results of repeat prostate biopsy
mentally fuse the MRI with ultrasound while aiming for
after an initial negative biopsy.36 However, there is a need
cancers that are often smaller than 1 cm and the inability to
for external validation in large cohorts of men on active
track the location of each biopsy site. Hard fusion enables
surveillance to find the optimal role of these genomic tests.
proper fusion of MRI information with the ultrasound image,
increasing precision of the biopsy.
A number of studies have investigated the value of fusion CONCLUSION
biopsies, using nonrandomized designs and nonscreening Screening guidelines regarding use of PSA have, for the most
populations. A recent meta-analysis of 16 studies compared part, moved to informed or shared decision making. The PSA
MRI-targeted biopsies (MRI-TBx) and ultrasound-targeted and the PCPTRC 2.0 are currently available for use and can be
biopsies (TRUS-Bx) for prostate cancer detection.35 MRI-TBx used in helping doctors and patients decide about biopsy
and TRUS-Bx did not significantly differ in overall prostate after screening results are available. More precise tests and
cancer detection (sensitivity, 0.85 and 0.81, respectively). decision tools will be available in the future. Indeed, the use
MRI-TBx had a higher rate of detection of significant prostate of new biomarkers, imaging, and genomic profiles in di-
cancer compared with TRUS-Bx (sensitivity, 0.91 vs. 0.76) agnosing high-risk prostate cancer is extremely promising.
and a lower rate of detection of insignificant prostate cancer There is an urgent need for evaluation of these new tests in
(sensitivity, 0.44 vs. 0.83). Subgroup analysis revealed an high-quality, large population-based studies. In addition to
improvement in significant prostate cancer detection by efficacy, these new modalities need to be assessed with a
MRI-TBx among men with previous negative biopsy, rather health-economic perspective. If not, we might find ourselves
than among men with initial biopsy. The authors concluded in the same situation as with the PSA test and most likely
that, due to underlying methodological flaws of MRI-TBx, the using these new diagnostic tools inefficiently, only driving
comparison must be regarded with caution. costs without any gain.

References
1. Wolf AM, Wender RC, Etzioni RB, et al; American Cancer Society Prostate Therapy Oncology Group Protocol 85-31. J Clin Oncol. 1997;15:
Cancer Advisory Committee. American Cancer Society guideline for the early 1013-1021.
detection of prostate cancer: update 2010. CA Cancer J Clin. 2010;60:70-98. 5. Bolla M, Gonzalez D, Warde P, et al. Improved survival in patients with
2. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin. locally advanced prostate cancer treated with radiotherapy and
2015;65:5-29. goserelin. N Engl J Med. 1997;337:295-300.
3. Center MM, Jemal A, Lortet-Tieulent J, et al. International variation in 6. Bill-Axelson A, Holmberg L, Ruutu M, et al; SPCG-4 Investigators. Radical
prostate cancer incidence and mortality rates. Eur Urol. 2012;61: prostatectomy versus watchful waiting in early prostate cancer. N Engl J
1079-1092. Med. 2011;364:1708-1717.
4. Pilepich MV, Caplan R, Byhardt RW, et al. Phase III trial of androgen 7. Bill-Axelson A, Holmberg L, Ruutu M, et al; Scandinavian Prostate
suppression using goserelin in unfavorable-prognosis carcinoma of the Cancer Group Study No. 4. Radical prostatectomy versus watchful
prostate treated with definitive radiotherapy: report of Radiation waiting in early prostate cancer. N Engl J Med. 2005;352:1977-1984.

e86 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


EVOLVING RECOMMENDATIONS ON PROSTATE CANCER SCREENING

8. Andriole GL, Crawford ED, Grubb RL III, et al; PLCO Project Team. 23. Do V, Choo R, De Boer G, et al. The role of serial free/total prostate-
Mortality results from a randomized prostate-cancer screening trial. specific antigen ratios in a watchful observation protocol for men with
N Engl J Med. 2009;360:1310-1319. localized prostate cancer. BJU Int. 2002;89:703-709.
9. Schroder FH, Hugosson J, Roobol MJ, et al; ERSPC Investigators. 24. Thompson IM, Ankerst DP, Chi C, et al. Assessing prostate cancer risk:
Screening and prostate-cancer mortality in a randomized European results from the Prostate Cancer Prevention Trial. J Natl Cancer Inst.
study. N Engl J Med. 2009;360:1320-1328. 2006;98:529-534.
10. Thompson IM, Goodman PJ, Tangen CM, et al. The influence of finasteride 25. Thompson IM, Pauler DK, Goodman PJ, et al. Prevalence of prostate
on the development of prostate cancer. N Engl J Med. 2003;349:215-224. cancer among men with a prostate-specific antigen level , or =4.0 ng
11. Wilt TJ, Brawer MK, Jones KM, et al; Prostate Cancer Intervention versus per milliliter. N Engl J Med. 2004;350:2239-2246.
Observation Trial (PIVOT) Study Group. Radical prostatectomy versus 26. Torre LA, Siegel RL, Ward EM, et al. Global cancer incidence and
observation for localized prostate cancer. N Engl J Med. 2012;367: mortality rates and trends-an update. Cancer Epidemiol Biomarkers
203-213. Prev. 2016;25:16-27.
12. Carter HB, Partin AW, Walsh PC, et al. Gleason score 6 adenocarcinoma: 27. Andriole GL, Crawford ED, Grubb RL III, et al; PLCO Project Team.
should it be labeled as cancer? J Clin Oncol. 2012;30:4294-4296. Prostate cancer screening in the randomized Prostate, Lung, Colorectal,
13. Welch HG, Albertsen PC. Prostate cancer diagnosis and treatment after and Ovarian Cancer Screening Trial: mortality results after 13 years of
the introduction of prostate-specific antigen screening: 1986-2005. follow-up. J Natl Cancer Inst. 2012;104:125-132.
J Natl Cancer Inst. 2009;101:1325-1329. 28. Etzioni RD, Thompson IM. What do the screening trials really tell
14. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin. us and where do we go from here? Urol Clin North Am. 2014;41:
2016;66:7-30. 223-228.
15. Hoffman RM, Stone SN, Hunt WC, et al. Effects of misattribution in 29. Stattin P, Carlsson S, Holmstrom B, et al. Prostate cancer mortality in
assigning cause of death on prostate cancer mortality rates. Ann Epi- areas with high and low prostate cancer incidence. J Natl Cancer Inst.
demiol. 2003;13:450-454. 2014;106:dju007.
16. Attard G, Parker C, Eeles RA, et al. Prostate cancer. Lancet. 2016;387: 30. Cuzick J, Thorat MA, Andriole G, et al. Prevention and early detection of
70-82. prostate cancer. Lancet Oncol. 2014;15:e484-e492.
17. U.S. Preventive Services Task Force. Screening for prostate cancer: U.S. 31. Smailyte G, Aleknaviciene B. Incidence of prostate cancer in Lithuania
Preventive Services Task Force recommendation statement. Ann Intern after introduction of the Early Prostate Cancer Detection Programme.
Med. 2008;149:185-191. Public Health. 2012;126:1075-1077.
18. Moyer VA; U.S. Preventive Services Task Force. Screening for prostate 32. Heidenreich A, Bastian PJ, Bellmunt J, et al; European Association of
cancer: U.S. Preventive Services Task Force recommendation state- Urology. EAU guidelines on prostate cancer. part 1: screening, diag-
ment. Ann Intern Med. 2012;157:120-134. nosis, and local treatment with curative intent-update 2013. Eur Urol.
19. Smith RA, Manassaram-Baptiste D, Brooks D, et al. Cancer screening in 2014;65:124-137.
the United States, 2015: a review of current American cancer society 33. Gronberg H, Adolfsson J, Aly M, et al. Prostate cancer screening in men
guidelines and current issues in cancer screening. CA Cancer J Clin. 2015; aged 50-69 years (STHLM3): a prospective population-based diagnostic
65:30-54. study. Lancet Oncol. 2015;16:1667-1676.
20. Schroder FH, Hugosson J, Roobol MJ, et al; ERSPC Investigators. 34. Barentsz JO, Weinreb JC, Verma S, et al. Synopsis of the PI-RADS v2
Prostate-cancer mortality at 11 years of follow-up. N Engl J Med. 2012; guidelines for multiparametric prostate magnetic resonance imaging
366:981-990. and recommendations for use. Eur Urol. 2016;69:41-49.
21. Schroder FH, Hugosson J, Roobol MJ, et al; ERSPC Investigators. 35. Schoots IG, Roobol MJ, Nieboer D, et al. Magnetic resonance imaging-
Screening and prostate cancer mortality: results of the European targeted biopsy may enhance the diagnostic accuracy of significant
Randomised Study of Screening for Prostate Cancer (ERSPC) at 13 years prostate cancer detection compared to standard transrectal
of follow-up. Lancet. 2014;384:2027-2035. ultrasound-guided biopsy: a systematic review and meta-analysis. Eur
22. Klotz L, Vesprini D, Sethukavalan P, et al. Long-term follow-up of a large Urol. 2015;68:438-450.
active surveillance cohort of patients with prostate cancer. J Clin Oncol. 36. Bostrom PJ, Bjartell AS, Catto JW, et al. Genomic predictors of outcome
2015;33:272-277. in prostate cancer. Eur Urol. 2015;68:1033-1044.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e87


CANCER PREVENTION, HEREDITARY GENETICS,
AND EPIDEMIOLOGY

Refining Breast Cancer Risk


Assessment: Additional Genes,
Additional Screening

CHAIR
Allison W. Kurian, MD, MSc
Stanford University School of Medicine
Stanford, CA

SPEAKERS
Antonis C. Antoniou, PhD
University of Cambridge
Cambridge, United Kingdom

Susan M. Domchek, MD
University of Pennsylvania Perelman School of Medicine
Philadelphia, PA
KURIAN, ANTONIOU, AND DOMCHEK

Refining Breast Cancer Risk Stratification: Additional Genes,


Additional Information
Allison W. Kurian, MD, MSc, Antonis C. Antoniou, PhD, and Susan M. Domchek, MD

OVERVIEW

Recent advances in genomic technology have enabled far more rapid, less expensive sequencing of multiple genes than was
possible only a few years ago. Advances in bioinformatics also facilitate the interpretation of large amounts of genomic data.
New strategies for cancer genetic risk assessment include multiplex sequencing panels of 5 to more than 100 genes (in which
rare mutations are often associated with at least two times the average risk of developing breast cancer) and panels of
common single-nucleotide polymorphisms (SNPs), combinations of which are generally associated with more modest cancer
risks (more than twofold). Although these new multiple-gene panel tests are used in oncology practice, questions remain
about the clinical validity and the clinical utility of their results. To translate this increasingly complex genetic information for
clinical use, cancer risk prediction tools are under development that consider the joint effects of all susceptibility genes,
together with other established breast cancer risk factors. Risk-adapted screening and prevention protocols are underway,
with ongoing refinement as genetic knowledge grows. Priority areas for future research include the clinical validity and
clinical utility of emerging genetic tests; the accuracy of developing cancer risk prediction models; and the long-term
outcomes of risk-adapted screening and prevention protocols, in terms of patients experiences and survival.

R ecent advances in genomic technology have enabled


far more rapid, less expensive sequencing of multi-
ple genes than was possible only a few years ago. Ad-
The genes included on most MSPs are displayed in
Table 1. 12-42
Another recent development are panels of several
vances in bioinformatics also facilitate the interpretation SNPs that correlate with breast cancer risk. Identified
of large amounts of genomic data.1 Massively parallel, largely through genome-wide association studies (GWAS)
next-generation sequencing allows the simultaneous that compared breast cancer cases and controls,43-46 SNPs
analysis of many different genes, 2,3 and the emerging most often have minor allele frequencies of greater than
clinical application of whole-genome sequencing.4-6 For 1% and are associated with a lower magnitude of cancer
clinical cancer risk assessment, this next-generation risk (e.g., odds ratios of 1.0-1.5). Recent studies have ex-
sequencing technology has been implemented primar- plored the effect of SNP combinations,47,48 and of SNP
ily as germline multiplex panels that analyze from 5 to interactions with BRCA1/2 mutations14,49-51 and other risk
more than 100 cancer-implicated genes. In general, factors.52,53 As described later, SNP panels are being used
pathogenic mutations in genes on these multiplex se- to improve the accuracy of breast cancer risk prediction
quencing panels confer a twofold or greater risk of de- models.
veloping cancer (considered in some settings to be a At this time, no single, clinically available panel combines
threshold value for intensified screening),7,8 and are rare all established breast cancerassociated genes and SNPs.
in the population (, 0.5% carrier frequency).9,10 These Practice guidelines have begun to extend management
genes are sometimes referred to as moderate to high recommendations to carriers of mutations in the higher-risk
penetrance cancer susceptibility genes. Focusing on genes on MSPs,12 but no guidelines currently exist for cancer
breast cancer risk, multiplex sequencing panels (MSPs) screening or prevention on the basis of the combined effects
represent an intermediate stage between the past of the common genetic variants (SNPs). Significant questions
practice of testing BRCA1 and BRCA2 (BRCA1/2) only, 11 remain about these emerging approaches to genetic risk
and a possible future of routine whole-genome sequencing. assessment.

From the Departments of Medicine and of Health Research and Policy, Stanford University School of Medicine, Stanford, CA; Centre for Cancer Genetic Epidemiology, Department of
Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom; Basser Research Center and Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Allison W. Kurian, MD, MSc, Divisions of Oncology and Epidemiology, Stanford University School of Medicine, 150 Governors Ln., Stanford, CA 94305-5405;
email: akurian@stanford.edu.

2016 by American Society of Clinical Oncology.

44 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


EXTENDING GERMLINE TESTING IN PATIENTS WITH BREAST CANCER

ADVANTAGES OF MULTIPLEX SEQUENCING (whether the test result enables better patient care and
PANELS outcomes).
Multiplex sequencing panels that analyze several breast With regard to clinical validity, a major challenge lies in
cancerrelated genes simultaneously offer several advan- the high rate of reported variants of uncertain significance
tages over the prior standard of testing only two genes (VUS) with MSPs. Variants of uncertain significance are
(BRCA1/2). In past years, the high cost of sequencing genetic alterations (frequently missense) whose disease
more than two genes was prohibitive and thus limited association is unknown, and uncertainty about their clinical
to patients with striking features of Cowden, Li Fraumeni, or relevance may frustrate patients and physicians. With
other rare syndromes. By contrast, MSPs of 5 to greater than MSPs, the prevalence of at least one VUS (among all se-
100 genes now cost approximately $1,500, less than half the quenced genes) is approximately 20% to 40%.54,57,59,64,68
cost of sequencing two genes in past years. With a turn- By contrast, VUS are rare (2%5%) when only BRCA1/2 are
around time of 3 to 4 weeks, MSPs are far more efficient than sequenced, because widespread BRCA1/2 testing in diverse
the previous standard of care. populations has better defined the spectrum of normal
Multiplex sequencing panels also provide an improved variation for these genes.69 Another challenge arises when
diagnostic yield. Among clinic-based studies that collectively two laboratories provide conflicting interpretations of the
assessed more than 10,000 patients who tested negative for same genetic alteration (e.g., VUS vs. deleterious).
BRCA1/2 mutations, mutation prevalence in other MSP Questions of clinical validity also concern apparently
genes ranged from 4% to 16%.54-64 Some mutations were pathogenic (often truncating) mutations in genes whose
clinically unexpected (e.g., a patient with triple-negative cancer risks are not well understood, yet are included on
breast cancer who was found to have a MSH6 mutation, MSPsfor example, whether a mutation in MRE11A confers
consistent with Lynch syndrome),56 prompting a change in clinically meaningful risk of breast cancer.9,66 There is no
cancer screening or prevention targeted to the identified transparent process for gene selection on many MSPs, and
mutation. There is growing recognition that the striking the combination of genes may seem arbitrary. Emerging
cancer phenotypes associated with certain high-risk mu- MSP delivery models that empower ordering clinicians to
tations may be too narrowly defined, and that muta- select genes for inclusion on a custom panel (and to ex-
tion penetrance may be more variable than previously clude other genes considered irrelevant to a patients care)
believed.65-67 may help, although this may require a level of genetic
knowledge that is unrealistic for most clinicians. A recent
review article designated 11 well-established breast
MULTIPLEX SEQUENCING PANELS cancer susceptibility genes (ATM, BRCA1, BRCA2, CDH1,
Questions of Clinical Validity CHEK2, NBN, NF1, PALB2, PTEN, STK11, TP53) and ad-
The benefits of MSPs are accompanied by potential risks and vised that mutations in other genes not be used in clinical
uncertainties. These concern questions of clinical validity breast cancer risk assessment.70
(such as whether the test result correctly predicts a clini-
cal phenotype [e.g., high cancer risk]) and clinical utility
Questions of Clinical Utility
The most pressing question about MSPs is their clinical utility.
Rather than the more typical process for a new intervention,
KEY POINTS in which proof of clinical utility precedes widespread
adoption, MSPs diffused into oncology practice ahead of
Recent advances in genomic technology have enabled
far more rapid, less expensive sequencing of multiple
any supporting evidence base.9,66 Causative forces include
genes than was possible only a few years ago. new genomic technology, a 2013 U.S. Supreme Court de-
Although these new multiple-gene panel tests are used cision against gene patenting that caused increased market
in oncology practice, questions remain about the clinical competition and reduced costs, and celebrity testing dis-
validity and the clinical utility of their results. closures that raised public awareness.71-74 In recent years,
Cancer risk prediction tools that consider the joint studies have evaluated the clinical experience of MSP
effects of all known susceptibility genes, together with testing in various populations, 54-60,62-64 with some re-
other established breast cancer risk factors, are under porting subsequent changes in screening and prevention
development. protocols.56,64 Practice guidelines including those of the
Risk-adapted screening and prevention protocols are National Comprehensive Cancer Network (NCCN) have added
underway, with ongoing refinement as genetic
management recommendations for genes with a better-
knowledge grows.
Priority research areas include the validity of emerging
established risk profile (e.g., ATM, CHEK2, PALB2),12,26 and
genetic tests; the accuracy of developing cancer risk in 2015 the American Society of Clinical Oncology (ASCO)
prediction models; and the long-term outcomes of risk- issued a position statement on extended germline testing.75,76
adapted screening and prevention protocols, in terms of However, uncertainty remains about patients preferences and
patients experiences and survival. experiences of MSPs; the optimal care delivery models; and
the effectiveness of risk-reducing interventions developed for

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 45


KURIAN, ANTONIOU, AND DOMCHEK

TABLE 1. Cancer Risks and Guidelines for Breast CancerAssociated Genes Often Included on Multiplex Panels
Established Breast Cancer Susceptibility Genes
Breast Cancer
Gene Relative Risk Other Cancer Risks and Syndromes Clinical Practice Guidelines Reference
ATM Two- to threefold Ataxia telangiectasia syndrome in National Comprehensive Cancer Network (NCCN): Screening 12,13
homozygotes; possibly with breast MRI
colon, pancreas
BRCA1 10-fold Ovarian American Cancer Society (ACS) and NCCN: Screening 8,12,14-16
breast MRI, recommend risk-reducing bilateral
salpingo-oophorectomy (RRBSO),
discuss risk-reducing mastectomy (RRM)
BRCA2 10-fold Ovarian, pancreatic, prostate; ACS and NCCN: Screening breast MRI, recommend RRBSO, 8,12,14-16
possibly melanoma discuss RRM
CDH1 Fivefold Gastric NCCN: Screening breast MRI, discuss RRM, discuss 12,17-19
risk-reducing gastrectomy
CHEK2 Two- to threefold Possibly colon, thyroid, lung NCCN: Screening breast MRI 12,20,21
NBN Two- to threefold Nijmegen breakage syndrome in None 22-24
homozygotes; possibly ovarian
NF1 Two- to threefold Central nervous system, No breast cancerrelated guidelines 25
peripheral nerve sheath
PALB2 Three- to fivefold Pancreas; possibly ovarian NCCN: Screening breast MRI, discuss RRM 12,26
PTEN At least fivefold Thyroid, endometrial NCCN: Screening breast MRI, discuss RRM 12,27,28
STK11 At least fivefold Pancreas, colon, ovarian sex NCCN: Screening breast MRI, discuss RRM 12,29
cord-stromal
TP53 At least 10-fold Multiple: sarcoma, leukemia, NCCN: Screening breast MRI, discuss RRM; 12,30
adrenocortical, brain, others whole-body MRI, colonoscopy, complete
blood count, and other tests

Genes With Undefined Risk of Breast Cancer Frequently Included on Multiplex Sequencing Panels
Breast Cancer
Gene Relative Risk Other Cancer Risks and Syndromes Clinical Practice Guidelines Reference
BARD1 Undefined Unknown None 31,32
BLM Undefined Blooms syndrome in homozygotes None 33
BRIP1 Undefined Ovarian NCCN: Consider RRBSO 12,34
FAM175A Undefined Fanconi anemia in homozygotes None 35
FANCC Undefined Fanconi anemia in homozygotes None 36
MRE11A Undefined Ataxia telangiectasialike disorder in None 22,23,37
homozygotes; possibly ovarian
RAD50 Undefined Possibly ovarian None 22,23
RAD51C Undefined Ovarian NCCN: Consider RRBSO 12,38
RAD51D Undefined Ovarian NCCN: Consider RRBSO 12,39-41
XRCC2 Undefined Unknown None 42

BRCA1/2 mutation carriers among carriers of other gene As discussed previously, studies have reported estimates
mutations. of the prevalence of other gene mutations among patients
tested for BRCA1/2 mutations.12,54-60,62-64 Although in-
formative and clinically relevant, most existing studies have
UNANSWERED QUESTIONS ABOUT MULTIPLEX
focused exclusively on patients with striking personal and/or
SEQUENCING
Genetic Epidemiology family histories of early onset breast/ovarian cancer, and on
Determining the clinical utility of MSPs requires research in two non-Hispanic whites. A key question is the prevalence of
major disciplines: epidemiology and health service research.66 mutations and VUS among patients at lower estimated risk,
First, we must understand the prevalence, penetrance, and including breast cancer patients unselected for age or family
phenotype of mutations in genes beyond BRCA1/2; second, we history; such data are essential to learning which pa-
must understand how identifying such mutations affects pa- tients may benefit from MSPs. Of similar importance is
tients outcomes, in terms of psychosocial symptoms, cancer defining the prevalence of mutations and VUS across
detection rate and stage shift, and, ultimately, survival. racial/ethnic groups, as done for BRCA1/2.77,78 Studies in

46 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


EXTENDING GERMLINE TESTING IN PATIENTS WITH BREAST CANCER

population-based datasets are currently underway to genetic susceptibility variants, together with other estab-
address these questions. lished risk factors. However, to construct such multifactorial
Understanding the variability in penetrance of mutations cancer risk prediction models, it is important first to un-
in genes beyond BRCA1/2 is necessary to enable accurate derstand the underlying models of genetic susceptibility for
patient counseling. Large, collaborative studies will be re- breast cancer.
quired to obtain sufficient numbers of mutation carriers:
ongoing consortium efforts include the PROMPT, Breast Models of Genetic Susceptibility to Breast Cancer
Cancer Risk after Diagnostic Gene Sequencing (BRIDGES), Epidemiologic studies have estimated that breast cancer is
and COMPLEXO studies (www.promptstudy.info; cordis. approximately twice as common among first-degree rela-
europa.eu/project/rcn/193315_en.html).79 Other crucial tives of breast cancer patients (defined as the familial
research questions include reclassifying clinically ambiguous relative risk).85 The most important breast cancer sus-
VUS to benign or deleterious, which is an aim of the ENIGMA ceptibility genes are BRCA1/2, mutations that account for
consortium80; identifying the pathologic and prognostic 17% to 20% of the excess familial risk of breast cancer.86-88
features of tumors associated with mutations in specific Rare mutations in the established breast cancer suscepti-
genes; and etiologic studies of cancer causation by muta- bility genes included on MSPs, such as PALB2, CHEK2, ATM,
tions, particularly those in genes whose association with TP53, PTEN, CHD1, STK11, NF1, and NBN (Table 1), account
breast cancer is less well proven. for approximately 5%,89 and more than 100 common SNPs
account for an additional 16% of familial breast cancer.90
Health Services Research Together, all known breast cancer susceptibility variants
There are several crucial topics for health services research on account for 38% to 41%; thus more than 50% of familial
MSPs. One important question is how best to deliver genetic clustering remains unexplained. Both known genetic vari-
counseling and testing services, in the challenging environ- ants and residual familial aggregation must be considered in
ment of increasingly complex test results and a shortage of calculating the risk of developing breast cancer.
licensed genetic counselors.81 New delivery models include a
tiered and binned approach to summarizing test options by Combined Single-Nucleotide Polymorphisms
risk categories.82 Long-distance counseling methods such as Associations
telemedicine are also under investigation. Individual common alleles confer modest risks and their
Another essential question is the patient experience of individual predictive utility is poor. Recently, Mavaddat
MSP testing. As described before, there is concern that the et al91 investigated all pairwise interactions between 77
volume, complexity, and uncertainty of results may over- breast cancerassociated SNPs and found no evidence of
whelm patients. One study reported that some patients deviation from a model in which SNPs combine multipli-
decline MSPs because of such concerns.83 In the ongoing catively. Common SNPs can thus be combined into a Poly-
Hereditary Cancer Panel prospective trial of MSPs at three genic Risk Score (PRS): considering 90 common susceptibility
academic cancer centers, most patients reported little loci, 5% of women have a greater than twofold increased risk
distress or uncertainty after receiving their test results.84 and 0.7% have a greater than threefold increased risk,
A related question is the impact of extended germline compared with the general populations risk of breast
sequencing on patients uptake of interventions such as cancer.90 In models that considered the joint effects of PRS
preventive surgery or more intensive screening (e.g., and family history, PRS stratifies risk in women without
magnetic resonance imaging), given the concern that VUS family history and further refines genetic risk assessment in
might be misinterpreted as cause for unwarranted in- women with family history.48,91-95 Ongoing large studies,
terventions. Although early results of the Hereditary Cancer such as the OncoArray experiment, are expected to refine
Panel trial offer no evidence that patients overuse pro- these models.
phylactic procedures based on test results,84 longer-term
follow-up is required. Answering these questions about the Single-Nucleotide Polymorphisms and BRCA1/2
delivery and outcomes of MSP testing will be required Mutations
to guide analyses of the incremental benefit of analyzing The International Consortium of Investigators of Modifiers
more versus fewer genes, both in terms of efficacy and of of BRCA1/2 (CIMBA) studied the effects of common SNPs on
cost-effectiveness. BRCA1/2-associated breast cancer risks,96 using different
approaches: GWAS of BRCA1/2 mutation carriers,49,97,98
meta-analysis of BRCA1/2-associated risks with other similar
POLYMORPHIC BREAST CANCER RISK FACTORS phenotypes, fine-scale mapping of known risk-modifying
AND NEW MODELS loci,43 and directly assessing the effects of established
Despite the widespread availability of MSPs and SNP arrays, breast cancer susceptibility alleles in mutation carriers.99-103
their utility is limited by the lack of risk prediction models CIMBA identified 24 SNPs that modify BRCA1-associated risk
that consider the multifactorial etiology of breast cancer (which have stronger associations with estrogen receptor
susceptibility. There is an urgent need for breast cancer risk [ER]negative breast cancer in the general population) and
prediction models that consider the joint effects of all known 20 SNPs that modify BRCA2-associated risk.104-106 A recent

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 47


KURIAN, ANTONIOU, AND DOMCHEK

study investigated the associations of common SNPs sub- BRCA1 and BRCA2 mutation carriers as in the general
divided by the ER status of BRCA1- and BRCA2-associated population.117 For example, later age of first birth is asso-
breast tumors,51,103 and found that differences in SNP as- ciated with lower breast cancer risk for BRCA1 mutation
sociations between BRCA1 and BRCA2 carriers and non- carriers, yet with higher breast cancer risk in the general
carriers are largely explained by differences in the population.117
prevalence of ER-positive and ER-negative tumors in mu-
tation carriers and the heterogeneity of the associations Implications for Cancer Risk Stratification
between common alleles and ER-positive or ER-negative Our growing understanding of the multifactorial etiology of
disease. These are in line with a general model of suscep- breast cancer suggests that much more powerful risk pre-
tibility under which BRCA1/2 mutations and common alleles diction can be achieved by combining data on all genetic,
combine multiplicatively once stratified by ER status. As a lifestyle, and hormonal risk factors,92,118 reaching stratifi-
result, jointly, common SNPs can result in large differences in cation levels that can guide individual decision making and
the absolute lifetime risks of breast cancer among BRCA1/2 population risk-reduction programs.93,95,119 However, the
mutation carriers.97,107 Risk stratification may improve as assumptions underlying these projections require testing in
enhanced PRS are developed that include a large number of empirical studies.
SNPs. Women with BRCA1/2 mutations may be among the
first to gain a clinically meaningful benefit from the use of
common SNPs for risk assessment; however, large studies Breast Cancer Risk Assessment Tools
are required to assess prospectively the enhanced PRS that Several breast cancer risk prediction models are available
are currently under development. for unaffected women; they differ in the risk factors they
consider and in their underlying statistical modeling ap-
Single-Nucleotide Polymorphisms and Other Rare proaches (Table 2). The two broad categories of models
Gene Mutations are empirical and genetic. Empirical models do not
Little is known about the combined effects of common SNPs assume a specific genetic model, but instead consider
and other rare mutations beyond BRCA1/2, mainly because of summary measures of family history: an example is the
the lack of large-scale cohorts tested with MSPs. However, widely used Gail model120 in the Breast Cancer Risk As-
breast cancer risks among PALB2 and CHEK2 mutation car- sessment Tool (www.cancer.gov/bcrisktool/). Genetic
riers appear to be modified by other familial factors, in- models make explicit assumptions about known and
creasing with family history and/or with bilateral breast unknown genetic effects, considering the mode of in-
cancer.26,108-110 These findings suggest a multiplicative model heritance, mutation frequency, and penetrance. A major
as with BRCA1/2, but confirmatory studies are needed. advantage of genetic models is that they can compute
risks for families of any disease structure; however, their
Combined Effects of Rare Gene Mutations adequacy depends on modeling all genetic effects, which
Recent evidence suggests that gene-gene interactions can be approximate only because all breast cancer sus-
among CHEK2, ATM, BRCA1, and BRCA2 may not be mul- ceptibility genes have not yet been discovered.121
tiplicative.111 Indeed, a multiplicative model would be im- Examples of genetic models include Claus,122,123
plausible for BRCA1 and BRCA2, because it would predict an BRCAPRO,124 IBIS,125 and BOADICEA (Table 2).112,126,127
extremely high breast cancer risk at very young ages. Pro- BRCAPRO, IBIS, and BOADICEA all consider the explicit
teins encoded by the majority of breast cancer genes play effects of BRCA1/2 mutations but differ in their modeling
roles in DNA repair pathways. If no elevated risks are ob- of residual familial aggregation: IBIS assumes a hypo-
served in the presence of two gene mutations, the data thetical third dominant gene, whereas BOADICEA
would be consistent with a model in which, if the pathway is assumes a polygenic component.
disrupted by a mutation, further disruption by a lower
penetrance mutation would not increase risk.112 Single-Nucleotide Polymorphisms in Risk Assessment
Tools
Combined Effects of Genetic, Lifestyle, and Hormonal Several studies have demonstrated improvement in the
Factors performance of some models through consideration of
Studies of common SNPs and other risk factors suggest that common SNPs.47,119,128,129 However, most such studies as-
they combine in a multiplicative fashion.113-116 There is sumed that the risks predicted by existing models (e.g., IBIS,
currently no evidence on how the risks of rare gene muta- BOADICEA) are multiplicative with those conferred by SNPs,
tions (other than BRCA1/2) combine with lifestyle and without adjusting for the fact that SNPs account for ap-
hormonal factors, but large ongoing studies (e.g., BRIDGES, proximately 15% of the familial aggregation of breast cancer.
cordis.europa.eu/project/rcn/193315_en.html), will report This approach warrants further validation before routine
on this question soon. The evidence for pathogenic variants clinical use. More accurate implementations will require the
in BRCA1 and BRCA2 is currently limited and conflicting, but underlying algorithms to be modified to ensure that the fa-
there are some suggestions that the relative risks of lifestyle/ milial risks predicted by the models on the basis of the known
hormonal factors may not necessarily be the same in both and unknown genetic effects are correctly specified.

48 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


EXTENDING GERMLINE TESTING IN PATIENTS WITH BREAST CANCER

TABLE 2. Breast Cancer Risk Assessment Tools Used in Clinical Practice: Components and Assumptions
Factor* Gail Claus BRCAPRO IBIS BOADICEA
Family history YES (descriptive) YES YES YES YES
BRCA1, BRCA2 mutations NO NO YES YES YES
Common low-risk alleles NO NO NO NO NO
Intermediate-high risk mutations (CHEK2, PALB2, ATM, etc.) NO NO NO NO YES**
Residual non-BRCA1/2 familial aggregation NO NO NO YES; dominant YES; polygenic
3rd gene
BRCA1/2 breast cancer pathology associations NO NO YES NO YES
BRCA1/2 risk modification by family history NO NO NO NO YES
Variants of uncertain significance NO NO NO NO NO
Predicting estrogen receptor (ER)-specific risks NO NO NO NO NO
Mammographic density NO NO NO NO NO
Hormonal, lifestyle, and reproductive risk factors YES NO NO YES; assumes same NO
effect on BRCA1/2
Other cancers (nonbreast or ovarian cancer) NO NO YES NO YES
Predicting second cancer risks (contralateral breast, ovarian cancer) NO NO NO NO YES
*Considered in the available tools for clinical use.
**Beta-version release.
Only pathology information for the proband.

Rare Gene Mutations in Risk Assessment Tools newly identified mutations for both mutation carriers and
Until recently, no model incorporated the effects of muta- their relatives.
tions in genes beyond BRCA1/2. A major difficulty had been
the lack of precise cancer risk estimates; however, a recent Unresolved Problems in Risk Modeling, Challenges,
extension to the BOADICEA risk assessment model includes and Future Directions
the effects of truncating mutations in PALB2, CHEK2, and Along with BRCA1, BRCA2, PALB2, ATM, and CHEK2, there are
ATM for which reliable risk estimates are now available.70,112 several other breast cancer susceptibility genes which could
A beta-testing version of this tool is available for clinical use be incorporated into risk prediction.70 However, more ac-
(ccge.medschl.cam.ac.uk/boadicea/). This extended BOA- curate estimates are required for the breast cancer risks of
DICEA model can be used to obtain cancer risks on the basis mutations in these genes. Similarly, little is known about the
of mutation screening in all five major breast cancer sus- joint effects of rare gene mutations, about the risks for carriers
ceptibility genes (BRCA1, BRCA2, PALB2, CHEK2, ATM) of of mutations in more than one breast cancer susceptibility
explicit family history information and of other risk factors gene, or how these genetic risks combine with other lifestyle
(Table 2). Figure 1A demonstrates that risks for carriers of and hormonal risk factors. A number of ongoing research
rare mutations (e.g., an ATM truncating variant) depend on programs will address these issues in the near future, such as
family history. Although the average lifetime breast cancer the PROMPT, BRIDGES, and CARRIERS programs. As discussed
risk for a carrier of an ATM truncating variant is approxi- before, another problem is VUS reported in BRCA1/2 and
mately 29%, the risk for an ATM carrier whose mother de- other genes. Several efforts are underway to reclassify these
veloped breast cancer at age 40 is estimated at VUS through consortia like ENIGMA80 and using approaches
approximately 44%. These differences may have implications such as the posterior probability of causality.131,132 Future
for screening and preventive interventions. Figure 1B shows modeling efforts should aim to include such evidence in order
predicted risks for a woman whose mother carried a BRCA1 to provide an integrated approach for counseling VUS carriers.
or ATM mutation and had breast cancer, but who has herself Breast cancer is a heterogeneous disease that differs
tested negative for the same mutation (known as a true based on tumor characteristics. Tumor characteristics vary
negative or noncarrier130). Compared to considering based on genetic background, and risk factors have different
family history alone, the reduction in breast cancer risk after associations with different tumor subtypes.133,134 A meta-
negative testing is greater when the mother carries a analysis of trials of selective estrogen receptor modulators
BRCA1 mutation (predicted risk approximately 15%) as breast cancer chemoprevention agents suggested a 38%
compared with an ATM mutation (predicted risk . 18%). risk reduction135; however, a critical gap for clinical trans-
These results suggest that negative predictive testing for lation is how to identify which women will develop ER-
ATM (or similarly moderate-risk mutations) may not imply positive breast cancer, because selective estrogen receptor
sufficient risk reduction to enable reclassification for modulators prevent only ER-positive disease.133,136 Thus
clinical management purposes, and emphasize the need for there is need of risk models to predict ER-specific risks. Data
further study of the breast cancer risks associated with such generated through the large scale B-CAST (Breast CAncer

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 49


KURIAN, ANTONIOU, AND DOMCHEK

FIGURE 1. Predicted Breast Cancer Risks Using the BOADICEA Model, Beta-Version 4.0

(A) Predicted breast cancer risk by ATM mutation status and family history. BOADICEA-predicted risks by ATM mutation status for a female in the United Kingdom born in 1995 (age
20). The three curves show the breast cancer risk in the population, the risk for the average carrier of an ATM mutation (without taking family history into account), and the risk for
a carrier of an ATM mutation whose mother also developed breast cancer at age 40.
(B) Negative predictive testing and predicted breast cancer risk. The predicted risk of breast cancer for a 20-year-old female born in 1995 in the United Kingdom, whose
mother developed breast cancer at age 40, according to her mothers mutation status. Risk estimates were obtained using the BOADICEA model, beta-version 4.0.

Stratification, http://www.b-cast.eu) program will yield valu- favor fewer mammograms, it becomes increasingly impor-
able data for modeling disease subtypespecific risks. tant to improve risk stratification using factors such as re-
Finally, it will be essential to validate risk-prediction productive and hormonal exposures, breast density, and
models in large, well-designed prospective studies, and to genetics; and to understand the complex interactions be-
develop user-friendly tools that meet the needs of clinicians tween these risk factors. Optimizing risk stratification is
at all care levels. essential to inform individualized breast cancer screening
recommendations.
RISK REFINEMENT AND STRATIFYING
SCREENING APPROACHES Breast MRI
Mammography At the current time for women at elevated risk, breast MRI is
Recent updates to guidelines have triggered ongoing con- often added to yearly screening mammogram. Breast MRI is
troversy regarding the age of initiation and interval timing more sensitive than mammogram for detecting cancers in
of screening mammography in women at average risk for BRCA1/2 mutation carriers and women with a family history
breast cancer.137-139 Although the specifics of guidelines of breast cancer.140 Compared with historical controls,
differ, the trend is for later initiation of routine mammo- breast cancers detected by MRI are diagnosed at an earlier
grams, with the possibility of spacing them to every other stage; historical comparisons and modeling analyses predict
year. Throughout this discussion, it has been recognized that that MRI screening may improve survival.141-145 No ran-
risk stratification is key to optimizing the clinical benefit of domized studies have been performed evaluating breast
breast cancer screening; that is, these new recommenda- MRI as an adjunct to screening mammography. Concerns
tions are for women at average risk, but women at higher risk about yearly breast MRI include cost, inconvenience (the
may need a more intensive approach. However, most test requires intravenous contrast) and specificity. Cost of
women who develop breast cancer are not considered to be breast MRI is particularly high in the United States. Although
at elevated risk before their diagnosis. Thus, as guidelines adding annual breast MRI from the ages of 35 to 54 to annual

50 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


EXTENDING GERMLINE TESTING IN PATIENTS WITH BREAST CANCER

mammograms in BRCA1 mutation carriers has been shown carriers of mutations in moderate penetrance genes
to be cost-effective at $100,000 per quality-adjusted life- should continue annual screening mammograms. Re-
year, the cost-effectiveness of MRI screening decreases at cently, Mandelblatt et al examined eight different
lower levels of risk. For example, Plevritis et al calculated a mammographic screening strategies and determined
cost-effectiveness ratio (CER) of $266,344/quality-adjusted that for women with a two- to fourfold increase over the
life-year for annual MRI in addition to mammogram screen- average breast cancer risk, annual screening from age
ing of BRCA2 mutation carriers from ages 35 to 54.146 CERs 40 years had similar harms and benefits as did screening
in women with mutations conferring a lower risk of breast average-risk women biennially from age 50 to 74 years.153
cancer will be even less favorable. Finally, emerging technologies such as breast tomosynthesis
Regardless of concerns about cost, multiple guidelines and rapid breast MRI may alter screening approaches in
recommend annual breast MRI in women at elevated risk the future.
of breast cancer.7,12,147-149 Guidelines differ in the
stipulated level of risk that should trigger the addition of Prevention Options
breast MRI, but in the United States, the American Women at elevated risk for breast cancer also have
Cancer Society, and NCCN recommend adding annual options for prevention. All women, regardless of esti-
breast MRI to mammogram in women with a lifetime risk mated risk level, should be counseled on the harmful
of greater than 20% based on a family history model8,12; impact that obesity, lack of exercise, and alcohol use
this recommendation exists despite the absence of have on breast cancer risk. In addition, pharmacologic
empirical evidence that breast MRI screening improves breast cancer risk reduction with tamoxifen, raloxifene,
outcomes. Existing guidelines do not specify what is or an aromatase inhibitor is available. In randomized
meant by lifetime riskwhether cumulative lifetime risk clinical trials, tamoxifen taken for 5 years confers a 30%
or remaining risk at a womans current age. Therefore, to 50% reduction in the risk of developing breast can-
depending on the models used, different recommen- cer, 136,154 with continued benefit at 16 years of follow-
dations may be made for the same woman.150 up (albeit no documented survival benefit to date). 136
Average lifetime breast cancer risks are estimated to Raloxifene also confers a reduction in the risk of breast
approach or exceed 30% with mutations in PALB2, ATM, cancer. When directly compared with tamoxifen in a
NBN, and CHEK2 (for CHEK2, excluding the p.I157T and p. randomized clinical trial, in long-term follow-up ralox-
S428F mutations). Therefore, carriers of mutations in ifene had 76% of the effectiveness of tamoxifen in
these genes could be considered for breast MRI screening preventing invasive breast cancer, yet far less toxicity in
on the basis of existing U.S. guidelines. Importantly, two terms of the risk of endometrial cancer.155 Exemestane
missense mutations in CHEK2, p.I157T and p.S428F, are (an aromatase inhibitor) has also been shown to reduce
associated with lower levels of breast cancer risk (relative breast cancer risk in a randomized controlled trial, with a
risk [RR] , 2), and the corresponding lifetime risks are hazard ratio of 0.35 (95% CI, 0.180.70; p = .002). 156
unlikely to exceed 20% in the absence of a family history of Although eligibility criteria differed somewhat for these
breast cancer.151,152 Likewise, there is insufficient evi- studies, most included women age 60 or older, those
dence at this time to recommend breast MRI screening in with a Gail modelbased estimate of 5-year risk of de-
carriers of mutations in BARD1, BRIP1, MRE11A, RAD51C, veloping breast cancer of greater than 1.66%, and/or
and RAD51D in the absence of a family history.70 In those those with lobular carcinoma in situ. Data are not
women who do meet existing guidelines for MRI sur- available regarding the effectiveness of chemoprevention
veillance, the appropriate age at which to initiate screen- among carriers of mutations in moderate penetrance
ing is currently unclear. Because of the lower risks and breast cancer susceptibility genes. CHEK2 mutations do
later median onset of breast cancer associated with mu- appear to be associated with ER-positive breast cancer,
tations in moderate penetrance genes compared with but data regarding chemoprevention in this setting are
BRCA1/2, it seems reasonable to initiate breast MRI unavailable.
screening in carriers of mutations in moderate penetrance There is no threshold risk that mandates risk-reducing
genes later than for BRCA1/2 mutation carriers, for whom mastectomy in any unaffected woman, regardless of her
MRI screening begins at age 25; however, screening mutation type. It is unlikely that mastectomy will provide a
recommendations for any individual patient will depend survival advantage to women with moderate penetrance
on family cancer history. mutations, given their level of risk and the effectiveness of
Currently, breast MRI screening often begins 5 to breast cancer screening and treatment. Elicitation of indi-
10 years before the earliest age at diagnosis of breast vidual preferences and communication about levels of risk
cancer in the family; however, a more rational approach are critical in these conversations.
may be to focus on the absolute risk of breast cancer over
the next 5 years. As evidence accumulates, the ages for Ovarian Cancer
initiation of surveillance and the type of surveillance Unfortunately, ovarian cancer screening has been shown not
must be re-examined. Because guidelines recommend to reduce mortality.157 Therefore, risk-reducing salpingo-
less for women at average risk, women identified as oophorectomy (RRSO) is the standard recommendation for

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 51


KURIAN, ANTONIOU, AND DOMCHEK

women with BRCA1/2 mutations. Large case-control studies Pancreas Cancer


have demonstrated that women with mutations in BRIP1, Mutations in PALB2 and ATM have been associated with
RAD51C, and RAD51D are at increased risk of ovarian cancer. familial pancreas cancer159,160; however, the absolute risks
The absolute risks associated with mutations in these genes of pancreas cancer associated with such mutations are
are intermediate between those of a woman with a BRCA1/2- unknown. There are no proven effective screening or pre-
negative first-degree relative with ovarian cancer (RR 2.2, vention measures for pancreas cancer, although there is
absolute lifetime risk 3%5%) and of a woman carrying much interest in endoscopic ultrasonography and magnetic
BRCA2 mutation (absolute lifetime risk approximately resonance cholangiopancreatography as potential screening
17%).15,158 Studies have not proven increased ovarian approaches. Carriers of ATM or PALB2 mutations who have a
cancer risks in carriers of mutations in other moderate family history of pancreas cancer may be candidates for
penetrance genes including PALB2, ATM, CHEK2, BARD1, clinical trials of screening strategies.
MRE11A, NBN, and RAD51B, although mutations in these
genes have been observed in families with both breast and Communication
ovarian cancers. Communicating information about breast cancer risk and
Risk-reducing salpingo-oophorectomy is not routinely the risks and benefits of management options can be
recommended for women whose only ovarian cancer risk challenging in the clinical setting. As knowledge increases
factor is an affected first-degree relative. A womans cu- about the genetic epidemiology of breast cancer risk,
mulative risk should therefore at least be similar to the risk communication becomes even more important and chal-
of a woman with an affected BRCA1/2-negative first-degree lenging. Partly owing to the op-ed pieces by celebrity
relative with ovarian cancer before RRSO is recommended. It Angelina Jolie in The New York Times, more women are
is unlikely that carriers of mutations in BRIP1, RAD51C, or aware of the implications of BRCA1/2 mutations. Physicians
RAD51D would cross this risk threshold before age 50. must be careful to explain how mutations in moderate
However, family history of ovarian cancer may influence this penetrance genes differ from BRCA1/2 mutations in terms of
risk and thus may alter decision making about RRSO timing. the magnitude of associated risks.

References
1. Collins FS, Varmus H. A new initiative on precision medicine. N Engl J 13. Renwick A, Thompson D, Seal S, et al; Breast Cancer Susceptibility
Med. 2015;372:793-795. Collaboration (UK). ATM mutations that cause ataxia-telangiectasia
2. Shendure J, Ji H. Next-generation DNA sequencing. Nat Biotechnol. are breast cancer susceptibility alleles. Nat Genet. 2006;38:873-875.
2008;26:1135-1145. 14. Mavaddat N, Peock S, Frost D, et al; EMBRACE. Cancer risks for BRCA1
3. Tucker T, Marra M, Friedman JM. Massively parallel sequencing: the and BRCA2 mutation carriers: results from prospective analysis of
next big thing in genetic medicine. Am J Hum Genet. 2009;85:142-154. EMBRACE. J Natl Cancer Inst. 2013;105:812-822.
4. Kidd JM, Cooper GM, Donahue WF, et al. Mapping and sequencing of 15. Antoniou A, Pharoah PD, Narod S, et al. Average risks of breast and
structural variation from eight human genomes. Nature. 2008;453:56-64. ovarian cancer associated with BRCA1 or BRCA2 mutations detected
5. Yang Y, Muzny DM, Xia F, et al. Molecular findings among patients in case Series unselected for family history: a combined analysis of 22
referred for clinical whole-exome sequencing. JAMA. 2014;312: studies. Am J Hum Genet. 2003;72:1117-1130.
1870-1879. 16. Chen S, Parmigiani G. Meta-analysis of BRCA1 and BRCA2 penetrance.
6. Dewey FE, Grove ME, Pan C, et al. Clinical interpretation and impli- J Clin Oncol. 2007;25:1329-1333.
cations of whole-genome sequencing. JAMA. 2014;311:1035-1045. 17. Pharoah PD, Guilford P, Caldas C; International Gastric Cancer Linkage
7. Mainiero MB, Lourenco A, Mahoney MC, et al. ACR Appropriateness Consortium. Incidence of gastric cancer and breast cancer in CDH1
Criteria Breast Cancer Screening. J Am Coll Radiol. 2013;10:11-14. (E-cadherin) mutation carriers from hereditary diffuse gastric cancer
8. Saslow D, Boetes C, Burke W, et al; American Cancer Society Breast families. Gastroenterology. 2001;121:1348-1353.
Cancer Advisory Group. American Cancer Society guidelines for breast 18. Kaurah P, MacMillan A, Boyd N, et al. Founder and recurrent CDH1
screening with MRI as an adjunct to mammography. CA Cancer J Clin. mutations in families with hereditary diffuse gastric cancer. JAMA.
2007;57:75-89. 2007;297:2360-2372.
9. Domchek SM, Bradbury A, Garber JE, et al. Multiplex genetic testing 19. van der Post RS, Vogelaar IP, Carneiro F, et al. Hereditary diffuse
for cancer susceptibility: out on the high wire without a net? J Clin gastric cancer: updated clinical guidelines with an emphasis on
Oncol. 2013;31:1267-1270. germline CDH1 mutation carriers. J Med Genet. 2015;52:361-374.
10. Kurian AW, Kingham KE, Ford JM. Next-generation sequencing for 20. Weischer M, Bojesen SE, Ellervik C, et al. CHEK2*1100delC genotyping
hereditary breast and gynecologic cancer risk assessment. Curr Opin for clinical assessment of breast cancer risk: meta-analyses of 26,000
Obstet Gynecol. 2015;27:23-33. patient cases and 27,000 controls. J Clin Oncol. 2008;26:542-548.
11. Couch FJ, Nathanson KL, Offit K. Two decades after BRCA: setting 21. Meijers-Heijboer H, van den Ouweland A, Klijn J, et al; CHEK2-Breast
paradigms in personalized cancer care and prevention. Science. 2014; Cancer Consortium. Low-penetrance susceptibility to breast cancer
343:1466-1470. due to CHEK2(*)1100delC in noncarriers of BRCA1 or BRCA2 muta-
12. Daly MB, Pilarski R, Axilbund JE, et al; National comprehensive cancer tions. Nat Genet. 2002;31:55-59.
network. Genetic/familial high-risk assessment: breast and ovarian, 22. Bartkova J, Tommiska J, Oplustilova L, et al. Aberrations of the MRE11-
version 1.2014. J Natl Compr Canc Netw. 2014;12:1326-1338. RAD50-NBS1 DNA damage sensor complex in human breast cancer:

52 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


EXTENDING GERMLINE TESTING IN PATIENTS WITH BREAST CANCER

MRE11 as a candidate familial cancer-predisposing gene. Mol Oncol. 43. Bojesen SE, Pooley KA, Johnatty SE, et al. Multiple independent
2008;2:296-316. variants at the TERT locus are associated with telomere length and
23. Hsu HM, Wang HC, Chen ST, et al. Breast cancer risk is associated with risks of breast and ovarian cancer. Nat Genet. 2013;45:371-384,
the genes encoding the DNA double-strand break repair Mre11/ 384e371-372.
Rad50/Nbs1 complex. Cancer Epidemiol Biomarkers Prev. 2007;16: 44. Garcia-Closas M, Couch FJ, Lindstrom S, et al. Genome-wide associ-
2024-2032. ation studies identify four ER negative-specific breast cancer risk loci.
24. Zhang G, Zeng Y, Liu Z, et al. Significant association between Nijmegen Nat Genet. 2013;45:392-398, 398e391-392.
breakage syndrome 1 657del5 polymorphism and breast cancer risk. 45. Ghoussaini M, Fletcher O, Michailidou K, et al; Netherlands Collab-
Tumour Biol. 2013;34:2753-2757. orative Group on Hereditary Breast and Ovarian Cancer (HEBON);
25. Seminog OO, Goldacre MJ. Age-specific risk of breast cancer in women Familial Breast Cancer Study (FBCS); Gene Environment Interaction of
with neurofibromatosis type 1. Br J Cancer. 2015;112:1546-1548. Breast Cancer in Germany (GENICA) Network; kConFab Investigators;
26. Antoniou AC, Casadei S, Heikkinen T, et al. Breast-cancer risk in Australian Ovarian Cancer Study Group. Genome-wide association
families with mutations in PALB2. N Engl J Med. 2014;371:497-506. analysis identifies three new breast cancer susceptibility loci. Nat
27. Tan MH, Mester JL, Ngeow J, et al. Lifetime cancer risks in individuals Genet. 2012;44:312-318.
with germline PTEN mutations. Clin Cancer Res. 2012;18:400-407. 46. Michailidou K, Hall P, Gonzalez-Neira A, et al. Large-scale genotyping
28. Pilarski R, Eng C. Will the real Cowden syndrome please stand up identifies 41 new loci associated with breast cancer risk. Nat Genet.
(again)? Expanding mutational and clinical spectra of the PTEN 2013;45:353-361, 361e351-352.
hamartoma tumour syndrome. J Med Genet. 2004;41:323-326. 47. Dite GS, MacInnis RJ, Bickerstaffe A, et al. Breast cancer risk prediction
29. Hearle N, Schumacher V, Menko FH, et al. Frequency and spectrum of using clinical models and 77 independent risk-associated SNPs for
cancers in the Peutz-Jeghers syndrome. Clin Cancer Res. 2006;12: women aged under 50 years: Australian Breast Cancer Family Registry.
3209-3215. Cancer Epidemiol Biomarkers Prev. 2015.
30. Gonzalez KD, Noltner KA, Buzin CH, et al. Beyond Li Fraumeni Syn- 48. Pharoah PD, Antoniou AC, Easton DF, et al. Polygenes, risk prediction,
drome: clinical characteristics of families with p53 germline muta- and targeted prevention of breast cancer. N Engl J Med. 2008;358:
tions. J Clin Oncol. 2009;27:1250-1256. 2796-2803.
31. Karppinen SM, Barkardottir RB, Backenhorn K, et al. Nordic col- 49. Antoniou AC, Wang X, Fredericksen ZS, et al; EMBRACE; GEMO Study
laborative study of the BARD1 Cys557Ser allele in 3956 patients Collaborators; HEBON; kConFab; SWE-BRCA; MOD SQUAD; GENICA. A
with cancer: enrichment in familial BRCA1/BRCA2 mutation-negative locus on 19p13 modifies risk of breast cancer in BRCA1 mutation
breast cancer but not in other malignancies. J Med Genet. 2006;43: carriers and is associated with hormone receptor-negative breast
856-862. cancer in the general population. Nat Genet. 2010;42:885-892.
32. Stacey SN, Sulem P, Johannsson OT, et al. The BARD1 Cys557Ser 50. Gaudet MM, Kuchenbaecker KB, Vijai J, et al; KConFab Investigators;
variant and breast cancer risk in Iceland. PLoS Med. 2006;3:e217. Ontario Cancer Genetics Network; HEBON; EMBRACE; GEMO Study
33. Broberg K, Huynh E, Schlawicke Engstrom K, et al. Association be- Collaborators; GENICA Network. Identification of a BRCA2-specific
tween polymorphisms in RMI1, TOP3A, and BLM and risk of cancer, a modifier locus at 6p24 related to breast cancer risk. PLoS Genet. 2013;
case-control study. BMC Cancer. 2009;9:140. 9:e1003173.
34. Seal S, Thompson D, Renwick A, et al; Breast Cancer Susceptibility 51. Mulligan AM, Couch FJ, Barrowdale D, et al; Breast Cancer Family
Collaboration (UK). Truncating mutations in the Fanconi anemia J gene Registry; EMBRACE; GEMO Study Collaborators; HEBON; kConFab
BRIP1 are low-penetrance breast cancer susceptibility alleles. Nat Investigators; Ontario Cancer Genetics Network; SWE-BRCA; CIMBA.
Genet. 2006;38:1239-1241. Common breast cancer susceptibility alleles are associated with tu-
35. Solyom S, Aressy B, Pylkas K, et al. Breast cancer-associated Abraxas mour subtypes in BRCA1 and BRCA2 mutation carriers: results from
mutation disrupts nuclear localization and DNA damage response the Consortium of Investigators of Modifiers of BRCA1/2. Breast
functions. Sci Transl Med. 2012;4:122ra23. Cancer Res. 2011;13:R110.
36. Barroso E, Pita G, Arias JI, et al. The Fanconi anemia family of genes and 52. Wacholder S, Hartge P, Prentice R, et al. Performance of common
its correlation with breast cancer susceptibility and breast cancer genetic variants in breast-cancer risk models. N Engl J Med. 2010;362:
features. Breast Cancer Res Treat. 2009;118:655-660. 986-993.
37. Walsh T, Lee MK, Casadei S, et al. Detection of inherited mutations for 53. Mealiffe ME, Stokowski RP, Rhees BK, et al. Assessment of clinical
breast and ovarian cancer using genomic capture and massively validity of a breast cancer risk model combining genetic and clinical
parallel sequencing. Proc Natl Acad Sci USA. 2010;107:12629-12633. information. J Natl Cancer Inst. 2010;102:1618-1627.
38. Meindl A, Hellebrand H, Wiek C, et al. Germline mutations in breast 54. Tung N, Battelli C, Allen B, et al. Frequency of mutations in individuals
and ovarian cancer pedigrees establish RAD51C as a human cancer with breast cancer referred for BRCA1 and BRCA2 testing using next-
susceptibility gene. Nat Genet. 2010;42:410-414. generation sequencing with a 25-gene panel. Cancer. 2015;121:25-33.
39. Loveday C, Turnbull C, Ramsay E, et al; Breast Cancer Susceptibility 55. Castera L, Krieger S, Rousselin A, et al. Next-generation sequencing for
Collaboration (UK). Germline mutations in RAD51D confer suscepti- the diagnosis of hereditary breast and ovarian cancer using genomic
bility to ovarian cancer. Nat Genet. 2011;43:879-882. capture targeting multiple candidate genes. Eur J Hum Genet. 2014;
40. Thompson ER, Rowley SM, Sawyer S, et al. Analysis of RAD51D in 22:1305-1313.
ovarian cancer patients and families with a history of ovarian or breast 56. Kurian AW, Hare EE, Mills MA, et al. Clinical evaluation of a multiple-
cancer. PLoS One. 2013;8:e54772. gene sequencing panel for hereditary cancer risk assessment. J Clin
41. Baker JL, Schwab RB, Wallace AM, et al. Breast cancer in a RAD51D Oncol. 2014;32:2001-2009.
mutation carrier: case report and review of the literature. Clin Breast 57. LaDuca H, Stuenkel AJ, Dolinsky JS, et al. Utilization of multigene
Cancer. 2015;15:e71-e75. panels in hereditary cancer predisposition testing: analysis of more
42. Kuschel B, Auranen A, McBride S, et al. Variants in DNA double-strand than 2,000 patients. Genet Med. 2014;16:830-837.
break repair genes and breast cancer susceptibility. Hum Mol Genet. 58. Maxwell KN, Wubbenhorst B, DAndrea K, et al. Prevalence of
2002;11:1399-1407. mutations in a panel of breast cancer susceptibility genes in

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 53


KURIAN, ANTONIOU, AND DOMCHEK

BRCA1/2-negative patients with early-onset breast cancer. Genet 79. Southey MC, Park DJ, Nguyen-Dumont T, et al; COMPLEXO.
Med. 2015;17:630-638. COMPLEXO: identifying the missing heritability of breast cancer via
59. Selkirk CG, Vogel KJ, Newlin AC, et al. Cancer genetic testing panels for next generation collaboration. Breast Cancer Res. 2013;15:402.
inherited cancer susceptibility: the clinical experience of a large adult 80. Spurdle AB, Healey S, Devereau A, et al; ENIGMA. ENIGMAevidence-
genetics practice. Fam Cancer. 2014;13:527-536. based network for the interpretation of germline mutant alleles: an
60. Couch FJ, Hart SN, Sharma P, et al. Inherited mutations in 17 breast international initiative to evaluate risk and clinical significance as-
cancer susceptibility genes among a large triple-negative breast sociated with sequence variation in BRCA1 and BRCA2 genes. Hum
cancer cohort unselected for family history of breast cancer. J Clin Mutat. 2012;33:2-7.
Oncol. 2015;33:304-311. 81. Stadler ZK, Schrader KA, Vijai J, et al. Cancer genomics and inherited
61. Churpek JE, Walsh T, Zheng Y, et al. Inherited predisposition to breast risk. J Clin Oncol. 2014;32:687-698.
cancer among African American women. Breast Cancer Res Treat. 82. Bradbury AR, Patrick-Miller L, Long J, et al. Development of a tiered
2015;149:31-39. and binned genetic counseling model for informed consent in the era
62. Cybulski C, Lubinski
J, Wokoorczyk D, et al. Mutations predisposing to of multiplex testing for cancer susceptibility. Genet Med. 2015;17:
breast cancer in 12 candidate genes in breast cancer patients from 485-492.
Poland. Clin Genet. 2015;88:366-370. 83. Bradbury AR, Patrick-Miller LJ, Egleston BL, et al. Patient feedback and
63. Thompson ER, Rowley SM, Li N, et al. Panel testing for familial breast early outcome data with a novel tiered-binned model for multiplex
cancer: calibrating the tension between research and clinical care. breast cancer susceptibility testing. Genet Med. 2016;18:25-33.
J Clin Oncol. Epub 2016 Jan 19. 84. Kurian AW, Idos GE, McDonnell K, et al. The patient experience in a
64. Desmond A, Kurian AW, Gabree M, et al. Clinical actionability of prospective trial of multiplex gene panel testing for cancer risk.
multigene panel testing for hereditary breast and ovarian cancer risk Abstract presented at the San Antonio Breast Cancer Symposium.
assessment. JAMA Oncol. 2015;1:943-951. 2015; San Antonio, Texas.
65. Idos GE, Kurian AW, McDonnell K, et al. Interim analysis of multiplex 85. Collaborative Group on Hormonal Factors in Breast Cancer. Familial
gene panel testing for inherited susceptibility to breast cancer. Ab- breast cancer: collaborative reanalysis of individual data from 52
stract presented at the San Antonio Breast Cancer Symposium; 2015; epidemiological studies including 58,209 women with breast cancer
San Antonio, Texas. and 101,986 women without the disease. Lancet. 2001;358:
66. Kurian AW, Ford JM. Multigene panel testing in oncology practice: 1389-1399.
how should we respond? JAMA Oncol. 2015;1:277-278. 86. Anglian Breast Cancer Study Group. Prevalence and penetrance of
67. Kurian AW, Ford JM. Multiple-gene panels and the future of genetic BRCA1 and BRCA2 mutations in a population-based series of breast
testing. Curr Breast Cancer Rep. 2015;7:98-104. cancer cases. Br J Cancer. 2000;83:1301-1308.
68. Lincoln SE, Kobayashi Y, Anderson MJ, et al. A systematic comparison 87. Peto J, Collins N, Barfoot R, et al. Prevalence of BRCA1 and BRCA2 gene
of traditional and multigene panel testing for hereditary breast and mutations in patients with early-onset breast cancer. J Natl Cancer
ovarian genes in more than 1000 patients. J Mol Diagn. 2015;17: Inst. 1999;91:943-949.
533-544. 88. Mavaddat N, Pharoah PD, Blows F, et al; SEARCH Team. Familial
69. Hall MJ, Reid JE, Burbidge LA, et al. BRCA1 and BRCA2 mutations in relative risks for breast cancer by pathological subtype: a population-
women of different ethnicities undergoing testing for hereditary based cohort study. Breast Cancer Res. 2010;12:R10.
breast-ovarian cancer. Cancer. 2009;115:2222-2233. 89. Bahcall OG. iCOGS collection provides a collaborative model. Fore-
70. Easton DF, Pharoah PD, Antoniou AC, et al. Gene-panel sequencing word. Nat Genet. 2013;45:343.
and the prediction of breast-cancer risk. N Engl J Med. 2015;372: 90. Michailidou K, Beesley J, Lindstrom S, et al; BOCS; kConFab In-
2243-2257. vestigators; AOCS Group; NBCS; GENICA Network. Genome-wide
71. Offit K, Bradbury A, Storm C, et al. Gene patents and personalized association analysis of more than 120,000 individuals identifies 15
cancer care: impact of the Myriad case on clinical oncology. J Clin new susceptibility loci for breast cancer. Nat Genet. 2015;47:373-380.
Oncol. 2013;31:2743-2748. 91. Mavaddat N, Pharoah PD, Michailidou K, et al. Prediction of breast
72. Evans DG, Barwell J, Eccles DM, et al; FH02 Study Group; RGC teams. cancer risk based on profiling with common genetic variants. J Natl
The Angelina Jolie effect: how high celebrity profile can have a major Cancer Inst. 2015;107:djv036.
impact on provision of cancer related services. Breast Cancer Res. 92. Garcia-Closas M, Gunsoy NB, Chatterjee N. Combined associations of
2014;16:442. genetic and environmental risk factors: implications for prevention of
73. Juthe RH, Zaharchuk A, Wang C. Celebrity disclosures and information breast cancer. J Natl Cancer Inst. 2014;106:dju305.
seeking: the case of Angelina Jolie. Genet Med. 2015;17:545-553. 93. Pashayan N, Guo Q, Pharoah PD. Personalized screening for cancers:
74. Lebo PB, Quehenberger F, Kamolz LP, et al. The Angelina effect should we consider polygenic profiling? Per Med. 2013;10(6).
revisited: Exploring a media-related impact on public awareness. 94. Hall AE, Chowdhury S, Hallowell N, et al. Implementing risk-stratified
Cancer. 2015;121:3959-3964. screening for common cancers: a review of potential ethical, legal and
75. Robson ME, Bradbury AR, Arun B, et al. American Society of Clinical social issues. J Public Health (Oxf). 2014;36:285-291.
Oncology Policy Statement Update: Genetic and Genomic Testing for 95. Burton H, Chowdhury S, Dent T, et al. Public health implications from
Cancer Susceptibility. J Clin Oncol. 2015;33:3660-3667. COGS and potential for risk stratification and screening. Nat Genet.
76. Yu PP, Vose JM, Hayes DF. Genetic cancer susceptibility testing: increased 2013;45:349-351.
technology, increased complexity. J Clin Oncol. 2015;33:3533-3534. 96. Chenevix-Trench G, Milne RL, Antoniou AC, et al; CIMBA. An in-
77. John EM, Miron A, Gong G, et al. Prevalence of pathogenic BRCA1 ternational initiative to identify genetic modifiers of cancer risk in
mutation carriers in 5 US racial/ethnic groups. JAMA. 2007;298: BRCA1 and BRCA2 mutation carriers: the Consortium of Investigators
2869-2876. of Modifiers of BRCA1 and BRCA2 (CIMBA). Breast Cancer Res. 2007;9:
78. Kurian AW. BRCA1 and BRCA2 mutations across race and ethnicity: 104.
distribution and clinical implications. Curr Opin Obstet Gynecol. 2010; 97. Couch FJ, Wang X, McGuffog L, et al; kConFab Investigators; SWE-
22:72-78. BRCA; Ontario Cancer Genetics Network; HEBON; EMBRACE; GEMO

54 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


EXTENDING GERMLINE TESTING IN PATIENTS WITH BREAST CANCER

Study Collaborators; BCFR; CIMBA. Genome-wide association study in 110. Fletcher O, Johnson N, Dos Santos Silva I, et al. Family history, genetic
BRCA1 mutation carriers identifies novel loci associated with breast testing, and clinical risk prediction: pooled analysis of CHEK2 1100delC
and ovarian cancer risk. PLoS Genet. 2013;9:e1003212. in 1,828 bilateral breast cancers and 7,030 controls. Cancer Epidemiol
98. Gaudet MM, Kirchhoff T, Green T, et al; GEMO Study Collaborators; Biomarkers Prev. 2009;18:230-234.
HEBON Study Collaborators; OCGN; kConFab; EMBRACE. Common 111. Turnbull C, Seal S, Renwick A, et al; Breast Cancer Susceptibility
genetic variants and modification of penetrance of BRCA2-associated Collaboration (UK), EMBRACE. Gene-gene interactions in breast
breast cancer. PLoS Genet. 2010;6:e1001183. cancer susceptibility. Hum Mol Genet. 2012;21:958-962.
99. Antoniou AC, Hardy R, Walker L, et al. Predicting the likelihood of 112. Lee A, Cunningham AP, Tischkowitz M, et al. Incorporating Truncating
carrying a BRCA1 or BRCA2 mutation: validation of BOADICEA, Variants in PALB2, CHEK2 and ATM into the BOADICEA Breast Cancer
BRCAPRO, IBIS, Myriad and the Manchester scoring system using data Risk Model. Genet Med. 2016. In press.
from UK genetics clinics. J Med Genet. 2008;45:425-431. 113. Nickels S, Truong T, Hein R, et al; Genica Network; kConFab; AOCS
100. Antoniou AC, Sinilnikova OM, McGuffog L, et al; Kathleen Cuningham Management Group. Evidence of gene-environment interactions
Foundation Consortium for Research into Familial Breast Cancer; between common breast cancer susceptibility loci and established
OCGN; HEBON; EMBRACE; GEMO; Breast Cancer Family Registry; environmental risk factors. PLoS Genet. 2013;9:e1003284.
CIMBA. Common variants in LSP1, 2q35 and 8q24 and breast cancer 114. Campa D, Kaaks R, Le Marchand L, et al. Interactions between genetic
risk for BRCA1 and BRCA2 mutation carriers. Hum Mol Genet. 2009;18: variants and breast cancer risk factors in the breast and prostate
4442-4456. cancer cohort consortium. J Natl Cancer Inst. 2011;103:1252-1263.
101. Antoniou AC, Kartsonaki C, Sinilnikova OM, et al; SWE-BRCA; HEBON; 115. Rudolph A, Fasching PA, Behrens S, et al. A comprehensive evaluation
EMBRACE; CEMO Study Collaborators; Breast Cancer Family Registry; of interaction between genetic variants and use of menopausal
kConFab investigators; CIMBA. Common alleles at 6q25.1 and 1p11.2 hormone therapy on mammographic density. Breast Cancer Res.
are associated with breast cancer risk for BRCA1 and BRCA2 mutation 2015;17:110.
carriers. Hum Mol Genet. 2011;20:3304-3321. 116. Rudolph A, Milne RL, Truong T, et al; kConFab Investigators; AOCS
102. Antoniou AC, Kuchenbaecker KB, Soucy P, et al; CIMBA, SWE-BRCA; Group; GENICA-Network. Investigation of gene-environment in-
HEBON; EMBRACE; GEMO Collaborators Study; kConFab In- teractions between 47 newly identified breast cancer susceptibility
vestigators. Common variants at 12p11, 12q24, 9p21, 9q31.2 and in loci and environmental risk factors. Int J Cancer. 2015;136:E685-E696.
ZNF365 are associated with breast cancer risk for BRCA1 and/or 117. Friebel TM, Domchek SM, Rebbeck TR. Modifiers of cancer risk in
BRCA2 mutation carriers. Breast Cancer Res. 2012;14:R33. BRCA1 and BRCA2 mutation carriers: systematic review and meta-
103. Kuchenbaecker KB, Neuhausen SL, Robson M, et al; Breast Cancer analysis. J Natl Cancer Inst. 2014;106:dju091.
Family Registry; EMBRACE Study; GEMO Study Collaborators; HEBON; 118. Sieh W, Rothstein JH, McGuire V, et al. The role of genome sequencing
KConFab Investigators; CIMBA. Associations of common breast cancer in personalized breast cancer prevention. Cancer Epidemiol Bio-
susceptibility alleles with risk of breast cancer subtypes in BRCA1 and markers Prev. 2014;23:2322-2327.
BRCA2 mutation carriers. Breast Cancer Res. 2014;16:3416. 119. Brentnall AR, Evans DG, Cuzick J. Distribution of breast cancer risk
104. Mavaddat N, Barrowdale D, Andrulis IL, et al; HEBON; EMBRACE; from SNPs and classical risk factors in women of routine screening age
GEMO Study Collaborators; kConFab Investigators; SWE-BRCA Col- in the UK. Br J Cancer. 2014;110:827-828.
laborators; Consortium of Investigators of Modifiers of BRCA1/2. 120. Gail MH, Brinton LA, Byar DP, et al. Projecting individualized prob-
Pathology of breast and ovarian cancers among BRCA1 and BRCA2 abilities of developing breast cancer for white females who are being
mutation carriers: results from the Consortium of Investigators of examined annually. J Natl Cancer Inst. 1989;81:1879-1886.
Modifiers of BRCA1/2 (CIMBA). Cancer Epidemiol Biomarkers Prev. 121. Antoniou AC, Easton DF. Risk prediction models for familial breast
2012;21:134-147. cancer. Future Oncol. 2006;2:257-274.
105. Lakhani SR, Van De Vijver MJ, Jacquemier J, et al. The pathology of 122. Claus EB, Risch N, Thompson WD. The calculation of breast cancer risk
familial breast cancer: predictive value of immunohistochemical for women with a first degree family history of ovarian cancer. Breast
markers estrogen receptor, progesterone receptor, HER-2, and p53 in Cancer Res Treat. 1993;28:115-120.
patients with mutations in BRCA1 and BRCA2. J Clin Oncol. 2002;20: 123. Claus EB, Risch N, Thompson WD. Autosomal dominant inheritance of
2310-2318. early-onset breast cancer. Implications for risk prediction. Cancer.
106. Lakhani SR, Reis-Filho JS, Fulford L, et al; Breast Cancer Linkage 1994;73:643-651.
Consortium. Prediction of BRCA1 status in patients with breast cancer 124. Parmigiani G, Berry D, Aguilar O. Determining carrier probabilities for
using estrogen receptor and basal phenotype. Clin Cancer Res. 2005; breast cancer-susceptibility genes BRCA1 and BRCA2. Am J Hum
11:5175-5180. Genet. 1998;62:145-158.
107. Antoniou AC, Beesley J, McGuffog L, et al; Ontario Cancer Genetics 125. Tyrer J, Duffy SW, Cuzick J. A breast cancer prediction model in-
Network; SWE-BRCA; HEBON; EMBRACE; GEMO; GEMO; Breast corporating familial and personal risk factors. Stat Med. 2004;23:
Cancer Family Registry; kConFab; CIMBA. Common breast cancer 1111-1130.
susceptibility alleles and the risk of breast cancer for BRCA1 and 126. Antoniou AC, Cunningham AP, Peto J, et al. The BOADICEA model of
BRCA2 mutation carriers: implications for risk prediction. Cancer Res. genetic susceptibility to breast and ovarian cancers: updates and
2010;70:9742-9754. extensions. Br J Cancer. 2008;98:1457-1466.
108. Meijers-Heijboer H, van den Ouweland A, Klijn J, et al; CHEK2-Breast 127. Lee AJ, Cunningham AP, Kuchenbaecker KB, et al; Consortium of
Cancer Consortium. Low-penetrance susceptibility to breast cancer Investigators of Modifiers of BRCA1/2; Breast Cancer Association
due to CHEK2(*)1100delC in noncarriers of BRCA1 or BRCA2 muta- Consortium. BOADICEA breast cancer risk prediction model: updates
tions. Nat Genet. 2002;31:55-59. to cancer incidences, tumour pathology and web interface. Br J
109. CHEK2 Breast Cancer Case-Control Consortium. CHEK2*1100delC and Cancer. 2014;110:535-545.
susceptibility to breast cancer: a collaborative analysis involving 128. Gail MH. Discriminatory accuracy from single-nucleotide polymor-
10,860 breast cancer cases and 9,065 controls from 10 studies. Am J phisms in models to predict breast cancer risk. J Natl Cancer Inst.
Hum Genet. 2004;74:1175-1182. 2008;100:1037-1041.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 55


KURIAN, ANTONIOU, AND DOMCHEK

129. Darabi H, Czene K, Zhao W, et al. Breast cancer risk prediction and 146. Plevritis SK, Kurian AW, Sigal BM, et al. Cost-effectiveness of screening
individualised screening based on common genetic variation and BRCA1/2 mutation carriers with breast magnetic resonance imaging.
breast density measurement. Breast Cancer Res. 2012;14:R25. JAMA. 2006;295:2374-2384.
130. Kurian AW, Gong GD, John EM, et al. Breast cancer risk for noncarriers 147. National Collaborating Centre for Cancer. Familial breast cancer:
of family-specific BRCA1 and BRCA2 mutations: findings from the Classification and care of people at risk of familial breast cancer and
Breast Cancer Family Registry. J Clin Oncol. 2011;29:4505-4509. management of breast cancer and related risks in people with a family
131. Easton DF, Deffenbaugh AM, Pruss D, et al. A systematic genetic history of breast cancer. Cardiff (U.K.): National Collaborating Centre
assessment of 1,433 sequence variants of unknown clinical signifi- for Cancer, United Kingdom; 2013.
cance in the BRCA1 and BRCA2 breast cancer-predisposition genes. 148. Netherlands Breast Cancer Screening Guidelines, version 2.0. 2012;
Am J Hum Genet. 2007;81:873-883. Integraal Kankercentrum Nederland.
132. Lindor NM, Guidugli L, Wang X, et al. A review of a multifactorial 149. Thomssen C, Harbeck N, on behalf of the AGO Breast Committee.
probability-based model for classification of BRCA1 and BRCA2 var- Update 2010 of the German AGO Recommendations for the Diagnosis
iants of uncertain significance (VUS). Hum Mutat. 2012;33:8-21. and Treatment of Early and Metastatic Breast Cancer - Chapter B:
133. Rosner B, Glynn RJ, Tamimi RM, et al. Breast cancer risk prediction Prevention, Early Detection, Lifestyle, Premalignant Lesions, DCIS,
with heterogeneous risk profiles according to breast cancer tumor Recurrent and Metastatic Breast Cancer. Breast Care (Basel). 2010;5:
markers. Am J Epidemiol. 2013;178:296-308. 345-351.
134. Yang XR, Chang-Claude J, Goode EL, et al. Associations of breast cancer risk 150. Quante AS, Whittemore AS, Shriver T, et al. Practical problems with
factors with tumor subtypes: a pooled analysis from the Breast Cancer clinical guidelines for breast cancer prevention based on remaining
Association Consortium studies. J Natl Cancer Inst. 2011;103:250-263. lifetime risk. J Natl Cancer Inst. 2015;107:djv124.
135. Cuzick J, Sestak I, Bonanni B, et al; SERM Chemoprevention of Breast 151. Han FF, Guo CL, Liu LH. The effect of CHEK2 variant I157T on cancer
Cancer Overview Group. Selective oestrogen receptor modulators in susceptibility: evidence from a meta-analysis. DNA Cell Biol. 2013;32:
prevention of breast cancer: an updated meta-analysis of individual 329-335.
participant data. Lancet. 2013;381:1827-1834. 152. Shaag A, Walsh T, Renbaum P, et al. Functional and genomic ap-
136. Cuzick J, Sestak I, Cawthorn S, et al; IBIS-I Investigators. Tamoxifen for proaches reveal an ancient CHEK2 allele associated with breast cancer
prevention of breast cancer: extended long-term follow-up of the IBIS- in the Ashkenazi Jewish population. Hum Mol Genet. 2005;14:
I breast cancer prevention trial. Lancet Oncol. 2015;16:67-75. 555-563.
137. Oeffinger KC, Fontham ET, Etzioni R, et al; American Cancer Society. 153. Mandelblatt JS, Stout NK, Schechter CB, et al. Collaborative modeling
Breast cancer screening for women at average risk: 2015 guideline of the benefits and harms associated with different U.S. breast cancer
update from the american cancer society. JAMA. 2015;314:1599-1614. screening strategies. Ann Oncol. 2016;164:215-225.
138. Nelson HD, Tyne K, Naik A, et al. Screening for breast cancer: an 154. Fisher B, Costantino JP, Wickerham DL, et al. Tamoxifen for prevention
update for the U.S. Preventive Services Task Force. Ann Intern Med. of breast cancer: report of the National Surgical Adjuvant Breast and
2009;151:727-737, W237-742. Bowel Project P-1 Study. J Natl Cancer Inst. 1998;90:1371-1388.
139. Myers ER, Moorman P, Gierisch JM, et al. Benefits and harms of breast 155. Vogel VG, Costantino JP, Wickerham DL, et al; National Surgical
cancer screening: A systematic review. JAMA. 2015;314:1615-1634. Adjuvant Breast and Bowel Project. Update of the National Surgical
140. Warner E, Messersmith H, Causer P, et al. Systematic review: using Adjuvant Breast and Bowel Project Study of Tamoxifen and Raloxifene
magnetic resonance imaging to screen women at high risk for breast (STAR) P-2 Trial: Preventing breast cancer. Cancer Prev Res (Phila).
cancer. Ann Intern Med. 2008;148:671-679. 2010;3:696-706.
141. Warner E, Hill K, Causer P, et al. Prospective study of breast cancer 156. Goss PE, Ingle JN, Ale s-Martnez JE, et al; NCIC CTG MAP.3 Study
incidence in women with a BRCA1 or BRCA2 mutation under sur- Investigators. Exemestane for breast-cancer prevention in post-
veillance with and without magnetic resonance imaging. J Clin Oncol. menopausal women. N Engl J Med. 2011;364:2381-2391.
2011;29:1664-1669. 157. Jacobs IJ, Menon U, Ryan A, et al. Ovarian cancer screening and
142. Evans DG, Kesavan N, Lim Y, et al; MARIBS Group. MRI breast mortality in the UK Collaborative Trial of Ovarian Cancer Screen-
screening in high-risk women: cancer detection and survival analysis. ing (UKCTOCS): a randomised controlled trial. Lancet. 2016;387:
Breast Cancer Res Treat. 2014;145:663-672. 945-956.
143. Heijnsdijk EA, Warner E, Gilbert FJ, et al. Differences in natural history 158. Jervis S, Song H, Lee A, et al. Ovarian cancer familial relative risks by
between breast cancers in BRCA1 and BRCA2 mutation carriers and tumour subtypes and by known ovarian cancer genetic susceptibility
effects of MRI screening-MRISC, MARIBS, and Canadian studies variants. J Med Genet. 2014;51:108-113.
combined. Cancer Epidemiol Biomarkers Prev. 2012;21:1458-1468. 159. Grant RC, Selander I, Connor AA, et al. Prevalence of germline mu-
144. Saadatmand S, Obdeijn I-M, Rutgers EJ, et al. Survival benefit in tations in cancer predisposition genes in patients with pancreatic
women with BRCA1 mutation or familial risk in the MRI screening cancer. Gastroenterology. 2015;148:556-564.
study (MRISC). Int J Cancer. 2015;137:1729-1738. 160. Zhen DB, Rabe KG, Gallinger S, et al. BRCA1, BRCA2, PALB2, and
145. Kurian AW, Sigal BM, Plevritis SK. Survival analysis of cancer risk reduction CDKN2A mutations in familial pancreatic cancer: a PACGENE study.
strategies for BRCA1/2 mutation carriers. J Clin Oncol. 2010;28:222-231. Genet Med. 2015;17:569-577.

56 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


CARE DELIVERY AND PRACTICE MANAGEMENT

Access to Cancer Therapeutics


in Low- and Middle-Income
Countries

CHAIR
Paul Ruff, MBBCh, MMed, FCP(SA)
University of Witwatersrand Faculty of Health Sciences
Johannesburg, South Africa

SPEAKERS
Lawrence N. Shulman, MD
Abramson Cancer Center of the University of Pennsylvania
Philadelphia, PA

Sana Al-Sukhun, MD, MSc


The University of Jordan
Amman, Jordan
RUFF ET AL

Access to Cancer Therapeutics in Low- and Middle-Income


Countries
Paul Ruff, MBBCh, MMed, FCP(SA), Sana Al-Sukhun, MD, MSc, Charmaine Blanchard, MPhil, MBBCh, BSc,
and Lawrence N. Shulman, MD

OVERVIEW

Cancer is rapidly becoming a major health care problem, especially in developing countries, where 60% of the worlds total
new cases are diagnosed. The success of new antineoplastic medicines and modern radiation devices to cure a good
proportion of patients with cancer and to alleviate the suffering of many more has been achieved at a dramatic cost.
Therefore, it has become mandatory for health care authorities and pharmaceutical companies to cooperate to use and
develop resources in an efficient manner to improve health care delivery to patients with cancer worldwide. Regulatory
harmonization is an important key to overcome delays in the approval process, whether for antineoplastic and pain control
medicines or for essential medical devices. More emphasis on the significant role of opiates in pain control among patients
with cancer is needed to overcome the ingrained belief in their potential for addiction. The World Health Organization (WHO)
serves an important role in guiding priorities for health care and efficiently allocating resources by providing essential
medicine lists (EMLs) and device lists. However, the financial challenge for access to health care is multi-tiered and requires
collaboration between key stakeholders including pharmaceutical industry, local national health authorities, WHO, and
other nonprofit, patient-oriented organizations.

S ince the turn of the century, we have seen a paradigm


shift in the way we treat cancer, with the advent of
targeted therapies, especially monoclonal antibodies and
available in the 1950s. Surgical care is limited by lack of skills
and facilities, whereas access to palliative medicines, es-
pecially for pain control, is limited by regulatory and legal
small-molecule kinase inhibitors. Indeed the development of restrictions, costs, and storage, as well as cultural attitudes
imatinib in chronic myeloid leukemia (CML) and rituximab in to end-of-life issues. Radiation machines should be procured
B-cell lymphomas have been considered among the greatest and maintained in LMICs at reasonable prices without
breakthroughs in cancer care in the past 50 years. Unfor- sacrificing safety and efficacy, whereas surgical skills should
tunately, these and many other therapies that have signif- be enhanced by improving local facilities and training cancer
icantly improved outcomes in patients with cancer are not surgeons on site or at international centers.
available to everyone who needs them, especially in low- There are no easy solutions to drug costs because phar-
and middle-income countries (LMICs). The cost of new maceutical companies should make profits to be able to
anticancer medicines is increasing, with the average monthly develop new medicines, knowing that not all medicines
cost of some newly released molecules being over $12,000 undergoing study come to market and that antineoplastic
per month for oral kinase inhibitors and $150,000 for agents, especially biologic medicines, are difficult and costly
a course of monoclonal antibodies. These costs have long to produce. The current model of oncology drug develop-
been out of the range of most LMICs and are now becoming ment, distribution, and marketing must change before it is
excessive even for European and North American patients. too late. There must be collaboration between academia and
The treatment of cancer has always been costly and dif- individual pharmaceutical companies, as well as with reg-
ficult to access, be it surgery, radiation therapy, systemic ulatory authorities and governments, to avoid the situation
medicines, or palliative care. Radiation machines are costly being experienced in renal cancer, where we now have more
and limited in access in LMICs, whereas chemotherapy drugs than 10 agents on the market from multiple pharmaceutical
have always been difficult to procure since they first became companies. Collaboration with regulatory authorities can

From the University of Jordan, Amman, Jordan; University of Witwatersrand Faculty of Health Sciences, Johannesburg, South Africa; Abramson Cancer Center, University of
Pennsylvania, Philadelphia, PA.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Paul Ruff, MBBCh, MMed, FCP(SA), University of Witwatersrand Faculty of Health Sciences, 7 York Rd., Johannesburg, 2193, South Africa; email: pruff@
iafrica.com.

2016 by American Society of Clinical Oncology.

58 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


CULTURAL AND REGULATORY BARRIERS TO TREATING CANCER PAIN

reduce research and development costs by streamlining the populations.2 For cancer, expected revenues in LMICs may
regulatory burden on clinical research and expediting reg- not justify investment because the target populations
istration of medicines with significant benefits over those average income is low. This lack of incentive compelling
with limited benefits. Development of medicines with regulation to register medicines results in excessive delays
limited benefits should be halted as early as possible. in access to newer medicines.
Evergreening (enhanced intellectual property protection) Once an application for a new medicine is filed, the
must also be prevented; extended patents are detrimental complex regulatory approval process starts to ensure
to underfunded patients in both high-income countries and availability of high-quality, safe, and effective medicines.3
LMICs. International collaboration in the development of Although some countries can assess a new drug themselves,
good quality generics and biosimilars will help make these based on the scientific dossier provided by the manufacturer
molecules available more rapidly to patients around the (usually high-income countries), middle-income countries
world. Development of EMLs, both in individual countries with varying levels of development and drug regulatory
and internationally by WHO, will make the use of cancer capabilities, or low-income countries with very limited or no
medicines more cost effective and less wasteful. drug regulatory capability cannot undertake full assess-
Unless we all work together to find a solution, cancer care ments of new pharmaceutical products. LMICs depend to
will only be available to the very wealthy. Indeed, we are differing extents on assessment made by a foreign drug
seeing the development of more and more exciting treat- regulatory authority, for example the U.S. Food and Drug
ments for fewer and fewer people.1 Administration (FDA) or European Medicines Agency (EMA).
Drug approval by one of these major authorities provides
DRUG ACCESS AND APPROVAL IN LOW- AND a valid basis for marketing a product in these LMICs. In an
MIDDLE-INCOME COUNTRIES attempt to facilitate drug registration in LMICs, WHO has
Cancer is rapidly becoming a major health care problem, developed a certification scheme to provide quality assur-
especially in LMICs. The success of antineoplastic medicines ance for imported medicines by means of standard forms
to cure patients and alleviate their suffering has been achieved at confirming the registration of the product in the country of
a dramatic cost. Therefore, it has become mandatory for health manufacture (Certificate of Pharmaceutical Product [CPP])
care authorities to cooperate to use resources efficiently to and approval of good manufacturing practice conditions
improve health care delivery to patients with cancer worldwide. based on inspections and quality analysis of the product.4,5
The initial step in the process of access to antineoplastic However, even after 2 decades of existence, the WHO
medicines is a pharmaceutical companys application for certification scheme is not widely used. Regulatory pro-
approval by the responsible local health regulatory au- cedures still vary significantly among nations, with certificate
thorities. The expected revenues of a medicine depend on format, conditions, and wording varying from one country to
the volume of drug sold and the average income of targeted another.6 Each country requires different documentation,
but almost all require at least one CPP from the manufacturing
site, the packaging and release site, or both. The process for
registering medicines remains complex and burdened with
KEY POINTS inefficiencies, duplications, delay, and, in some instances,
corruption.7,8 For instance, the manufacturing process may
The majority of patients with cancer reside in LMICs with take place in several countries, complicating the process of
limited access to essential medicines and services, obtaining a satisfactory CPP for the regulatory authorities in
resulting in a disproportionate increase in cancer
importing countries. This will potentially delay regulatory ap-
mortality.
Obstacles to cancer care are numerous and include
proval if the regulatory authority requests different CPPs than
costs, regulatory and cultural barriers, and limited what was already provided. Some products are manufac-
availability of health care practitioners. tured and approved in a certain country for export only,
The financial challenge for access to care is multi-tiered leading to concern that the competent authorities in the
and requires collaboration between all stakeholders manufacturing country are less stringent with regard to
including the pharmaceutical industry, national and products not to be used in their own territory. It is logical for
local health authorities, nongovernmental some importing countrys regulatory authority to request
organizations, and the World Health Organization. more reassuring documentation (e.g., formal declaration
Regulatory, financial, and cultural barriers limit access to signed by the manufacturers responsible good manufacturing
chemotherapy agents, supportive care medicines, practice person in addition to the Export CPP), to ensure
radiation therapy, and complementary and alternative
follow-up on complaints from the importing countrys au-
medicines required for good cancer pain management in
LMICs.
thority regarding product defects or inaccuracy of the dec-
Collaborative partnerships and regulatory larations contents. Still, the regulatory authority could refuse
harmonization can overcome delays in treatment access to register that product.9,10 To further illustrate the complexity
and approval and facilitate the development of quality of implementing the WHO certification scheme, assess-
cancer care delivery. ment of quality of active pharmaceutical ingredients was
also recommended by the International Conference of

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 59


RUFF ET AL

Drug Regulatory Authorities.7,11 Such recommendations relevant to medicines, in addition to long-term plans to control
added to the confusion and complexity of drug approval the price increase as a result of that commitment. Compulsory
processes in LMICs, especially because many do not yet licensing allowing generic versions of medications to be
have the regulatory capacity required to fully implement produced despite the existence of a patent to provide med-
the WHO certification scheme, let alone comply with icines of need in a society at affordable prices, is a plausible
those recommendations.12 policy already adopted in India, South Africa, and Thailand.22
After a medicines approval by EMA or FDA, it takes a year Direct price control measures could help reduce the price
or more to register that medicine in LMICs. Some countries by an average of 20%.23 Control measures could be price
adopt fast-track for the registration of priority medicines setting included in registration and procurement through
fulfilling an unmet medical need.13 Even in North America, it a central government agency. Because most of antineo-
was estimated that more than 250,000 life-years are lost per plastic medicines are not procured centrally, they are only
year of delay in access to drugs that were shown to prolong available in the private sector for out-of-pocket purchase at
overall survival in phase III clinical trials.14 Even more life- much higher prices.7 If price is adjusted for affordability,
years are lost if we consider the possible estimate in LMICs LMICs pay a much higher price compared with high-income
where the delay is more of a challenge and the disease countries.24 The prices set for the recently developed bi-
burden is escalating. Regulatory harmonization (i.e., either ologics are prohibitive not only for out-of-pocket purchase
allowing for one centralized approval for drug registration in but also for central procurement, creating the case for
multiple countries, or mutual recognition once a drug has differential international pricing. Concerns related to this
been registered in certain countries) is the key to using approach (e.g., parallel trading and use in reference pricing)
resources more efficiently and especially to speeding up the do seem legitimate, but the experience with HIV treatment
process to facilitate access to patients.7 does not support those concerns.22 However, this differ-
Once a medicine is registered, access varies from one ential pricing is beneficial if the different pricing levels in-
country to another. Rarely, registration entails availability deed reflect the ability of the target population to pay. This is
for patients treated in public health care systems. In some amplified by The World Banks country income classifica-
countries, even if the medicine were to be available, it takes tion; although it was designed for World Bank lending
an additional 1 to 2 years to obtain.13 In most countries, decisions, its improper adoption to inform health-related
medicines will only be available for those who can pay out of decisions increases the cost of medicines. It does not reflect
pocket, creating a financial access obstacle, probably the health system capacity of governments to invest more in
most challenging in the access process. In many LMICs, as health, and it does not take into account income inequality
much as 90% of the population purchase medicines on an within a nation. Many countries have graduated to become
out-of-pocket basis,15,16 with spending often dispropor- upper-middle income resulting from statistical recalcula-
tionate to personal or family income. The recently updated tions, although 15% to 55% live at or below national poverty
WHO EML for cancer could potentially serve as a valuable lines.25 This graduation will further complicate drug pricing
asset for advocacy to influence the local health authorities in policies and availability of expanded drug access programs
LMICs to provide those medicines free of charge or make for patients. Regulatory harmonization is the key to over-
them available for patients at affordable cost.17 come delays in the approval process and efficiently use
The financial access challenge creates another serious resources. The financial challenge for access is multitiered
concern of drug quality. Many reports have described pa- and requires collaboration between key stakeholders
tients receiving substandard or counterfeit drugs.18,19 This is including pharmaceutical industry, local national health
mainly the result of importing cheaper, poorly manufactured authorities, WHO, and other nonprofit, patient-oriented
rogue generics. Although some governments have begun organizations.
to promote the development of generic drug manufacturing
capacity within their own bordersas in India, Brazil, South
Africa, and, recently, Jordanmany countries rely solely on CULTURAL AND REGULATORY BARRIERS TO
importing drugs. In its effort to combat this challenge faced PAIN MANAGEMENT INCLUDING ANTICANCER
in every field of medicine, WHO developed a prequalification DRUGS, RADIOTHERAPY, AND SUPPORTIVE
program for pharmaceuticals and active pharmaceutical MEDICINES IN LOW- AND MIDDLE-INCOME
ingredients,20 initially developed to help international COUNTRIES
procurement agencies, but lately used by LMICs to guide More than half of all patients with cancer and more than
bulk purchase of medicines. Drugs passing quality control three-quarters of patients with advanced disease will suffer
appear on the WHO prequalification website; however, from pain at some time in the course of the illness. The pain is
antineoplastic medicines are not there yet. Conversely, usually moderate to severe, affecting the patients quality of
delayed market entry of generics because of evergreening in life, and it can become chronic in long-term cancer survivors.26
countries that joined the World Trade Organization further The management of cancer pain is a multidisciplinary
increases cost.21 Such countries should consider increasing process that has many barriers confronting patients and
spending on public health to offset the adverse impact on their families, as well as health care professionals. There are
patients of strengthening its intellectual property protection many difficult aspects to the management of cancer pain in

60 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


CULTURAL AND REGULATORY BARRIERS TO TREATING CANCER PAIN

both high-income countries and LMICs developing. Despite


the high prevalence of cancer pain, it is often poorly treated SIDEBAR. Medicines Used for Management of
for several reasons. To manage cancer pain effectively, it is Cancer Pain
important to assess the type of pain and its effect on quality
Disease-Modifying Agents
of life and to decide on a management plan using appropriate Chemotherapeutic agents
interventions, analgesics, and supportive care as required. Analgesics
Interventions can include disease-modifying therapies such as Paracetamol
chemotherapy, radiotherapy, or surgery. Analgesics include Nonsteroidal anti-inflammatory drugs
nonopioid and opioid drugs, as well as co-analgesic (adjuvant) Weak opioids: codeine, tramadol.
drugs for pain that is either not responsive or partially re- Strong opioids: morphine, fentanyl
sponsive to opioids. Interventions and analgesics have side Local anesthesia
effects, which should be treated with further medications. Co-analgesics (Adjuvants)
Many societies and cultures use traditional or comple- Tricyclic antidepressants
mentary and alternative medications and nondrug mea- Anticonvulsants
sures to control pain.
a2d ligands: gabapentin and pregabalin
Baclofen
Benzodiazepines
Other Essential Medications
ORTHODOX MEDICINES Laxatives
Cancer chemotherapy plays a critical role in reducing pain in Antiemetics
patients with cancer by shrinking and, in certain malig- Proton-pump inhibitors
nancies, curing the cancer. As mentioned before, access to Corticosteroids
cancer chemotherapy has always been limited by costs and Bisphosphonates and RANK-ligand inhibitors
by slow regulatory approval in many LMICs. In South Africa,
approval of a new chemical entity or generic medicine may
take as many as 3 years unless a fast-track status is obtained,
owing to an unmet medical need or public health require- of bone pain. Unfortunately, their availability is also limited
ment, which will reduce the time to about 18 months. by slow regulatory approvals and high costs (Sidebar).
Dossiers for registration have to go through three to four
committees, including the Naming and Scheduling Commit-
tee, the Pharmaceutical and Analytic Committee, the Central RADIATION THERAPY FACILITIES: LINEAR
Clinical Committee, and, in some cases, the Biologic Com- ACCELERATOR VERSUS COBALT 60
mittee, all of which are understaffed. Many doctors in LMICs Linear accelerator access is limited by high up-front and
are reluctant to refer patients for chemotherapy because of maintenance costs, with less effective but low maintenance
myths concerning the side effects, and patients themselves and cheaper cobalt 60 being supported in some LMICs by the
have similar views often perpetuated by the media. International Atomic Energy Agency. In addition, cultural
Opioid analgesics are the mainstay of cancer pain con- beliefs and use of traditional medicines likely affect the ac-
trol but are frequently inadequately used because of ceptability of radiation therapy for patients in LMICs. A study
doctors and patients fears concerning addiction and abuse. in San Francisco found that Chinese and other Asian women
Longer-acting opioids including slow-release morphine and received less radiation or other adjuvant treatment of breast
fentanyl patches are limited by slow regulatory approval cancer than Japanese or white women, most likely because of
of generics and high costs of the originator. Opiophobia cultural beliefs about disease and death, body image, med-
is a significant barrier to pain control with opioid analge- ical decision making, and use of alternative medicines.31
sics in LMICs. 27 Patient-related factors include fear of
psychological dependence and stigma related to opioid TRADITIONAL OR COMPLEMENTARY AND
use, especially where opioids are associated with crim- ALTERNATIVE MEDICINES
inal activity and gang violence.28,29 Health professional The use of traditional or complementary and alternative medi-
related opiophobia, including beliefs that opioids cause cines (TCAMs) varies across the world from 7% to 64%.32 In LMICs,
addiction, tolerance, or difficult-to-control side effects, is TCAMs are a natural element of traditional health practice.
also a significant barrier to adequate pain treatment with TCAMs are categorized by the National Centre for Comple-
opioids.30 mentary and Alternative Medicine in the United States (Table 1).33
Tricyclics, anticonvulsants, and particularly a2d ligands There is limited evidence for the effectiveness of TCAMS in
gabapentin and pregabalin play an important role in the treating cancer pain. Differing responses to multidisciplinary
treatment of neuropathic pain but are often not available in pain programs have been noted where the mood symptoms
LMICs because of slow regulation of generics and high costs improved in all patient groups but improvement in pain
of originators. was culturally determined.34 The effectiveness of TCAMs in
Osteoclast inhibitors including bisphosphonates and RANK- managing cancer pain is dependent on cultural beliefs and
ligand inhibitors play an important role in the management expectations. However, reliance on TCAMs and resistance to

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 61


RUFF ET AL

TABLE 1. Categories of Traditional or Complementary cultural barriers to pain management: (1) differences in
and Alternative Medicines perception of pain, health beliefs, and the meaning of pain
with normalizing of the cancer experience; (2) reluctance to
Category Example report pain because of negative stigma associated with
Alternative medical systems Traditional Chinese, Ayurvedic, cancer progression and fatalism, and a belief in accepting
and African traditional pain with stoicism; and (3) different coping practices with
medicine
belief in traditional remedies, faith healing, prayer, or
Mind-body (spirit) Meditation, prayer, faith healing, positive thinking.
interventions support groups, music therapy,
and hypnosis There appear to be differences in the experience of
Biologically based therapies Herbal medicines, dietary physical pain, in the reporting of pain, and in the emotional
supplements, vitamins response to pain, which may reflect differences in the
Manipulation and TENS, acupuncture massage, meaning of pain in different cultures. There are cultural
body-based therapies chiropractic, osteopathy, taboos and fears that limit access to pain-relieving medi-
and reflexology
cations, but there are also traditional practices that are used
Energy therapies Qi Gong, Reiki, and magnetic field to manage pain. Although it is important to ensure that pain
therapy
medications and modalities to treat pain are accessible to all,
it is important to consider different cultural attitudes toward
illness and pain and its management.
conventional interventions and opioids may exacerbate pain
in LMICs.
WORLD HEALTH ORGANIZATION LIST OF
ESSENTIAL MEDICINES AND DEVICES:
CANNABINOIDS MAXIMIZING CANCER CARE VALUE AND
Historically, the use of cannabis as medicine dates back to SUPPORTING THE DEVELOPMENT OF CANCER
before the Christian era in Asia and spread to the Middle East DELIVERY PROGRAMS
in the 10th century, to Africa in the 15th century, to South Cancer is a group of very disparate malignant diseases, with
America in the 16th century, and to Europe and the United the diagnostics and treatment varying widely among them.
States in the 19th century.35 A meta-analysis of several In addition, many patients require sophisticated surgery,
controlled clinical trials supports the use of cannabinoids radiation, and systemic therapies. High-quality pathology is
(dronabinol and nabilone) for chemotherapy-induced nau- required to make accurate diagnoses in almost all patients,
sea and vomiting, but not for pain. There is no good evidence and imaging is frequently necessary for cancer staging and
supporting the use of inhaled or oral extracts of cannabis for assessment of treatment response and follow-up. There are
any cancer-related side effects.36 Country- and state-specific many essential cancer medications with different cancers
barriers to legalizing cannabis for symptom control exist requiring different noninterchangeable agents. Many
largely because of lack of adequate controls and concerns countries and ministries of health face daunting obstacles in
about abuse. establishing functional and quality services in all of these
areas. Toward this end, the WHO has several processes
CULTURAL BARRIERS TO CANCER PAIN aimed at aiding countries in the establishment and en-
MANAGEMENT IN LOW- AND MIDDLE-INCOME hancement of cancer services.
COUNTRIES
Many studies on cultural barriers, reported in English, are The Essential Medicines List
largely associated with pain management in Western In 1977 WHO published its first EML addressing medicines
countries with cultural minorities, usually of ethnic groups felt to be critical to treating many diseases, including cancer.
originating from LMICs. These minority groups are often In the intervening years, additional medicines were evalu-
marginalized and from lower socioeconomic levels, which ated and added to the EML. In the case of cancer medicines,
may confound the results because these groups often do not agents were added one at a time, without clear instruction
have equitable access to health care. In these countries, about where the benefit for the medication existed. In 2012,
language barriers may also play a role in inaccurate pain the Union International for Cancer Control and the Dana-
assessment. It has been shown that nurses who share the Farber Cancer Institute filed applications with WHO to add
same language as Arabic patients when assessing pain trastuzumab and imatinib to the EML for cancer. At that
usually assigned similar ratings to the patients compared time, there were no targeted therapies or on-patent drugs
with non-Arabicspeaking nurses.37 However, with this in on the EML for cancer. The applications argued that these
mind, these studies may provide a proxy for studying cultural two medicines dramatically alter the outcomes and survival
aspects of cancer pain management in LMICs. rates for patients with HER2-positive breast cancer and CML,
Two reviews of ethnic and cultural differences in pain and respectively, and that there were no less-costly alternatives
pain management by Kwok and Bhuvanakrishna38 and by having similar benefits. WHO deferred judgment and asked
Campbell and Edwards34 reveal the following potential Union International for Cancer Control and Dana-Farber

62 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


CULTURAL AND REGULATORY BARRIERS TO TREATING CANCER PAIN

Cancer Institute to lead a comprehensive review of the it to 85%, and the addition of adjuvant chemotherapy might
cancer EML. add 5% more to the long-term remission rate, bringing it
In 2014, a process was undertaken to identify cancers to 90%.
where systemic therapies had significant impact. Cancers The disease-based documents contained specific infor-
were chosen if they were of high burden, if systemic ther- mation on dosing of each medication as well as schedule and
apies had at least some benefit (such as nonsmall cell lung duration of therapy. With this information, ministries of
cancer) and if they had lower disease burden (such as CML), health could estimate annual drug needs by knowing the
and if systemic therapies (e.g., imatinib) had a major impact number of patients with each disease and the dose of each
on survival. In all, 27 diseases were evaluated, with separate drug needed to treat a patient.
applications for early-stage and metastatic disease for breast Drafts of the 29 disease-based documents were reviewed by
cancer and colorectal cancer. Nearly 100 oncologists from all an expert committee at an in-person meeting at WHO in
continents were recruited to help in the process. For each November 2014, and finalized documents were submitted to
disease, a document was created that included: (1) an exec- WHO in December 2014. They were posted on the WHO
utive summary; (2) public health relevance; (3) requirements website for public comment in January 2015. In April 2015,
for diagnosis, treatment, and monitoring; (4) overview of a WHO Expert Committee reviewed all documents, and in May
regimens; (5) review of benefits and harms (including refer- 2015 made public their decisions. The documents had pro-
ences and systematic reviews); and (6) recommendations for vided support for the 30 existing medications on the EML and
additions to the EML. recommended the addition of 22 medicines. WHO approved
Each disease-based document was initially written by the 30 existing medications and added 16 new medicines to
a cancer expert or team of experts and reviewed and cri- the list, whereas six were rejected. Approved medications
tiqued by at least two other experts or teams. A central included, for the first time, patented, costly agents (i.e.,
committee then collated the work of all three groups to form trastuzumab and imatinib). The rejected medicines included
a consensus-based document. The central committee added two second-generation tyrosine kinase inhibitors for CML
the section on public health relevance and references fo- (insufficient data to support them as more essential than
cusing on important phase III studies and systematic re- imatinib), two EGFR antagonists for the treatment of non
views, and in some cases costing information. Importantly, small cell lung cancer (molecular testing frequently unavail-
each document contained a section on diagnostics and able with modest incremental benefit), arsenic trioxide for the
specific needs for treatment and monitoring of patients. treatment of acute promyelocytic leukemia (insufficient data
The documents were designed to provide critical in- to support it as an essential medication), and diethylstilbestrol
formation to governments and ministries to understand for the treatment of metastatic prostate cancer (mostly un-
better what was needed to treat the particular disease available and toxic).39
and make administration of the listed medications safe The final documents and the revised EML for cancer are
and effective. now available on the WHO website. Each medicine on the
The approach in each document was regimen-based, EML can be referenced back to a disease-based document
rather than based on individual medications. For some supporting and delineating its contribution to treatment of
diseases, such as CML, the regimen was a single drug; in this the disease, something not previously available on the EML.
case imatinib, but for many diseases the regimen consisted Planning is underway to develop a mechanism for periodic
of multiple drugs such as doxorubicin, bleomycin, vinblas- reviews of the EML for cancer as new scientific data become
tine, and dacarbazine (ABVD) for Hodgkin lymphoma. In available.
addition, the incremental benefit of the medicines was
quantitated. As an example for diffuse large B-cell lym-
phoma, where surgery is required for biopsy and diagnosis ESSENTIAL MEDICAL DEVICES FOR CANCER
but does not add at all to remission or cure rates, the In April 2015, WHO convened a group of experts with the
medicines alone accounted for all benefits. In this case, the goal of detailing needs for services critical to the diagnosis,
administration of cyclophosphamide, doxorubicin, vincris- evaluation, and treatment of patients with cancer. Based on
tine, and prednisone (CHOP) would result in a 55% long-term the advice of this group, a steering committee of experts was
remission rate, whereas the addition of rituximab to this formed and met at WHO in September 2015 to better outline
regimen (R-CHOP) would provide an incremental benefit of a plan to accomplish the work. Committees were formed to
15%, bringing the long-term remission rate to 70%. In the delineate needs for pathology and laboratory services, ra-
case of early-stage breast cancer, where surgery is required diology services, surgical services, administration of systemic
and will result in cure for many patients, the medicines therapies, and radiation therapy. Committee members
would have incremental benefit above surgery. In a hypo- were recommended by the steering committee and met
thetical patient with node-negative, estrogen receptor via teleconference throughout 2015.
positive, HER2-negative breast cancer, surgery alone might This work is also disease-based, using a subset of cancers
result in a long-term remission rate of 70%. The addition of to complete lists of essential medical devices for each of the
hormone therapy such as tamoxifen might give 15% in- categories listed before. The work is still underway, with
cremental benefit to the long-term remission rate, bringing plans to complete the lists in the first half of 2016.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 63


RUFF ET AL

THE ROLE OF WHO IN DEVELOPMENT OF raised for the Global Fund to Fight AIDS, Tuberculosis, and
CANCER TREATMENT PROGRAMS Malaria.
WHO exists to serve its member states in the arena of health All of this work aims at bringing affordable and high-quality
care. In many LMICs, ministries of health struggle with health cancer care to those who currently have little or no access.
care priorities as advances in control of infectious diseases Patients who have treatable cancers with a high likelihood
and other important areas such as maternal-child health, for cure in high-income countries are, in many locations,
noncommunicable diseases become increasing health bur- dying without treatment or with inadequate and poor-
dens. In acknowledgment of this, the United Nations con- quality treatment. We should work together to erase this
vened a high-level meeting in September 2011 to address social and medical inequity.
the needs of noncommunicable diseases. The work of WHO
flows from the recommendations of this meeting and follow- CONCLUSION
up meetings. Improving patient outcomes is not only achieved by the
success of medicines or procedures in large phase III
THE ROLE OF COUNTRIES AND MINISTRIES clinical trials. It is achieved when patients worldwide have
WHO is providing guidance to countries and ministries on ready access to those successful interventions and the
essential medicines and devices for the diagnosis and infrastructure and human capacity to use those inter-
treatment of cancer. It remains, though, for individual ventions safely and effectively. The challenges to such
countries to develop cancer plans specific to their needs and access are many, with regulatory, financial, and cultural
environment. In some LMICs, this is well underway, and in barriers among the most important barriers to overcome.
others, much remains to be done. Many countries have little Success can only be accomplished by constant collabo-
or no in-country cancer care expertisefew, if any, pa- ration between key stakeholders, including the phar-
thologists, medical oncologists, radiation oncologists, and maceutical industry, local and national health authorities,
skilled nurses. Many countries have very limited resources the WHO, and other nonprofit, patient-oriented orga-
and must prioritize their needs as best they can. Partnerships nizations. The oncology community in high-income
between cancer specialists from high-income countries and countries should have a humanitarian obligation to ac-
LMICs can help. External funding would help immensely to company those in LMICs to achieve these goals to the
move the global cancer agenda forward, funding such as was betterment of all.

References
1. Ruff P. Achieving treatment for all. ASCO Connection: The Best of ASCO 9. Ministry of Public Health. Guidelines on Good Storage and Distribution
Connection Commentary 2015; Special Edition 2014-2015: 35. Practices of Pharmaceutical Products in Lebanon. http://www.moph.
2. Villa S, Compagni A, Reich MR. Orphan drug legislation: lessons for gov.lb/Drugs/Documents/Guidelines-GSDPLebanon%28English%29.pdf.
neglected tropical diseases. Int J Health Plann Manage. 2009;24:27-42. Accessed January 10, 2016.
3. World Health Organization. Policy Perspectives on Medicine. http:// 10. World Health Organization. Proceedings of the Ninth International
whqlibdoc.who.int/hq/2003/WHO_EDM_2003.2.pdf. Accessed Janu- Conference of Drug Regulatory Authorities (ICDRA) - Berlin, Germany
ary 5, 2016. 25-April 29, 1999. http://apps.who.int/medicinedocs/en/d/Js4925e/.
4. World Health Organization. Model Certificate of a Pharmaceutical Accessed January 10, 2016.
Product. http://www.who.int/medicines/areas/quality_safety/ 11. World Health Organization. 15th International Conference of Drug
regulation_legislation/certification/modelcertificate/en/. Accessed Regulatory Authorities (ICDRA) - Tallinn, Estonia, 23-October 26, 2012,
January 5, 2016. http://apps.who.int/medicinedocs/documents/s20142en/s20142en.
5. World Health Organization. Certification scheme on the quality of pdf. Accessed January 10, 2016.
pharmaceutical products moving in international commerce. http:// 12. Doua JY, Van Geertruyden JP. Registering medicines for low-income
www.who.int/medicines/areas/quality_safety/regulation_legislation/ countries: How suitable are the stringent review procedures of the World
certification/en/. Accessed January 5, 2016. Health Organisation, the US Food and Drug Administration and the
6. World Health Organization. Guidelines on the implementation of the European Medicines Agency? Tropic Med Int Health. 2014;19:23-36.
WHO certification scheme on the quality of pharmaceutical products 13. Registration Requirements of the Pharmaceutical Products with Jordan
moving in international commerce. http://www.who.int/medicines/areas/ FDA. http://www.nobles.com.jo/PharmaceuticalProducts/FormNo2-
quality_safety/regulation_legislation/certification/guidelines/en/index1. 1Product/reg_req_of_the_pharmacutical_products_2-1.pdf. Accessed
html. Accessed January 5, 2016. January 8, 2016.
7. World Health Organization. The World Medicines Situation 2011. 14. Stewart DJ, Stewart AA, Wheatley-Price P, et al. Impact of time to drug
http://apps.who.int/medicinedocs/en/m/abstract/Js20054en/. approval on potential years of life lost: The compelling need for im-
Accessed January 5, 2016. proved trial and regulatory efficiency. 16th World Conference on Lung
8. World Health Organization. Measuring transparency in medicines reg- Cancer. Presented September 7, 2015.
istration, selection and procurement: four country assessment stud- 15. Cameron A, Ewen M, Ross-Degnan D, et al. Medicine prices, availability,
ies. http://www.who.int/medicines/areas/policy/goodgovernance/ and affordability in 36 developing and middle-income countries:
Transparency4CountryStudy.pdf. Accessed January 6, 2016. a secondary analysis. Lancet. 2009;373:240-249.

64 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


CULTURAL AND REGULATORY BARRIERS TO TREATING CANCER PAIN

16. ODonnell O, van Doorslaer E, Rannan-Eliya RP, et al. Who pays for 28. Bagivan G, Tosun N, Komurcu S, et al. Analysis of patient-related
health care in Asia? J Health Econ. 2008;27:460-475. barriers in cancer pain management in Turkish patients. J Pain Symptom
17. World Health Organization. WHO Model List of Essential Medicines. Manage. 2009;38:727-737.
2015; http://www.who.int/entity/medicines/publications/essential- 29. Kim YC, Ahn JS, Calimag MM, et al; ACHEON Working Group. Current
medicines/en/index.html. Accessed January 15, 2016. practices in cancer pain management in Asia: a survey of patients and
18. Caudron JM, Ford N, Henkens M, et al. Substandard medicines in physicians across 10 countries. Cancer Med. 2015;4:1196-1204.
resource-poor settings: a problem that can no longer be ignored. Trop 30. Peker L, Celebi N, Canbay O, et al. Doctors opinions, knowledge and
Med Int Health. 2008;13:1062-1072. attitudes towards cancer pain management in a university hospital.
19. Gautam CS, Utreja A, Singal GL. Spurious and counterfeit drugs: Agri. 2008;20:20-30.
a growing industry in the developing world. Postgrad Med J. 2009;85: 31. Prehn AW, Topol B, Stewart S, et al. Differences in treatment patterns
251-256. for localized breast carcinoma among Asian/Pacific islander women.
20. World Health Organization. Prequalification Programme. A United Cancer. 2002;95:2268-2275.
Nations Programme Managed by WHO. http://apps.who.int/prequal/. 32. Cassileth BR. Complementary therapies: overview and state of the art.
Accessed January 4, 2016. Cancer Nurs. 1999;22:85-90.
21. Abbott RB, Bader R, Bajjali L, et al. The price of medicines in Jordan: 33. Molassiotis A, Fernadez-Ortega P, Pud D, et al. Use of complementary
the cost of trade-based intellectual property. J Generic Med. 2012;9: and alternative medicine in cancer patients: a European survey. Ann
75-85. Oncol. 2005;16:655-663.
22. Hoen Et, Law L-M. Access to cancer treatment. http://apps.who.int/ 34. Campbell CM, Edwards RR. Ethnic differences in pain and pain man-
medicinedocs/documents/s21758en/s21758en.pdf. Accessed March 2, agement. Pain Manag. 2012;2:219-230.
2016. 35. Zuardi AW. History of cannabis as a medicine: a review. Rev Bras
23. Schut FT, Van Bergeijk PA. International price discrimination: the Psiquiatr. 2006;28:153-157.
pharmaceutical industry. World Dev. 1986;14:1141-1150. 36. National Cancer Institute. Cannabis and CannabinoidsHealth Pro-
24. Machado M, OBrodovich R, Krahn M, et al. International drug price fessional Version http://www.cancer.gov/about-cancer/treatment/
comparisons: quality assessment. Rev Panam Salud Publica. 2011;29:46-51. cam/hp/cannabis-pdq. Accessed January 23, 2016.
25. The World Bank. Data. http://data.worldbank.org/country. Accessed 37. Al-Atiyyat N. Cultural diversity and cancer pain. J Hosp Palliat Nurs.
January 10, 2016. 2009;11:154-164.
26. Blanchard C, Chetty S, Ganca L, et al. Guide to the treatment of cancer 38. Kwok W, Bhuvanakrishna T. The relationship between ethnicity and the
pain in South Africa. http://dx.doi.org/10.6084/m9.figshare.1612170. pain experience of cancer patients: a systematic review. Indian J Palliat
Accessed March 2, 2016. Care. 2014;20:194-200.
27. Krakauer EL, Wenk R, Buitrago R, et al. Opioid inaccessibility and its 39. Shulman LN, Wagner CM, Barr R, et al. Proposing essential medicines to
human consequences: reports from the field. J Pain Palliat Care treat cancer: Methodologies, processes, and outcomes. J Clin Oncol.
Pharmacother. 2010;24:239-243. 2016;34:69-75.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 65


CARE DELIVERY AND PRACTICE MANAGEMENT

Integrating Patient-Reported
Outcomes Into Real-Life Medical
Decisions

CHAIR
Paul G. Kluetz, MD
U.S. Food and Drug Administration
Kensington, MD

SPEAKERS
Diana T. Chingos, MS, MFA
Patient Advocate
Studio City, CA

Ethan M. Basch, MD, MSc


The University of North Carolina at Chapel Hill
Chapel Hill, NC

Sandra A. Mitchell, PhD, CRNP, FAAN


National Cancer Institute
Rockville, MD
MEASURING SYMPTOMATIC ADVERSE EVENTS WITH PRO-CTCAE

Patient-Reported Outcomes in Cancer Clinical Trials:


Measuring Symptomatic Adverse Events With the National
Cancer Institutes Patient-Reported Outcomes Version of
the Common Terminology Criteria for Adverse Events
(PRO-CTCAE)
Paul G. Kluetz, MD, Diana T. Chingos, MS, MFA, Ethan M. Basch, MD, MSc, and
Sandra A. Mitchell, PhD, CRNP, FAAN

OVERVIEW

Systematic capture of the patient perspective can inform the development of new cancer therapies. Patient-reported outcomes
(PROs) are commonly included in cancer clinical trials; however, there is heterogeneity in the constructs, measures, and analytic
approaches that have been used making these endpoints challenging to interpret. There is renewed effort to identify rigorous
methods to obtain high-quality and informative PRO data from cancer clinical trials. In this setting, PROs are used to address specific
research objectives, and an important objective that spans the product development life cycle is the assessment of safety and
tolerability. The U.S. Food and Drug Administrations (FDA) Office of Hematology and Oncology Products (OHOP) has identified
symptomatic adverse events (AEs) as a central PRO concept, and a systematic assessment of patient-reported symptomatic AEs can
provide data to complement clinician reporting. The National Cancer Institutes Patient-Reported Outcomes version of the Common
Terminology Criteria for Adverse Events (PRO-CTCAE) is being evaluated by multiple stakeholders, including the FDA, and is
considered a promising tool to provide a standard yet flexible method to assess symptomatic AEs from the patient perspective. In this
article, we briefly review the FDA OHOPs perspective on PROs in cancer trials submitted to the FDA and focus on the assessment of
symptomatic AEs using PRO-CTCAE. We conclude by discussing further work that must be done to broaden the use of PRO-CTCAE as a
method to provide patient-centered data that can complement existing safety and tolerability assessments across cancer clinical trials.

T he intent of this educational manuscript is to discuss the im-


portance of PRO assessments in cancer trials, identify strengths
and limitations of currently used PRO strategies, and focus on
extensive work as an advisor to cancer studies brings a unique
combination of patient focus and understanding of the com-
plexities of clinical trial design and conduct.
the potential utility of a rigorous and systematic assessment of
symptomatic AEs as a component of a broader PRO strategy.
As part of this effort, we have been fortunate to have a patient THE PATIENT PERSPECTIVE ON PATIENT-
advocate included to introduce the manuscript by providing her REPORTED OUTCOMES IN CANCER TRIALS
personal perspective on the inclusion of PROs in cancer trials. Patients, and human beings more generally, are accustomed
Diana Chingos is a 20-year survivor of early onset breast cancer to providing our views in many aspects of our lives. Until
whose advocacy work extends to membership on the National recently, the act of seeking out feedback from those who
Cancer Institutes (NCI) Investigational Drugs Steering Com- use the health care system was rare. One can argue that the
mittee and the National Clinical Trials Networks Core Correl- health care user experience should reign supreme over all
ative Sciences Committee, as well as participation on a data and other contexts, when the quality and quantity of our lives is
safety monitoring board for the California Cancer Consortium at stake, sometimes at great financial and logistical expense.
(a phase I/II clinical trials group) and an institutional review Gratefully, at least from this patients view, PRO ques-
board. Her experience as a patient and caregiver coupled with tionnaires have been developed to bring our perspective

From the Office of Hematology and Oncology Products, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD; Independent Cancer Patient
Advocate, Los Angeles, CA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC; Outcomes Research Branch, National Cancer Institute, Rockville, MD.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Sandra Mitchell, PhD, CRNP, FAAN, Division of Cancer Control and Population Sciences, Outcomes Research Branch, National Cancer Institute, 9609 Medical
Center Blvd., Rockville, MD 20850; email: mitchlls@mail.nih.gov.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 67


KLUETZ ET AL

into the research setting more systematically. In my opinion, I welcome the ability to report the side effects I have
soliciting patient reports espouses equity and autonomy, experienced, and I encourage others to do so. However, it is
enabling patients to speak for themselves, without the filter important to mention that although most patients want to
of health care providers. Capturing a patients self-report describe their experiences, it can become tedious, depending
using well-validated measures offers a direct indicator of on the time required. Survey fatigue is real and is magnified by
change in symptoms, function, or well-being during treat- medication-related fatigue, cancer-related fatigue, and the
ment, providing additional information to supplement the unfortunate synergy of physical, emotional, psychologic, and
clinicians evaluation of tumor response and toxicity. environmental strains involved with being a patient with cancer
Many patients with cancer are willing to play their part in undergoing therapy. Given this, it is important to consider the
research through answering questionnaires. Unless patients length of the survey, the relevance of the questions, and the
are very ill and/or have cognitive deficits, many patients with time points for assessment, while trying to reduce the re-
cancer like the opportunity to contribute to research, es- dundancy of questions.
pecially meaningful research that can improve care for Some patients need encouragement and validation to
patients in the future. It can be difficult for some patients to participate in this process, that their voice is valued and
assess the value of specific research studies, but everyone integral to the clinical trial. Patient engagement helps the
understands treatment toxicity and side effects. You live it systemit does not hinder it. Empowering patients to ar-
and usually have something to say about it. ticulate their experiences can provide value to those who
From the informed patients perspective, the patients develop cancer therapies by more accurately charac-
voice has been a key missing element in the current system terizing a drugs effect on the patient and also may enhance
of drug safety assessment. Assessing patient-reported research participation, a necessity if we are to speed up the
symptomatic AEs can fill this need. Published studies have rate of knowledge generation.
demonstrated discordance between physician and patient
reports, with underreporting of patients symptoms and
their severity being common.1,2 Something is getting lost Past Efforts at Collecting Patient-Reported Outcomes
in the translation. PRO measures provide an opportunity in Cancer Trials
for the patient to directly report side effects and their in- To date, the most common PRO strategy for oncology has
tensity from the perspective of the person experiencing it. been to assess the broad multidomain concept of health-
The PRO-CTCAE has generated considerable interest in the related quality of life (HRQOL), utilizing instruments largely
broader stakeholder community as a PRO tool that could be developed in a different therapeutic era.35 These existing
used across the therapeutic development process to address HRQOL measures have strengths, including translations
important questions related to the tolerability profile of a across multiple languages and a familiarity with their use
specific therapy. among the cancer therapeutic development community.
Many instruments have also been expanded to include
disease-specific modules in an effort to better capture
disease- and treatment-related symptoms.6,7 Substantial
KEY POINTS data have been accumulated using many of these measures,
and some offer the advantage of well-established cut scores,
There is a need to strengthen the rigor of PROs in cancer minimally important difference thresholds, and normative
trials. values that can aid in interpretation.
The FDAs Office of Hematology and Oncology Products
However, although the PRO measures commonly used in
has identified the systematic assessment of
symptomatic adverse events reported by patients as
oncology trials to date address a broad range of impor-
an opportunity to better describe the safety and tant and common symptoms and functional domains, they
tolerability of an investigational product across the typically include the same questions irrespective of disease
drug development life cycle. stage or the therapy under study. This can lead to questions
The NCIs PRO-CTCAE is a PRO measurement system that may be less relevant to the trial context and/or miss the
that includes a library of questions that measure assessment of important symptoms (e.g., toxicities not
symptomatic adverse events from the patient currently included in existing static instruments). This limi-
perspective. tation is becoming more evident in the current drug devel-
PRO-CTCAE holds promise as a rigorous and flexible opment era of molecularly targeted agents with wide-ranging
approach to the longitudinal assessment of side effect profiles. Investigators could benefit from a more
symptomatic adverse events in cancer clinical trials.
flexible toolbox of PRO measures that can adapt to differing
Challenges and knowledge gaps exist with respect to
trial designs, implementation, and interpretation of
disease and treatment contexts.
PRO-CTCAE. Effort is ongoing to identify the best From the FDA OHOP perspective, all PRO data will be taken
approaches to make PRO-CTCAE scores available, into consideration as part of the overall data package to
alongside CTCAE grading, in published reports and inform the benefits and risks of a therapy under review.
FDA drug labels. However, not all data reviewed in an application can be
included in the FDA drug label. The FDA is tasked with

68 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


MEASURING SYMPTOMATIC ADVERSE EVENTS WITH PRO-CTCAE

providing information in the product label that is useful to PRO strategy may be more appropriate and efficient in
prescribers in treating their patients and must ensure that early clinical development. This is an important distinction,
the information is easily interpreted, unbiased, and not as the classic phases of drug development are blurring, and
misleading. Broad concepts such as HRQOL are more precision medicine trials may have multiple study arms
challenging to define, with some domains such as social well- designed to simultaneously gauge antitumor activity11
being farther removed from a therapys direct effect on the with some single-arm cohorts potentially demonstrating
patient. Submitted HRQOL data can be further complicated notable antitumor activity suitable for accelerated ap-
by trial design limitations, missing data, and lack of pre- proval.12 Safety is an important trial objective in all phases
specified analyses. For this reason, PRO data have rarely of therapeutic product development. Inclusion of a PRO
been included in FDA labeling of cancer therapies. When measure of symptomatic AEs can improve our understanding
PRO data have been included in the FDA product label for of safety, tolerability, and dose selection and thus is applicable
cancer products, it has predominantly relied on well-defined to a broad range of clinical trial contexts.
measures of specific symptoms or functional measures that
relate directly to the disease under study.8
There continues to be wide variability in the type and THE USE OF PATIENT-REPORTED OUTCOMES TO
quality of PRO data acquired from cancer trials. This lack INFORM TREATMENT TOLERABILITY
of standardization includes heterogeneity in the PRO in- Safety assessment in cancer clinical trials has been stan-
struments used, as well as the assessment frequencies, and dardized through the use of the CTCAE.13 Currently, in
the approach to data analysis and reporting. High levels of version 4, the CTCAE provides a widely accepted lexicon
missing data have also been a common challenge that can and associated criteria for grading AEs of cancer treatment.
adversely affect the interpretability of PRO findings. Many CTCAE is routinely updated and used in conjunction with
stakeholders, including the FDA OHOP, have actively en- the Medical Dictionary for Regulatory Activities by regu-
gaged the oncology drug development community to latory agencies. Using CTCAE, AEs are graded on a scale
evaluate existing instruments and identify emerging op- from grade 1 to 5, in which, by convention, grade 5 is death,
portunities to improve the PRO strategy in cancer trials to and grade 4 reflects toxicities that are life-threatening and
provide more rigorous patient-centered data to all those warrant urgent intervention. Grades 13 represent pro-
who weigh the risks and benefits of cancer therapies.9 gressive worsening in severity or frequency of the toxicity,
interference with self-care and the performance of daily
activities, and the need for clinical intervention. Although
FOCUSING ANALYSES ON CORE this method of safety reporting provides for standardiza-
PATIENT-REPORTED OUTCOME CONCEPTS tion and efficiency in data collection and analysis, the
INCLUDING SYMPTOMATIC ADVERSE EVENTS assessment of AEs using CTCAE is elicited by health care
In most late-phase randomized clinical trials, concepts providers, including symptomatic AEs such as nausea or
such as HRQOL and social, emotional, and cognitive do- sensory neuropathy. It has been stated by the FDA and
mains are expected by many stakeholders to provide a others that the patient is best positioned to report his or
reasonably comprehensive picture of the patients ex- her own symptoms.14 Thus, the systematic assessment of
perience of their disease and treatment. These data will symptomatic AEs using a PRO provides additional in-
be reviewed by FDAs OHOP as important supportive data. formation that is complementary to existing safety as-
However, in an effort to increase the amount of patient- sessments reported by clinicians using the CTCAE.
centered data in FDA labeling, OHOP has recently pro- The assessment of symptomatic AEs may be of increasing
posed that our key PRO analyses focus on three core importance as we enter into a new therapeutic era in ma-
symptom and functional concepts (Fig. 1).10 Symptomatic lignant hematology and oncology. An expanding number of
AEs, physical function, and disease-related symptoms are mechanistic drug classifications have produced a more di-
considered by OHOP to be more well-defined and closer to verse range of potential toxicities.15 Many of the molecularly
the effect of a therapy on the patient and their disease. targeted agents are administered orally, often require
The careful collection and analysis of these core concepts prolonged treatment duration, and may produce less severe
can provide PRO data that may be more consistent with but more chronically bothersome side effects.16 A system-
FDA requirements for labeling. atic longitudinal assessment of relevant symptomatic AEs
It should be acknowledged that cancer trials are using a PRO measure may provide informative patient-
designed for differing purposes along the therapeutic centered data on symptomatic side effects that may oth-
development continuum from first in-human exploration erwise have been considered low grade by standard clinician
of dose and safety, to exploratory therapeutic trials, to report.17
trials designed to demonstrate substantial evidence of Existing HRQOL measures and their disease modules
safety, tolerability, and efficacy to support a regulatory evaluate a more limited range of side effects, many of
submission. Whereas a comprehensive PRO strategy may which were selected based on therapies that were used at
be expected to address the needs of multiple stakeholders the time they were developed (e.g., cytotoxic chemo-
in the later phases of therapeutic development, a focused therapies). Given the different therapeutic landscape

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 69


KLUETZ ET AL

FIGURE 1. U.S. Food and Drug Administration Core Concepts for Patient-Reported Outcomes Analysis in Cancer Trials

In an effort to increase the amount of informative patient-centered data in product labeling, FDA OHOP focuses its PRO analyses on the core concepts of symptomatic
AEs, physical function, and disease-related symptoms. Although these well-defined concepts are more in line with the regulatory framework of the FDA for labeling
considerations, all submitted PRO data will be taken into consideration as important supportive data. The three core concepts are not the only PRO measures to assess
in a trial to support drug approval, as broader domains and HRQOL remain important exploratory measures. Reprinted from Kluetz et al.10
Abbreviations: OHOP, Office of Hematology and Oncology Products; PRO, patient-reported outlets; AEs, adverse events; HRQOL, health-related quality of life.

today, this can lead to measurement of irrelevant symp- to provide a standard yet flexible tool to assess symp-
toms not considered part of the toxicity profile of the tomatic AEs.
newer drug and/or potentially miss the assessment of
important unique side effects of contemporary therapies.
Furthermore, the limited assessment frequency used to Development of the NCI PRO-CTCAE Measurement
date in many PRO corollary studies may not be optimal to System
adequately gauge tolerability. The PRO-CTCAE measurement system has been developed
Contemporary drug development requires a more flexible by NCI as a companion to the CTCAE. It was designed to
PRO approach to ensure an unbiased assessment of the most improve the validity, reliability, and precision with which
important symptomatic treatment side effects based on the symptomatic adverse effects of treatment are evaluated
anticipated toxicity profile of the therapies under study. in patients on cancer clinical trials. PRO-CTCAE was developed
Selection of symptomatic AEs from a large library of options by a multidisciplinary team of trialists, methodologists, cli-
would therefore be desirable. Recently, the NCI has de- nicians, informatics experts, patients, and regulators18 and
veloped and tested a measurement system to capture has been tested and refined in a consortium of aca-
symptomatic toxicities directly from patients.18 Com- demic and community-based cancer-treatment sites and
prised of both a library of 124 questions reflecting 78 in the NCI-sponsored clinical trials network (NCI contracts
symptomatic toxicities drawn from the CTCAE and an HHSN261200800043C and HHSN261200800063C) and by
electronic system for survey administration, reminders, more than 120 early adopters in 10 countries. The PRO-CTCAE
central monitoring, and alerts, NCI PRO-CTCAE is designed item library consists of 124 discrete items representing 78

70 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


MEASURING SYMPTOMATIC ADVERSE EVENTS WITH PRO-CTCAE

symptomatic AEs that are common in oncology clinical trials have generally been specified using an approach similar to
and included in the CTCAE. PRO-CTCAE items were created that used to define the AE surveillance plan for the trial
with substantial input from patients, clinicians, and PRO more broadly. That is, the study team reviews published
methodologists and underwent refinement through cognitive data, as well as data from earlier phase trials or animal
interviews with patients to establish content validity.19 Sub- models, if available, and incorporates information about
sequently, the quantitative measurement properties, in- the known or anticipated on- and off-target effects of
cluding validity, reliability, and responsiveness, were evaluated agents in a similar mechanistic class to identify those
in a large and diverse sample of patients receiving cancer symptomatic AEs likely to be associated with the regimens
treatment in six sites around the United States.20 in the trial.24 PRO-CTCAE items corresponding to these
Each of the 78 symptom terms included in the PRO-CTCAE symptomatic AEs are loaded into a software platform.
item library is assessed relative to one or more distinct Patients are trained to use the software and are asked to
attributes, including presence/absence, frequency, severity, self-report either from home on a regular basis or at clinic
and/or interference with usual or daily activities.18 Re- visits. After establishing a pretreatment baseline, more
sponses are provided on a five-point Likert scale. The generic frequent PRO-CTCAE administration is generally war-
PRO-CTCAE item structures for the frequency, severity, and ranted during the first few cycles of therapy (e.g., weekly
interference attributes are listed below: reporting during the initial several months of ther-
Frequency item: How OFTEN did you have __________?
(Never / Rarely / Occasionally / Frequently / Almost
apy). Thereafter, the assessment intervals may be extended
(e.g., monthly or quarterly, depending on the regimen
constantly) under study), particularly in trial contexts in which the
Severity item: What was the SEVERITY of your
__________ at its WORST?
duration of investigational treatment is prolonged. How-
ever, the time points of measurement should reflect the
(None / Mild / Moderate / Severe / Very severe) anticipated pattern of toxicity and scientific objectives of
Interference item: How much did __________ INTERFERE
with your usual or daily activities?
the trial.
Moving forward, several challenges and knowledge gaps
(Not at all / A little bit / Somewhat / Quite a bit / Very must be addressed. Although multiple translations are in
progress (for example, Italian, Korean, Chinese, and Swed-
much)
The standard PRO-CTCAE recall period is the past 7 days. ish), and linguistically validated language versions are
available in Spanish, German, Japanese, and Danish,25-27
A recent study suggests that longer recall periods (2-, 3-,
translation and linguistic validation of the PRO-CTCAE item
or 4-week recall) are associated with small but succes-
library in other languages is needed. Second, as is the case
sively increasing measurement error, which must be
for the collection of PRO data in any trial, there are per-
considered if recall periods longer than 1 week are used
sonnel and infrastructure requirements. Investments will
in a trial for logistical reasons (TR Mendoza, AV Bennett,
be required to develop and refine strategies that achieve
SA Mitchell et al, unpublished data, March 2016). Ad-
efficient PRO data collection and ensure data completeness
ministration of PRO-CTCAE via different modes including
web, interactive voice response, and paper offers flexi- (e.g., central monitoring and backup data collection). Po-
tential concerns about workload for clinical research staff
bility for patients and study operations personnel, and
will also need to be addressed, and analyses are in prog-
there is psychometric evidence to justify comparison of
ress that will yield specific estimates of the resource re-
results and pooled analyses across studies that use dif-
quirements associated with collecting and analyzing PRO-CTCAE
ferent PRO-CTCAE modes of administration.21 A pediatric
data in NCI-sponsored clinical trials. Third, there is limited ex-
version of PRO-CTCAE is also currently in development.22
perience with respect to how to optimally analyze and interpret
For more information about PRO-CTCAE, visit http://
patient-reported symptomatic AE data. Efforts are underway to
healthcaredelivery.cancer.gov/pro-ctcae.
determine how PRO-CTCAE scores should be interpreted to
assign a corresponding CTCAE grade. Additional work will
Early Adoption of PRO-CTCAE and Lessons Learned be required to identify the most informative ways to display
The PRO-CTCAE has been implemented in multiple cancer symptomatic toxicity scores descriptively alongside CTCAE
clinical trials. Patients are generally willing and able to self- grades in both published reports as well as potential product
report this information weekly via the web or automated labels.
telephone systems though electronic reminders; central Notably, FDA OHOP is committed to working with the
monitoring and personnel for backup data collection were NCI as well as commercial sponsors early in programs to
also needed to optimize response rates. Clinicians note identify opportunities to include PRO-CTCAE in clinical
finding this information to be meaningful and valuable for trials. Data generated using PRO-CTCAE offer comple-
clinical decision-making and AE reporting.23 mentary descriptive patient-reported information about
Selecting which symptomatic AEs to assess and de- symptomatic side effects that may further inform patients
termining the time points for measurement are critical and providers. There is also interest in using PRO-CTCAE
trial-design decisions. In trials developed to date, items as a component to support comparative tolerability
from the PRO-CTCAE and time points of measurement trial designs. More work must be done to explore this

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 71


KLUETZ ET AL

opportunity, including identifying an appropriate approach in product labels, complementing the standard safety as-
to capture the overall side effect burden of treatment. In sessment of a therapy.
addition, FDA OHOP, in collaboration with Clinical Outcome
Assessment staff and the Office of Biostatistics, has initiated
several internal working groups to explore different analysis CONCLUSION
and data presentation methods. Patients and their clinicians would benefit from improved
There is a reasonable concern that stacking new in- data about the effects of anticancer therapy on how an in-
struments on top of existing lengthy HRQOL and disease dividual feels and functions. Traditional PRO strategies are
modules as well as utility measures could lead to duplication being revisited as patient-focused drug development has
and pose additional respondent burden. FDA OHOP is fos- generated multistakeholder interest to optimize the collec-
tering international collaboration to review existing HRQOL tion and interpretation of PRO data to satisfy the needs of the
instruments and their disease modules to identify a col- many end users of this information. As a key component of a
lection of new and existing or modified existing instruments broader PRO strategy, the systematic longitudinal assessment
to meet the needs of all those who will use these data to of patient-reported symptomatic AEs can provide additional
make treatment, regulatory, and health policy decisions. The complementary tolerability data to inform dose selection and
goal remains to identify approaches that increase the rel- the overall benefit: risk assessment of a cancer therapy. The
evance and interpretability of PRO data while minimizing the PRO-CTCAE has been developed as a standardized mea-
burden to patients to ensure that the patient can be queried surement system that can provide a flexible fit-for-purpose
at an assessment frequency that provides the best picture of approach to assess relevant symptomatic AEs across a broad
the patient experience while on therapy. range of cancer therapies. It is anticipated that the NCI PRO-
There has been a call to improve the quality of PRO data CTCAE item library will continue to be iteratively refined as
captured from cancer clinical trials and integrate more of novel symptomatic toxicities are identified and a deeper
these data in the FDA product label.28 To realize this goal, understanding of its measurement properties emerges. There
more attention must be paid to PRO measures in both trial is vigorous and ongoing international collaboration among
design and conduct to improve overall data quality, re- trialists, methodologists, regulators, and patients to address
gardless of the symptoms or domains being measured. The these and other challenges in study design, implementation,
assessment of symptomatic AEs using a rigorously de- and interpretation to evolve a standard method to obtain
veloped item library such as the PRO-CTCAE can increase the well-defined, descriptive patient-centered data on the safety
likelihood of inclusion of descriptive patient-centered data and tolerability of cancer therapies.

References
1. Basch E, Iasonos A, McDonough T, et al. Patient versus clinician 9. Basch E, Geoghegan C, Coons SJ, et al. Patient-reported outcomes in
symptom reporting using the National Cancer Institute Common Ter- cancer drug development and US regulatory review: perspectives from
minology Criteria for Adverse Events: results of a questionnaire-based industry, the Food and Drug Administration, and the patient. JAMA
study. Lancet Oncol. 2006;7:903-909. Oncol. 2015;1:375-379.
2. Xiao C, Polomano R, Bruner DW. Comparison between patient-reported 10. Kluetz PG, Slagle A, Papadopoulos E, et al. Focusing on core patient-
and clinician-observed symptoms in oncology. Cancer Nurs. 2013;36: reported outcomes in cancer clinical trials: symptomatic adverse
E1-E16. events, physical function, and disease-related symptoms. Clin Cancer
3. Cella DF, Tulsky DS, Gray G, et al. The Functional Assessment of Cancer Res. 2016;22:1553-1558.
Therapy scale: development and validation of the general measure. 11. Biankin AV, Piantadosi S, Hollingsworth SJ. Patient-centric trials for
J Clin Oncol. 1993;11:570-579. therapeutic development in precision oncology. Nature. 2015;526:
4. Niezgoda HE, Pater JL. A validation study of the domains of the core 361-370.
EORTC quality of life questionnaire. Qual Life Res. 1993;2:319-325. 12. Theoret MR, Pai-Scherf LH, Chuk MK, et al. Expansion cohorts in first-in-
5. Srensen JB, Klee M, Palshof T, et al. Performance status assessment in human solid tumor oncology trials. Clin Cancer Res. 2015;21:4545-4551.
cancer patients. An inter-observer variability study. Br J Cancer. 1993; 13. U.S. Department of Health and Human Services NIH, National Cancer
67:773-775. Institute. Common Terminology Criteria for Adverse Events (CTCAE)
6. European Organization for Research and Treatment of Cancer (EORTC) Version 4.0. http://evs.nci.nih.gov/ftp1/CTCAE/CTCAE_4.03_2010-06-
Disease Specific Modules. http://groups.eortc.be/qol/why-do-we- 14_QuickReference_5x7.pdf. Accessed February 27, 2016.
need-modules. Accessed February 25, 2016. 14. U.S. Food and Drug Administration. Guidance for Industry Patient-
7. Functional Assessment of Cancer Therapy (FACT) Cancer Specific Reported Outcome Measures: Use in Medical Product Development to
Measures. http://www.facit.org/FACITOrg/Questionnaires. Accessed Support Labeling Claims. http://www.fda.gov/downloads/Drugs/
February 25, 2016. Guidances/UCM193282.pdf. Accessed February 27, 2016.
8. Rock EP, Kennedy DL, Furness MH, et al. Patient-reported outcomes 15. Michot JM, Bigenwald C, Champiat S, et al. Immune-related adverse
supporting anticancer product approvals. J Clin Oncol. 2007;25: events with immune checkpoint blockade: a comprehensive review. Eur
5094-5099. J Cancer. 2016;54:139-148.

72 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


MEASURING SYMPTOMATIC ADVERSE EVENTS WITH PRO-CTCAE

16. Klastersky JA. Adverse events of targeted therapies. Curr Opin Oncol. validation study among pediatric oncology clinicians. Pediatr Blood
2014;26:395-402. Cancer. 2013;60:1231-1236.
17. Thanarajasingam G, Hubbard JM, Sloan JA, et al. The imperative for a 23. Bruner DW, Hanisch LJ, Reeve BB, et al. Stakeholder perspectives on
new approach to toxicity analysis in oncology clinical trials. J Natl Cancer implementing the National Cancer Institutes patient-reported out-
Inst. 2015;107:djv216. comes version of the Common Terminology Criteria for Adverse Events
18. Basch E, Reeve BB, Mitchell SA, et al. Development of the National (PRO-CTCAE). Transl Behav Med. 2011;1:110-122.
Cancer Institutes patient-reported outcomes version of the common 24. Cella D, Wagner L. Re-personalizing precision medicine: is there a role
terminology criteria for adverse events (PRO-CTCAE). J Natl Cancer Inst. for patient-reported outcomes? J Community Support Oncol. 2015;13:
2014;106:dju244. 275-277.
19. Hay JL, Atkinson TM, Reeve BB, et al; NCI PRO-CTCAE Study Group. 25. Arnold B, Lent L, Mendoza T, et al; PRO-CTCAE Spanish Translation
Cognitive interviewing of the US National Cancer Institutes Patient- and Linguistic Validation Study Group. Linguistic validation of
Reported Outcomes version of the Common Terminology Criteria for the Spanish version of the National Cancer Institutes Patient-
Adverse Events (PRO-CTCAE). Qual Life Res. 2014;23:257-269. Reported Outcomes version of the Common Terminology Criteria
20. Dueck AC, Mendoza TR, Mitchell SA, et al; National Cancer Institute for Adverse Events (PRO-CTCAE). Support Care Cancer. Epub 2016
PRO-CTCAE Study Group. Validity and reliability of the US National Feb 2.
Cancer Institutes Patient-Reported Outcomes version of the Common 26. Kirsch M, Mitchell SA, Dobbels F, et al. Linguistic and content validation
Terminology Criteria for Adverse Events (PRO-CTCAE). JAMA Oncol. of a German-language PRO-CTCAE-based patient-reported outcomes
2015;1:1051-1059. instrument to evaluate the late effect symptom experience after al-
21. Bennett AV, Dueck AC, Mitchell SA, et al; National Cancer Institute PRO- logeneic hematopoietic stem cell transplantation. Eur J Oncol Nurs.
CTCAE Study Group. Mode equivalence and acceptability of tablet 2015;19:66-74.
computer-, interactive voice response system-, and paper-based ad- 27. Baeksted C, Pappot H, Bidstrup PE, et al. Danish translation and lin-
ministration of the U.S. National Cancer Institutes Patient-Reported guistic validation of the Patient-Reported Outcomes version of the
Outcomes version of the Common Terminology Criteria for Adverse Common Terminology Criteria for Adverse Events (PRO-CTCAE). J Pain
Events (PRO-CTCAE). Health Qual Life Outcomes. 2016;14:24. Symptom Manage. In press.
22. Reeve BB, Withycombe JS, Baker JN, et al. The first step to integrating 28. Basch E. Toward patient-centered drug development in oncology.
the childs voice in adverse event reporting in oncology trials: a content N Engl J Med. 2013;369:397-400.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 73


CARE DELIVERY AND PRACTICE MANAGEMENT

Optimizing Team-Based Oncology


Care: Building the Village

CHAIR
Lee Schwartzberg, MD, FACP
The West Cancer Center
Memphis, TN

SPEAKERS
Lillie D. Shockney, RN, BS, MAS
Johns Hopkins Breast Center
Baltimore, MD

Wendy H. Vogel, MSN, FNP, AOCNP


Wellmont Cancer Institute
Kingsport, TN
INFORMING THE TEAM WITH PATIENT-REPORTED OUTCOMES

Electronic Patient-Reported Outcomes: The Time Is Ripe for


Integration Into Patient Care and Clinical Research
Lee Schwartzberg, MD, FACP

OVERVIEW

In the emerging team-based approach to delivering cancer care, collecting patient-reported outcomes (PROs) provides
longitudinal monitoring of treatment adverse effects, disease complications, functional statuses, and psychological states
throughout the cancer continuum for all providers to use. Electronic systems offer added capabilities, including easy
quantitation of individual symptom items and aggregated scales, standardization, and longitudinal tracking of patient
surveys for trend analysis over time. An ideal electronic PRO (ePRO) platform is clinically relevant, validated, and reliable and
would offer patient usability. Additionally, it should allow for automated responses to and from patients, have scheduling
functionality, and send real-time alerts to site personnel and patients. Clinical interfaces should be easy to read and in-
tegrated into the electronic medical record. Multiple ePRO systems, often using electronic tablets, have been created and are
beginning to be widely deployed. The Patient Care Monitor is one example of a system that has evolved into a com-
prehensive patient engagement platform, with a complete review of systems survey and capabilities for mobile health
usage. Recent clinical trials have established ePRO systems as an effective method of providing information, which aids
improved patient outcomes, including reduced health resource utilization and longer time on therapy. ePROs are also
increasingly incorporated into clinical trials, where they can provide more thorough reporting of adverse events than can be
captured by alternative methods. Mobile devices have the potential to become the method by which all members of the
provider team communicate with patients both at the point-of-care and between clinic visits to optimize care delivery.

A chieving optimal patient-centered cancer care depends


in large part on the crucial clinician-patient interaction
at the point of care. In an era of increasing demands on the
without interpretation by a clinician or any other individual.
Collecting PROs allows clinicians to longitudinally monitor a
patients tolerance of therapy, response to therapy, and
physicians time, declining reimbursement, a surge in doc- symptoms that result from the underlying disease or
umentation requirements, and ever-increasing patient treatment. Psychological and functional statuses can be
volumes, the very nature of the patient-provider relation- assessed in the same fashion, which allows examination of
ship is threatened. One fundamental goal of the patient the interplay between treatment and global quality of life
interview is to obtain relevant information about the present and identification of issues that require referral to other
health status to guide treatment and symptom control. A members of the team. The importance of subjective mon-
thorough review of systems and evaluation of psychosocial itoring is firmly established; pain as the fifth vital sign is an
status and physical and social functioning is ideally ac- integral assessment measure for the Quality Oncology
complished at the clinic visit. Practice Initiative program, and the Commission on Cancer
The complexities of modern oncology care require a team now requires distress screening as the sixth vital sign to be
approach to deliver quality management of these multiple measured and addressed at every visit. A systematic ap-
dimensions. Having a record of PROs, which are quantitative, proach to assess distress can be enhanced through the use of
validated, easily captured, standardized, and recorded in the an ePRO system, which allows self-identification of the
electronic health record, has the potential to add huge value. problem without prejudice.
All of these characteristics can be achieved by using a variety In a distributed care model, a permanent, searchable
of electronic systems that have been developed to enhance record of the longitudinal health status allows all members
the interaction between the clinical staff and the patient. of the care team to access, interpret, and act on the findings.
A patient-reported outcome is defined as a measurement It is well documented that patients with cancer are often
of the patients condition, reported directly by the patient reluctant to discuss important toxicities with providers for a

From The West Cancer Center, Memphis, TN.

Disclosures of potential conflicts of interest provided by the author are available with the online article at asco.org/edbook.

Corresponding author: Lee Schwartzberg, MD, FACP, 7945 Wolf River Blvd., Germantown, TN 38138; email: lschwartzberg@westclinic.com.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e89


LEE SCHWARTZBERG

variety of reasons.1 Completion of an electronic survey staff, typically by email or text message. Many of the newer
appears to reduce that reluctance.2 systems are designed for long-term monitoring across the
Empowering patients to self-report provides the best in- cancer care continuum. Flexibility of system reporting allows
formation on subjective symptoms. Multiple investigations ePROs to be used effectively by various stakeholders of the
have clearly documented under-reporting when information care team, including clinicians, navigators, psychologists,
is provided by clinicians, although nurses appear better at care support counselors, and financial aid associates. One
eliciting symptoms than physicians.3,4 Reasons for under- limitation of most ePRO systems to date is a lack of the ability
reporting of toxicities may include a judgment call by the of patients to add self-identified concerns, which would
clinician when the symptom is detected but not reported. require the use of free text and patient input into selection of
This circumstance might occur when symptoms are pre- the items of most importance to them at a given moment in
existing, attributable to the disease, or mild and not re- the cancer trajectory.
quiring intervention.5 Symptoms that are not reported In order for an ePRO system to be widely disseminated and
because of inadequate communication are potentially more routinely used, value to both the provider and the patient
damaging. With the advent of many new drugs, some with must be demonstrated. Evidence exists to support in-
novel toxicities, the possibility of missing unusual but im- tegration of ePROs into clinical workflow in a manner that is
portant adverse effects that are not familiar to the provider relatively unobtrusive and that provides the potential for
looms large in the presence of poor symptom communi- improved symptom control and patient-clinician in-
cation. Standardized routine assessment is recognized teraction.8 Table 1 displays a proposed feature set that
among medical policymakers as a key component of en- serves as a good framework for elements of optimal
suring high-quality patient-centered care.6 platforms.

USE OF ePRO SYSTEMS DEVELOPMENT OF AN ePRO SYSTEM


A review of ePRO systems used in cancer clinical care was Our group began developing an electronic tablet-based
recently published.7 Thirty-three systems were identified; system and an instrument to routinely monitor patient
the majority are used in the United States, and one-third are symptoms and health statuses more than 15 years ago.
used in a single academic institution, with the majority used Assessment of items pertaining to physical symptoms,
at a single clinical entity. Most systems were developed to be emotional reactions, and functional statuses were identified
used in the clinic at the point of care, and most are web- by a team of medical oncologists, clinical psychologists
based. The focus of these systems is use predominantly working in oncology, oncology nurses, and patients. Items
during treatment and follow-up care, to monitor chemo- were assembled by reviewing previously developed quality-
therapy and other cancer therapy. Only half of the systems of-life instruments from the research setting and by de-
had greater than 500 users. The majority of systems sent termining the importance via face validity in routine patient
real-time alerts of the patients response to providers and care. Each item was presented as a 0-to-10 Likert scale of
severity, in which 0 represented no severity and 10 repre-
sented the most severe, with a timeframe of the past 7 days.
The instrument, termed the Patient Care Monitor (PCM;
KEY POINTS initially called Cancer Care Monitor) consisted of 38 items,
grouped into seven constructs of physical symptoms,
PROs provide the most thorough and reliable treatment adverse effects, distress, despair, impaired am-
information on subjective symptoms, overcoming the
bulation, impaired performance, and global quality of life.
well-documented under-reporting by clinicians.
ePRO systems, which are web-based and are typically
Validation studies established psychometric validity, scale
delivered on tablets, computers, or other mobile consistency, and reliability.9 Alternative form reliability
devices, are now readily available. that was based on comparisons with paper forms was high,
One example of an ePRO system, the Patient Care and patient preference was for the tablet-administered
Monitor, is a full patient engagement system, which version. In subsequent studies, item validity by PCM was
includes a complete review of system survey and a compared with a blinded structured interview conducted by
flexible architecture that allows deployment of custom an experienced oncology nurse, and good concordance was
surveys and patient alerts to relevant clinical staff. established between the structured interview and patient
The use of an ePRO survey for patients with advanced self-report.10
cancer has been shown in a randomized controlled trial An expanded version of the PCM, termed PCM 2.0, was
to improve health-related quality of life, reduce
subsequently created to address clinicians desires to have a
emergency department visits, and improve 1-year
survival compared with usual clinical care.
complete review of systems provided by patient self-report.
A subset of the Common Terminology Criteria for The gender-specific survey consisted of 86 items (79 for
Adverse Events library is now available as an ePRO men) that encompassed 13 organ sitespecific systems (e.g.,
platform and has been validated as an alternative gastrointestinal, cardiovascular). A validation study was
method of eliciting symptom reports in clinical trials. performed for PCM 2.0 and showed good correlation be-
tween patient- and nurse-reported severities.11 The median

e90 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


INFORMING THE TEAM WITH PATIENT-REPORTED OUTCOMES

TABLE 1. Desirable Features of ePRO Systems in interface. In the EMR, the items formed part of the per-
Clinical Care manent record located on the flowsheet for a given out-
patient visit. The paper version displayed demographics,
Elements Attributes columns of the last three surveys of each item, and visual
Symptom Questions/ Demonstrated validity and reliability prompts to allow the clinician to quickly scan the report for
Items severe endorsements and potentially clinically significant
Known clinical significance of score changes
Relevance in the selected patient population differences ($ 3-point change) from the last survey, signified
and clinical context by up and down arrows. The surveys are transmitted to the
Appropriate recall period (no longer than clinician immediately after completion and serve the
1 week) function of focusing the patient interview on the most
Patient Interface Demonstrated usability in patients severe/changed item endorsements.
Web-based and available in multiple modes PCM 2.0 was installed in a major academic cancer center in
for data entry via internet, handheld de- 2006. A pilot trial established patient satisfaction with PCM
vice, tablet app
usage and acknowledgment that the system facilitated
Allowance for in-clinic and/or between-visit symptom reporting to the clinicians.12 Validation was ac-
reporting
complished in a larger trial of 275 patients with breast,
Inclusion of a scheduling/calendar compo-
nent to specify when patients are expected gastrointestinal, or lung cancer.13 Good internal consistency
to report was demonstrated; Cronbachs a coefficients were similar to
Automated reminders to patients to report those determined in the community oncology validation
(e.g., email, text message, automated study. Pearson correlatives for PCM subscales were good,
telephone call)
and there was good factor concordance with the Functional
Allowance for use of simple usernames/ Assessment of Cancer TherapyGeneral (FACT-G), Func-
passwords that are not overly complex or
cumbersome to patients tional Assessment of Chronic Illness Therapy-Fatigue (FACIT-F),
Availability of staff to train and assist patients
and MD Anderson Symptom Inventory (MDASI) instruments
widely used in the research setting. PCM 2.0 thus served a dual
Allows patients to request additional help
functionality as a clinical and a research tool, which allowed
Clinician Interface Includes displays of longitudinal patient-
reported symptoms numerically and individual clinicians to use the data to inform the patient visit
graphically and became part of a longitudinal repository of standardized
Integrated with electronic health record PRO items well suited to research. A larger data set of 5,624
Allows clinicians to report symptoms/ patients who completed the PCM survey was recently ana-
comment on patient reports lyzed. The instrument exhibited high internal consistency, and
Alerts/Notifications Sends real-time automated alerts (e.g., email, investigators were able to accurate distribute the items into
text message) to clinicians when patients three constructs of emotional function, physical function, and
report symptom severities above
predetermined thresholds or experience
physical symptoms.14 This large-scale experience firmly
symptom worsening that exceeds a established the practicality of systematic collection of ePROs
predetermined amount as part of the standard of care.
Triggers automated notifications to site The widespread adoption of smartphones, tablets, and
personnel when patients do not report at other portable computing devices led to the development
a scheduled time point
of a new architecture for the PCM system in 2014, when it
Provides automated messages to patients
and triggers automated patient education
was redeployed as a cloud-based, web-enabled application
when symptoms exceed predetermined with an open architecture to meet the growing need for
absolute thresholds or score-change additional surveys at the practice level. The core instrument
thresholds
was condensed somewhat to 56 items on the basis of the
Back-up Data Includes predetermined methods to contact evaluation of rarely reported symptoms and those that did
Collection patients who do not self-report on
schedule not contribute meaningfully to the aggregate scale scores.
Abbreviations: ePRO, electronic patient-reported outcome. A complete 13-system review to meet Centers for Medicare
Adapted from Basch and Abernethy.8 & Medicaid Services guidelines for the highest-level visit
was retained.
time for patients to complete PCM 2.0 was 11 minutes. This This revised PCM core version is backward compatible with
version of the platform, which included a wireless tablet earlier versions and has concordance with several other
system linked to a central server, was eventually deployed in commonly used instruments for core symptoms. The new
more than 100 community practice sites for use in practice system is delivered via standard commercial tablets pro-
performance improvement projects and as part of routine vided by the oncology practice rather than as the previously
clinical care. used self-contained dedicated system. Acceptance by pa-
PCM 2.0 results were available both in a paper format tients and providers has been excellent. All told, greater than
and integrated into the electronic medical record (EMR) 100,000 patients have completed PCM surveys since its
of commercial oncology EMR products through an HL-7 inception. Additional modules that have been added to the

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e91


LEE SCHWARTZBERG

core survey include a radiation therapyspecific survey, a FIGURE 1. Cumulative Incidence of Emergency
falls risk assessment and learning needs assessment survey, Department Visits
and a tobacco use and readiness to quit module.

BENEFIT OF ePROS
Despite the value attributed to PROs in reporting and
addressing routine symptoms, the impact of collecting this
data has not been settled. Systematic reviews have largely
failed to demonstrate a clear benefit of collecting PROs,
largely because of methodologic issues with the ways
studies are structured, such as including of several sources of
bias or widely varying endpoints.15 A prospective random-
ized controlled trial conducted at two academic medical
centers met its primary endpoint of demonstrating that self-
reporting of ePROs available to the clinician at the clinic visit
resulted in more discussion of the problem with the patient
than usual care when the symptom intensity was higher
than a predefined threshold.16 Patient-provider visits were
not significantly longer in duration despite having the PROs
to address, and most clinicians surveyed as part of the study
felt that the PROs served a clinical utility function.
A recent multicenter randomized controlled trial reported
by Basch et al17 assigned patients who received chemo-
therapy to report a small number of symptoms via tablet-
computer with weekly email prompts or to receive usual
care. Symptom assessment was delivered either to providers
at the outpatient visit or to the nursing staff via email when
patients reported severe or worsening symptoms. The pri-
mary endpoint of the study was change in health-related
quality of life (QOL) at 6 months compared with baseline.
Secondary endpoints included emergency department visits,
hospitalizations, and overall survival. Patients were addi-
tionally separated into subgroups on the basis of their prior
experiences with computer systems.
The trial revealed that more patients in the ePRO group at
6 months had improvements in health-related QOL (34% vs.
18%), fewer had a decrease (38% vs. 53%, p , .001), and the
mean health-related QOL score declined less in the ePRO group
than in the routine care group (1.4 vs. 7.1, p , .001). Im-
portantly, significantly fewer patients who routinely reported
PROs had an emergency department visit during the period of
study (Fig. 1), and those who used the electronic system re-
ceived chemotherapy longer. Overall survival at 1 year was
better for the ePRO group than the usual care group (75% vs.
69%, p = .05). Interestingly, computer-inexperienced patients
appeared to derive more benefit from the system with regard
to reduction in emergency department visits, perhaps because
this subgroup was older, less educated, and may have been
under-reporting symptoms previously for cultural reasons.
This study highlights the power of ePRO reporting to in-
fluence tangible outcome measures for patients with ad-
vanced cancer, including reduced resource utilization, better
quality of life, and evenpossiblyimproved survival. Why
The incidence of patients visiting the emergency department is shown, with
would this be? Routine reporting of cancer symptoms and a death as a competing event: all patients (A); computer-experienced patients (B);
system of planned interactions to address them appear to and computer-inexperienced patients (C).
Abbreviation: STAR, Symptom Tracking and Reporting (a web-based self-reporting
improve patient well-being and allow patients to tolerate system as the study intervention).
treatment better, which might influence survival. In the trial Adapted and used with permission.17

e92 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


INFORMING THE TEAM WITH PATIENT-REPORTED OUTCOMES

conducted by Basch et al,17 time on therapy was approxi- and on collecting information as frequently as necessary
mately 2 months longer in the ePRO group, which supports for the proposed research without overburdening patients.
this assertion. The results also mirror those of patients with In using PROs for comparative effectiveness research, the
lung cancer who were referred early to palliative care,18 in part evidence guidance document recommended a priori power
because patients symptoms in both instances were taken calculations of PRO endpoints, a plan for handling missing
seriously and addressed. The patient-centered medical home data, and a plan for reporting individual measure changes
model, endorsed by the Centers for Medicare & Medicaid proportionally and as mean group measure changes. The
Services (via its Oncology Care Model), calls for improved document also endorsed simultaneous publication of the
interactions between the clinic staff and patients, including PRO data analysis with the other endpoints of the re-
mechanisms for patients to report problems in a real-time way search trial.
without having to resort to hospitalization or emergency Standard adverse event reporting in clinical trials is assessed
department visits. An ePRO system is an excellent tool to assist by the Common Terminology Criteria for Adverse Events
implementation of the Oncology Care Model program. (CTCAE), created by the National Cancer Institute. Adverse
events may reflect laboratory abnormalities, physical exami-
PROS IN CLINICAL RESEARCH nation changes, functional status variations, and patient-
Inclusion of patients self-reported symptoms and adverse elicited toxicities. Traditionally, symptoms are prompted
events is fundamental to evaluating new drugs in oncology. from the patient by a clinical observer involved in the trial.
The use of PROs is endorsed by all major entities involved in However, evidence suggests that adverse event symptoms are
conducting clinical trials.19 A multidisciplinary group was often under-reported by clinical investigators.21,22 The Na-
charged by the Center for Medical Technology Policy to tional Cancer Institute has developed a library of items, termed
create an evidence guidance document with recommen- the PRO-CTCAE, which consists of 124 PROs that reflect 78
dations for PRO use in comparative effectiveness research.20 different adverse events. Recently, a validity and reliability
The group devised 15 recommendations related to three assessment of the PRO-CTCAE tool has been published.23 The
broad domains of PROs: measure selection, implementation study included 975 adult patients receiving chemotherapy or
measures, and data analysis/reporting. Chief among these radiation therapy who completed the instrument on tablets in
were the importance of selecting a validated tool that is fit clinic waiting rooms at nine U.S. cancer centers and com-
for the purpose of the study. Twelve symptoms that share munity oncology practices. PRO-CTCAE were compared with
commonality among PRO instruments were highlighted as the clinician-reported Eastern Cooperative Oncology Group
having particular value in assessing wellbeing and health- (ECOG) performance status and a reference instrument, the
related QOL (Table 2). Implementation recommendations European Organization for Research and Treatment of Cancer
focused on obtaining information rapidly, preferably elec- Core Quality of Life Questionnaire (QLQ-C30).
tronically, when this method had been adequately com- A broad sample of patients was enrolled, including many
pared with other standard methods such as paper forms, who had ECOG performance statuses of 2 to 4. Symptom

TABLE 2. Availability in Existing PRO Instruments of Common Cancer Symptoms

PRO Instrument*
Symptom ESAS FACT LASA MDASI MSAS PCM PRO-CTCAE PROMIS QLQ-C30 RSCL SDS
Anorexia X X X X X X X X X
Anxiety X X X X X X X X X
Constipation X X X X X X X
Depression X X X X X X X X X
Diarrhea X X X X X X X X
Dyspnea X X X X X X X X X
Fatigue X X X X X X X X X X X
Insomnia X X X X X X X X X X X
Nausea X X X X X X X X X
Pain X X X X X X X X X X X
Neuropathy X X X X X X
Vomiting X X X X X X
Abbreviations: CER, comparative effectiveness research; ESAS, Edmonton Symptom Assessment Scale; FACIT, Functional Assessment of Chronic Illness Therapy; FACT-G, Functional
Assessment of Cancer Therapy-General; LASA, Linear Analog Self-Assessment; MDASI, MD Anderson Symptom Inventory; MSAS, Memorial Symptom Assessment Scale; PCM, Patient
Care Monitor; PRO, patient-reported outcome; PRO-CTCAE, PRO version of the Common Terminology Criteria for Adverse Events; PROMIS, PRO Measurement Information System;
QLQ-C30, European Organization for Research and Treatment of Cancer Quality of Life Questionnaire; RSCL, Rotterdam Symptom checklist; SDS, Symptom Distress Scale.
Adapted and used with permission.20
*Instruments are listed alphabetically. Dash signifies symptom not measured by that instrument; X, measured by that instrument. Most measurement systems include additional
symptom items beyond these 12 symptoms.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e93


LEE SCHWARTZBERG

endorsement was compared with several clinical anchors, disease progression or various chemotherapy regimens on
including performance status, type of therapy, supportive PROs in metastatic breast cancer, prostate cancer, and lung
care measures initiated, expert consensus, and patient in- cancer.24-27
put. A median (range) of 23 (091) PROs were endorsed.
Ninety-six percent of patients reported five or more
symptoms at the first visit, and 768 (81.7%) of 940 patients EXPANDING ePROS TO FULL PATIENT
reported at least one symptom as frequent, severe, and/or ENGAGEMENT PLATFORM
interfering quite a bit with daily activities. Validity of the A 2013 Institute of Medicine report, Delivering High-Quality
instrument was achieved; all 124 items were endorsed in the Cancer Care: Charting a New Course for a System in Crisis,
same direction as the QLQ-C30 reference instrument, and stated that cancer care is often not as patient-centered,
114 items demonstrated meaningful correlations, with a accessible, coordinated, or evidence-based as it should be.28
Pearson coefficient r of 0.1 or greater. Of these 114 items, Part of the solution is to actively engage patients to interact
111 were significant (p , .05). with an adequately staffed and trained workforce. To meet
Responsiveness of items to the patient-reported global the needs of an increasing number of patients, cancer clinics
impression of change (GIC) from visit one to two was must embrace and expand nonphysician members of the
evaluated in a core set of 20 symptomatic adverse events care team to deliver high-quality multidimensional care.
with high face validity for relation to change in QOL, physical Enhancing the patient-clinician interaction is another aspect
condition, and/or emotional state. Thirteen items achieved of best practice cancer care organizations.
excellent correlation with GIC scores, which validated this The electronic platforms that serve as the basis for ePRO
symptom set as critical in approximating patient-reported acquisitions are being expanded to address the growing
health statuses. This trial represents the culmination of more needs of the cancer patient. Flexible system design can begin
than a decade of study comparing patient assessment of to deliver functionality beyond information gathering.
toxicity to clinician assessment in clinical trials. The challenge Configured with advanced scheduling functions, branching
now will be to incorporate these assessments into routine question capabilities, easy loading of additional surveys, and
adverse event reporting when new therapeutic entities are two-way communication loops between patient and prac-
evaluated. tice, patient engagement platforms can facilitate patient
Longitudinal collection of ePROs offers opportunities to decision support and shared decision making. These plat-
integrate symptom attribution into retrospective studies of forms can triage patients automatically to appropriate
treatment patterns in the real-world setting. Such analyses members of the care delivery team with minimal in-
have been performed with the PCM to address the effect of terruption of normal clinic processes.

FIGURE 2. Workflow for Smoking Cessation Program


Tobacco Use Cessaon
PCM Screening May we contact you to
support your eorts to quit
PCM A Psycho-oncology Behavioral
using tobacco? Health Iniave
PCM will present NEW
paents with quesons If NO, quesons will be
about tobacco use at the Yes No presented again in 6
rst visit following NCCN months and at 1 year.
guidelines for smoking
cessaon. If YES, paent will be contacted via
phone, or e-mail or mail and will be provided
with resources via phone, email ,or postal
PCM will present all other mail
paents the tobacco use
quesons on a rolling basis Invitaon to aend weekly informaon
over the first year to create sessions to receive informaon about
a sustainable work ow. smoking, resources, and support

Screen for Case Finding


Individual assessments
The opportunity to have an individual
Currently use Tobacco session where personalized quit plans
will be provided
Informaon on pharmacotherapy
Frequency and quanty
Resources on Quit lines
Of tobacco use
A dedicated email and phone will for the
Ready to Quit smoking cessaon team will be provided as
a way for those interested to contact for
addional informaon
Request for Informaon

The workflow is based on patient engagement platforms to survey patients on tobacco use.
Abbreviation: PCM, Patient Care Monitor.

e94 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


INFORMING THE TEAM WITH PATIENT-REPORTED OUTCOMES

Our institution has recently launched a smoking cessation emails/texts to nursing or navigator staff, who then contact
project that identifies patients through a series of questions the patient directly. The goal is to alleviate severe toxicities
on the PCM about their current and past tobacco use sta- that emerge between visits by using electronic communi-
tuses and their readiness to quit (Fig. 2). If the patient is cation with the platform.
smoking but does not self-identify as ready to quit, they will
be scheduled to be automatically recontacted at 6-month
intervals. If ready to quit and requesting more information, CONCLUSION
an email message is sent to members of the psychology ePROs have emerged as an effective method of capturing
team, who then contact the patient and enroll them in an patient symptoms that can lead to a more thorough and
evidence-based smoking cessation program. Tobacco use consistent evaluation of patient physical and emotional
habits can also be appropriately coded to trigger email alerts health status and QOL. Numerous studies have validated
to clinicians for consideration of low-dose CT screening for multiple assessment instruments and platforms that can
early detection of lung cancer. now be used in routine clinical practice. New data support an
With the advent of mobile technology, studies have ad- ePRO solution to collecting patient symptoms consistent
dressed postoperative symptom severity after cancer sur- with regulatory criteria in the clinical trial assessment of new
gery at home, and an automated system has shown therapies. The widespread use of mobile devices provides an
superiority in controlling symptom threshold events in a excellent opportunity to expand patient engagement be-
randomized trial.29 Multiple pilot studies are underway to yond the point of care to routinely monitor patients for
address adherence to oral medications, such as aromatase emerging problems with minimal investment of time and
inhibitors in early-stage breast cancer and tyrosine kinase resources by the care delivery system, thus enhancing the
use in advanced colorectal cancer, through frequent overarching goal of high-quality patient-centered cancer
mobile surveying of emerging symptoms with triggers of care.

References
1. Forcina JM. Re: National Institutes of Health State-of-the-Science 11. Schwartzberg L, Fortner B, Houts A. Use of technology to enhance
Conference Statement: Symptom Management in Cancer: Pain, De- doctor-patient interactions. Am Soc Clin Oncol Educ Book. 2007;27:686-
pression, and Fatigue, July 15-17, 2002. J Natl Cancer Inst. 2004;96: 691.
1109-1110, author reply 1110. 12. Abernethy AP, Herndon JE II, Wheeler JL, et al. Feasibility and
2. Gwaltney CJ, Shields AL, Shiffman S. Equivalence of electronic and acceptability to patients of a longitudinal system for evaluating cancer-
paper-and-pencil administration of patient-reported outcome mea- related symptoms and quality of life: pilot study of an e/tablet data-
sures: a meta-analytic review. Value Health. 2008;11:322-333. collection system in academic oncology. J Pain Symptom Manage. 2009;
3. Basch E, Iasonos A, McDonough T, et al. Patient versus clinician 37:1027-1038.
symptom reporting using the National Cancer Institute Common Ter- 13. Abernethy AP, Zafar SY, Uronis H, et al. Validation of the Patient Care
minology Criteria for Adverse Events: results of a questionnaire-based Monitor (Version 2.0): a review of system assessment instrument for
study. Lancet Oncol. 2006;7:903-909. cancer patients. J Pain Symptom Manage. 2010;40:545-558.
4. Cirillo M, Venturini M, Ciccarelli L, et al. Clinician versus nurse symptom 14. Samsa GP, Wolf S, LeBlanc TW, et al. An exploratory factor analysis of
reporting using the National Cancer Institute Common Terminology the scale structure of the Patient Care Monitor Version 2.0. J Pain
Criteria for Adverse Events during chemotherapy: results of a com- Symptom Manage. 2016;51:776-783.
parison based on patients self-reported questionnaire. Ann Oncol. 15. Kotronoulas G, Kearney N, Maguire R, et al. What is the value of the
2009;20:1929-1935. routine use of patient-reported outcome measures toward improve-
5. Di Maio M, Basch E, Bryce J, et al. Patient-reported outcomes in the ment of patient outcomes, processes of care, and health service
evaluation of toxicity of anticancer treatments. Nat Rev Clin Oncol. outcomes in cancer care? A systematic review of controlled trials. J Clin
Epub 2016 Jan 20. Oncol. 2014;32:1480-1501.
6. Bergeson SC, Dean JD. A systems approach to patient-centered care. 16. Berry DL, Blumenstein BA, Halpenny B, et al. Enhancing patient-provider
JAMA. 2006;296:2848-2851. communication with the electronic self-report assessment for cancer:
7. Jensen RE, Snyder CF, Abernethy AP, et al. Review of electronic patient- a randomized trial. J Clin Oncol. 2011;29:1029-1035.
reported outcomes systems used in cancer clinical care. J Oncol Pract. 17. Basch E, Deal AM, Kris MG, et al. Symptom monitoring with patient-
2014;10:e215-e222. reported outcomes during routine cancer treatment: a randomized
8. Basch E, Abernethy AP. Supporting clinical practice decisions with real- controlled trial. J Clin Oncol. 2015;34:557-565.
time patient-reported outcomes. J Clin Oncol. 2011;29:954-956. 18. Temel JS, Greer JA, Muzikansky A, et al. Early palliative care for patients with
9. Fortner B, Okon T, Schwartzberg L, et al. The Cancer Care Monitor: metastatic nonsmall-cell lung cancer. N Engl J Med. 2010;363:733-742.
psychometric content evaluation and pilot testing of a computer ad- 19. Lipscomb J, Gotay CC, Snyder CF. Patient-reported outcomes in cancer:
ministered system for symptom screening and quality of life in adult a review of recent research and policy initiatives. CA Cancer J Clin. 2007;
cancer patients. J Pain Symptom Manage. 2003;26:1077-1092. 57:278-300.
10. Fortner B, Baldwin S, Schwartzberg L, et al. Validation of the Cancer Care 20. Basch E, Abernethy AP, Mullins CD, et al. Recommendations for in-
Monitor items for physical symptoms and treatment side effects using expert corporating patient-reported outcomes into clinical comparative ef-
oncology nurse evaluation. J Pain Symptom Manage. 2006;31:207-214. fectiveness research in adult oncology. J Clin Oncol. 2012;30:4249-4255.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e95


LEE SCHWARTZBERG

21. Di Maio M, Gallo C, Leighl NB, et al. Symptomatic toxicities experienced 25. Walker MS, Masaquel AS, Kerr J, et al. Early treatment discontinuation
during anticancer treatment: agreement between patient and physician and switching in first-line metastatic breast cancer: the role of patient-
reporting in three randomized trials. J Clin Oncol. 2015;33:910-915. reported symptom burden. Breast Cancer Res Treat. 2014;144:673-681.
22. Fromme EK, Eilers KM, Mori M, et al. How accurate is clinician reporting 26. Stepanski EJ, Houts AC, Schwartzberg LS, et al. Second- and third-line
of chemotherapy adverse effects? A comparison with patient-reported treatment of patients with non-small-cell lung cancer with erlotinib in
symptoms from the Quality-of-Life Questionnaire C30. J Clin Oncol. the community setting: retrospective study of patient healthcare uti-
2004;22:3485-3490. lization and symptom burden. Clin Lung Cancer. 2009;10:426-432.
23. Dueck AC, Mendoza TR, Mitchell SA, et al; National Cancer Institute 27. Walker MS, Reyes C, Kerr J, et al. Treatment patterns and outcomes
PRO-CTCAE Study Group. Validity and Reliability of the US National among patients with metastatic melanoma treated in community
Cancer Institutes patient-reported outcomes version of the Common practice. Int J Dermatol. 2014;53:e499-e506.
Terminology Criteria for Adverse Events (PRO-CTCAE). JAMA Oncol. 28. Levit L, Balogh E, Nass S, et al. Delivering High-Quality Cancer Care:
2015;1:1051-1059. Charting a New Course for a System in Crisis. Washington, DC, Institute
24. Schwartzberg LS, Cobb P, Walker MS, et al. Patient and practice impact of Medicine, National Academies Press; 2013.
of capecitabine compared to taxanes in first-/second-line chemo- 29. Cleeland CS, Wang XS, Shi Q, et al. Automated symptom alerts reduce
therapy for metastatic breast cancer. Support Care Cancer. 2009;17: postoperative symptom severity after cancer surgery: a randomized
1081-1088. controlled clinical trial. J Clin Oncol. 2011;29:994-1000.

e96 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


ADVANCED PRACTITIONERS IN ONCOLOGY CARE

Oncology Advanced Practitioners Bring Advanced Community


Oncology Care
Wendy H. Vogel, MSN, FNP, AOCNP

OVERVIEW

Oncology care is becoming increasingly complex. The interprofessional team concept of care is necessary to meet projected
oncology professional shortages, as well as to provide superior oncology care. The oncology advanced practitioner (AP) is a
licensed health care professional who has completed advanced training in nursing or pharmacy or has completed training
as a physician assistant. Oncology APs increase practice productivity and efficiency. Proven to be cost effective, APs may
perform varied roles in an oncology practice. Integrating an AP into an oncology practice requires forethought given to the
type of collaborative model desired, role expectations, scheduling, training, and mentoring.

2,400 pharmacists who specialized in oncology.1 The Na-


S uperior oncology care requires a superior team. As on-
cology care becomes increasingly complex, the inter-
professional team concept and collaborative care is no
tional Commission on Certification of Physician Assistants4
Statistical Profile of Recently Certified Physician Assistants
longer an option but a necessity. A team is a group of two or noted over 102,000 certified physician assistants in the
more people who function interdependently to achieve a United States, across all heath fields. Data extrapolated from
shared goal. Oncology APs are an integral part of the on- the American Academy of Physician Assistants Physician
cology team. Assistants Annual Survey Report estimate that about 2.4% of
physician assistants work in oncology (approximately 2,000).
DEFINITION OF ADVANCED PRACTITIONER Based on these data, there could be over 11,000 oncology
An AP is defined as a licensed health care professional who APs, but it is impossible to give an exact number because
has completed advanced training in nursing (such as a nurse some oncology APs may belong to multiple societies, not all
practitioner or clinical nurse specialist) or pharmacy or has APs have certification in oncology, and not all APs are
completed training as a physician assistant.1 Table 1 lists the members of one of the previously mentioned societies.
various APs and educational requirements.
SCOPE OF PRACTICE
STATISTICS Advanced practitioners legal scope of practice varies from
The American Society of Clinical Oncologys (ASCO) The State state to state,3 as noted in Table 1. The scope of practice may
of Cancer Care in America: 2015 notes that the numbers of also differ because of the APs type, level of experience,
APs in oncology are increasing. This may assist in meeting the and facility.1,5 The majority of APs have prescriptive
growth in demand for oncologists despite the fact that the privileges, with differing requirements/rules for controlled
number of oncologists remains static.2 However, it is difficult substances.3
to estimate the exact number of APs in oncology. The 28th
Annual Advanced Practice Registered Nurse Legislative COLLABORATIVE CARE MODELS IN ONCOLOGY
Update3 notes there are more than 306,000 advanced The majority of oncology care (80%) is provided in the
practice nurses in the United States, across all medical fields. community setting.6 With the expected shortfall of oncol-
ASCO2 estimates that there are about 3,000 APs working in ogists by 2020, collaborative practice will be required to
oncology and that this number is growing. The Oncology meet the demands of oncology care.7,8 Collaborative
Nursing Society counts 2,601 nurse practitioners (NPs) and practice requires the involvement of all interdisciplinary
1,173 clinical nurse specialists in the membership.1 The team members to maximize positive patient outcomes.1
Hematology/Oncology Pharmacy Organization notes about Advanced practitioners are an integral part of the oncology

From the Wellmont Cancer Institute, Kingsport, TN.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Wendy H. Vogel, MSN, FNP, AOCNP, WMA Hematology & Oncology at Kingsport, Wellmont Cancer Institute, 4485 West Stone Dr., Suite 200, Kingsport, TN
37660; email: wendyvogel55@gmail.com.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e97


WENDY H. VOGEL

TABLE 1. Education, Licensure, Professional Organizations, and Certification of Advanced Practitioners in Oncology
Advanced Entry-Level Board Certification Professional Prescriptive
Practice Role Degree Licensure and Certifying Body Organizations Authority Eligibility*
Nurse Doctorate (DNP or PhD) State Board of Nursing General certification: American AANP, ANA, Scope defined by
Practitioner preferred, previously NP: adult, pediatric, Nurses Association and State APSHO, ONS individual states
masters level or acute care, primary Board of Nursing Oncology
postmasters care, family practice Certification: AOCNP-ONCC
certificate AOCN-ONCC
Physician Masters degree State Board requires PA-C, NCCPA AAPA, APAO, Yes, requires
Assistant PA National Certifying APSHO supervising
Examination, physician
administered by NCCPA agreement
Clinical 4-year professional State Board The Board of Pharmacy Specialties ACCP, APSHO, Scope defined by
Pharmacist degree (Doctor of Pharmacy provides testing and maintains ASHP, HOPA individual states
of Pharmacy) the Board Certified Oncology
Pharmacist status
Clinical Nurse Masters degree State Board of Nursing State Board of APSHO, ONS Scope defined by
Specialist Advanced Practice Nurse Nursing: AOCNS-ONS individual states**
Abbreviations: DNP, Doctor of Nursing Practice; AOCNP, advanced oncology certified nurse practitioner; ONS, Oncology Nursing Society; AOCN, advanced oncology certified nurse;
AANP, American Academy of Nurse Practitioners; ANA, American Nurses Association; APSHO, Advanced Practitioner Society for Hematology and Oncology; NCCPA, National
Commission on Certification of Physician Assistants; APAO, Association of Physician Assistants in Oncology; AAPA, American Academy of Physician Assistants; ACCP, American College
of Clinical Pharmacy; ASHP, American Society of Health-System Pharmacists; HOPA, Hematology/Oncology Pharmacy Organization; AOCNS, advanced oncology clinical nurse
specialist.
*Variability by state.
**From the National Council of State Boards of Nursings APRN campaign for Consensus: Moving Toward Uniformity in State Laws (https://www.ncsbn.org/5410.htm).
Adapted from Vogel5 and from Kurtin SE et al.1

team. The ASCO Workforce Advisory Group suggested that practices. The collaborative model and billing practices in
increasing the use of and maximizing the potential of APs academic oncology practices are likely to be different.10 In
would improve practice efficiency.7 Since this 2007 publi- the ASCO study, both the incident-to model and the in-
cation, other studies and publications have examined var- dependent model consist of the physician and AP working
ious collaborative oncology practice models.8 independently; however, the primary difference is billing, the
Three collaborative practice models have been described in amount of consultation required, and the autonomy of the
oncology practices: the incident-to visit model, the shared AP. Other disciplines, such as primary care, describe practice
visit model, and the independent visit model (Table 2).8,9 The models of the AP (an NP) working completely independent
ASCO study of collaborative practice arrangements8 noted of a physician. Currently in 15 states, NPs are regulated by a
that more than 75% of practices use the incident-to visit Board of Nursing and have full autonomous practice and
model. This is likely to maximize reimbursement. Most of the prescriptive authority without requirement for physician
practices in this study (73%) were physician-owned private supervision, delegation, consultation, or collaboration.3
In some oncology practices, the AP works exclusively with
one oncologist; in other practices, the AP works with all
practice oncologists.8 The ASCO Study of Collaborative
Practice agreements noted that about 60% of APs work with
KEY POINTS
all practice oncologists and not exclusively with one on-
An advanced practitioner is defined as a licensed health cologist. Interestingly, the practices with APs working with
care professional who has completed advanced training all practice oncologists had higher productivity.8
in nursing or pharmacy or has completed training as a When choosing a collaborative practice model, the
physician assistant. practice needs and patient volume must be considered, as
Advanced practitioner legal scope of practice varies well as both the physician and APs training, strengths, skills,
from state to state, but most have prescriptive and expert knowledge. The ability to meet regulatory
privileges, with differing requirements/rules for constraints for AP billing must be carefully considered to
controlled substances. avoid fraud. If the AP is new to oncology, a thorough training
Three collaborative care models are identified in and mentoring program must be planned.1,5,11
oncology practices: incident-to visit model, the shared
Barriers to efficient practice must also be considered. A
visit model, and the independent visit model.
Barriers to efficient collaborative practice include
recent survey of APs in oncology noted the most common
disallowing the AP the full scope of practice and barrier to the AP role was time spent on tasks that could
inadequate training and mentoring. be delegated to non-AP staff.1,12 Other common barriers
Advanced practitioners are cost effective and an integral included insufficient support, mentoring, and insufficient
part of the future of oncology care. autonomy and training to meet role expectations. Most APs
are trained as generalists with limited time devoted to

e98 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


ADVANCED PRACTITIONERS IN ONCOLOGY CARE

TABLE 2. Collaborative Practice Models8-10,18


Model Description
Incident-To (Mixed) Visit Model AP sees patient independently, following physician plan of care; physician consult available if needed;
physician generally in office suite; billing is incident-to at full physician fee schedule; however,
must meet CMS regulations on alternate visit schedules
Shared Visit Model Both the AP and the physician see patient. The physician completes the visit note and bills for services.
Independent Visit Model AP sees patient independently, consulting at critical treatment decision points (such as initial treatment
plans, at progression, end-of-life decisions); billed under NPP own provider number, at 85% of
physician fee schedule
Abbreviations: AP, advanced practitioner; CMS, Center for Medicaid & Medicare Services; NPP, nonphysician provider.

oncology.1,13 In a survey of physician assistants in oncology practice and prescriptive authority in some states.5 Physi-
practices,13 the majority reported that it took 1 to 2 years of cians and APs must work together to change restrictive
practice to become fully competent in the practice of legislation.
oncology. PAs in this survey also described the most im-
portant mode of obtaining their oncology knowledge base
was direct physician mentoring. The second most important MYTHS DEBUNKED
mode was self-study. A similar study of oncology NPs re- There was a fear among physicians that patients might not
ported that the majority of NPs felt inadequately prepared to be aware that their care was provided by an AP. However,
provide cancer care.14 These barriers are substantial, noting the ASCO Study of Collaborative Practice Arrangements8
that most oncology practices use informal, on-the-job found that 98% of patients with cancer were aware that an
training for new APs.8 Sidebar 1 lists organizations that AP was providing their care. There was also a belief that
provide AP oncology education. patients would be unhappy with the care of the AP. In fact,
Adequate and thorough training and direct mentoring by
the oncologist would enable the oncology AP to become
much more efficient and productive. Ensuring that the AP is
functioning at their highest scope of practice will maximize
SIDEBAR 2. Oncology Advanced Practitioner
the skills of the AP, allowing maximum productivity. It is
Functions and Roles*1,5,8,13
important to understand that adequate AP productivity History taking
measurements are lacking. To only measure AP productivity Physical examination
by relative value units (RVUs) will underestimate the value Diagnostic testing: ordering, interpretation, discussion
the AP brings to a practice.15 Treatment selection
Prescribing (including chemotherapy)
Cancer risk assessment, screening, and management
ROLES OF ADVANCED PRACTITIONERS IN Symptom management
ONCOLOGY Medication management
Advanced practitioners in oncology perform a variety of
In-patient hospital rounds
On-call services
functions (Sidebar 2). Roles vary according to legal scope of Patient education
practice, practice policy, and experience/expertise of the Staff/peer education
AP. To maximize role productivity and efficiency, the AP Survivorship care
must be allowed to work at their full scope of practice as Palliative care
independently as possible. The biggest obstacle to the most Hospice care
effective and efficient utilization of the AP is the lack of full Pain management
Research (including roles as principal or subinvestigator)
Emergent care
Case management
SIDEBAR 1. Organizations Providing Oncology Cancer patient navigator
Education for the Advanced Practitioner Genetic services
Association of Physician Assistants in Oncology
Lung nodule clinic
(www.apao.org)
Community education
American Society of Clinical Oncology (www.asco.org)
Faculty positions outside institution of employment
Advanced Practitioner Society for Hematology and On-
Procedures including (but not limited to) bone marrow
cology (www.apsho.org) aspiration/biopsy, paracentesis, thoracentesis, central line
National Cancer Institute (www.cancer.gov) placement, lumbar puncture, intrathecal chemotherapy
Oncology Nursing Society (www.ons.org) administration, punch biopsy, suturing, surgical assist
*This list is not all inclusive.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e99


WENDY H. VOGEL

this same study found that 92.5% of patients were extremely AP RVUs). Productivity measures should also include value-
satisfied with AP services. Other disciplines have docu- added functions (that might be nonrevenue generating),
mented similar results.16,17 patient satisfaction, and professional satisfaction.
One of the most common reasons that oncology practices Proactive and thoughtful scheduling of patient visits will
have cited as reasons for not adding an AP to the practice is enable the oncologist to offer new patients rapid access.18
physician lack of interest in working with an AP.8 However, Planning for urgent care time slots decreases the use of the
data show that not only is practice productivity increased emergency department, thus increasing patient satisfaction.
with the addition of an AP, professional satisfaction of the Efficient scheduling takes planning in advance and thorough
collaborative oncologist increases as well.8,15 training of scheduling staff and patient education, as well as
There is also misunderstanding about the cost effectiveness the flexibility of the AP and oncologist.
of an AP, especially in terms of AP billing. APs can see and bill Adequate training and mentoring of the AP will enable the
for new patients and can bill all levels of services. Data highest level of productivity in the timeliest manner. This will
consistently show the cost effectiveness of the AP.16,17 also lower burnout rates and increase professional satis-
faction. Thoughtful preplanning for this education and
INTEGRATING THE ADVANCED PRACTITIONER mentoring is required. It is also important to plan for the
INTO THE ONCOLOGY TEAM lengthy time period that could be required for license and
Oncology team members have a shared goal of improving credentialing. Ongoing education is critical for the AP to stay
patient outcomes. Other goals of collaboration might in- current in this rapidly changing field.
clude increased productivity of the practice, improved pa-
tient access to timely care, among others.18 Successful
collaboration takes time and deliberate planning to fully CONCLUSION
engage each members unique strengths and skills. Mea- Collaborative care is required to meet the increasing de-
surement of productivity must include not only individual mands of oncology care. The oncology AP is a cost-effective,
RVUs, but also team-based RVUs (combined physician and integral part of the professional oncology team.

References

1. Kurtin SE, Peterson M, Goforth P, et al. White Paper: The Advanced clinics at national comprehensive cancer network institutions. J Oncol
Practitioner and Collaborative Practice in Oncology. J Adv Pract Oncol. Pract. 2010;6:182-187.
2015;6:515-527. 11. Kurtin S, Viale P, Hylton H, et al. The APSHO practice survey. Paper
2. American Society of Clinical Oncology. The State of Cancer Care presented at: 3rd JADPRO LIVE at Advanced Practitioner Society for
in America: 2015. http://www.asco.org/sites/www.asco.org/files/ Hematology and Oncology Annual Meeting; November 2015; Phoenix,
2015ascostateofcancercare.pdf. Accessed January 16, 2016. AZ Poster JL316.
3. Phillips SJ. 28th annual APRN legislative update: advancements con- 12. McCorkle R, Engelking C, Knobf MT, et al. Transition to a new cancer
tinue for APRN practice. Nurse Pract. 2016;41:21-48. care delivery system: opportunity for empowerment of the role of the
4. National Commission on Certification of Physician Assistants. 2014 advanced practice provider. J Adv Pract Oncol. 2012;3:34-42.
Statistical Profile of Recently Certified Physician Assistants. https:// 13. Ross AC, Polansky MN, Parker PA, et al. Understanding the role of
www.nccpa.net/Uploads/docs/RecentlyCertifiedReport2014.pdf. physician assistants in oncology. J Oncol Pract. 2010;6:26-30.
Accessed January 16, 2016. 14. Nevidjon B, Rieger P, Miller Murphy C, et al. Filling the gap: devel-
5. Vogel WH. Advanced practitioners in oncology: meeting the challenges. opment of the oncology nurse practitioner workforce. J Oncol Pract.
J Adv Pract Oncol. 2010;1:13-18. 2010;6:2-6.
6. Levit L, Smith AP, Benz EJ, et al. Ensuring quality cancer care through the 15. Kosty M, Acheson AK, Tetzlaff ED. Clinical oncology practice 2015:
oncology workforce. J Oncol Pract. 2010;6:7-11. preparing for the future. Am Soc Clin Oncol Educ Book. 2015;35:e622-
7. Erikson C, Salsberg E, Forte G, et al. Future supply and demand for e627.
oncologists : challenges to assuring access to oncology services. J Oncol 16. Martin-Misener R, Harbman P, Donald F, et al. Cost-effectiveness of
Pract. 2007;3:79-86. nurse practitioners in primary and specialised ambulatory care: sys-
8. Towle EL, Barr TR, Hanley A, et al. Results of the ASCO Study of Col- tematic review. BMJ Open. 2015;5:e007167.
laborative Practice Arrangements. J Oncol Pract. 2011;7:278-282. 17. Donald F, Kilpatrick K, Reid K, et al. A systematic review of the cost-
9. Buswell LA, Ponte PR, Shulman LN. Provider practice models in ambu- effectiveness of nurse practitioners and clinical nurse specialists: what is
latory oncology practice: analysis of productivity, revenue, and provider the quality of the evidence? Nurs Res Pract. 2014;2014:896587.
and patient satisfaction. J Oncol Pract. 2009;5:188-192. 18. Shulman LN. Efficient and effective models for integrating advanced
10. Hinkel JM, Vandergrift JL, Perkel SJ, et al. Practice and productivity of practice professionals into oncology practice. Am Soc Clin Oncol Educ
physician assistants and nurse practitioners in outpatient oncology Book. 2013;33:e377-e379.

e100 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


CARE DELIVERY AND PRACTICE MANAGEMENT

Quality and Value: Measuring and


Utilizing Both in Your Practice

CHAIR
Lowell E. Schnipper, MD
Beth Israel Deaconess Medical Center
Boston, MA

SPEAKERS
Anne C. Chiang, MD, PhD
Yale Cancer Center
New Haven, CT

Steven L. DAmato, BCOP, RPh


New England Cancer Specialists
Scarborough, ME
ANNE C. CHIANG

Why the Quality Oncology Practice Initiative Matters: Its Not


Just About Cost
Anne C. Chiang, MD, PhD

OVERVIEW

The nature and cost of cancer care is evolving, affecting more patients and often involving expensive treatment options. The
upward cost trends also coincide with a national landscape of increasing regulatory mandates that may demand improved
outcomes and value, but that often require significant up-front investment in infrastructure to achieve safety and quality.
Oncology practices participating in the American Society of Clinical Oncology (ASCO) Institute for Qualitys Quality Oncology
Practice Initiative (QOPI) and the QOPI Certification Program (QCP) continue to grow in number and reflect changing
demographics of the provision of cancer care. QOPI and QCP benchmarking can be used to achieve quality improvement and
to build collaborative quality communities. These programs may be useful tools for oncology practices to comply with new
legislation such as the Medicare Access and CHIP Reauthorization Act (MACRA).

T he cost of cancer in the United States has risen signifi-


cantly over the past decade. According to the Centers
for Disease Control and Prevention, 8.5% of the adult U.S.
comprehensive detail in the recently released Chapter 800
regarding not only storage and preparation but also the
administration of hazardous drugs.5 The American College
population has been diagnosed with cancer, totaling over of Surgeons has recently mandated implementation of
20 million patients in 2012 and more than 29 million distress screening and survivorship care plans for hospitals
ambulatory visits to physician offices, hospital outpatient to achieve and maintain accreditation of their cancer
offices, and emergency departments.1 The estimated direct programs.6
medical costs for cancer care in the United States in 2011 More recently, MACRA was passed to reform Medicare
were $88.7 billion, with 50% of that cost for physician or payments. This act is intended to focus on payment for value
hospital outpatient office visits, 35% for inpatient stays, and and better care by incentivizing providers to participate in
11% for prescription drugs.2 Projections of growth from 2010 quality programs through either a Merit-Based Incentive Pay-
to 2020 based on 13.8 and 18.1 million cancer survivors, re- ment System (MIPS) or Alternative Payment Model (APM). The
spectively, estimated the costs associated with cancer care to MIPS program involves three components: the physician
be $124.57 and $157.77 billion, respectively, reflecting a 27% quality reporting system (PQRS), the value modifier (value-
increase in medical costs.3 based payment modifier), and the Medicare EHR incentive
The increase in costs is likely multifactorial, involving program. Alternative Payment Models include accountable
more patients, more expensive drugs, and changes in care organizations, patient-centered medical homes, and
cancer care delivery. The upward cost trends also coincide bundled payment models.7
with a national landscape of increasing government and In such an evolving landscape, it is critical to foster a
regulatory mandates that often require significant up- culture of self-examination and quality improvement to
front investment in equipment and infrastructure to maintain a focus on achieving quality and patient-centered
achieve. Such government mandates include the Ameri- care. To this end, the principles and evolution of ASCOs
can Recovery and Reinvestment Act of 2009 enacted on quality programs, QOPI and QCP, make them attractive tools
February 17, 2009, that established incentive payments to for practices to comply with legislation, such as MACRA, that
promote the adoption and meaningful use of qualified require not only benchmarking of standards but also built-in
electronic health records (EHRs) and interoperable health quality improvement mechanisms. QOPI is deemed by the
information technology.4 The U.S. Pharmacopeia Chapter Centers for Medicare & Medicaid Services (CMS) as a
797 describes enforceable sterile compounding standards Qualified Clinical Data Registry (QCDR), with clinical mea-
that apply to chemotherapy preparation, with even more sures that are highly relevant to contemporary ambulatory

From the Yale University School of Medicine, New Haven, CT.

Disclosures of potential conflicts of interest provided by the author are available with the online article at asco.org/edbook.

Corresponding author: Anne C. Chiang, MD, PhD, Yale University School of Medicine, 200 York St., New Haven, CT 06520; email: anne.chiang@yale.edu.

2016 by American Society of Clinical Oncology.

e102 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


WHY QOPI MATTERS: ITS NOT JUST ABOUT COST

practice.8 Participants abstract and submit data for a set of the mean number of medical oncologists per practice in-
core measures as well as additional modules (e.g., disease- creased from 6.7 to 7.8. The participating practices had
specific) that they select. They receive detailed reports of similar geographic distribution across the United States, but
their performance on quality metrics, which allows them to overall, there was an increase in the number of practices that
compare with national benchmark data, to identify areas for are high volume (e.g., with more than 1,500 new patients
improvement and develop focused improvement plans. seen per year). Interestingly, the number of QOPI practices
While QOPI focuses on quality metrics, QCP validates a with employed physicians increased from 15% (82) to 28.7%
practices processes, demonstrating to patients, payers, and (273), and the number of private independent physician
colleagues a commitment to quality. Through practice and practices decreased from 63.8% (344) to 47.9% (455). The
policy review, including an on-site survey, QCP verifies percentage of practices identifying as academic full time
practice standards for safe administration of chemotherapy. increased slightly from 7.2% (39) to 10.5% (100), while the
Some health plans recognize QOPI certification. QCP uses percentage of practices with academic affiliation was stable
national benchmarking standards developed by ASCO, the at 7%. Roughly 6% of practices have a fellowship program.
Oncology Nursing Society (ONS), and other organizations QOPI participation correlates with improvement in scores
that safeguard the practice and patients. over time.9 The expectations of the nature of cancer care is
constantly changing, as evidenced by new standards man-
dated by the American College of Surgeons regarding dis-
CURRENT STATE OF QOPI tress screening and use of survivorship care plans as well as
QOPI has been ASCOs signature quality program for almost the CMS requirement that practices deliver care using an
10 years, and since 2006, over half of all U.S.-based medical electronic medical record (EMR). These mandates parallel
oncologists have participated, numbering over 1,000 prac- the shift from oncologists being self-employed to employed;
tices representing 7,000 oncologists. In addition, international hospital-affiliated practices certainly may have increased ac-
practices within the European Union, Argentina, Brazil, India, cess to other services. In fact, patient access to these in-
and Saudi Arabia were recently allowed to participate in QOPI, terdisciplinary services such as social work increased from 45%
with 16 practices participating since fall 2015. A total of 994 (188) to 55.5% (466), nutrition from 38.2% (159) to 52.3%
practices submitted over 32,000 charts for the three most (324), and genetic counseling from 31.9% (132) to 38.7% (324).
recent abstraction rounds (fall 2013, spring 2014, and spring Furthermore, 90.7% (564) of practices now have an EMR.
2015). These results were compared with data from 5 years
ago (fall 2009, spring 2009, and fall 2010) and demonstrate
notable changes in the demographics, the practice model of FUTURE STATE OF QOPI
employment, and the range of services provided (Table 1). QOPI continues to offer new opportunities and enhance-
Of note, the QOPI fall 2014 round did not take place because ments in its services to aid practices in assessing quality and
of a change in vendor. to comply with new federal requirements. CMS has approved
Several interesting trends were noted when comparing QOPI as such a registry for oncologists to satisfy PQRS mea-
data from 2009 to 2010 with data from 2013 to 2015. First, sures. Oncology practices registered with QOPI can fulfill CMS
PQRS requirements by manually or electronically submitting
data through QOPI for the oncology measures group or by
submitting data through QOPI via a QCDR reporting pathway,
KEY POINTS which offers 20 measures chosen by ASCO.10 Since QOPI has
been deemed as a QCDR, it is anticipated that QOPI will
ASCOs quality programsQOPI and QOPI QCPare
attractive tools for practices to comply with new
become the primary mechanism for ASCO members to report
legislation, such as MACRA, that require not only their quality as required by MIPS and in APMs.
benchmarking of standards but also built-in quality In fall 2015, ASCO began piloting eQOPI to decrease the
improvement mechanisms. burden of manual chart abstraction by implementing
Oncology practices participating in QOPI and QCP technology to automate data abstraction. The technology
continue to grow in number and reflect changing enables the required data elements in the EHR to be mapped
demographics of the provision of cancer care. and extracted (or in some cases, data can be pushed); this
QOPI and QCP benchmarking can be used to identify allows the majority of the elements required for the clinical
areas for improvement, provide comparative metrics for quality measures to be captured automatically, leaving
quality initiative projects, and build collaborative quality only a small percentage for manual abstraction. The dif-
communities.
ferences in the scope and reporting pathways for classic
QCP provides hands-on expert gap analysis assessment
of a practice and a road map toward quality through
QOPI and eQOPI are highlighted in Table 2. Many of the
ASCO certification status. performance measures in QOPI are already based on ASCO
ASCO has multiple offerings as vehicles for diverse clinical practice guidelines, and as guideline scope and
practices to join a quality community working toward volume increase, so will the available measures yet to be
improving cancer care. incorporated into QOPI, with an increased emphasis on out-
comes measures and measures that reflect patient experience,

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e103


ANNE C. CHIANG

TABLE 1. ASCO 2015 QOPI Practice Demographic Data

Fall 2009Fall 2010 (609) Fall 2013Spring 2015 (994)


Mean (SD) or N (%) Mean (SD) or N (%)
Number of Medical Oncologists 6.7 (10.0) 7.8 (13.6)
Number of New Patients
< 500 96 (31.8) 251 (31.5)
501-1,500 136 (45.0) 326 (40.8)
> 1,500 70 (23.2) 221 (27.7)
Census Region
Midwest 198 (32.5) 302 (30.4)
Northeast 111 (18.2) 192 (19.3)
South 195 (32.0) 308 (31.0)
West 105 (17.3) 192 (19.3)
Site Affiliation
Academic Full Time 39 (7.2) 100 (10.5)
Private With Academic Affiliation 40 (7.4) 67 (7.1)
Employee 82 (15.2) 273 (28.7)
Private Independent 344 (63.8) 455 (47.9)
Fellowship Program 34 (6.4) 55 (5.8)
Social Work 188 (45.1) 466 (55.5)
Dietician/Nutritionist 159 (38.2) 442 (52.3)
Genetic Counselor 132 (31.9) 324 (38.7)
Quality Improvement Training 320 (52.5)
Electronic Medical Records 564 (90.7)

as well as patient-reported outcomes. For instance, QOPI began A total of 298 practices have achieved QOPI certification,
pilot testing value measures in 2015, based on lists of tests out of 525 applicants representing 336 unique practices.
and procedures ASCO developed as part of its participation Subsequently, 189 practices have applied for recertifica-
in the Choosing Wisely campaign that are not evidence-based. tion, with 96% receiving certification. Most certified practices
As the technology that powers QOPI becomes more sophis- are small to midsized with one to three full-time equivalents
ticated to decrease the workload on practices required for (FTEs; 30%) or four to seven FTEs (35%). The rest of the
quality reporting, QOPI-based quality assessment will become practices are larger with seven to 15 FTEs (23%), 25 to 50
a key and value-added component of ASCOs rapid-learning FTEs (4%), and even 50 to 100 FTEs (1%). Only 25% of the
system, CancerLinQ, where abstraction of data elements from practices identify themselves as private independent and
the EHR will be fully automated.11,12 17% as academic full-time practices. The largest group of
practices (35%) identify themselves as employed, and an
QOPI CERTIFICATION PROGRAM additional 14% are hospital-employed. The practices are
Although QOPI focuses on data regarding individual patients distributed across the United States, with 23.4% in the
and providers only, certification of practices by ASCO South, 28.2% in the East, 33.1% in the Midwest, and
requires a minimum QOPI Quality Score, submission of 15.2% in the West (Fig. 1; unpublished data, T. Gilmore,
documentation for 20 standards, and an on-site survey.13 ASCO).
The certification process provides a comprehensive evalu- In general, most practices performed well, with an average
ation of patient care including examination of documen- of 4.1 missed standards out of 20. Eight standards dem-
tation, policies and procedures, and administration of onstrated 100% concordance in the last round. In rounds 9,
chemotherapy. A surveyor travels to multiple sites in the 10, and 11, the top six missed standards were qualifications,
practice and has the opportunity to observe and evaluate chart documentation, double check in administration,
processes directly. The surveyor and ASCO QCP staff have documentation for each visit, oral chemotherapy monitor-
the opportunity to share feedback and best practices be- ing, and patient education. The top six most commonly met
tween sites to help address standards that are initially not in standards include both process and policy measures: con-
accordance. The benefits of participating in the QCP pro- sent policy, double check of order, day of treatment as-
gram include identifying areas for quality improvement and sessments, referrals, toxicity assessment available prior to
achieving QOPI certification status by ASCO, as well as payor writing chemotherapy order, and cumulative dosing (un-
benefits in some cases. published data, T. Conti-Kalchik, ASCO).

e104 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


WHY QOPI MATTERS: ITS NOT JUST ABOUT COST

TABLE 2. Differences in Scope and Reporting and a positive effect on practices. To understand the effect
Pathways for Classic QOPI and eQOPI on practices, a survey in 2013 was sent to 1,450 participants
at 850 practices to assess which measure modules/standards
Classic QOPI were selected by QOPI/QCP participants as the basis for local
191 Measures quality initiative efforts and to understand the nature of the
7 Modules (Existing) improvement initiatives.14
1 Module (New) Out of 89 respondents, 96% (85/89) reported that QOPI/
3 Palliative Care
QCP led to quality improvement efforts. Respondents were
asked to select module(s) that spurred subsequent quality
3 Reporting Pathways (New)
improvement activities: core measures (57%; 45), symptom/
3 QOPI Certification Program
toxicity management (48%; 38), end-of-life care (38%; 29),
3 PQRS Oncology Measures Group breast cancer (13%; 10), colorectal cancer (10%; 8), non-
3 Patient-Centered Oncology Practice Hodgkin lymphoma (6%; 5), and nonsmall cell lung cancer
eQOPI (4%; 3). Related to the QCP structural safety standards,
38 Measures participants reported quality improvement projects as fol-
6 Modules (Existing) lows: chemotherapy planning/chart documentation (39%;
2 Reporting Pathways (New) 31), general chemotherapy standards (30%; 24), monitoring
3 QOPI Certification Program and assessment (29%; 23), chemotherapy administration
(27%; 21), chemotherapy orders (23%; 18), staffing (16%;
3 Qualified Clinical Data Registry
13), and drug preparation (9%; 7).
Practices reported that QOPI measures improved in
ASCO continues to expand and improve the QCP program subsequent rounds as a result of specific projects (22/25;
by exploring medical liability insurance discounts and new 88%); 100% felt that these quality improvement projects
measures to respond to MACRA MIPS/APM requirements affected their practices for the better. Quality improvement
and a pilot program to allow international practices to apply project results were presented primarily in practice meet-
for QOPI certification for the first time in 2016. ings (74%; 26), hospital or community forums (17%; 6), ASCO
Quality Care Symposium, or other meetings (9%; 3). No
projects reached publication. Of note, 17 of 31 respondents
QOPI IN ACTION who reported practice status indicated achieving QOPI
QOPI and QCP have succeeded in spurring local quality certification. Specific quality improvement projects included
improvement efforts that have led to score improvements, building oral chemotherapy order sets and modified anti-
achievement of standards, increased discussion of quality, emetic regimens in chemotherapy protocols, improving

FIGURE 1. QOPI Certification Program Growth, December 2015

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e105


ANNE C. CHIANG

documentation of distress, pain, performance status, smoking ambulatory clinics throughout Smilow to comply with the
status and referrals, chemotherapy consent, and intent of American College of Surgeons mandate for psychosocial
treatment. Practices also improved safety by initiating distress assessment.18 Other areas of noncompliance have
semiannual review and gap analysis of adherence to ASCO/ been identified and addressed. An oral chemotherapy ad-
ONS Chemotherapy Safety Standards, improving the medi- herence program including specialty pharmacy and EMR
cation reconciliation process, and training for oral chemotherapy. workflows was recently established and implemented
ASCO continues to build quality improvement resources through all clinics (unpublished data, K. Adelson). QOPI
and practical training, such as the Quality Training Program, certification was achieved for the Smilow Cancer Hospital
launched in 2013 as part of its Institute for Quality.15 The network and continues to spur quality improvement pro-
Quality Training Program uses in-person training sessions jects (e.g., pain assessment and immunotherapy toxicity
and coaching to teach quality improvement curriculum and education and management).
skills throughout an improvement project and to develop
quality leaders and champion quality improvement. Since
2013, 60 individuals have participated in this program CONCLUSION
(unpublished data, E. Holton, ASCO). QOPI and QCP continue to be ASCOs signature quality
programs, with continuing growth in the number and extent
of participating practices. With the increasing emphasis on
A QUALITY IMPROVEMENT COMMUNITY value because of the rising costs of cancer care, it is essential
QOPI has served as a platform to create a quality community to have constant development and validation of quality
in two published reports by the Michigan Oncology Quality measures that reflect the changing nature of oncology (e.g.,
Consortium and the National Cancer Institute Community oral chemotherapy adherence, Choosing Wisely measures,
Cancer Centers Program (NCCCP). In each case, a network of and response to the demands of MACRA). QOPI bench-
diverse practices agreed to benchmark their aggregate QOPI marking can be used to identify areas for improvement,
and collaborate on quality improvement initiatives to im- provide comparative metrics for quality improvement
prove quality of care and to identify best practices.16,17 The projects, and build collaborative quality communities.
NCCCP practices demonstrated a measurable increase in Widespread EMR adoption will allow eQOPI to help reduce
each sites QOPI scores from fall 2010 to fall 2013, with the the time burden of abstraction, and delivery of the QOPI
exception of one parameter. quality assessments through CancerLinQ will take advantage
Another example illustrates how QOPI can be used to of the fully automated data abstraction capabilities of that
achieve hospital integration of a network of community platform. QCP provides hands-on expert gap analysis as-
practices. The Smilow Cancer Hospital provides cancer sessment of a practice and a road map toward quality
services by Yale Cancer Center clinicians in New Haven, through ASCO certification status. The ASCO Institute for
Connecticut, or in one of nine Care Center community lo- Quality also offers the Quality Training Program to help train
cations. Started in 2012 by the acquisition of community and coach interdisciplinary teams in quality improvement
practices, all Smilow Cancer Hospital Care Centers are fully initiatives and develop leadership. In summary, ASCO has
integrated practice sites using a provider-based model and multiple offerings as vehicles for diverse practices to join a
unified EMR baseline data across all centers. They created a quality community working toward improving cancer care.
quality council to focus on clinical quality and patient safety
and to coordinate quality improvement activities. In spring
2012, 49% (217/445) of the patients at Smilow Cancer ACKNOWLEDGMENT
Hospital Care Centers had documented emotional assess- I would like to acknowledge ASCO staff members Terry
ments, with compliance in two sites below 12%. Distress Gilmore, Tara Conti-Kalchik, Elaine Holton, Stephanie Crist,
screening was implemented in these two clinics through a and Robert S. Miller, MD, for their help in providing data and
Quality Training Program pilot by using quality improvement in reviewing the manuscript. I would to acknowledge Joe
tools and subsequently expanded in 2015 to 2016 to all Jacobson, MD, for his help in reviewing the manuscript.

References
1. Centers for Disease Control and Prevention. Cancer. http://www.cdc. 4. Centers for Medicare & Medicaid Services. Electronic Health Records
gov/nchs/fastats/cancer.htm. Accessed February 10, 2016. (EHR) Incentive Programs. https://www.cms.gov/Regulations-and-Guid-
2. American Cancer Society. Economic Impact of Cancer. http://www. ance/Legislation/EHRIncentivePrograms/index.html?redirect=/ehrincen-
cancer.org/cancer/cancerbasics/economic-impact-of-cancer. Accessed tiveprograms. Accessed February 10, 2016.
February 10, 2016. 5. U.S. Pharmacopeial Convention. USP Compounding Standards & Resources.
3. Mariotto AB, Yabroff KR, Shao Y, et al. Projections of the cost of cancer care http://www.usp.org/usp-healthcare-professionals/compounding. Accessed
in the United States: 2010-2020. J Natl Cancer Inst. 2011;103:117-128. February 10, 2016.

e106 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


WHY QOPI MATTERS: ITS NOT JUST ABOUT COST

6. American College of Surgeons. Cancer Program Standards (2016 13. Gilmore TR, Schulmeister L, Jacobson JO. Quality Oncology Practice
Edition). https://www.facs.org/quality%20programs/cancer/coc/ Initiative Certification Program: measuring implementation of che-
standards. Accessed February 10, 2016. motherapy administration safety standards in the outpatient oncology
7. Hahn J, Blom KB. H.R. 2: The Medicare Access and CHIP Reauthorization Act setting. J Oncol Pract. 2013;9(2 Suppl)14s-18s.
of 2015. Congressional Research Service. http://democrats.waysandmeans. 14. Chiang AC, McNiff K, Kadlubek P, et al. Assessment of quality im-
house.gov/sites/democrats.waysandmeans.house.gov/files/documents/ provement efforts by ASCOs Quality Oncology Practice Initiative
CRS%20report%20on%20HR%202.pdf. Accessed February 10, 2016. (QOPI) participants. J Clin Oncol. 2013;31:15s (suppl; abstr 55).
8. Centers for Medicare & Medicaid Services. Qualified Clinical Data 15. Kamal, AH, Quinn, D, Gilligan TD, et al. Feasibility and effectiveness of a
Registry Reporting. https://www.cms.gov/medicare/quality-initiatives- pilot program to facilitate quality improvement learning in oncology:
patient-assessment-instruments/pqrs/qualified-clinical-data-registry- experience of the American Society of Clinical Oncology Quality Training
reporting.html. Accessed February 10, 2016. Program. J Oncol Pract. Epub 2015 Aug 18.
9. Neuss MN, Malin JL, Chan S, et al. Measuring the improving quality of 16. Blayney DW, Stella PJ, Ruane T, et al. Partnering with payers for success:
outpatient care in medical oncology practices in the United States. J Clin quality oncology practice initiative, blue cross blue shield of michigan,
Oncol. 2013;31:1471-1477. and the michigan oncology quality consortium. J Oncol Pract. 2009;5:
10. ASCO Institute for Quality. QOPI Certification Standards. http://www. 281-284.
instituteforquality.org/qopi/qopi-certification-standards. Accessed February 17. Siegel RD, Castro KM, Eisenstein J, et al. Quality improvement in the
10, 2016. national cancer institute community cancer centers program: the
11. Jacobson JO, Neuss MN, Hauser R. Measuring and improving value of care in quality oncology practice initiative experience. J Oncol Pract. 2015;11:
oncology practices: ASCO programs from Quality Oncology Practice Initiative e247-e254.
to the rapid learning system. Am Soc Clin Oncol Educ Book. 2012;e70-e76. 18. Chiang AC, Buia Amport S, Corjulo D, et al. Incorporating patient-
12. Shah A, Stewart AK, Kolacevski A, et al. Building a rapid learning health reported outcomes to improve emotional distress screening and as-
care system for oncology: why CancerLinQ collects identifiable health sessment in an ambulatory oncology clinic. J Oncol Pract. 2015;11:
information to achieve its vision. J Clin Oncol. 2016;34:756-763. 219-222.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e107


CARE DELIVERY AND PRACTICE MANAGEMENT

The Oncology Care Model: The


Man Behind the Curtain

CHAIR
Christian A. Thomas, MD
New England Cancer Specialists
Scarborough, ME

SPEAKERS
Jeffery C. Ward, MD
Swedish Cancer Institute Edmonds
Edmonds, WA

Ronald Michael Kline, MD


Center for Medicare & Medicaid Innovation
Washington, DC
THE ONCOLOGY CARE MODEL: A CRITIQUE

The Oncology Care Model: A Critique


Christian A. Thomas, MD, and Jeffrey C. Ward, MD

OVERVIEW

Rapidly increasing national health care expenditures are a major area of concern as threats to the integrity of the health care
system. Significant increases in the cost of care for patients with cancer are driven by numerous factors, most importantly the
cost of hospital care and escalating pharmaceutical costs. The current fee-for-service system (FFS) has been identified as a
potential driver of the increasing cost of care, and multiple stakeholders are interested in replacing FFS with a system that
improves the quality of care while at the same time reducing cost. Several models have been piloted, including a Center for
Medicare & Medicaid Innovation (CMMI)sponsored medical home model (COME HOME) for patients with solid tumors
that was able to generate savings by integrating a phone triage system, pathways, and seamless patient care 7 days a week to
reduce overall cost of care, mostly by decreasing patient admissions to hospitals and referrals to emergency departments.
CMMI is now launching a new pilot model, the Oncology Care Model (OCM), which differs from COME HOME in several
important ways. It does not abolish FFS but provides an additional payment in 6-month increments for each patient on active
cancer treatment. It also allows practices to participate in savings if they can decrease the overall cost of care, to include all
chemotherapy and supportive care drugs, and fulfill certain quality metrics. A critical discussion of the proposed model,
which is scheduled to start in 2016, will be provided at the 2016 American Society of Clinical Oncology (ASCO) Annual Meeting
with practicing oncologists and a Centers for Medicare & Medicaid Services (CMS) representative.

N ational health care expenditures (HCEs) are a substantial


part of the national spending and currently account for
17.1% of the U.S. gross domestic product, significantly more
There is broad consensus that this trend is not sustainable
at the current rate. Private and public stakeholders are
discussing mechanisms to curb spending while at the same
than other developed countries, which typically spend be- time maintaining high-quality care. Because physician be-
tween 8% and 12% of their gross domestic product on health havior can be influenced by financial incentives,4 it has been
care. Despite high spending, several important health out- postulated that tying payment directly to the quantity of
comes measuresfor example, infant mortalitydo not re- performed services, the backbone of the FFS system, in-
flect the current level of health care spending, supporting the centivizes physicians to perform more services to improve
notion that there is a disconnect between money spent and financial gain. The flipside of the argument is the Health
the quality of care produced by the system. When national Maintenance Organization experience of the early 1990s
health care systems are compared according to country, the
where FFS was replaced by a fixed monthly payment for each
United States currently ranks 54th when factors such as effi-
patient but not tied to patient encounters or specific ser-
ciency, life expectancy, and health care cost are considered.1
vices. This led to inadequate access to care with poor patient
A major concern is the significant growth rate of HCEs that
care and satisfaction.
outpace other national financial benchmarks and continue
So how is a rational system of providing the right health
to rise at annual levels of greater than 5%. The inflation of
care to the right patient at the right time to be designed?
health carerelated expenditures is mainly driven by two
If FFS is abandoned for an improved way to deliver and pay
major factors: hospital-related expenditures and drug prices.
for health care, the following elements have to be met:
U.S. expenditures on pharmaceuticals are more than twice
that of any other member country of the Organisation for 1. A payment system that adequately covers the true
Economic Cooperation and Development.2 cost of carethe current Medicare fee schedule is
Almost no other area of medicine is affected more by the underfunding the cost of care by 20% to 30%, forcing
steep increase in the cost of pharmaceuticals than cancer providers to use other resources to cover the cost of
care where drug prices can easily top $10,000 per month per care for Medicare beneficiaries such as increasing their
patient.3 fees for private payers;

From the New England Cancer Specialists, Scarborough, ME; Swedish Cancer Institute, Edmonds, WA.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Christian A. Thomas, MD, New England Cancer Specialists, 100 Campus Dr., Suite 108, Scarborough, ME 04074; email: thomac@newecs.org.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e109


THOMAS AND WARD

2. A system that allows providers and their staff to care department referrals for patients who could be ade-
for patients in a variety of ways including phone calls, quately cared for in the office.5 This example of increasing
emails, and other forms of contact in addition to the value of care by reducing cost while at the same time
traditional face-to-face encounters; improving the quality of care and leading to higher patient
3. A system that allows easy and unrestricted access for satisfaction is an encouraging example of positive health
patients; care reform.
4. A system that ties successful adherence to nationally Unfortunately, CMMI decided to discontinue further fund-
accepted quality standards and patient satisfaction ing for COME HOME and has presented a new model aimed
directly to physician reimbursement; at reducing cost of care for medical oncology patients while
5. A system that provides a mechanism for controlling at the same time attempting to improve the quality of care,
the cost for drug and hospital spending. the Oncology Care Model (OCM). This model uses two types
of additional payments in addition to the usual FFS pay-
ments and other payments, for example, for chemotherapy
THE ONCOLOGY CARE MODEL administration: a performance-based incentive as well as
Federal legislation now mandates CMS to fund alternate bundled monthly payments for patients undergoing che-
payment models to deliver health care, and CMS has es- motherapy. In exchange, oncology practices are obligated to
tablished the Center for Medicare & Medicaid Innovation establish systems aimed at providing a higher level of care
(CMMI) to explore options. From 2013 to 2015, a $20 million with additional reporting requirements to CMMI. As detailed
grant was provided to 12 medical oncology practices for further below, practices will receive an additional payment
the COME HOME project, which used three novel mecha- of $160 per beneficiary per month (the bundle) for any 6-
nisms to control the cost of care for patients with common month period during which a patient is undergoing active
cancers: chemotherapy treatment in a medical oncology practice.
1. The systematic use of a phone triage system to de- In return, practices agree to strict reporting and quality
termine the acuity of a patients complaint and offer standards:
them one of three options: advise over phone for
minor complaints, a same-day appointment in the
Provide patient navigation;
Document a care plan that contains the 13 components
medical oncology office for more urgent issues, or a as outlined by the Institute of Medicine;
direct referral to the nearest emergency department
for the sickest patients;
Provide 24-hour-a-day, 7-day-a-week patient access to
an appropriate clinician who has real-time access to the
2. Extended office hours on evenings and weekends so practices medical records;
that patients requiring an assessment or office services
such as antiemetics, hydration, antibiotics, or other
Treat patients with therapies consistent with nationally
recognized clinical guidelines;
injections would not have to visit a hospital or emer-
gency department;
Use data to drive continuous quality improvement;
Use an ONC-certified electronic health record and attest
3. The use of treatment pathways that were created by to stage 2 of meaningful use by the end of the third
the participating practices to create predictable, cost- model performance year.
effective, but high-quality care. In addition, private payers were invited to apply, extending
Although savings varied geographically, as a whole the potential reach of the project to patients not covered
participating COME HOME practices were able to show by CMS.
substantial savings with lower costs largely attributable The OCM is often characterized as an episode-of-care or
to avoiding costly hospital admissions and emergency bundled reimbursement model but is in reality a FFS and
shared savings model. The shared savings are predicated
on a calculated bundle, but the reimbursement application
KEY POINTS would more honestly be called an Oncology Accountable
Care Organization.
The rapidly increasing cost of health care threatens to There are theoretical advantages to a true bundle for both
destabilize the health care system. payers and providers. The payers costs are predictable for
New systems are needed, and CMS has proposed a new an individual patient for a set period time and the economic
pilot model, the Oncology Care Model, that provides incentive toward high-cost, high-quantity care, inherent in
payments to practices in addition to the traditional FFS if FFS medicine, is replaced by physician accountability for
practices can decrease overall cost of care and fulfill
costs and quality they can control. The provider has freedom
quality criteria.
This article and its corresponding Education Session at from payer micromanagement and, ideally, adequate and
the 2016 ASCO Annual Meeting are aimed at providing predictable payments. Then, released from the constraints
a critical overview of the model by practicing of practicing to billing codes, practices have the flexibility
physicians and a CMS representative. necessary to seek high-quality care with fewer resources
through innovative care redesign. But the key to these

e110 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


THE ONCOLOGY CARE MODEL: A CRITIQUE

advantages is that the bundle has replaced FFS reimburse- sequence of shorter repeating bundles. ASCO has put
ment.6 Unfortunately, OCM fails to do that. forward a monthly bundle in its own payment reform
The Accountable Care Organization model has yet to proposal.8
deliver on its promise of high-quality, low-cost care,7 per- The all-inclusive nature of the bundle has also been
haps because simply adding incentives to decrease costs on controversial. Conceptually, the aim of a bundle is to transfer
top of the existing payment system that incents increased risks associated with high levels of spending from payers to
costs does not solve the problem. At the best, it requires a the providers doing the spending. Presumably, the provider
very complicated balancing act of incentives and quality then decreases the spending. However, within an all-
accountability. At its worst, it is the hope that two wrongs inclusive bundle, there is spending, or risks, that the pro-
make a right. Skeptics might suggest that participants, vider can control and risks that he cannot. Economists refer
constrained by continued dependence on FFS reimburse- to these two categories of financial risk as technical risk and
ment from capitalizing on a redesign of the way they deliver probability risk.
services, will be best served financially by collecting their FFS Technical risk is a function of the practices clinical skill and
billings and management fee, go through the motions of efficiency in managing the diagnosis and treatment or other
CMS dictates, but avoid real innovative efforts and hope that aspects of care that are under the physicians control and
health care inflation and a bit of luck will result in some that he is able to impact either by controlling utilization or
additional booty. cost, preferably both. Probability risks are random or un-
predictable events that we classically buy insurance to
mitigate. In theory, a bundle should only include technical
LOGISTICAL HURDLES OF THE ONCOLOGY CARE risks and the probability risks should remain with the payer.
MODEL BUNDLE Practically, this can be very difficult.
However, it is the bundle construct that makes OCM both When ASCOs payment reform work group attempted to
novel and controversial. Additionally, because one can easily split patient care into oncology related versus other medical
envision that the CMMI pilot model is the precursor of a real care, it quickly became apparent that adjudicating which CPT
bundled reimbursement model, it requires careful scrutiny. and ICD-10 codes should be attributed to the oncologist
The OCM takes a simple approachall spending on all care was simply impractical. Furthermore, several pilot projects
provided to the patient with a particular cancer is in a bundle have now demonstrated that medical oncology homes
that starts with the initiation of chemotherapy and lasts for can dramatically lower emergency department and hospital
6 months. This construct has raised a number of concerns. costs,9-11 broadly indicating that there is substantial tech-
The OCM bundle reimbursement is constructed based on nical risk within patient care. CMS has indicated that the
an individual practices historical costs when available and probability risk that does exista patient with higher than
on regional or national data adjusted for geographic vari- average comorbidities or the patient who is in an auto-
ation when historical costs are not available. The focus on mobile accident on the way to their chemotherapy ap-
competing against your own data guarantees that over time pointment, for examplecan be mitigated through risk
opportunities for new cost sharing diminishes. Although adjustments based on patient and service characteristics
CMMI may not choose to ratchet down the reimbursement in the first example and through reinsurance or stop-loss
for a bundle during the pilot model, the history of Medicares provisions in the latter case. Of course, the most important
oldest bundle, dialysis reimbursement, strongly suggests mitigation of risk in the pilot model is the provision of one-
that is exactly what will happen. sided risk, clearly a carrot that will ultimately be replaced
The choice of 6 months is arbitrary. In other specialties, with a stick.
CMS and commercial payers have defined an episode of
care to be a full course of treatment of a specified treat-
ment. In this scenario, adjuvant concurrent fluorouracil THE ECONOMICS OF DRUGS IN A BUNDLE
(5-FU) and radiation for rectal cancer is a shorter episode and The RAND Corporation estimated that 25% to 40% of the
FOLFOX (folinic acid, 5-FU, and oxaliplatin) for colon cancer OCM bundle is chemotherapy and supportive care drugs.12
is a longer episode. In the OCM model, the first patient is Among key opinion leaders and policymakers, the inclusion
paid a 6-month bundle for 5 weeks of treatment and the of drugs in the oncology bundle seems a foregone conclu-
second patient is paid the same bundle for 24 weeks of sion. It is assumed that (1) physicians, not patients, control
therapy. Unless, of course, the second patient has some demand; (2) practices generate substantial revenue from
delays in treatment, then the practice may be paid two drugs via the buy-and-bill system; (3) if oncologists are at
6-month bundles for 27 weeks of therapy. A study com- financial risk for the drugs, they will choose the least costly
missioned by CMS and performed by the RAND Corporation regimens when efficacies are similar; and (4) because on-
demonstrated that gaps of this sort in courses of the same cologists will become more price sensitive, they will seek out
therapy are common. One can envision that the longer the better prices through negotiations with manufacturers
bundle the greater the variation of practice costs within a and group purchasing organizations. Although theoretically
bundle and greater the opportunity for selection bias, either compelling, there is reason to question the practical validity
intentionally or inadvertently. Critiques have suggested a of these claims.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e111


THOMAS AND WARD

If drugs are to be appropriately included in a bundle, Today, that means developing bundles based on the genetic
analysis should conclude that drug spending is a technical profiles of cancers. In a fairly short time, nonsmall cell lung
risk. Empirical evidence of this is limited. One often-cited cancer has evolved from one monolithic cancer and four
example of drugs being a technical risk is the Zaltrap story.13 equally efficacious treatment regimens to at least four
Zaltrap was launched in 2012 at twice the price of Avastin, a different cancers with very different therapies and associ-
drug with similar outcomes. Physicians at Memorial Sloan ated personalized costs. Unfortunately, it would be naive to
Kettering Cancer Center announced in a New York Times assume that any payment system will be able to keep up with
Op-Ed piece that they would not prescribe Zaltrap, and its the personalized medicine revolution. It is little wonder that
price was reduced by 50%. Although this may have em- UHC and The University of Texas MD Anderson Cancer
boldened oncologists who have increasingly decried high- Center have chosen head and neck cancer as their first
priced drugs, the average monthly launch prices of cancer effort at a bundled payment contract. It is a disease for
drugs since then suggest that the pharmaceutical industry which drug therapy and treatment options are limited.
has been unfazed. Even then, they have eight distinct treatment/payment
Another expectation if drugs are a technical risk is that bundles.15 Due to the impracticality of managing granular
practices should have leverage over the acquisition prices of bundles, it is anticipated that the OCM bundles will be
pharmaceuticals. Although there are examples of volume fairly generic buckets.
discounts for large purchasers and group purchasing orga- In that circumstance, the other way to manage the risk
nizations, this applies only to generic medications and the of the bundle will be for practices to get big. Large volumes
infrequent circumstance in which there are multiple infus- of patients can then mitigate the risks associated with any
able competitors with similar efficacy within a therapeutic one individual. This would be expected to further the con-
class. Oncology practices have no control over the price of solidation of oncology into larger groups through mergers,
novel agents, and oral drug prices are negotiated by payers. acquisitions, and affiliations both in independent and hospital-
For these reasons, the prices of new cancer drugs, those that based practices. This may prove acceptable to health policy
drive costs the most, should be considered a probability risk. makers but could have important unintended consequences;
Proponents of drugs in the bundle will argue that the there is very limited evidence that consolidation improves
inability to control price is outweighed by the oncologists either quality or access to care.
ability to impact utilization. They argue that the 20% to 30% The third way to mitigate the risk of drugs in the bundle is
of annual spending on off-label drugs represents tremen- to move beyond efficiency in drug choice and utilization to
dous waste in the system, driven by FFS medicine and buy- the realm of stenting on care. OCM promises to monitor
and-bill drug reimbursement. The evidence for this is sparse. quality of care and to hold practices accountable for it, but
Brooks et al14 reported in an analysis of FFS Medicare data the task is not a simple one. It is hypocritical to believe that
that only 10% of regional variation in spending was a result of buy-and-bill drives overutilization and to ignore the in-
drug choice, and that drug costs were only 16% of the total centive to underuse inherent in drugs in a bundle.
spend. This was dwarfed by the 67% of regional spending
variation related to acute hospital care, accounting for 48%
of the total spend. THE ALTERNATIVE: PATHWAYS
A real-life example of the principles of probability and An alternative to drugs in the bundle, one that may still
technical risks as they pertain to oncology may be found in assure payers that drugs are being used appropriately, are
the recently reported United Health Care (UHC) dem- value-based treatment pathways.16 Evidence-based with
onstration project. Margins on drugs were replaced by a the purpose of improving value, quality, and safety, path-
management fee to remove profit via drug choice, and ways have been shown to reduce variance in drug regimens,
practices were incentivized to decrease hospital and doses, schedules, and treatment duration. Several studies
emergency department services through shared savings have shown savings with the utilization of what must be
and a bundling of inpatient evaluation and management considered rudimentary pathways and less than stellar path-
services. Drug costs increased, but the demonstration was way compliance.17,18 One project reduced the 12-month
determined a success because of impressive decreases in average costs for chemotherapy and supportive care drugs
acute-care costs.9 One interpretation of these data is that for patients with lung cancer by 37% or $9,747.18
the probability risks (drug costs) could not be controlled, but Another advantage to pathways over bundles is that,
the technical risks associated with hospital and emergency as opposed to bundles developed by payers, pathways can
department services were. be developed by oncologists through transparent, peer-
reviewed, committee-selected regimens that incorporate
efficacy, toxicity, and cost considerations. ASCO recently
MANAGING A BUNDLE ON DRUGS published a policy statement providing direction and rec-
The risks of a bundle are dependent on its variability. There ommendations regarding the development and utilization of
are three ways to manage the variable risk. One way is pathways in clinical practice.19 As pathway development ma-
to define bundles with enough granularity to minimize tures, they will incorporate more sophisticated value models
the variability between patients and their associated costs. that will account not only for drug costs but also the total

e112 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


THE ONCOLOGY CARE MODEL: A CRITIQUE

costs of a treatment regimen to include toxicity, ancillary brings together a true bundle without FFS to reimburse the
support services, acute care, and patient productivity loss. care that an attentive and skilled physician can control and a
robust pathways program that will negate a perceived need
ONCOLOGY CARE MODEL II: A PATHWAY/BUNDLE to change the way we pay for drugs.
HYBRID ASCO is to be commended to convene an Education
Criticisms of OCMs reliance on competition against oneself, Session at its 2016 Annual Meeting during which CMMI will
an arbitrary 6-month episode, and fuzzy details on estab- be able to present its model and have an open discussion
lishing limits on financial risk are details that can most cer- with oncology practice representatives from various back-
tainly be overcome if CMMI is introspective in its evaluation grounds about the models feasibility. It is clear that any
and willing to make incremental improvements. practices participating in the CMS model will have to in-
However, the model has two Achilles heels that it must stitute significant structural changes to fulfill these criteria
consider if OCM is to become a win-win-win for patients, and only the future will tell if the investment of time and
providers, and payers. First, it must divorce itself from the resources will benefit patients while at the same time
reliance on FFS as its backbone of reimbursement. Only then bending the cost curve for oncology. In reality, the model is
can it align incentives with innovative solutions that can not designed to achieve at least the second goal, reducing
produce both high-quality and efficient cancer care. the cost of care. This is because the main drivers of ex-
Then it must consider that drugs in a bundle may mitigate pensive care are not addressed in the model (i.e., the cost
cost and variability for payers, but this is only accomplished of hospital care and the ever-increasing cost of cancer
by putting patients and practices at unacceptable risks. drugs). In addition, other expensive cost centers for patients
Bundled drugs are structured to require the doctor to run a with cancer such as biopsies, surgical procedures, or ra-
gauntlet between rational care and rationed care through diation therapy are also not part of the model, making it
the use of hidden incentives. Pathways, alternatively, use impossible for medical oncology practices to affect the
transparency, consensus, and evidence-based practice overall cost of care.
benchmarks to assist the doctor. Unless CMS addresses all price centers contributing to the
OCM II should not wait 7 years for completion of the escalating cost of cancer care, its reforms will fail to make
current OCM pilot model. It should be a hybrid model that good on their promise.

References
1. Chen S, Wong S. Singapore Beats Hong Kong in Health Efficiency: Cancer Care. http://www.chqpr.org/downloads/ASCO_Patient-centered_
Southeast Asia. http://www.bloomberg.com/visual-data/best-and- Oncology_Payment.pdf. Accessed March 29, 2016.
worst/most-efficient-health-care-2014-countries. Accessed March 9. Newcomer LN, Gould B, Page RD, et al. Changing physician incentives for
29, 2016. affordable, quality cancer care: results of an episode payment model.
2. Organisation for Economic Cooperation and Development. OECD J Oncol Pract. 2014;10:322-326.
Health Statistics 2014. How Does the United States Compare? http:// 10. Association for Value-Based Cancer Care. Value-Based Cancer Care:
www.oecd.org/unitedstates/Briefing-Note-UNITED-STATES-2014.pdf. Oncology Medical Home Shows Significant Cost-Savings, Improved
Accessed March 29, 2016. Care Delivery. http://www.valuebasedcancer.com/vbcc-issues/2015/
3. Light DW, Kantarjian H. Market spiral pricing of cancer drugs. Cancer. december-2015-vol-6-no-11/26472-oncology-medical-home-shows-
2013;119:3900-3902. significant-cost-savings-improved-care-delivery. Accessed March 29,
4. Lungren MP, Amrhein TJ, Paxton BE, et al. Physician self-referral: 2016.
frequency of negative findings at MR imaging of the knee as a marker 11. Sprandio JD. Oncology patient-centered medical home. J Oncol Pract.
of appropriate utilization. Radiology. 2013;269:810-815. 2012;8:47s-49s (suppl 3).
5. Burns J. COME HOME Program Set to Save $33.5M Over 3 Years. http:// 12. CMS Alliance to Modernized Healthcare. Specialty Payment Model
www.onclive.com/publications/oncology-business-news/2014/August- Opportunities and Assessment: Oncology Design Report. Santa Monica,
2014/COME-HOME-Program-Set-to-Save-335M-Over-3-Years. Accessed CA: RAND Corporation; 2014.
March 29, 2016. 13. Conti RM, Berndt ER. Winners and losers of the Zaltrap price discount.
6. American Medical Association. Center for Healthcare Quality and Health Affairs weblog. February 22, 2013. http://healthaffairs.org/blog/
Payment Reform: A Guide to Physician Focused Alternative Payment 2013/02/20/winners-and-losers-from-the-zaltrap-price-discount-
Models. http://www.chqpr.org/downloads/Physician-FocusedAlternative unintended-consequences/. Accessed March 29, 2016.
PaymentModels.pdf?utm_source=Distribute+Physician-Focused+APM+ 14. Brooks GA, Li L, Uno H, et al. Acute hospital care is the chief driver of
Report&utm_campaign=Bundling+Badly&utm_medium=email. Accessed regional spending variation in Medicare patients with advanced cancer.
March 29, 2016. Health Aff (Millwood). 2014;33:1793-1800.
7. Burns LR, Pauly MV. Accountable care organizations may have difficulty 15. The University of Texas MD Anderson Cancer Center. MD Anderson,
avoiding the failures of integrated delivery networks of the 1990s. UnitedHealthcare Launch New Cancer Care Payment Model. MD
Health Aff (Millwood). 2012;31:2407-2416. Accessed March 29, 2016. Anderson News Release December 16, 2014. http://www.mdanderson.
8. American Society of Clinical Oncology. Patient-Centered Oncology org/newsroom/news-releases/2014/md-anderson-unitedhealthcare-
Payment. Payment Reform to Support Higher Quality, More Affordable new-cancer-payment.html. Accessed March 29, 2016.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e113


THOMAS AND WARD

16. Conti R, Ward JC, Page R, et al. A pathway through the bundle jungle. 18. Neubauer MA, Hoverman JR, Kolodziej M, et al. Cost effectiveness of
J Oncol Pract. In press. evidence-based treatment guidelines for the treatment of non-small-
17. Kreys ED, Koeller JM. Documenting the benefits and cost savings of a cell lung cancer in the community setting. J Oncol Pract. 2010;6:12-18.
large multistate cancer pathway program from a payers perspective. 19. Zon RT, Frame JN, Neuss MN, et al. American Society of Clinical Oncology Policy
J Oncol Pract. 2013;9:e241-e247. Statement on Clinical Pathways in Oncology. J Oncol Pract. 2016;12:261-266.

e114 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


CENTRAL NERVOUS SYSTEM TUMORS

Multidisciplinary Management of
Brain Metastases

CHAIR
Manmeet S. Ahluwalia, MD
Cleveland Clinic
Cleveland, OH

SPEAKERS
Anna S. Berghoff, MD, PhD
University of Vienna
Vienna, Austria

Jeffrey Scott Wefel, PhD


The University of Texas MD Anderson Cancer Center
Houston, TX

Morris D. Groves, MD
Texas Oncology/The US Oncology Network
Austin, TX

Paul D. Brown, MD
The University of Texas MD Anderson Cancer Center
Houston, TX

Riccardo Soffietti, MD
University and San Giovanni Battista Hospital
Torino, Italy
BERGHOFF ET AL

Immune Checkpoint Inhibitors in Brain Metastases: From


Biology to Treatment
Anna S. Berghoff, MD, PhD, Vyshak A. Venur, MBBS, Matthias Preusser, MD, and
Manmeet S. Ahluwalia, MD

OVERVIEW

Cancer immunotherapy has been a subject of intense research over the last several years, leading to new approaches for
modulation of the immune system to treat malignancies. Immune checkpoint inhibitors (antiCLTA-4 antibodies and
antiPD-1/PD-L1 antibodies) potentiate the hosts own antitumor immune response. These immune checkpoint inhibitors
have shown impressive clinical efficacy in advanced melanoma, metastatic kidney cancer, and metastatic nonsmall cell lung
cancer (NSCLC)all malignancies that frequently cause brain metastases. The immune response in the brain is highly
regulated, challenging the treatment of brain metastases with immune-modulatory therapies. The immune microenvi-
ronment in brain metastases is active with a high density of tumor-infiltrating lymphocytes in certain patients and, therefore,
may serve as a potential treatment target. However, clinical data of the efficacy of immune checkpoint inhibitors in brain
metastases compared with extracranial metastases are limited, as most clinical trials with these new agents excluded
patients with active brain metastases. In this article, we review the current scientific evidence of brain metastases biology
with specific emphasis on inflammatory tumor microenvironment and the evolving state of clinical application of immune
checkpoint inhibitors for patients with brain metastases.

B rain metastases are a long-known devastating compli-


cation of advanced malignancies that lead to substantial
morbidity. As patients with metastatic cancer are expected
THE INFLAMMATORY MICROENVIRONMENT OF
BRAIN METASTASES
Immune responses in the brain parenchyma are tightly reg-
to live longer with newer systemic therapeutic agents and ulated to prevent overwhelming and potentially destroying
with better and more sensitive imaging studies, it is expected immune reactions in this organ, which has little recovery
that more patients will be diagnosed with brain metastases. In capacity.2 Importantly, the brain parenchyma is not an im-
the past, traditional treatment approaches included surgery mune privileged organ that actively suppresses any immune
or radiation therapy, or a combination of the two. Chemo- response, but, rather, it initiates and regulates immune
therapeutic agents were generally used for refractory disease. responses. Therefore, targeting the inflammatory tumor
However, in the last few years, several treatment advances in microenvironment of brain metastases as a therapeutic
targeted therapy and immunotherapy have changed the land- target must consider several unique factors compared with
scape of the management of brain metastases. Recently, the the inflammatory tumor microenvironment of extracranial
American Society of Clinical Oncology (ASCO) announced cancer malignancies.3
immunotherapy as the advance of the year,1 and its role in Immune escape is an emerging hallmark of cancer.4 A brain
the management of brain metastases is undergoing active metastasisinitiating cell has to facilitate the process of
investigation. There has been a paucity of research focused on immune escape several times, first during initiation of the
brain metastases; as a result, there are several unanswered primary tumor, then during travel through the bloodstream,
questions. In this article, we present the available evidence and, finally, during the extravasation and metastatic out-
regarding the interactions between the immune system and growth process within the brain parenchyma. The process of
brain parenchyma in brain metastases, the clinical application metastatic spread may actually be supported by the immune
of immune checkpoint inhibitors in the management of brain system, as tumor-associated macrophages were shown to
metastases, and the potential of combining radiation with facilitate intravasation as well as extravasation from the
immune checkpoint inhibitors. vascular system.5,6 Further, cytokines promote site-specific

From the German Cancer Research Center, University of Heidelberg, Heidelberg, Germany; Department of Medicine I, Medical University of Vienna, Vienna, Austria; Comprehensive
Cancer Center Vienna CNS Unit, Vienna, Austria; Division of Hematology and Oncology, Department of Internal Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA; Burkhardt
Brain Tumor and Neuro-Oncology Center, Neurologic Institute, Cleveland Clinic, Cleveland, OH; Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Manmeet S. Ahluwalia, MD, Cleveland Clinic, 9500 Euclid Ave., S73, Cleveland, OH 44195; email: ahluwam@ccf.org.

2016 by American Society of Clinical Oncology.

e116 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


IMMUNE CHECKPOINT INHIBITIORS IN BRAIN METASTASES

metastatic behavior as the chemokine pair CXCR4/CXCL12 compared with the matched primary tumor.15 Further, the
was shown to promote adhesion of the brain metastasis composition of TIL subtypes differs. Infiltration with immune
initiating cells to brain vessels and further facilitated the suppressive FOXP3+ TILs, as well as so-called exhausted
migration through endothelial cells.7,8 PD-1+ TILs, has been observed in a large portion of speci-
In established brain macrometastases, the inflammatory mens.14 Characteristics of the primary tumor probably in-
tumor microenvironment is composed of the innate fluence the T cell response in brain metastases, as a denser
immune system, namely microglia and blood-derived myeloid TIL infiltration was observed in melanoma-derived brain
cells/macrophages, and the adaptive immune system, which metastases compared with those that are derived from
is mainly represented by T cells.9 breast cancer.14 Importantly, patients with brain metas-
The density of microglia in and around human and in vivo tases who present with dense infiltration with effector
brain metastases were shown to be highly heterogeneous.10,11 CD3+, cytotoxic CD8+, or memory CD45RO+ TILs had an
Importantly, the differences in microglia accumulation were improved survival prognosis from diagnosis compared
observed early during the process of extravasation in pre- with patients with little or absent infiltration of TILs.14 This
clinical models, indicating that microglia may be involved in finding is well in line with the effect of the inflammatory
the process of extravasation and survival in the perivascular tumor microenvironment on survival prognosis in extra-
niche.11 The functions of microglia within the brain metastasis cranial malignancies, as high density of TILs is associated
tumor microenvironment include antigen presentation, cyto- with improved survival prognosis in various malignancies
toxicity via expression of nitric oxide (NO) and superoxide, and that frequently cause brain metastases, such as triple-negative
phagocytic function.12 Expression of HMGB1 by microglia breast cancer, NSCLC, or melanoma.16-18
cells, a factor involved in antigen presentation and activation Importantly, patients with extracranial metastatic mela-
of the adaptive immune system, was widely observed, indi- noma and an active inflammatory tumor microenvironment,
cating that in general the tumor microenvironment is able to as defined by dense infiltration with TILs, have shown in-
alter an adaptive immune response. However, microglia and creased response rates to CTLA-4 immune checkpoint in-
macrophages were also shown to express immunosuppres- hibitors.19 Further, expression of PD-L1 on tumor cells may
sive factors like PD-L1.13 be associated with a higher chance of response to a PD-
Infiltration with T cells or tumor-infiltrating T-lymphocytes 1axis modulating immune checkpoint inhibitor.20 There-
(TILs) was shown to be highly heterogeneous, varying from fore, the potential tumor microenvironment preconditions
total absence to very dense infiltration.10,14 Therefore, the for response to an immune checkpoint inhibitor are present
inflammatory microenvironment of brain metastases differs in brain metastases, underscoring the importance of further
between patients, showing active tumor-directed function clinical investigation of immune checkpointbased therapy
in some, but not all, cases. Here, expression of the immune strategies in patients with brain metastases.
suppressive factor PD-L1 could be one mechanism whereby The inflammatory microenvironment is further influenced
cancer cells facilitate an immune escape. PD-L1 tends to by the vascular properties in the brain parenchyma. The
have higher expression in NSCLC-derived brain metastases blood-brain barrier is composed of endothelial cells with
tight junctions and astrocytes. Here, astrocytes are known
to facilitate pro-inflammatory as well as anti-inflammatory
aspects.21 However, data of the interaction of astrocytes
KEY POINTS and TILs in the inflammatory microenvironment of brain
metastases is yet not available. Further, the vascular endo-
The inflammatory tumor microenvironment of brain
thelium regulates the infiltration of T cells. Presence of so-
metastases differs from extracranial metastases, as the
brain is a highly regulated organ in terms of immune
called high endothelial venules, which are specialized blood
response, and is active in the majority of cases with vessels for lymphocyte extravasation, in melanoma spec-
dense infiltration of tumor-infiltrating lymphocytes. imens were shown to be associated with dense infiltration
The blood-brain barrier presents different challenges of TILs.22 Although neovascularization is a characteristic of
and unique opportunities for the treatment of patients brain metastases, with differing extent between primary
with brain metastases. tumor types, no functional studies have yet investigated the
Immune checkpoint inhibitors have shown intracranial interaction of the blood-tumor barrier or the blood-brain
activity in early clinical trials for patients with brain barrier in the infiltration zone of a brain metastasis con-
metastases from melanoma and nonsmall cell lung cerning their restriction of T-cell infiltration.
cancer.
Several retrospective case series suggest immune
checkpoint inhibitors can be safely combined with
radiation therapies.
CLINICAL EFFICACY OF IMMUNE THERAPY IN
As the prevalence of brain metastases increases, CANCER
concerted research in this field of limited knowledge is The clinical utility of immune-based therapies in the fight
of essence. Meanwhile, a multidisciplinary approach for against cancer has been a fascinating story with ebb and
the management of brain metastases is recommended. flow. Historically, melanoma and kidney cancer were the two
major malignancies that were thought to be susceptible to

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e117


BERGHOFF ET AL

immune modulation. Indeed, melanoma has high muta- CA 184-045, 165 patients with stable brain metastases were
tional burden, with production of several neoantigens, treated with 10 mg/kg of ipilimumab. The preliminary results
making it an ideal candidate for immunotherapy. Interleukin from this study reported a 1-year OS rate of 20%, similar to that
(IL)-2 was one of the first immunotherapeutic agents to be reported in the open-label, phase II study.31
used in melanoma. IL-2 is a cytokine that is produced by Another open-label, single-arm, phase II trial evaluated the
human T-lymphocytes and plays a pivotal role in expansion combination of ipilimumab and fotemustine in advanced
and activation of T cells.23 In the 1990s, high-dose IL-2 was metastatic melanoma with and without brain metastases
studied in advanced melanoma and renal cell carcinomas (NIBIT-M1).32 This Italian study enrolled a total of 83 pa-
with modest success.24 An extracranial complete response tients; 20 of whom had asymptomatic brain metastases.
rate of 5% was seen, and some patients experienced sub- Eleven of the 20 patients completed the induction phase
stantial adverse effects, including death.24 Two single in- of ipilimumab (four doses of 10 mg/kg every 3 weeks).
stitution retrospective experiences have shown intracranial Immune-related disease control was observed in 10 pa-
activity of IL-2; however, because of concerns of vascular tients; five had undetectable brain lesions and five had
leak syndrome and potential life-threatening thrombocy- stable disease. The median progression-free survival (PFS) in
topenia, prospective studies excluded patients with brain patients with brain metastases was 3.0 months (range, 0 to
metastases.25,26 10.9 months). Adverse reactions were reported in 18 out
of 20 patients with brain metastases. Central nervous
system events like intracranial hemorrhage, seizures, and
AntiCTLA-4 Antibody: Ipilimumab headaches were reported in five patients, all of whom had
With the advent of immune checkpoint inhibitors, immu- intracranial disease progression. A 3-year follow-up of this
notherapy has made substantial inroads in the management trial was presented at the European Society for Medical
of advanced malignancies. Ipilimumab is a fully human Oncology (ESMO) Congress in 2015, and the group with brain
monoclonal antibody against CTLA-4, which plays a pivotal metastases had a median OS of 12.7 months (95% CI,
role in downregulating the production of cytotoxic T cells.27 2.722.7 months) and a 3-year survival rate of 27.8%.33 With
The intracranial activity of ipilimumab was first noted in the the promising results from the phase II trial, a phase III trial
post hoc analysis of a phase III trial of ipilimumab with or of patients with melanoma-derived brain metastases
without gp100 peptide vaccine compared with gp100 vac- (NIBIT-M2) is planned.34 The trial was initially designed with
cine alone.28 This double-blinded randomized trial included two arms; arm A with single-agent fotemustine and arm B
82 patients with asymptomatic central nervous system (CNS) with fotemustine and ipilimumab. However, with recent
metastases, of whom, 73 had received prior treatment for reports of high clinical activity with a combination of ipi-
CNS metastases. Ipilimumab was administered to 61 of the limumab and the antiPD-1 monoclonal antibody nivo-
82 patients; 46 patients also received gp100 and 15 received lumab, the protocol was amended to include a third arm,
ipilimumab alone. The hazard ratio (HR) for death in patients arm C, with a combination of nivolumab and ipilimumab.35
with brain metastases was 0.70 (95% CI, 0.411.20) in the Table 1 reviews the published studies of immune check-
ipilimumab plus vaccine group compared with 0.76 (95% CI, point inhibitors in brain metastases.
0.381.54) in the ipilimumab alone group. This led to a
single-agent, open-label phase II study of ipilimumab for
patients with melanoma-derived brain metastases.29 Ipili- AntiPD-1/PD-L1 Antibodies
mumab was administered at a dose of 10 mg/kg given every Another promising immune checkpoint target is the
3 weeks as a 12-week induction phase, followed by main- PD-1/PD-L1 pathway. AntiPD-1/PD-L1 antibodies such as
tenance therapy every 12 weeks. A total of 72 patients were nivolumab and pembrolizumab have been approved for
enrolled across 10 centers in the United States.30 The trials advanced melanoma, kidney cancer, and NSCLC. All of the
comprised of two cohorts: cohort A included 51 asymp- early trials of antiPD-1/PD-L1 agents excluded patients with
tomatic patients with active brain metastases who were not brain metastases. In 2015, interim results of a single-center,
on corticosteroids, and cohort B included 21 patients with phase II study of pembrolizumab of patients with melanoma
symptomatic melanoma-derived brain metastases taking a and NSCLC with brain metastases was presented at the 2015
corticosteroid. A response rate of 18% (nine out of 51 pa- ASCO Annual Meeting.36,37 Patients with newly diagnosed
tients) in cohort A and 5% (one out of 21 patients) in cohort B asymptomatic or progressing brain metastases who did not
were reported using modified World Health Organization require immediate treatment with steroids are being en-
(WHO) criteria, but, when immune-related response criteria rolled in this study. By December 2014, 17 patients with
were used, the response rate improved to 25% (13 patients) metastatic melanoma and 10 patients with NSCLC were
and 10% (two patients) in cohorts A and B, respectively. The enrolled. These patients were treated with 10 mg/kg of
median overall survival (OS) was 7.0 months in cohort A and pembrolizumab administered every 2 weeks. Among the
3.7 months in cohort B. Ipilimumab was well tolerated and 12 evaluable patients in the melanoma arm, three were
no unexpected adverse events were noted. Fatigue, nausea, observed to have partial response and two had stable disease
vomiting, diarrhea, and rash were the common side effects with a 6-month OS of 47%. In the NSCLC arm, nine patients
noted in the study. As a part of the expanded access program, were evaluated for response and four had partial response.

e118 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


IMMUNE CHECKPOINT INHIBITIORS IN BRAIN METASTASES

TABLE 1. Studies With Immunotherapy in Melanoma Patients With Brain Metastases


Concurrent
Local Intracranial
Special Trial Therapy Radiation Response OS
Study Design Characteristics Tested Treatment No. of Patients Rate PFS (Months)
Margolin et al29 Phase II Cohort A: no Ipilimumab None Cohort A: 51 24% NR 7.0
steroids
Cohort B: Cohort B: 21 10% 3.7
steroids
Di Giacoma Phase II Asymptomatic Ipilimumab/ None 20 50% NR 12.7
et al32 fotemustine
Patel R et al49 Retrospective SRS vs. SRS/ Ipilimumab SRS (given SRS only: 34 NR 1-year 12-month:
comparative ipilimumab within LC: 38%
analysis 4 months of 92.3%
initiation of
SRS/ 1-year 12-month:
ipilimumab)
ipilimumab: LC: 37.1%
20 71.4%
Weber et al50 Retrospective Ipilimumab/ Ipilimumab None 12 41.6% NR 14.0
analysis of budesonide
phase II trial (asymptomatic
BM)
Knisely et al41 Retrospective SRS vs. SRS/ Ipilimumab SRS SRS: 17 NR NR 4.9
comparative ipilimumab
SRS/ 21.3
analysis
ipilimumab:
11
Silk et al40 Retrospective SRS vs. SRS/ Ipilimumab SRS/WBRT No NR 3.3 5.3
comparative ipilimumab ipilimumab:
analysis 37
Ipilimumab: 33 2.7 18.3
42
Mathew et al Retrospective SRS vs. SRS/ Ipilimumab SRS SRS: 33 NR NR 6-month: 56%
comparative ipilimumab
SRS/ 6-month:45%
analysis
ipilimumab:
25
Ahmed et al43 Retrospective SRS/nivolumab Nivolumab SRS 26 NR NR 11.8
analysis
Abbreviations: BM, brain metastases; PFS, progression-free survival; OS, overall survival; NR, not reported; SRS, stereotactic radiosurgery; LC, local control; WBRT, whole-brain
radiotherapy.

Only one patient in both arms experienced grade 3 adverse whole-brain radiation therapy (WBRT) with or without sur-
events related to pembrolizumab (liver function abnormali- gery plays a crucial role in the management of symptomatic
ties). The investigators presented updated results at the brain metastases.
16th World Conference on Lung Cancer in September 2015; Several retrospective single-center case series have shown
18 patients had sufficient follow-up time for response eval- that stereotactic radiation and ipilimumab can be combined
uation. The median OS was 7.7 months (95% CI, 3.5 months safely in the management of brain metastases derived from
to not reached).38 These early results appear promising for melanoma.40-42 The safety of combining nivolumab with
the utility of pembrolizumab in patients with asymptomatic stereotactic radiation was reported in a recent single-center
brain metastases. Another multicenter phase II clinical trial study.43 This retrospective report included patients who
of ipilimumab plus nivolumab for patients with advanced were treated in the context of a clinical trial of nivolumab
melanoma with brain metastases (NCT02320058) is currently for advanced melanoma. Twenty-six patients with brain me-
accruing patients.39 This study is planned to treat patients tastases were treated with stereotactic radiation, with, prior to,
with asymptomatic brain metastases from melanoma with an or after nivolumab. The combination of nivolumab and ste-
induction phase of four cycles of 1 mg/kg of nivolumab and reotactic radiation was well tolerated. Distant intracranial
3 mg/kg of ipilimumab every 3 weeks, followed by mainte- recurrence and OS were improved compared with historical
nance therapy of 3 mg/kg of nivolumab every 2 weeks. Table 2 controls treated with radiation alone.
provides a list of other important clinical trials in this setting. There are three important aspects to combining radiation
with immune checkpoint inhibitors:
Immunotherapy and Radiation 1. Can immunotherapy potentiate radiation therapy
Radiation therapy has been the cornerstone of treat- (radio-sensitizing effect) leading to better intracranial
ment for brain metastases. Stereotactic radiosurgery and disease control?

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e119


BERGHOFF ET AL

TABLE 2. Select Clinical Trials Currently Accruing Patients


Estimated
Study Design Therapy tested Primary Cancer Enrollment
NCT0246006839 Multicenter, randomized, phase III Fotemustine Melanoma 168
vs. fotemustine/ipilimumab
vs. ipilimumab/nivolumab
NCT0262151551 Multicenter, phase II Nivolumab in symptomatic brain metastases Melanoma 70
NCT0232005852 Multicenter, phase II Nivolumab/ipilimumab followed by nivolumab Melanoma 110
NCT0237424253 Multicenter, phase II Nivolumab/ipilimumab for melanoma brain metastases Melanoma 75
NCT0208507054 Single-center, phase II Pembrolizumab Melanoma, 64
nonsmall cell
lung cancer

2. Will the combination of radiation therapy and immu- single largest extracranial lesion after stereotactic radiation
notherapy lead to better extracranial disease control to the brain metastases and ipilimumab.44
(abscopal effect) and, thus, improved OS? The third aspect of combining radiation and immuno-
3. If the radiation potentiates the immunotherapy, what therapy and choosing the preferred fractionation schedule
is the best fractionation schedule to achieve the most is being evaluated in preclinical models. Single session and
benefit? fractioned radiation schedules with and without immuno-
The first question is relatively straightforward, and several therapy have been tested in a preclinical mouse model.48
clinical trials are currently underway to answer the potential TSA breast carcinoma and MCA38 colon cancer mouse
intracranial responses of combining immunotherapy and models were used to test the various radiation fractionation
radiation. The major confounder in this endeavor is the use schemes with and without antiCTLA-4 antibody, 9H10. The
of steroids. Relatively high doses of steroids have been used mice were injected with tumor cells at two sites; a primary
with radiation, especially WBRT, to reduce the intracranial and a secondary site. They were then randomly assigned
edema. The detrimental effect of the concomitant use of to receive radiation to the primary site at three different
steroids and immunotherapy was observed in the phase II fractionation schedules (one cycle of 20 Gy, three cycles
trial of ipilimumab for patients with asymptomatic, of 8 Gy, or five cycles of 5 Gy on consecutive days), im-
melanoma-derived brain metastases.29 Since this study, the munotherapy alone, or a combination of radiation and
required steroid dose used with stereotactic radiation immunotherapy. Only fractionated radiation with immu-
therapy has decreased, and they are used often for a shorter notherapy to the primary site produced abscopal effect at
duration. In addition, many centers are now moving away the secondary site. However, strong clinical data to sup-
from using steroids routinely for all patients with brain port this preclinical model are lacking. Innovative clinical
metastases.44 trials are needed to improve our understanding of the in-
This second aspect of combining radiation and immuno- teraction of radiation and immunotherapy and the abscopal
therapy, the abscopal effect, is interesting and more effect.
challenging to evaluate in clinical trials. It has been well
established that radiation causes DNA damage that leads to CONCLUSION
cell death. However, the interaction of radiation and the In summary, some brain metastases harbor an immune
immune system is not well understood. Several preclinical active microenvironment that in theory can be targeted by
data have reported that cancer cells release numerous immune-modulating therapies like immune checkpoint in-
chemokines and cytokines in response to radiation. The hibitors. However, a deeper insight of the specific mecha-
damaged cancer cells have also been shown to upregulate nisms in the highly regulated microenvironment of the brain
MHC-1 expression, hence increasing the interaction with is needed to understand and overcome potential resistance
CD8 T cells.45 In addition, radiation leads to the upregulation mechanisms. Early results of immune checkpoint inhibitors
of PD-L1 on cancer cells, limiting their interaction with CD8 in clinical trials have shown intracranial activity in brain
T cells.46,47 Nevertheless, the activated CD8 T cells can elicit a metastases from melanoma and NSCLC. There are several
systemic antitumor effect, thereby leading to reduction in ongoing clinical trials investigating the role of immune
the tumor burden. Methods to reliably produce and po- checkpoint blockade in brain metastases. Numerous retro-
tentiate this abscopal effect are a subject of intense re- spective case series suggest immune checkpoint inhibitors
search. However, increasing the interaction of CD8 T cells can be safely combined with radiation. Prospective studies
with radiated tumor cells by the use of immune checkpoint are needed to further confirm the safety of such approaches
inhibitors may enhance the abscopal effect. A retrospective and define the timing and the dose of the optimal radiation
case series tried to evaluate this effect by following the modality.

e120 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


IMMUNE CHECKPOINT INHIBITIORS IN BRAIN METASTASES

References
1. Dizon DS, Krilov L, Cohen E, et al. Clinical cancer advances 2016: annual 21. Gimsa U, Mitchison NA, Brunner-Weinzierl MC. Immune privilege as
report on progress against cancer from the American Society of Clinical an intrinsic CNS property: astrocytes protect the CNS against T-cell-
Oncology. J Clin Oncol. 2016;34:987-1011. mediated neuroinflammation. Mediators Inflamm. 2013;2013:320519.
2. Galea I, Bechmann I, Perry VH. What is immune privilege (not)? Trends 22. Martinet L, Le Guellec S, Filleron T, et al. High endothelial venules
Immunol. 2007;28:12-18. (HEVs) in human melanoma lesions: Major gateways for tumor-
3. Berghoff AS, Preusser M. The inflammatory microenvironment in brain infiltrating lymphocytes. OncoImmunology. 2012;1:829-839.
metastases: potential treatment target? Chin Clin Oncol. 2015;4:21. 23. Rosenberg SA. IL-2: the first effective immunotherapy for human
4. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. cancer. J Immunol. 2014;192:5451-5458.
Cell. 2011;144:646-674. 24. Atkins MB, Lotze MT, Dutcher JP, et al. High-dose recombinant in-
5. Noy R, Pollard JW. Tumor-associated macrophages: from mechanisms terleukin 2 therapy for patients with metastatic melanoma: analysis of
to therapy. Immunity. 2014;41:49-61. 270 patients treated between 1985 and 1993. J Clin Oncol. 1999;17:
6. Wyckoff JB, Wang Y, Lin EY, et al. Direct visualization of macrophage- 2105-2116.
assisted tumor cell intravasation in mammary tumors. Cancer Res. 25. Guirguis LM, Yang JC, White DE, et al. Safety and efficacy of high-dose
2007;67:2649-2656. interleukin-2 therapy in patients with brain metastases. J Immunother.
7. Hinton CV, Avraham S, Avraham HK. Role of the CXCR4/CXCL12 signaling 2002;25:82-87.
axis in breast cancer metastasis to the brain. Clin Exp Metastasis. 2010; 26. Powell S, Dudek AZ. Single-institution outcome of high-dose interleukin-
27:97-105. 2 (HD IL-2) therapy for metastatic melanoma and analysis of favorable
8. Lee BC, Lee TH, Avraham S, et al. Involvement of the chemokine re- response in brain metastases. Anticancer Res. 2009;29:4189-4193.
ceptor CXCR4 and its ligand stromal cell-derived factor 1 alpha in breast 27. Weber J. Review: anti-CTLA-4 antibody ipilimumab: case studies of
cancer cell migration through human brain microvascular endothelial clinical response and immune-related adverse events. Oncologist.
cells. Mol Cancer Res. 2004;2:327-338. 2007;12:864-872.
9. Fridman WH, Pages ` F, Sautes-Fridman
` C, et al. The immune contexture 28. Hodi FS, ODay SJ, McDermott DF, et al. Improved survival with ipilimumab
in human tumours: impact on clinical outcome. Nat Rev Cancer. 2012; in patients with metastatic melanoma. N Engl J Med. 2010;363:711-723.
12:298-306. 29. Margolin K, Ernstoff MS, Hamid O, et al. Ipilimumab in patients with
10. Berghoff AS, Lassmann H, Preusser M, et al. Characterization of the melanoma and brain metastases: an open-label, phase 2 trial. Lancet
inflammatory response to solid cancer metastases in the human brain. Oncol. 2012;13:459-465.
Clin Exp Metastasis. 2013;30:69-81. 30. Margolin K. Ipilimumab in a phase II trial of melanoma patients with
11. Lorger M, Felding-Habermann B. Capturing changes in the brain mi- brain metastases. Oncoimmunology. 2012;1:1197-1199.
croenvironment during initial steps of breast cancer brain metastasis. 31. Heller KN, Pavlick AC, Hodi FS, et al. Safety and survival analysis of
Am J Pathol. 2010;176:2958-2971. ipilimumab therapy in patients with stable asymptomatic brain me-
12. Graeber MB, Streit WJ. Microglia: biology and pathology. Acta Neu- tastases. J Clin Oncol. 2011;29 (suppl; abstr 8581).
ropathol. 2010;119:89-105. 32. Di Giacomo AM, Ascierto PA, Pilla L, et al. Ipilimumab and fotemustine in
13. Berghoff AS, Kiesel B, Widhalm G, et al. Programmed death ligand 1 patients with advanced melanoma (NIBIT-M1): an open-label, single-
expression and tumor-infiltrating lymphocytes in glioblastoma. Neuro arm phase 2 trial. Lancet Oncol. 2012;13:879-886.
Oncol. 2015;17:1064-1075. 33. Di Giacomo AM, Ascierto PA, Queirolo P, et al. Three-year follow-up
14. Berghoff AS, Fuchs E, Ricken G, et al. Density of tumor-infiltrating of advanced melanoma patients who received ipilimumab plus fote-
lymphocytes correlates with extent of brain edema and overall mustine in the Italian Network for Tumor Biotherapy (NIBIT)-M1 phase
survival time in patients with brain metastases. OncoImmunology. II study. Ann Oncol. 2015;26:798-803.
2015;5:e1057388. 34. Lyle M, Long GV. The role of systemic therapies in the management of
15. Berghoff AS, Inan C, Ricken G, et al. Tumor-infiltrating lymphocytes melanoma brain metastases. Curr Opin Oncol. 2014;26:222-229.
(TILs) and PD-L1 expression in non-small cell lung cancer brain me- 35. Di Giacomo AM, Margolin K. Immune checkpoint blockade in patients
tastases (BM) and matched primary tumors (PT). Ann Oncol. 2014;25: with melanoma metastatic to the brain. Semin Oncol. 2015;42:
iv426-iv470 (suppl 4; abstr 1324P). 459-465.
16. Adams S, Gray RJ, Demaria S, et al. Prognostic value of tumor-infiltrating 36. Kluger HM, Goldberg SB, Sznol M, et al. Safety and activity of pem-
lymphocytes in triple-negative breast cancers from two phase III ran- brolizumab in melanoma patients with untreated brain metastases.
domized adjuvant breast cancer trials: ECOG 2197 and ECOG 1199. J Clin J Clin Oncol. 2015;33 (suppl; abstr 9009).
Oncol. 2014;32:2959-2966. 37. Goldberg SB, Gettinger SN, Mahajan A, et al. Activity and safety of
17. Tao H, Mimura Y, Aoe K, et al. Prognostic potential of FOXP3 expression pembrolizumab in patients with metastatic non-small cell lung cancer
in non-small cell lung cancer cells combined with tumor-infiltrating with untreated brain metastases. J Clin Oncol. 2015;33 (suppl; abstr
regulatory T cells. Lung Cancer. 2012;75:95-101. 8035).
18. Clemente CG, Mihm MC Jr, Bufalino R, et al. Prognostic value of tumor 38. Seto T, Shukuya T, Yamanaka T, et al. Chemotherapy developments for
infiltrating lymphocytes in the vertical growth phase of primary cu- lung cancer. J Thorac Oncol. 2015;10:S173-S260.
taneous melanoma. Cancer. 1996;77:1303-1310. 39. Margolin KA, Tawbi HA-H, Ernstoff MS, et al. A multi-center phase II
19. Ji RR, Chasalow SD, Wang L, et al. An immune-active tumor microen- open-label study (CheckMate 204) to evaluate safety and efficacy of
vironment favors clinical response to ipilimumab. Cancer Immunol nivolumab (NIVO) in combination with ipilimumab (IPI) followed by
Immunother. 2012;61:1019-1031. NIVO monotherapy in patients (pts) with melanoma (MEL) metastatic to
20. Taube JM, Klein A, Brahmer JR, et al. Association of PD-1, PD-1 ligands, the brain. J Clin Oncol. 2015;33 (suppl; abstr TPS9080).
and other features of the tumor immune microenvironment with re- 40. Silk AW, Bassetti MF, West BT, et al. Ipilimumab and radiation therapy
sponse to anti-PD-1 therapy. Clin Cancer Res. 2014;20:5064-5074. for melanoma brain metastases. Cancer Med. 2013;2:899-906.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e121


BERGHOFF ET AL

41. Knisely JP, Yu JB, Flanigan J, et al. Radiosurgery for melanoma brain when combined with anti-CTLA-4 antibody. Clin Cancer Res. 2009;15:
metastases in the ipilimumab era and the possibility of longer survival. 5379-5388.
J Neurosurg. 2012;117:227-233. 49. Patel KR, Shoukat S, Oliver DE, et al. Ipilimumab and stereotactic
42. Mathew M, Tam M, Ott PA, et al. Ipilimumab in melanoma with limited radiosurgery versus stereotactic radiosurgery alone for newly di-
brain metastases treated with stereotactic radiosurgery. Melanoma agnosed melanoma brain metastases. Am J Clin Oncol. Epub 2015
Res. 2013;23:191-195. May 16.
43. Ahmed KA, Stallworth DG, Kim Y, et al. Clinical outcomes of melanoma 50. Weber JS, Amin A, Minor D, et al. Safety and clinical activity of ipilimumab
brain metastases treated with stereotactic radiation and anti-PD-1 in melanoma patients with brain metastases: retrospective analysis
therapy. Ann Oncol. 2016;27:434-441. of data from a phase 2 trial. Melanoma Res. 2011;21:530-534.
44. Schoenfeld JD, Mahadevan A, Floyd SR, et al. Ipilimumab and cranial 51. NCT02621515. Nivolumab in Symptomatic Brain Metastases (CA209-
radiation in metastatic melanoma patients: a case series and review. 322). https://clinicaltrials.gov/ct2/show/record/NCT02621515. Accessed
J Immunother Cancer. 2015;3:50. February 20, 2016.
45. Reits EA, Hodge JW, Herberts CA, et al. Radiation modulates the peptide 52. NCT02320058. A Multi-Center Phase 2 Open-Label Study to Evaluate
repertoire, enhances MHC class I expression, and induces successful Safety and Efficacy in Subjects With Melanoma Metastatic to the Brain
antitumor immunotherapy. J Exp Med. 2006;203:1259-1271. Treated With Nivolumab in Combination With Ipilimumab Followed by
46. Dovedi SJ, Adlard AL, Lipowska-Bhalla G, et al. Acquired resistance to Nivolumab Monotherapy (CheckMate 204). https://clinicaltrials.gov/
fractionated radiotherapy can be overcome by concurrent PD-L1 ct2/show/NCT02320058. Accessed February 20, 2016.
blockade. Cancer Res. 2014;74:5458-5468. 53. NCT02374242. Anti-PD 1 Brain Collaboration for Patients With Mela-
47. Deng L, Liang H, Burnette B, et al. Irradiation and anti-PD-L1 treatment noma Brain Metastases (ABC). https://clinicaltrials.gov/ct2/show/
synergistically promote antitumor immunity in mice. J Clin Invest. 2014; NCT02374242. Accessed February 20, 2016.
124:687-695. 54. NCT02085070. MK-3475 in Melanoma and NSCLC Patients With
48. Dewan MZ, Galloway AE, Kawashima N, et al. Fractionated but not Brain Metastases. https://clinicaltrials.gov/ct2/show/NCT02085070.
single-dose radiotherapy induces an immune-mediated abscopal effect Accessed February 20, 2016.

e122 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


TARGETED THERAPY IN BRAIN METASTASES

Targeted Therapy in Brain Metastases: Ready for Primetime?


Vyshak A. Venur, MD, and Manmeet S. Ahluwalia, MD

OVERVIEW

Brain metastasis is a serious complication of cancer that causes significant morbidity for patients. Over the last decade,
numerous new driver somatic mutations have been recognized and targeted therapies are changing the landscape of
treatment in lung cancer, breast cancer, and melanoma, which are also the three most common cancers that result in
brain metastases. The common actionable mutations include the EGFR mutation and anaplastic lymphoma kinase (ALK)
translocations in nonsmall cell lung cancer, the HER2 mutation in breast cancer, and the BRAF mutation in melanoma.
However, most of the early trials with targeted agents excluded patients with brain metastases. With a better understanding
of the biology, several recent trials of targeted therapy that focus on brain metastases have been reported and others are
ongoing. Novel agents with better penetration across the bloodbrain barrier are currently being investigated for patients
with brain metastases. In this review, we discuss the current state of use and future directions of targeted therapies in brain
metastases.

T he discovery of driver mutations and agents targeting these


mutations and pathways has revolutionized the treatment
of patients with advanced malignancies in the past decade. The
cancer develop brain metastases fairly early in the course of
the disease. They are usually treated with WBRT. Patients
with nonsmall cell lung cancer (NSCLC) are a more hetero-
incidence of brain metastases has increased, although newer geneous group, and 25%40% of patients with NSCLC will
targeted agents have improved the systemic control of malig- develop a brain metastasis during the course of the disease.3
nancy and hence survival. It is estimated that approximately 40% The common actionable mutations in nonsquamous NSCLC
of patients with metastatic cancer will develop a symptomatic include EGFR mutation and ALK gene rearrangements.
brain metastasis.1 Lung cancer, breast cancer, and melanoma are The frequency of somatic EGFR mutations varies from 30% to
the three common malignancies that lead to brain metastases. In 50% among East Asians to approximately 10% among Cauca-
the past, the cornerstone for treatment of brain metastases in- sians.4 Exon 19 (in-frame) deletions and point mutations (L858R)
cluded whole-brain radiation therapy (WBRT), stereotactic radia- at exon 21 comprise 90% of all known EGFR mutations in
tion (SRS), and surgery. The evolution of systemic therapy in the NSCLC.5 Studies suggest that patients with an EGFR mutation
management of brain metastases has been restricted by the develop brain metastases more frequently, have several small
presence of the bloodbrain barrier and efflux pumps. Most initial lesions with minimal peritumoral edema, and may have better
studies of drugs for the management of advanced cancer excluded overall survival (OS) compared with patients with lung cancer
patients with brain metastases, further limiting progress. Hence, brain metastases without any mutation.6 Erlotinib and gefi-
chemotherapy has had a limited role in the management of brain tinib are two first-generation tyrosine kinase inhibitors (TKIs)
metastases. Several new targeted agents with bloodbrain barrier that target EGFR mutations and are approved by the U.S. Food
penetration are being developed, with emphasis on treatment and Drug Administration (FDA) for use in lung cancer treatment.7
patients with brain metastases.2 In this article, we review the Both erlotinib and gefitinib have distinct pharmacologic char-
current evidence on the role of agents targeting the various driver acteristics in the central nervous system (CNS). [11C]erlotinib
mutations and pathways in management of brain metastases from was shown to have effective CNS distribution in the presence
lung cancer, breast cancer, and melanoma (Table 1). of brain metastases but not in the normal brain.8 However,
OSI-420, an active metabolite of erlotinib, is a substrate of
TARGETED THERAPY FOR LUNG CANCER WITH P-glycoprotein (P-gp), a drug efflux protein of the ATP-binding
BRAIN METASTASES cassette transporter family at the brain endothelial cells of the
Lung cancer is the most common cancer that results in brain bloodbrain barrier.9 Gefitinib is known to inhibit P-gp activity,
metastases. A high proportion of patients with small cell lung and none of its known metabolites are substrates of P-gp.10

From the Division of Hematology and Oncology, Department of Internal Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA; Burkhardt Brain Tumor and Neuro-Oncology
Center, Department of Medicine, Neurologic Institute, Cleveland Clinic, Cleveland, OH; Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Manmeet. S. Ahluwalia, MD, Cleveland Clinic, 9500 Euclid Ave., S73, Cleveland, OH 44195; email: ahluwam@ccf.org.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e123


VENUR AND AHLUWALIA

However, both of these drugs have limited bloodbrain barrier significant difference in PFS and OS was noted between the
penetration, and limited cerebral spinal fluid concentrations of two groups.
erlotinib (approximately 5%) and gefitinib (approximately 2.5%) The second-generation EGFR TKI afatinib is an irreversible
have been reported.11,12 inhibitor of EGFR.18 It also inhibits HER2 and ERBB4 re-
A Japanese phase II study evaluated the efficacy of gefi- ceptors and was initially designed to overcome T790M
tinib among 41 patients with brain metastases.13 The overall mutation.18 One-hundred patients with brain metastases
response rate was 87.8%, with a progression-free survival from EGFR-mutant NSCLC received afatinib in a compas-
(PFS) of 14.5 months (95% CI, 10.218.3 months) and a sionate use program.19 All of the patients had been treated
median OS of 21.9 months (95% CI, 18.530.1 months).13 previously with either erlotinib or gefitinib. For the 35
Patients with an EGFR exon 19 deletion had significantly evaluable patients, time to tumor progression of 3.6 months
better PFS and OS compared with those with an exon 21 with an overall cerebral response rate of 35% was reported.
L858R mutation. In one study, 31 Korean patients who had However, the CNS efficacy of afatinib has not been com-
never smoked and had EGFR-mutant NSCLC and synchro- pared with either erlotinib or gefitinib.
nous asymptomatic brain metastases were treated with A number of third-generation EGFR TKIs, including osi-
gefitinib or erlotinib.14 The objective response rate was mertinib (AZD-9291), rociletinib (CO-1686), ASP-8273, and
96.9%, PFS was 7.1 months, and OS was 18.8 months. HM-61713, are in various phases of clinical investigation.20-23
Several preclinical studies with human cell lines have in- These agents spare wild-type EGFRs and target mutant EGFRs,
dicated that EGFR TKIs enhance radiation therapy.15 Nu- including T790M.24 The activity of these newer agents against
merous clinical trials and retrospective reports have shown brain metastases is currently being investigated. Initial studies
increased efficacy of combining TKIs with radiation therapy with the third-generation EGFR TKIs included several patients
in the context of brain metastases from EGFR-mutant NSCLC. with brain metastases. For example, 162 of 411 patients in the
In a prospective phase II study, 40 patients with brain phase I/II trial of AZD-9291 had CNS metastases,25 and 186 of
metastases received erlotinib for 1 week prior to WBRT, 450 patients (41%) enrolled in the phase I/II trial of rociletinib
concurrently with WBRT followed by maintenance erloti- had a history of CNS involvement.26 In a report of patients who
nib.16 Nine of the 40 patients carried an EGFR mutation, and were treated with osimertinib after progression on rociletinib,
an overall response rate of 89.9% with a median OS of all three patients with brain metastases showed a response.27
19.1 months was noted in that cohort. In a randomized AZD-3759 is a novel EGFR TKI designed to cross the blood
phase II study, 80 patients with brain metastases from NSCLC brain barrier; however, it lacks activity against T790M mu-
received either erlotinib or placebo with WBRT.17 In the tation.2 Preclinical studies show that AZD-3759 is not a sub-
erlotinib arm, 35 of 40 patients had wild-type EGFR; no strate of P-gp efflux pumps and has significantly higher
penetration across the bloodbrain barrier. Preliminary
results of the ongoing phase I trial of AZD-3759 demon-
strated that the drug was well tolerated, with early evidence
of intracranial activity.28
KEY POINTS Approximately 3%7% of patients with nonsquamous NSCLC
have ALK gene rearrangement, resulting in a novel fusion gene
Brain metastases are a major cause of morbidity for containing portions of echinoderm microtubule-associated
patients with advanced malignancies, and the incidence protein like-4 and the ALK gene.29 This rearrangement leads
of brain metastases is increasing. to constitutive activity of the kinase domain of the ALK, leading
For patients with nonsmall cell lung cancer with a EGFR
to activation of the phosphoinositide 3-kinase and RAS path-
mutation and an anaplastic lymphoma kinase mutation
with brain metastases, several agents have shown
ways.30 Crizotinib is a first-generation ALK inhibitor that also
intracranial activity. Newer EGFR-targeting agents with has activity against ROS1 and MET tyrosine kinases. Although
better central nervous system penetration are in crizotinib does not have good bloodbrain barrier penetration,
development. CNS activity has been seen in several case reports and in
Lapatinib in combination with capecitabine and retrospective subanalyses of clinical trials. In a retrospective
neratinib has been tested in clinical trials for patients pooled analysis of 388 patients enrolled in the PROFILE 1005-
with brain metastases from breast cancer harboring a 1007 clinical trial of crizotinib, 275 patients had asymptomatic
HER2 mutation. brain metastases.31 Of the 275 patients with brain metastases,
Approximately 50% of patients with metastatic 166 had received prior treatment. The intracranial disease
melanoma carry a BRAF mutation. Small control rate was similar among those with prior treatment
moleculetargeted therapies such as vemurafenib and
compared with treatment-naive patients. However, the me-
dabrafenib have shown efficacy in BRAFV600-positive
melanoma brain metastases.
dian intracranial time to tumor progression was 7 months for
Innovative drug development and clinical trials for brain patients with previously untreated brain metastases compared
metastases are needed to improve treatment options with 13.2 months for patients with previously treated brain
and consequently outcomes of patients with brain metastases. In addition, the patients who continued crizotinib
metastases. beyond progression had better OS compared with those
who discontinued therapy at progression. A multi-institutional

e124 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


TARGETED THERAPY IN BRAIN METASTASES

TABLE 1. Selected Studies With Targeted Therapy in Brain Metastases


Intracranial
Concurrent Intracranial Progression- Overall
Primary Specific Trial Therapy Local No. of Response Free Survival Survival
Study Malignancy Design Characteristics Tested Treatment Patients Rate (%) (Months) (Months)
16
Welsh et al NSCLC Phase II Erlotinib started Erlotinib WBRT 40 86 8 11.8
a week prior to
and continued
through and
after WBRT
Lee et al17 NSCLC Phase II A randomized trial Erlotinib WBRT Arm A: 40 1.6 2.9
predominantly
Arm B: 40 1.6 3.4
among patients
with the EGFR
wild type; arm A:
WBRT alone;
arm B: WBRT plus
erlotinib
Ceresoli et al81 NSCLC Phase II Single-arm study, Gefitinib None 41 27 3 5
included both
newly diagnosed
and old BMs
Sperduto et al82 NSCLC Phase 3-arm study; arm A: Erlotinib WBRT plus Arm A: 44 8.1 13.4
(RTOG-0320) III WBRT plus SRS; SRS
Arm B: 40 4.6 6.3
arm B: WBRT plus
SRS plus temo- Arm C: 41 4.8 6.1
zolomide; arm
C: WBRT plus SRS
plus erlotinib
Bachelot et al59 HER2- Phase II Newly diagnosed Lapatinib None 45 65.9 5.5 17
(LANDSCAPE) positive BMs plus
breast capecitabine
cancer
Corte s et al61 HER2- Phase II 3-arm study; arm A: Afatinib None Arm A: 40 30 (12 of 40
positive afatinib; arm B: patients)*
breast afatinib plus
Arm B: 38 34.2 (13 of 38
cancer vinorelbine;
patients)*
arm C: investiga-
tors choice Arm C: 43 41.9 (18 of 43
patients)*
Freedman et al63 HER2- Phase II Single-arm study Neratinib None 40 1.9 8.7
positive in previously
breast treated patients
cancer
Long et al71 BRAF- Phase II Cohort A: no prior Dabrafenib None Cohort A, 39.2 4 (16.1 weeks) 8.2 (33.1
positive treatment of V600E: 74 weeks)
melanoma BMs; cohort B:
Cohort A, 6.7 2 (8.1 weeks) 7.7 (31.4
previously
V600K: 15 weeks)
treated BMs
Cohort B, 30.8 4.1 (16.6 4 (16.3
V600E: 65 weeks) weeks)
Cohort B, 22.2 4 (15.9 weeks) 5.2 (21.9
V600K: 18 weeks)
Kefford et al73 BRAF- Phase II Cohort 1: previously Vemurafenib None Cohort 1: 90 18 3.7 7
positive untreated;
Cohort 2: 56 20 3.94 9.53
melanoma cohort 2: previ-
ously treated
Abbreviations: NSCLC, nonsmall cell lung cancer; WBRT, whole-brain radiation therapy; BM, brain metastasis; SRS, stereotactic radiation.
*Patient benefit (defined as intracranial or extracranial progression-free survival, new neurologic signs or symptoms related to tumor, or increased corticosteroid use) at
12 weeks.

retrospective study included 90 patients with brain metastases brain metastasis was reported. The investigators concluded
from ALK-positive lung cancer.32 The majority of patients (84 of that absence of extracranial disease, a Karnofsky performance
90) received radiation therapy. Crizotinib was administered to score (KPS) of 90 or greater, and no prior ALK inhibitor therapy
84 patients and a second-generation ALK inhibitor was given to were independent prognostic factors for patients with brain
41 patients (21 received ceritinib, 16 were treated with bri- metastases from ALK-positive lung cancer. Several recent
gatinib, two received alectinib, and two were administered studies have suggested CNS treatment failure and CNS-
X-396). A median OS of 49.5 months from the diagnosis of a predominant progression among patients treated with

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e125


VENUR AND AHLUWALIA

crizotinib.33-35 Aggressive tumor biology, poor CNS penetration radiolabeled trastuzumab have shown increased CNS bio-
of crizotinib, and mutation in the binding domain of crizotinib availability.51,52 Although no prospective clinical trial has yet
have been suggested as the likely reasons.36,37 reported on the intracranial activity of trastuzumab, several
Ceritinib and alectinib are second-generation ALK in- retrospective studies have shown superior outcomes with
hibitors that can be used to overcome drug resistance to trastuzumab after radiation.53 In one such retrospective
crizotinib.38,39 The phase I trial of ceritinib (ASCEND-1) ac- study, the median OS of patients with HER2-overexpressing
crued 246 patients, 124 of which had brain metastases at breast cancer brain metastases was 21 months with WBRT
baseline.40 Ninety-eight patients had received ALK inhibitors followed by trastuzumab, compared with 9 months with
in the past, whereas 26 patients were ALK-inhibitor naive. Of WBRT followed by chemotherapy.53 Two-thirds of patients
the 14 patients with measurable brain metastases, seven with brain metastases still die of the systemic disease, and
patients showed an intracranial response and three patients better control of the primary cancer and extracranial me-
had stable disease. Unlike crizotinib and ceritinib, alectinib is tastases may be the reason for this observed survival benefit
not a substrate for P-gp efflux pumps, which may result in with the use of trastuzumab. Hence, for a patient with
a higher brain to plasma concentration of alectinib.41 The isolated CNS progression, it is reasonable to continue anti-
phase I/II study of alectinib enrolled 47 patients who pro- HER2 therapy as long as the extracranial disease is well
gressed or were intolerant to crizotinib.42 Eleven of the 21 controlled.
patients with asymptomatic brain metastases had an ob- HER2 (ERBB2) is shown to enhance EGFR signaling and vice
jective response (six complete responses, five partial re- versa.54,55 Therefore, it is postulated that dual inhibition
sponses). Brigatinib is another ALK inhibitor that has shown of HER2 and EGFR by agents such as lapatinib may confer
intracranial activity in an early phase I/II trial.43 A phase I trial improved outcomes among this patient population.56
of brigatinib included 46 patients with brain metastases. Lapatinib in combination with capecitabine is approved for
Thirteen patients had evaluable brain metastases; nine treatment of patients with HER2-overexpressing metastatic
patients (69%) had regression of the intracranial lesions, breast cancer who have progressed while taking trastuzu-
including four patients with a complete response and two mab. Lapatinib is a small molecule inhibitor; however, its
with a partial response.44 The median intracranial PFS was bioavailability in the CNS is limited by breast cancer re-
97 weeks. The phase II trial of this agent is currently accruing sistance protein and P-gp efflux pumps.57 Lapatinib was
patients and includes a cohort of patients with brain evaluated in an initial phase II study of patients with pro-
metastases.44 gressive brain metastases from HER2-positive breast can-
cer.58 The trial accrued 242 patients and all had received
prior radiation therapy and trastuzumab. The median PFS
TARGETED THERAPY FOR BREAST CANCER WITH and OS were 2.4 months and 6.37 months, respectively. This
BRAIN METASTASES study was amended to allow 50 additional patients who
Breast cancer is the second leading cause of brain metas- were treated with lapatinib and capecitabine after pro-
tases. The propensity of brain metastases varies with gression with lapatinib. In this cohort, the median PFS was
subtypes of breast cancer. Patients with metastatic triple- 3.65 months and a response rate of 20% was seen. The
negative breast cancer typically have brain metastases early pivotal study for the utility of lapatinib for patients with
in the course of their disease, and they frequently present breast cancer with brain metastases was the LANDSCAPE
with simultaneous progression of intracranial and extra- trial, which investigated the combination of lapatinib and
cranial disease.45,46 By contrast, patients with HER2 over- capecitabine as a first-line combination therapy prior to
expression are often treated with targeted therapies that radiation.59 An intracranial response of 66% was seen among
include monoclonal antibodies, and they develop CNS me- the 45 patients enrolled in this phase II trial. The WBRT was
tastases relatively later in the disease process, often with delayed by 8.6 months for patients with oligometastatic
stable extracranial disease. Approximately 30%55% of asymptomatic brain metastases treated with lapatinib and
patients with HER2-positive breast cancer develop brain capecitabine. A median OS of 17.0 months was reported
metastases.47,48 The increased incidence of brain metasta- among 44 patients who had measurable intracranial disease.
ses in this subtype of breast cancer is attributed to two main There is an ongoing randomized cooperative group study
reasons. First, anti-HER2 agents such as trastuzumab are evaluating WBRT in combination with lapatinib for patients
effective in controlling the systemic disease, prolonging with brain metastases from HER2-positive breast cancer
survival, and, in turn, unmasking otherwise asymptomatic (NCT01622868).
brain metastases. Second, the monoclonal antibodies di- The drug-antibody conjugate T-DM1 (trastuzumab plus
rected against HER2, like trastuzumab, have poor CNS emtansine) is approved by the FDA for treatment of pa-
penetration, especially for patients with an intact blood tients with HER2-positive breast cancer. In a recent ret-
brain barrier, thereby leading to brain metastases without rospective report, an intracranial response rate of 20% was
systemic progression of the disease.49 In the absence of seen for 10 patients with asymptomatic or progressive
brain metastases, the CNS concentration of trastuzumab is brain metastases.60 Prospective trials are needed to better
low compared with systemic concentrations.50 However, understand the clinical activity of T-DM1 among patients
among patients with brain metastases, studies using with brain metastases.

e126 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


TARGETED THERAPY IN BRAIN METASTASES

Afatinib, a second-generation inhibitor of the ERBB family, seen for approximately 50% of patients with metastatic mel-
is approved by the FDA for treatment of patients with NSCLC anoma and plays an important role in the MAPK (RAS-RAF-MEK-
harboring an EGFR mutation.18 In a phase II study of 121 ERK) pathway. Dabrafenib and vemurafenib are agents active
patients with HER2-overexpressing breast cancer with brain against BRAF V600E or V600K mutation that are approved by
metastases, 40 patients were treated with afatinib, 38 re- the FDA for treatment of advanced metastatic melanoma. In the
ceived afatinib and vinorelbine, and another 43 were treated phase I trial with dabrafenib, 8 of 10 patients with untreated
with the regimen of the investigators choice.61 Trastuzumab asymptomatic brain metastases showed an intracranial
and vinorelbine (11 of 43 patients) and lapatinib plus cape- response (four had a complete response and four had a partial
citabine (eight patients) were the most common treatments response).70 This initial observation led to a phase II multi-
chosen by the treating investigator. The primary study end- center open-label study of dabrafenib among 172 patients with
point (absence of CNS or extracranial disease progression, asymptomatic brain metastases with a BRAFV600E or BRAFV600K
new neurologic symptoms, or new corticosteroid use [termed mutation and at least one measurable brain metastasis (BREAK-
as patient benefit]) at 12 weeks was seen in 12 of 40 patients MB).71 Cohort A consisted of 89 patients who had never re-
(30%) in the afatinib arm, 13 of 38 patients (34%) in the ceived any local treatment and cohort B comprised 83 patients
afatinib plus vinorelbine arm, and 18 of 43 patients (42%) in with a history of prior radiation therapy for brain metastases.
the investigator choice arm. A modified version of the RECIST criteria with up to five in-
Neratinib is a potent irreversible inhibitor of EGFR, HER2, tracranial and extracranial index lesions was used to measure
and ERBB4 transmembrane tyrosine kinase receptors.62 A the response to treatment. In cohorts A and B, patients with a
phase II single-arm study evaluated neratinib among pa- BRAFV600E mutation had intracranial response rates of 39%
tients with HER2-overexpressing breast cancer with brain (29 of 74) and 31% (20 of 65), respectively. Progression-free
metastases.63 The primary trial endpoint was intracranial survival of 16.1 weeks and 16.6 weeks and OS of 33.1 and
response rate, which also included neurocognitive and quality- 31.4 weeks were noted in cohorts A and B, respectively. Pa-
of-life (QoL) assessments. The intracranial response rate among tients with a BRAFV600K mutation had lower intracranial re-
patients enrolled in the study was 8% (3 of 40 patients failed to sponse rates of 7% (1 of 15) and 22% (4 of 18) in cohorts A and B,
meet the primary endpoint of success). The rapid accrual of respectively. This trial demonstrated that dabrafenib is active in
patients and the neurocognitive and QoL endpoints of the trial BRAF-mutant melanoma brain metastases, specifically among
were noteworthy. patients with a BRAFV600E mutation with acceptable toxicity.
Pertuzumab, an antibody against the extracellular di- Vemurafenib was noted to have intracranial activity in an open-
merization domain of the HER2 receptor, has been shown to label pilot trial of 24 untreated patients with melanoma brain
increase efficacy when used with trastuzumab and docetaxel metastases harboring a BRAFV600 mutation.72 Of 19 patients with
among patients with untreated HER2-overexpressing meta- measurable intracranial disease, seven had tumor regression of
static breast cancer.64 A case report has shown intracranial more than 30% and three achieved a partial response. Median
activity from the combination of pertuzumab, trastuzumab, PFS and OS were 3.9 and 5.3 months, respectively. Initial results
and docetaxel.65 of a phase II study of 146 patients with BRAFV600 melanoma brain
Patients with luminal breast cancer have a lower fre- metastases treated with vemurafenib were reported at the 2013
quency of brain metastases.66 It has been suggested that Society of Melanoma Research Congress.73 Cohort 1 consisted of
hormone receptors are lost or altered during the process of 90 patients with newly diagnosed melanoma brain metastases
acquiring brain metastases.67 Newer agents such as mTOR and cohort 2 included 56 patients with melanoma brain me-
inhibitors and CDK4/6 have shown extracranial activity in tastases who progressed after initial local therapy. Both cohorts
advanced luminal breast cancer; however, their role in the had similar intracranial response rates at 18% and 20%, re-
treatment of patients with brain metastases is yet to be spectively. Median PFS and OS were 3.7 and 6.5 months in cohort
defined. There have been limited novel agents available for 1 and 4.0 and 6.4 months in cohort 2, respectively.
the treatment of patients with triple-negative breast cancer The feasibility, safety profile, and effectiveness of com-
with brain metastases who have a dismal prognosis. bining BRAF inhibitors and radiation have not yet been
tested in a prospective setting. There is concern regarding an
increased incidence of dermatitis with concurrent use of
TARGETED THERAPY FOR BRAIN METASTASES radiation and BRAF inhibitors, specifically in the extracranial
FROM MELANOMA setting.74 A retrospective analysis evaluated the effectiveness
Targeted agents against driver oncogenic mutations among cell of combining vemurafenib and radiation in BRAFV600 mela-
signaling pathways have shown benefit in metastatic mela- noma brain metastases.75 Six patients were treated with SRS:
noma. BRAF, NRAS, and KIT are the mutually exclusive driver two received WBRT, one received SRS and WBRT, and the
mutations commonly seen in metastatic melanoma.68 The remaining three received surgery and radiation. All 12 patients
presence of these mutations increases the risk of CNS metas- received vemurafenib. Of the 48 index lesions, 36 showed a
tases at the diagnosis of stage IV melanoma. In a 2012 study, a response, with 23 (48%) complete responses and 13 (27%)
higher prevalence of melanoma brain metastases was observed partial responses. Several other small retrospective case series
in BRAF- and NRAS-mutant melanoma (24% and 23%, re- have been reported on outcomes of the combination of tar-
spectively) compared with wild type (12%).69 BRAF mutation is geted agents with SRS/WBRT.76-78

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e127


VENUR AND AHLUWALIA

CONCLUSION metastases, despite limitations of the bloodbrain barrier and


Targeted therapies have improved survival in a subset of efflux pumps. Over the last few years, our understanding of
patients with NSCLC, breast cancer, and melanoma with molecular drivers of brain metastases, the bloodbrain bar-
actionable driver mutations. For patients with brain metas- rier, and CNS penetration of drugs has improved tremen-
tases, these agents provide not only intracranial control of the dously.80 Improved understanding of biology, multimodality
disease, but they also help manage systemic cancer. The treatment, and innovative drug development can help lead to
clinical and pathobiology of brain metastases for patients with improvement in the management of brain metastases. In the
mutations such as EGFR may differ from other patients with future, increasingly targeted therapies will have a significant
NSCLC without such mutations.79 Some of the newer targeted role in the treatment of patients with brain metastases with
agents have shown promising results in the treatment of brain actionable mutations.

References
1. Tabouret E, Chinot O, Metellus P, et al. Recent trends in epidemiology of 15. Dempke WC, Edvardsen K, Lu S, et al. Brain metastases in NSCLC - are
brain metastases: an overview. Anticancer Res. 2012;32:4655-4662. TKIs changing the treatment strategy? Anticancer Res. 2015;35:
2. Kim DW, Yang JCH, Chen K, et al. AZD3759, an EGFR inhibitor with blood 5797-5806.
brain barrier (BBB) penetration for the treatment of non-small cell lung 16. Welsh JW, Komaki R, Amini A, et al. Phase II trial of erlotinib plus
cancer (NSCLC) with brain metastasis (BM): preclinical evidence and concurrent whole-brain radiation therapy for patients with brain
clinical cases. J Clin Oncol. 2015;33 (suppl; abstr 8016). metastases from non-small-cell lung cancer. J Clin Oncol. 2013;31:895-
3. DAntonio C, Passaro A, Gori B, et al. Bone and brain metastasis in lung 902.
cancer: recent advances in therapeutic strategies. Ther Adv Med Oncol. 17. Lee SM, Lewanski CR, Counsell N, et al. Randomized trial of erlotinib plus
2014;6:101-114. whole-brain radiotherapy for NSCLC patients with multiple brain me-
4. Shigematsu H, Lin L, Takahashi T, et al. Clinical and biological features tastases. J Natl Cancer Inst. 2014;106:dju151.
associated with epidermal growth factor receptor gene mutations in 18. Sequist LV, Yang JC, Yamamoto N, et al. Phase III study of afatinib or
lung cancers. J Natl Cancer Inst. 2005;97:339-346. cisplatin plus pemetrexed in patients with metastatic lung adenocar-
5. Lynch TJ, Bell DW, Sordella R, et al. Activating mutations in the epi- cinoma with EGFR mutations. J Clin Oncol. 2013;31:3327-3334.
dermal growth factor receptor underlying responsiveness of non-small- 19. Hoffknecht P, Tufman A, Wehler T, et al. Efficacy of the irreversible ErbB
cell lung cancer to gefitinib. N Engl J Med. 2004;350:2129-2139. family blocker afatinib in epidermal growth factor receptor (EGFR)
6. Eichler AF, Kahle KT, Wang DL, et al. EGFR mutation status and survival tyrosine kinase inhibitor (TKI)-pretreated non-small-cell lung cancer
after diagnosis of brain metastasis in nonsmall cell lung cancer. Neuro patients with brain metastases or leptomeningeal disease. J Thorac
Oncol. 2010;12:1193-1199. Oncol. 2015;10:156-163.
7. Tan CS, Cho BC, Soo RA. Next-generation epidermal growth factor 20. Cross DA, Ashton SE, Ghiorghiu S, et al. AZD9291, an irreversible EGFR
receptor tyrosine kinase inhibitors in epidermal growth factor receptor TKI, overcomes T790M-mediated resistance to EGFR inhibitors in lung
-mutant non-small cell lung cancer. Lung Cancer. 2016;93:59-68. cancer. Cancer Discov. 2014;4:1046-1061.
8. Weber B, Winterdahl M, Memon A, et al. Erlotinib accumulation in brain 21. Sequist LV, Soria JC, Goldman JW, et al. Rociletinib in EGFR-mutated
metastases from non-small cell lung cancer: visualization by positron non-small-cell lung cancer. N Engl J Med. 2015;372:1700-1709.
emission tomography in a patient harboring a mutation in the epidermal 22. Sakagami H, Konagai S, Yamamoto H, et al. ASP8273, a novel mutant-
growth factor receptor. J Thorac Oncol. 2011;6:1287-1289. selective irreversible EGFR inhibitor, inhibits growth of non-small cell
9. Elmeliegy MA, Carcaboso AM, Tagen M, et al. Role of ATP-binding lung cancer (NSCLC) cells with EGFR activating and T790M resistance
cassette and solute carrier transporters in erlotinib CNS penetration and mutations. Cancer Res. 2014;74 (suppl; abstr 1728).
intracellular accumulation. Clin Cancer Res. 2011;17:89-99. 23. Lee KO, Cha MY, Kim M, et al. Discovery of HM61713 as an orally
10. Kitazaki T, Oka M, Nakamura Y, et al. Gefitinib, an EGFR tyrosine kinase available and mutant EGFR selective inhibitor. Cancer Res. 2014;74
inhibitor, directly inhibits the function of P-glycoprotein in multidrug (suppl; abstr LB-100).
resistant cancer cells. Lung Cancer. 2005;49:337-343. 24. Janne PA, Yang JC, Kim DW, et al. AZD9291 in EGFR inhibitor-resistant
11. Togashi Y, Masago K, Fukudo M, et al. Cerebrospinal fluid concentration non-small-cell lung cancer. N Engl J Med. 2015;372:1689-1699.
of erlotinib and its active metabolite OSI-420 in patients with central 25. Ahn MJ, Tsai CM, Yang JCH, et al. 3083 AZD9291 activity in patients with
nervous system metastases of non-small cell lung cancer. J Thorac EGFR-mutant advanced non-small cell lung cancer (NSCLC) and brain
Oncol. 2010;5:950-955. metastases: data from phase II studies. Eur J Cancer. 2015;51:
12. Wakeling AE, Guy SP, Woodburn JR, et al. ZD1839 (Iressa): an orally S625-S626.
active inhibitor of epidermal growth factor signaling with potential for 26. Bedikian AY, Papadopoulos NE, Kim KB, et al. A phase IB trial of in-
cancer therapy. Cancer Res. 2002;62:5749-5754. travenous INO-1001 plus oral temozolomide in subjects with unre-
13. Iuchi T, Shingyoji M, Sakaida T, et al. Phase II trial of gefitinib alone sectable stage-III or IV melanoma. Cancer Invest. 2009;27:756-763.
without radiation therapy for Japanese patients with brain metasta- 27. Sequist LV, Piotrowska Z, Niederst MJ, et al. Osimertinib responses after
ses from EGFR-mutant lung adenocarcinoma. Lung Cancer. 2013;82: disease progression in patients who had been receiving rociletinib.
282-287. JAMA Oncol. 2016;2:541-543.
14. Kim JE, Lee DH, Choi Y, et al. Epidermal growth factor receptor tyrosine 28. Zeng Q, Wang J, Cheng Z, et al. Discovery and evaluation of clinical
kinase inhibitors as a first-line therapy for never-smokers with ade- candidate AZD3759, a potent, oral active, central nervous system-penetrant,
nocarcinoma of the lung having asymptomatic synchronous brain epidermal growth factor receptor tyrosine kinase inhibitor. J Med Chem.
metastasis. Lung Cancer. 2009;65:351-354. 2015;58:8200-8215.

e128 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


TARGETED THERAPY IN BRAIN METASTASES

29. Bohn JP, Pall G, Stockhammer G, et al. Targeted therapies for the incidence, treatment, and survival in patients from registHER. Clin
treatment of brain metastases in solid tumors. Target Oncol. Epub 2016 Cancer Res. 2011;17:4834-4843.
Jan 29. 48. Olson EM, Abdel-Rasoul M, Maly J, et al. Incidence and risk of central
30. Zhang I, Zaorsky NG, Palmer JD, et al. Targeting brain metastases in nervous system metastases as site of first recurrence in patients with
ALK-rearranged non-small-cell lung cancer. Lancet Oncol. 2015;16: HER2-positive breast cancer treated with adjuvant trastuzumab. Ann
e510-e521. Oncol. 2013;24:1526-1533.
31. Costa DB, Shaw AT, Ou SH, et al. Clinical experience with crizotinib in 49. Pestalozzi BC, Holmes E, de Azambuja E, et al. CNS relapses in patients
patients with advanced ALKS-rearranged non-small-cell lung cancer and with HER2-positive early breast cancer who have and have not received
brain metastases. J Clin Oncol. 2015;33:1881-1888. adjuvant trastuzumab: a retrospective substudy of the HERA trial (BIG
32. Johung KL, Yeh N, Desai NB, et al. Extended survival and prognostic 1-01). Lancet Oncol. 2013;14:244-248.
factors for patients with ALK-rearranged non-small-cell lung cancer and 50. Stemmler HJ, Kahlert S, Siekiera W, et al. Characteristics of patients with
brain metastasis. J Clin Oncol. 2016;34:123-129. brain metastases receiving trastuzumab for HER2 overexpressing
33. Maillet D, Martel-Lafay I, Arpin D, et al. Ineffectiveness of crizotinib on metastatic breast cancer. Breast. 2006;15:219-225.
brain metastases in two cases of lung adenocarcinoma with EML4-ALK 51. Dijkers EC, Oude Munnink TH, Kosterink JG, et al. Biodistribution
rearrangement. J Thorac Oncol. 2013;8:e30-e31. of 89Zr-trastuzumab and PET imaging of HER2-positive lesions in
34. Riess JW, Nagpal S, Neal JW, et al. A patient with anaplastic lymphoma patients with metastatic breast cancer. Clin Pharmacol Ther. 2010;
kinase-positive non-small cell lung cancer with development of lep- 87:586-592.
tomeningeal carcinomatosis while on targeted treatment with crizo- 52. Tamura K, Kurihara H, Yonemori K, et al. 64Cu-DOTA-trastuzumab PET
tinib. J Natl Compr Canc Netw. 2013;11:389-394. imaging in patients with HER2-positive breast cancer. J Nucl Med. 2013;
35. Chun SG, Choe KS, Iyengar P, et al. Isolated central nervous system 54:1869-1875.
progression on crizotinib: an Achilles heel of non-small cell lung cancer 53. Bartsch R, Rottenfusser A, Wenzel C, et al. Trastuzumab prolongs overall
with EML4-ALK translocation? Cancer Biol Ther. 2012;13:1376-1383. survival in patients with brain metastases from Her2 positive breast
36. Costa DB, Kobayashi S, Pandya SS, et al. CSF concentration of the cancer. J Neurooncol. 2007;85:311-317.
anaplastic lymphoma kinase inhibitor crizotinib. J Clin Oncol. 2011;29: 54. Worthylake R, Opresko LK, Wiley HS. ErbB-2 amplification inhibits
e443-e445. down-regulation and induces constitutive activation of both ErbB-2 and
37. Choi YL, Soda M, Yamashita Y, et al; ALK Lung Cancer Study Group. epidermal growth factor receptors. J Biol Chem. 1999;274:8865-8874.
EML4-ALK mutations in lung cancer that confer resistance to ALK in- 55. Graus-Porta D, Beerli RR, Daly JM, et al. ErbB-2, the preferred hetero-
hibitors. N Engl J Med. 2010;363:1734-1739. dimerization partner of all ErbB receptors, is a mediator of lateral sig-
38. Shaw AT, Kim DW, Mehra R, et al. Ceritinib in ALK-rearranged non-small- naling. EMBO J. 1997;16:1647-1655.
cell lung cancer. New Engl J Med. 2014;370:1189-1197. 56. Konecny GE, Pegram MD, Venkatesan N, et al. Activity of the dual
39. Seto T, Kiura K, Nishio M, et al. CH5424802 (RO5424802) for patients kinase inhibitor lapatinib (GW572016) against HER-2-overexpressing
with ALK-rearranged advanced non-small-cell lung cancer (AF-001JP and trastuzumab-treated breast cancer cells. Cancer Res. 2006;66:
study): a single-arm, open-label, phase 1-2 study. Lancet Oncol. 2013; 1630-1639.
14:590-598. 57. Polli JW, Olson KL, Chism JP, et al. An unexpected synergist role
40. Shaw A, Mehra R, Tan DSW, et al. BM-32 Ceritinib (LDK378) for of P-glycoprotein and breast cancer resistance protein on the
treatment of patients with ALK-rearranged (ALK+) non-small cell lung central nervous system penetration of the tyrosine kinase inhibitor
cancer (NSCLC) and brain metastases (BM) in the ASCEND-1 trial. Neuro lapatinib (N-3-chloro-4-[(3-fluorobenzyl)oxy]phenyl-6-[5-([2-(methylsulfonyl)
Oncol. 2014;16:v39. ethyl]aminomethyl)-2-furyl]-4-quinazolinamine; GW572016). Drug Metab
41. Kodama T, Hasegawa M, Takanashi K, et al. Antitumor activity of the Dispos. 2009;37:439-442.
selective ALK inhibitor alectinib in models of intracranial metastases. 58. Lin NU, Dieras V, Paul D, et al. Multicenter phase II study of lapatinib in
Cancer Chemother Pharmacol. 2014;74:1023-1028. patients with brain metastases from HER2-positive breast cancer. Clin
42. Gadgeel SM, Gandhi L, Riely GJ, et al. Safety and activity of alectinib Cancer Res. 2009;15:1452-1459.
against systemic disease and brain metastases in patients with crizotinib- 59. Bachelot T, Romieu G, Campone M, et al. Lapatinib plus capecitabine in
resistant ALK-rearranged non-small-cell lung cancer (AF-002JG): results patients with previously untreated brain metastases from HER2-positive
from the dose-finding portion of a phase 1/2 study. Lancet Oncol. 2014; metastatic breast cancer (LANDSCAPE): a single-group phase 2 study.
15:1119-1128. Lancet Oncol. 2013;14:64-71.
43. Camidge DR, Bazhenova L, Salgia R, et al. Safety and efficacy of brig- 60. Bartsch R, Berghoff AS, Vogl U, et al. Activity of T-DM1 in Her2-positive
atinib (AP26113) in advanced malignancies, including ALK+ non-small breast cancer brain metastases. Clin Exp Metastasis. 2015;32:729-737.
cell lung cancer (NSCLC). J Clin Oncol. 2015;33 (suppl; abstr 8062). 61. Cortes J, Dieras V, Ro J, et al. Afatinib alone or afatinib plus vinorelbine
44. Kerstein D, Gettinger S, Gold K, et al. LBA4: evaluation of anaplastic versus investigators choice of treatment for HER2-positive breast
lymphoma kinase (ALK) inhibitor brigatinib [AP26113] in patients with cancer with progressive brain metastases after trastuzumab, lapatinib,
ALK+ non-small cell lung cancer (NSCLC) and brain metastases. Ann or both (LUX-Breast 3): a randomised, open-label, multicentre, phase 2
Oncol. 2015;26:i60-i61. trial. Lancet Oncol. 2015;16:1700-1710.
45. Dawood S, Lei X, Litton JK, et al. Incidence of brain metastases as a first 62. Zhang X, Munster PN. New protein kinase inhibitors in breast cancer:
site of recurrence among women with triple receptor-negative breast afatinib and neratinib. Expert Opin Pharmacother. 2014;15:1277-1288.
cancer. Cancer. 2012;118:4652-4659. 63. Freedman RA, Gelman RS, Wefel JS, et al. Translational Breast Cancer
46. Lin NU, Claus E, Sohl J, et al. Sites of distant recurrence and clinical Research Consortium (TBCRC) 022: a phase II trial of neratinib for
outcomes in patients with metastatic triple-negative breast cancer: high patients with human epidermal growth factor receptor 2-positive breast
incidence of central nervous system metastases. Cancer. 2008;113: cancer and brain metastases. J Clin Oncol. 2016;34:945-952.
2638-2645. 64. Swain SM, Baselga J, Kim SB, et al; CLEOPATRA Study Group. Pertu-
47. Brufsky AM, Mayer M, Rugo HS, et al. Central nervous system me- zumab, trastuzumab, and docetaxel in HER2-positive metastatic breast
tastases in patients with HER2-positive metastatic breast cancer: cancer. N Engl J Med. 2015;372:724-734.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e129


VENUR AND AHLUWALIA

65. Senda N, Yamaguchi A, Nishimura H, et al. Pertuzumab, trastuzumab 2, multicenter study. Paper presented at: 2013 Society for Melanoma
and docetaxel reduced the recurrence of brain metastasis from breast Research Congress; November 2013; Philadelphia, PA.
cancer: a case report. Breast Cancer. 2016;23:323-328. 74. Satzger I, Degen A, Asper H, et al. Serious skin toxicity with the com-
66. Arvold ND, Oh KS, Niemierko A, et al. Brain metastases after breast- bination of BRAF inhibitors and radiotherapy. J Clin Oncol. 2013;31:
conserving therapy and systemic therapy: incidence and characteristics e220-e222.
by biologic subtype. Breast Cancer Res Treat. 2012;136:153-160. 75. Narayana A, Mathew M, Tam M, et al. Vemurafenib and radiation
67. Duchnowska R, Dziadziuszko R, Trojanowski T, et al. Conversion of therapy in melanoma brain metastases. J Neurooncol. 2013;113:
epidermal growth factor receptor 2 and hormone receptor expression 411-416.
in breast cancer metastases to the brain. Breast Cancer Res. 2012;14: 76. Ahmed KA, Freilich JM, Sloot S, et al. LINAC-based stereotactic radio-
R119. surgery to the brain with concurrent vemurafenib for melanoma metas-
68. Long GV, Margolin KA. Multidisciplinary approach to brain metastasis tases. J Neurooncol. 2015;122:121-126.
from melanoma: the emerging role of systemic therapies. Am Soc Clin 77. Wolf A, Zia S, Verma R, et al. Impact on overall survival of the com-
Oncol Educ Book. 2013;33:393-398. bination of BRAF inhibitors and stereotactic radiosurgery in patients
69. Jakob JA, Bassett RL Jr, Ng CS, et al. NRAS mutation status is an in- with melanoma brain metastases. J Neurooncol. Epub 2016 Feb 6.
dependent prognostic factor in metastatic melanoma. Cancer. 2012; 78. Patel BG, Ahmed KA, Johnstone PA, et al. Initial experience with combined
118:4014-4023. BRAF and MEK inhibition with stereotactic radiosurgery for BRAF mutant
70. Falchook GS, Long GV, Kurzrock R, et al. Dabrafenib in patients with melanoma brain metastases. Melanoma Res. Epub 2016 Feb 29.
melanoma, untreated brain metastases, and other solid tumours: a phase 79. Eichler AF, Chung E, Kodack DP, et al. The biology of brain metastases-
1 dose-escalation trial. Lancet. 2012;379:1893-1901. translation to new therapies. Nat Rev Clin Oncol. 2011;8:344-356.
71. Long GV, Trefzer U, Davies MA, et al. Dabrafenib in patients with 80. Berghoff AS, Preusser M. The future of targeted therapies for brain
Val600Glu or Val600Lys BRAF-mutant melanoma metastatic to the metastases. Future Oncol. 2015;11:2315-2327.
brain (BREAK-MB): a multicentre, open-label, phase 2 trial. Lancet Oncol. 81. Ceresoli GL, Cappuzzo F, Gregorc V, et al. Gefitinib in patients with brain
2012;13:1087-1095. metastases from non-small-cell lung cancer: a prospective trial. Ann
72. Dummer R, Goldinger SM, Turtschi CP, et al. Vemurafenib in patients Oncol. 2004;15:1042-1047.
with BRAF(V600) mutation-positive melanoma with symptomatic brain 82. Sperduto PW, Wang M, Robins HI, et al. A phase 3 trial of whole brain
metastases: final results of an open-label pilot study. Eur J Cancer. 2014; radiation therapy and stereotactic radiosurgery alone versus WBRT and
50:611-621. SRS with temozolomide or erlotinib for non-small cell lung cancer and 1
73. Kefford R, Malo M, Arance A, et al. Vemurafenib in metastatic mela- to 3 brain metastases: Radiation Therapy Oncology Group 0320. Int J
noma patients with brain metastases: an open label, single arm, phase Radiat Oncol Biol Phys. 2013;85:1312-1318.

e130 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


CENTRAL NERVOUS SYSTEM TUMORS

The Future of Immunotherapy in


Glioblastoma

CHAIR
E. Antonio Chiocca, MD, PhD
Brigham and Womens Hospital
Boston, MA

SPEAKERS
Michael Lim, MD
The Johns Hopkins Hospital
Baltimore, MD

Michael Weller, MD
University Hospital Zurich
Zurich, Switzerland
LIM, WELLER, AND CHIOCCA

Current State of Immune-Based Therapies for Glioblastoma


Michael Lim, MD, Michael Weller, MD, and E. Antonio Chiocca, MD, PhD

OVERVIEW

Glioblastoma is one of the most aggressive solid tumors, and, despite treatment options such as surgery, radiation, and che-
motherapy, its prognosis remains grim. Novel approaches are needed to improve survival. Immunotherapy has proven efficacy for
melanoma, lung cancer, and kidney cancer and is now a focus for glioblastoma. In this article, glioblastoma-mediated immu-
nosuppression will be discussed and two exciting immune approaches, checkpoint inhibitors and viral-based therapies, will be
reviewed.

G lioblastoma has been studied as a paradigmatic tumor


for cancer-associated immunosuppression for more
than 3 decades. Initial observations included the charac-
indirectly mediates systemic immunosuppression. In fact, it
has recently been recognized that glioblastoma cells are
capable of reshaping the phenotype of tumor-infiltrating
terization of decreased immune responsiveness of periph- host cells, which comprise a large proportion of cells within
eral blood cells harvested from patients with glioblastoma.1 the microenvironment of glioblastoma, to support their
This remote effect of a locally growing neoplasm could only growth and maintain any immunosuppressive milieu.3
be explained by soluble factors released by the tumor in In addition to the soluble immunosuppressive molecule
sufficient quantities to induce systemic immunosuppression candidates mentioned above, immune-relevant molecules
(Fig. 1). The search for soluble factors produced by cultured expressed at the cell surface of glioblastoma have attracted
glioblastoma cells resulted in the identification of, among interest because there is major infiltration of glioblastomas
others, transforming growth factor (TGF)-b as a key im- by host cells, although the cells are mainly of macrophage-
munosuppressive cytokine that has remained a therapeutic monocyte lineage. These surface molecules include the CD95
target until today. Yet, local application of antisense oli- ligand, which may induce apoptosis in susceptible cells that
gonucleotides failed presumably because of poor target express the receptor, CD95, previously referred to as Fas or
coverage, whereas systemic application of TGF-b receptor APO-1. In the context of glioblastoma, cellular targets for
antagonists has its limits in nonhematologic toxicity. TGF-b CD95-mediated apoptosis are probably mainly T cells. Addi-
is a member of a large family of cytokines that interacts with tional candidate cell surface molecules with immunosup-
heterodimeric receptors. There are three TGF-b isoforms in pressive properties, but with uncertain significance in
humans that have nonoverlapping functions, at least in glioblastoma, include regeneration and tolerance factor (RTF),
development, as demonstrated in knockout mouse models. lectin-like transcript 1, and HLA-E and HLA-G.4
In contrast, the three isoforms are coregulated in glioblas- More recently, major emphasis has been placed on the
tomas, and no isotype-specific roles have been identified so aberrant expression of PD-L1 by glioblastoma cells. PD-L1 is
far in the biology of glioblastoma.2 Other soluble immu- the ligand for PD-1, a cell surface molecule expressed mainly
nosuppressive factor candidates attributed a role in shaping on T cells now referred to as an immune checkpoint, to-
the immunosuppressive microenvironment in glioblastoma gether with CTLA-4. These two molecules, PD-1 and CTLA-4,
include interleukin 10 and prostaglandin E2. mediate inactivation of T cells, and antibody-mediated
Admittedly, it has not been clarified definitively that the therapeutic neutralization of these molecules with agents
peripheral immunosuppression encountered among patients such as nivolumab or pembrolizumab that target PD-1, or
with glioblastoma is caused exclusively or even mainly by el- ipilimumab that targets CTLA-4, can be considered the
evated levels of soluble mediators systemically. Alternatively, greatest innovation in medical oncology in the past decade.5
it is conceivable that immune modulatory cells that encoun- They are currently being explored as novel agents across the
ter the glioblastoma microenvironment are forced to de- full spectrum of human cancers and have already been in-
velop into an immunosuppressive immune cell population that tegrated into the standard of care for malignant melanoma.

From The Johns Hopkins University, Baltimore, MD; University Hospital Zurich, Zurich, Switzerland; Institute for the Neurosciences at the Brigham and Womens/Faulkner Hospital,
Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, MA; The University of Chicago, Chicago, IL.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Michael Lim, MD, Johns Hopkins Hospital, Neurosurgery-Phipps 123, 600 North Wolfe St., Baltimore, MD 21287; email: mlim3@jhmi.edu.

2016 by American Society of Clinical Oncology.

e132 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


CURRENT STATE OF IMMUNE-BASED THERAPIES FOR GLIOBLASTOMA

Furthermore, antibodies targeting the ligand PD-L1 rather decreased potential for immune cell re-education by bulky
than the receptor, such as atezolizumab, also are being ex- tumor. Accordingly, most recent immunotherapy trials have
plored in various tumor entities. The extent that glioblastomas been and are now being conducted in highly selected patient
express PD-L1 in vivo has remained controversial,6 but PD-1 populations in the newly diagnosed setting and among
and CTLA-4 inhibition are remarkably active in mouse glioma patients with little or no residual tumor after concomi-
models, both as single agents and in combination. Impor- tant temozolomide chemoradiotherapy. Given these de-
tantly, inhibition of immune checkpoints probably confers a velopments, it can be assumed that neurosurgery will be
nonspecific state of immune activation that will be of benefit attributed a larger role not only in newly diagnosed patients
to patients if their particular cancers carry a high mutational but also in the setting of recurrent disease, if the first de-
load, rendering them potentially recognizable as altered. finitively positive immunotherapy data become available.
Whether the observed benefit can be translated to glio- Although removing the source of immunosuppression sur-
blastoma remains to be clarified. Several clinical trial initiatives gically seems to be the most straightforward approach to
to explore immune checkpoint inhibitors in glioblastoma are overcome immunosuppression, additional strategies include
now underway. neutralizing TGF-b, at least transiently, or employing im-
Because of disappointing results with traditional cancer mune adjuvants, such as granulocyte macrophage colony-
therapy (radiotherapy, chemotherapy, and angiogenesis stimulating factor or toll-like receptor 2 agonists. The most
inhibition by promising agents, such as VEGF pathway in- powerful combinatorial strategy for tumor-specific vaccines,
hibitors bevacizumab and cediranib) in this disease, various however, is likely to be the checkpoint inhibitors, as outlined
approaches to immunotherapy have gained a lot of interest. above. Such additional efforts at boosting immune re-
As of early 2016, several phase II and phase III clinical trials of sponses may be essential for a benefit from upcoming
immunotherapy are underway or have already been com- strategies of immunotherapy in broader populations of
pleted, including but not limited to (1) the rindopepimut patients, including patients with bulky disease, heavily
vaccine that targets epidermal growth factor variant III, pretreated patients, and older patients who are likely to
with a completed phase III trial in the newly diagnosed exhibit impaired immune responsiveness.
setting (ACT IV) and encouraging activity in phase II in the
recurrent setting (Re-ACT); (2) the six-candidate peptide
cocktail ICT-107, which is used to generate a vaccine by CHECKPOINT INHIBITORS
ex vivo stimulation of patient-derived dendritic cells, with a An important component of tumor-induced immunosup-
completed randomized phase II trial that indicated activity in pression involves the costimulatory interaction. Normally,
patients who were HLA-A2 positive and a phase III program when a T cell encounters an antigen-presenting cell (APC)
scheduled to start in early 2016; and (3) the DCVax phase III expressing the appropriate antigen, a second interaction
program that is near completion, with high logistical with a checkpoint molecule is required to either activate or
complexityit uses autologous tumor to stimulate autol- suppress the T cell (Fig. 2).7,8 This second interaction plays
ogous dendritic cells. an important role in modulating an immune response.
These treatments likely are most effective with minimal Furthermore, there are multiple costimulatory molecules,
residual tumor burden, which should be associated with which suggest that a hierarchy of activation status exists. In
decreased levels of soluble immunosuppressive factors and addition, this interaction is not unique to APCs and T cells;
other immune cells, such as natural killer cells and regulatory
T cells (Tregs), also have costimulatory molecules.8 Hence,
this suggests a hierarchy of immune cell activation as well as
an ability to attenuate an immune response.
KEY POINTS Checkpoint inhibitors are a class of antibodies that are
designed to interrupt or activate these costimulatory mol-
Immunotherapy has proven efficacy for other solid
ecules. Intense investigation is underway for the utilization
tumors and is now a focus for glioblastoma.
An important component of glioblastoma-mediated
of checkpoint inhibitors for the treatment of solid tumors.
immunosuppression stems from soluble factors such as CTLA-4 was one of the first molecules to be studied. Leach
TGF-b. et al9 found that they could induce an antitumor immune
Checkpoint inhibition probably confers a nonspecific response within a murine model for melanoma by using an
state of immune activation that is a promising approach antiCTLA-4 antibody.9 The second checkpoint inhibitor that
to glioblastoma, given the success of checkpoint has been intensely studied is antiPD-1. AntiPD-1 has been
inhibitors in melanoma, lung cancer, and renal cancers. shown to induce an antitumor immune response in multiple
Current strategies for biomarkers have focused on solid tumors, including glioblastoma.10,11
protein expression, immune cell characterization, and
mutational burden.
Tumor-selective viruses and viral-based vectors have Proven Efficacy of Checkpoint Inhibitors
shown increasingly promising results as easy-to-use Several large clinical trials in humans verified the observed
immunostimulants. efficacy of checkpoint inhibitors in the preclinical models. In
2010, Hodi et al12 ran a large phase III trial that demonstrated

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e133


LIM, WELLER, AND CHIOCCA

improved survival among patients with melanoma who were NCT02337491


treated with antiCTLA-4. The investigators also found that a NCT02337491 is phase II trial based on the premise that anti-
subset of patients were long-term survivors.12 Topalian et al13 VEGF therapy could be synergistic with immunotherapy.15
then published their experience in treating multiple solid This trial is a single-institution study sponsored by Merck using
tumors with antiPD-1, in which they found that antiPD-1 Mercks antiPD-1 drug. The study will measure progression-
improved survival for patients with melanoma, renal cell free survival at 6 months. The trial has two arms: antiPD-1
carcinoma, and nonsmall cell lung cancer. These and other alone and antiPD-1 with bevacizumab.
important studies resulted in U.S. Food and Drug Adminis-
tration (FDA) approval of checkpoint inhibitors. Checkpoint
inhibitors first gained FDA approval for use in patients with NCT02336166
melanoma in 2011, with the approval of antiCTLA-4. The FDA NCT02336166 is a phase II trial sponsored by the Ludwig
approved antiPD-1 in September 2014 for melanoma. Foundation to study the antiPD-1 therapy developed by
Shortly after, antiPD-1 was approved for lung cancer (both AstraZeneca. The trial has multiple objectives. The first
adenocarcinoma and squamous cell carcinoma) and renal cell cohort (cohort A) will study the efficacy of antiPD-L1 for
cancer. Hence, the enthusiasm for the use of checkpoint patients with newly diagnosed glioblastoma that has
inhibitors in glioblastoma is high. unmethylated O(6)-methylguanine DNA methyltransferase.
The remarkable factor in this arm is that temozolomide
(TMZ) will be withheld; patients will receive only antiPD-L1
Checkpoint Inhibitors for Glioblastoma and radiation. The rationale behind this design is that ra-
There are promising preclinical data suggesting that diation and antiPD-L1 are synergistic, so systemic che-
checkpoint inhibitors may promote an antitumor immune motherapy may be counterproductive. The second cohort
response. Fecci et al14 demonstrated improved survival (cohort B) will assess the efficacy of antiPD-L1 alone for
mediated by a CD4+ T-cell immune response in a murine patients who have recurrent glioblastoma. The third cohort
glioma model treated with antiCTLA-4. Zeng et al and (cohort C) will administer antiPD-L1 to patients who have
others have shown that antiPD-1 monotherapy improved recurrent glioblastoma that is progressing with bevacizumab
survival in a murine model for glioblastoma. Interestingly, treatment.
antiPD-L1 alone did not result in much survival im-
provement. With antiPD-1 use, the CD8+ T cells appear to
be responsible for the antitumor immune response.10,11 NCT02617589
NCT02617589 is a phase III trial sponsored by Bristol Myers
Squibb to assess the efficacy of antiPD-1 with radiation
CURRENT TRIALS FOR GLIOBLASTOMA among patients who have newly diagnosed glioblastoma.
Because we observed improved survival with the use of The comparator arm will enroll patients for treatment with
checkpoint inhibitors in the previously mentioned solid radiation and temozolomide (standard of care).
tumors, these compounds are now being applied to glio-
blastoma. Several clinical trials are currently underway, with
NCT02313272
encouraging preclinical results.
NCT02313272 is a phase I trial to assess the safety of an
antiPD-1 drug developed by Merck with bevacizumab and
NCT02017717 hypofractionated stereotactic irradiation for patients who
NCT02017717 is a large, randomized, phase III open-label have recurrent high-grade gliomas.
trial sponsored by Bristol Myers Squibb for patients with a
first-time recurrence of glioblastoma that used antiPD-1 as
NCT02530502
the treatment backbone to assess its safety and, ultimately,
NCT02530502 is a phase I/II trial for patients with newly
its efficacy. The study began with a small safety run-in,
diagnosed glioblastoma. Phase I will assess the safety of
during which patients were treated with antiPD-1 alone
combining an antiPD-1 drug developed by Merck with
or antiPD-1 with antiCTLA-4. Interim safety data that were
radiation and temozolomide for patients with newly di-
presented at the 2015 American Society of Clinical Oncology
agnosed glioblastoma. Phase II will compare the efficacy of
Annual Meeting showed that the rates of severe adverse
adding antiPD-1 to radiation and temozolomide for pa-
events were significantly higher among patients who re-
tients treated with radiation and temolozomide.
ceived the combination of antiPD-1 and antiCTLA-4; 40%
of patients had to discontinue therapy (10 patients per arm).
Thus, a decision was made to expand the antiPD-1 cohort NCT02311582
and to use bevacizumab as the comparator arm. Although NCT02311582 is a phase I, open-label, randomized safety
this was a study of safety, a partial response in one patient study to assess the addition of MK-3475, an antiPD-1
who was treated with anti-PD-1 alone, and a few cases of drug developed by Merck, for treatment among patients
pseudoprogression, was observed. The trial has finished who are being treated with laser ablation for recurrent
accrual. glioblastoma.

e134 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


CURRENT STATE OF IMMUNE-BASED THERAPIES FOR GLIOBLASTOMA

BIOMARKERS threatening. Treatments often require stopping the treat-


What we have learned from checkpoint inhibitor trials in ment, starting high-dose corticosteroids, and possibly ad-
other tumors is that the overall response rate is between ministering infliximab.12,13,23
20% and 30%. Therefore, it is important to identify patients
who would not respond and to minimize toxicities and treat IMAGING
them with other therapies. Current strategies for biomarkers Determining response to immunotherapy with imaging has
have focused on protein expression, immune cell charac- become an area of intense interest. In the trial reported by
terization, and mutational burden. Hodi et al,12 a large number of patients with melanoma who
The melanoma and lung cancer trials have focused on the received antiCTLA-4 experienced pseudoprogression.
expression of PD-L1 on tumor cells as a biomarker to predict Furthermore, the researchers found that pseudoprogression
response. Investigators have found that tumors that express could take months to resolve.24,25 As a result, many trials
PD-L1 were more likely than PD-L1negative tumors to have built in lag times for imaging to allow patients to
respond (overall response rate) to antiPD-1 therapy.16 continue therapy and to avoid considering the treatment a
Interestingly, the expression of PD-L1 was not important failure. In glioblastoma, Okada et al26 have developed an
in the trial of patients with melanoma who received the iRANO protocol specifically tailored for immunotherapy.
concurrent combination of antiPD-1 and antiCTLA-4.
Some theorized that an adaptive immune response oc-
curred with the combination therapy and that combination
COMBINATION THERAPY
As previously mentioned, the response rates for patients who
caused tumor cells to express PD-L1 as a defense mecha-
receive immunotherapy has ranged from 20% to 30%. Studies
nism.17 In an interesting twist, when the therapies were
are investigating ways to combine checkpoint inhibitors with
sequencedpatients received antiCTLA-4 first, followed by
other modalities, such as radiation, bevacizumab, and de-
antiPD-1the overall response rate again correlated to the
vices. Zeng et al10 demonstrated in a preclinical glioblastoma
PD-L1 status of the tumor.18
model that the combination of focused radiation with
Another approach has been to assess the activation status
antiPD-1 is synergistic. Clinical trials (e.g., NCT02313272) are
of various immune cells as a predictor of response. As an
looking at stereotactic radiation use in combination with
example, investigators assessed the activation status of CD8+
checkpoint inhibitors. Bevacizumab also may work syner-
T cells by measuring eomesodermin for patients with mela-
gistically with immunotherapy.20 Preclinical data suggest
noma after they received treatment with antiCTLA-4 and
that this approach could be effective, and a phase II trial
found that the eomesodermin status predicted relapse-free
(NCT02337491) is looking at the combination of bevacizumab
survival. Other markers of interest are interferon gamma
and antiPD-1 for patients with glioblastoma. Finally, another
expression, Helios expression, and various other checkpoint
interesting approach is the combination of laser ablation with
molecules.19
antiPD-1. A phase I trial (NCT02313272) is looking at the
Last, investigators have assessed mutational burden as a
combination of antiPD-1 with laser ablation.
predictor of response. Chan et al20 correlated mutational
In conclusion, checkpoint inhibitors have shown great
burden among patients with lung cancer to response rates
promise in other solid tumors. There is much excitement
among patients who received antiPD-1 therapy. They
about checkpoint inhibitors in the setting of glioblastoma.
found that patients with lung cancer who had a history of
Several large trials are underway to assess the efficacy of
smoking had higher mutational burdens than patients with
checkpoint inhibitors in glioblastoma.
lung cancer who did not smoke and that the higher number
of mutations correlated to improved survival.20 Le et al21
also studied patients with colon cancer and found that VIRAL- AND GENE-MEDIATED
patients who had a defective DNA repair gene had a higher IMMUNOTHERAPIES FOR GLIOBLASTOMA
number of mutations in their tumor, and this again corre- We have discussed the various types of immunotherapy for
lated to improved survival. Some theorize that the reason for glioblastoma. Now, we discuss one additional mode that
the observed improved antitumor immune responses in involves the use of genetically engineered viruses to deliver
patients with tumors that had more mutations was that cytotoxic/immunostimulatory genes into tumors.27 Two
the tumors expressed a higher number of target antigens for main types of viruses are used in this technology: replication-
the immune system.21,22 defective vectors, in which viral genes have been removed so
there is no expression of viral genes or generation of progeny
viruses, but there is expression of an immunostimulatory
TOXICITY and/or cytotoxic gene, and tumor replicationselective vi-
Immune-related toxicities are an important issue for pa- ruses (oncolytic viruses), in which a viral pathogen is engi-
tients who receive immunotherapy. Most of the toxicities neered so that its pathogenicity is now targeted to tumor
are related to autoimmune reactions. The most common cells and not normal cells.28 It is recognized now that the
toxicities include colitis, pneumonitis, hepatitis, pancreatitis, presence of viral genes and viral proteins in both of these
dermatitis, hypophysitis, and thyroiditis. If the toxicities are technologies can elicit powerful anticancer immune re-
not recognized early, these reactions could become life sponses, which are a major component of efficacy.29

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e135


LIM, WELLER, AND CHIOCCA

FIGURE 1. Immunosuppressive Properties of Glioma Stem Cells

Abbreviation: TGF-b, transforming growth factor-beta.


Modified from Hatiboglu et al.42

Characteristics that differentiate this immunotherapy 1. The delivered TK gene product phosphorylates the ad-
from others are the following: this immunotherapy in- ministered valacyclovir drug, which becomes incorporated
volves a neurosurgical component of direct injection into at sites where DNA is becoming repaired or where DNA is
glioblastoma, either by stereotaxy or free-hand injection replicating. This leads to termination of DNA repair and/or
during a craniotomy; is off-the-shelf, in that manipulation of replication, leading to immunogenic cell death.
cells from the patient before or after treatment is not in- 2. Standard of care also leads to cytotoxicity, and
volved; and does not require a priori knowledge of the radiation-induced DNA damage leads to additional
identity of tumor-specific antigens that require targeting unsuccessful and cytotoxic attempts at DNA repair
and, at least in theory, the cytotoxic action of the viral using the phosphorylated valacyclovir pools.
vector-delivered cytotoxic gene or of the replicating onco- 3. The delivered TK antigen and viral vector proteins act
lytic viruses will expose the repertoire of all tumor-specific as superantigens, providing an immunostimulatory
antigens to the immune system. stimulus in the glioblastoma microenvironment.
There are more than 2 decades of clinical trial experiences 4. The cytotoxic death of glioblastoma cells is also im-
that used various types of replication-defective vectors to munogenic, releasing and exposing multiple glioma
deliver various types of cytotoxic/immunostimulatory genes,
antigens to the immune system.
but none have resulted in an FDA-approved clinical product,
Our published mature phase II data appear to show en-
and two phase III clinical trialsa retroviral vector to deliver the couraging, albeit not definite, results in terms of possible
cytotoxic/immunostimulatory herpes thymidine kinase (TK)
efficacy on the basis of the extent of residual tumor burden:
gene for recurrent glioblastoma30 and an adenoviral vector to
the median overall survival (OS) durations for patients who
deliver TK in newly diagnosed glioblastoma (ASPECT)have
underwent gross total resection were 25.0 and 16.9 months
failed.31 However, we recently reported the results of a phase II
trial in which aglatimagene besadenovec (AdV-tk), a non- (a difference of 8.1 months) for GMCI/standard of care
replicating adenovirus expressing the herpesvirus TK gene, and standard of care, respectively (hazard ratio 0.59; 95%
was free-hand injected in a resected, newly diagnosed, ma- CI, 0.350.998; p = .0492); for patients who underwent
lignant glioma cavity.32,33 The patient then received the oral subtotal resections, the difference was only 1 month (13.5
antiherpetic prodrug (valacyclovir) while undergoing standard- vs. 12.5 months for GMCI/standard of care vs. standard of
of-care radiochemotherapy (gene-mediated cytotoxic immu- care; p = .4584).33 To further improve this therapy, we have
notherapy [GMCI]). There are multiple modes of cytotoxic and returned to the laboratory. The current theory is that to
immunostimulatory anticancer action (Fig. 3): be effective, an anticancer immune response by cytotoxic

e136 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


CURRENT STATE OF IMMUNE-BASED THERAPIES FOR GLIOBLASTOMA

FIGURE 2. Examples of Activating (CD28) and Inhibiting (PD-1) Immune Checkpoints

T cells requires removal of immune checkpoint signaling networks does lead to even more encouraging antigliomal
mediated by PD-1/PD-L1,34-36 the CTLA-4/B7 family,37,38 and effects of GMCI.
other molecules. We thus hypothesize that a combination of For the second type of viral-mediated therapy, multiple
GMCI and checkpoint inhibitors may lead to more effective types of oncolytic viruses have been tested in clinical trials of
immunotherapy.8 In fact, we are finding that the application glioblastoma, all in the recurrent setting. No trial for glio-
of GMCI in mouse gliomas does lead to increase signaling of blastoma has progressed to phase III, but there are two
immune checkpoint networks and that inhibition of these oncolytic viruses (an oncolytic adenovirus from DNAtrix

FIGURE 3. Schematics of the Published Clinical Trials and Different Modes of Anticancer Action of Gene-Mediated
Cytotoxic Immunotherapy

(Top panel) Schematic of the published clinical trials of gene-mediated cytotoxic immunotherapy (GMCI). On the day of surgery, the adenoviral vector that delivers TK (AdV-
tk) is injected in the resected newly diagnosed glioblastoma tumor cavity. The oral agent (valacyclovir) is administered to the patient on days 1 to 14. Standard-of-care
radiation and temozolomide are also administered as per the Stupp regimen. (Lower panel) Schematic of the different modes of anticancer action of GMCI.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e137


LIM, WELLER, AND CHIOCCA

and an oncolytic retrovirus from Tocagen) that have have been obtained to justify filing of an investigational
completed phase II evaluation and planning for advanced new drug application with the FDA for a planned first-
phase III trials. Commercial interest in this area of in-human clinical trial among patients with recurrent
oncolytic virusbased immunotherapy has been resur- glioblastoma.
rected recently by the FDA approval of a herpes simplex In summary, the use of tumor-selective viruses and viral-
virus (HSV) oncolytic virus for melanoma.29,39 Our group based vectors is increasingly delivering promising results as
has been involved in the preclinical and clinical devel- an easy to use immunostimulant approach, but only the
opment of an oncolytic virus, based on HSV1, that successful completion of phase III trials for several of these
has been engineered to selectively replicate and de- products will show if the treatments have achieved their
stroy gliomas on the basis of tumor deregulation of the promise as anticancer agents or if additional laboratory
p16 tumor suppressor pathway and expression of the development is required.
glioma stem-cell marker, nestin.40,41 This oncolytic virus
(rQNestin34.5v.2) shows potency in animal models of
gliomas compared with older, clinical trialtested versions ACKNOWLEDGMENT
of herpes oncolytic viruses. Extensive preclinical data We thank Eileen Kim for her help with Fig. 2.

References
1. Roszman T, Elliott L, Brooks W. Modulation of T-cell function by gliomas. 17. Grosso J, Horak CE, Inzunza D, et al. Association of tumor PD-L1 ex-
Immunol Today. 1991;12:370-374. pression and immune biomarkers with clinical activity in patients (pts)
2. Frei K, Gramatzki D, Tritschler I, et al. Transforming growth factor-b with advanced solid tumors treated with nivolumab (anti-PD-1;BMS-
pathway activity in glioblastoma. Oncotarget. 2015;6:5963-5977. 936558;ONO-4538). J Clin Oncol. 2013;31(suppl; abstr 3016).
3. Nduom EK, Weller M, Heimberger AB. Immunosuppressive mechanisms 18. Callahan MK, Postow MA, Wolchok JD. CTLA-4 and PD-1 pathway
in glioblastoma. Neuro Oncol. 2015;17:vii9-vii14. blockade: combinations in the clinic. Front Oncol. 2014;4:385.
4. Weiss T, Weller M, Roth P. Immunotherapy for glioblastoma: concepts 19. Wang W, Yu D, Sarnaik AA, et al. Biomarkers on melanoma patient
and challenges. Curr Opin Neurol. 2015;28:639-646. T cells associated with ipilimumab treatment. J Transl Med. 2012;10:
5. Preusser M, Lim M, Hafler DA, et al. Prospects of immune checkpoint mod- 146.
ulators in the treatment of glioblastoma. Nat Rev Neurol. 2015;11:504-514. 20. Rizvi NA, Hellmann MD, Snyder A, et al. Cancer immunology: mutational
6. Preusser M, Berghoff AS, Wick W, et al. Clinical neuropathology mini- landscape determines sensitivity to PD-1 blockade in nonsmall-cell
review 6-2015: PD-L1emerging biomarker in glioblastoma? Clin lung cancer. Science. 2015;348:124-128.
Neuropathol. 2015;34:313-321. 21. Le DT, Uram JN, Wang H, et al. PD-1 blockade in tumors with mismatch-
7. Sharma P, Allison JP. Immune checkpoint targeting in cancer therapy: toward repair deficiency. N Engl J Med. 2015;372:2509-2520.
combination strategies with curative potential. Cell. 2015;161:205-214. 22. Diaz LA Jr, Le DT. PD-1 blockade in tumors with mismatch-repair de-
8. Pardoll DM. The blockade of immune checkpoints in cancer immu- ficiency. N Engl J Med. 2015;373:1979.
notherapy. Nat Rev Cancer. 2012;12:252-264. 23. Weber JS, Kahler KC, Hauschild A. Management of immune-related
9. Leach DR, Krummel MF, Allison JP. Enhancement of antitumor im- adverse events and kinetics of response with ipilimumab. J Clin Oncol.
munity by CTLA-4 blockade. Science. 1996;271:1734-1736. 2012;30:2691-2697.
10. Zeng J, See AP, Phallen J, et al. Anti-PD-1 blockade and stereotactic 24. Hodi FS, Ribas A, Daud A, et al. Patterns of response in patients with
radiation produce long-term survival in mice with intracranial gliomas. advanced melanoma treated with pembrolizumab (MK-3475) and
Int J Radiat Oncol Biol Phys. 2013;86:343-349. evaluation of immune-related response criteria (irRC). J Immunother
11. Reardon DA, Freeman G, Wu C, et al. Immunotherapy advances for Cancer. 2014;2:P103.
glioblastoma. Neuro Oncol. 2014;16:1441-1458. 25. Topalian SL, Sznol M, McDermott DF, et al. Survival, durable tumor
12. Hodi FS, ODay SJ, McDermott DF, et al. Improved survival with ipili- remission, and long-term safety in patients with advanced melanoma
mumab in patients with metastatic melanoma. N Engl J Med. 2010;363: receiving nivolumab. J Clin Oncol. 2014;32:1020-1030.
711-723. 26. Okada H, Weller M, Huang R, et al. Immunotherapy response assess-
13. Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune ment in neuro-oncology: a report of the RANO working group. Lancet
correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366: Oncol. 2015;16:e534-e542.
2443-2454. 27. Kaufmann JK, Chiocca EA. Glioma virus therapies between bench and
14. Fecci PE, Ochiai H, Mitchell DA, et al. Systemic CTLA-4 blockade bedside. Neuro Oncol. 2014;16:334-351.
ameliorates glioma-induced changes to the CD4+ T cell compartment 28. Chiocca EA, Rabkin SD. Oncolytic viruses and their application to cancer
without affecting regulatory T-cell function. Clin Cancer Res. 2007;13: immunotherapy. Cancer Immunol Res. 2014;2:295-300.
2158-2167. 29. Lawler SE, Chiocca EA. Oncolytic virus-mediated immunotherapy:
15. Hodi FS, Lawrence D, Lezcano C, et al. Bevacizumab plus ipilimumab in a combinatorial approach for cancer treatment. J Clin Oncol. 2015;33:
patients with metastatic melanoma. Cancer Immunol Res. 2014;2: 2812-2814.
632-642. 30. Rainov NG. A phase III clinical evaluation of herpes simplex virus type 1
16. Taube JM, Klein A, Brahmer JR, et al. Association of PD-1, PD-1 ligands, thymidine kinase and ganciclovir gene therapy as an adjuvant to surgical
and other features of the tumor immune microenvironment with re- resection and radiation in adults with previously untreated glioblas-
sponse to anti-PD-1 therapy. Clin Cancer Res. 2014;20:5064-5074. toma multiforme. Hum Gene Ther. 2000;11:2389-2401.

e138 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


CURRENT STATE OF IMMUNE-BASED THERAPIES FOR GLIOBLASTOMA

31. Westphal M, Yla-Herttuala S, Martin J, et al; ASPECT Study Group. 37. Lane P. Regulation of T and B cell responses by modulating interactions
Adenovirus-mediated gene therapy with sitimagene ceradenovec fol- between CD28/CTLA4 and their ligands, CD80 and CD86. Ann N Y Acad
lowed by intravenous ganciclovir for patients with operable high-grade Sci. 1997;815:392-400.
glioma (ASPECT): a randomised, open-label, phase 3 trial. Lancet Oncol. 38. Korman A, Yellin M, Keler T. Tumor immunotherapy: preclinical and
2013;14:823-833. clinical activity of anti-CTLA4 antibodies. Curr Opin Investig Drugs. 2005;
32. Chiocca EA, Aguilar LK, Bell SD, et al. Phase IB study of gene-mediated 6:582-591.
cytotoxic immunotherapy adjuvant to up-front surgery and intensive 39. Andtbacka RH, Kaufman HL, Collichio F, et al. Talimogene laherparepvec
timing radiation for malignant glioma. J Clin Oncol. 2011;29:3611-3619. improves durable response rate in patients with advanced melanoma.
33. Wheeler LA, Manzanera AG, Bell SD, et al. Phase 2 multicenter study of J Clin Oncol. 2015;33:2780-2788.
gene-mediated cytotoxic immunotherapy as adjuvant to surgical resection 40. Aghi M, Visted T, Depinho RA, et al. Oncolytic herpes virus
for newly diagnosed malignant glioma. Neuro Oncol. Epub 2016 Feb 2. with defective ICP6 specifically replicates in quiescent cells with
34. Barber DL, Wherry EJ, Masopust D, et al. Restoring function in exhausted homozygous genetic mutations in p16. Oncogene. 2008;27:
CD8 T cells during chronic viral infection. Nature. 2006;439:682-687. 4249-4254.
35. Freeman GJ, Long AJ, Iwai Y, et al. Engagement of the PD-1 immu- 41. Kambara H, Okano H, Chiocca EA, et al. An oncolytic HSV-1 mutant
noinhibitory receptor by a novel B7 family member leads to negative expressing ICP34.5 under control of a nestin promoter increases sur-
regulation of lymphocyte activation. J Exp Med. 2000;192:1027-1034. vival of animals even when symptomatic from a brain tumor. Cancer
36. Mahoney KM, Rennert PD, Freeman GJ. Combination cancer immu- Res. 2005;65:2832-2839.
notherapy and new immunomodulatory targets. Nat Rev Drug Discov. 42. Hatiboglu MA, Wei J, Wu AS, et al. Immune therapeutic targeting of
2015;14:561-584. glioma cancer stem cells. Target Oncol. 2010;5:217-227.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e139


CENTRAL NERVOUS SYSTEM TUMORS

Treatment of Low-Grade Gliomas


in the Era of Genomic Medicine

CHAIR
Minesh P. Mehta, MD, MBChB, FASTRO
University of Maryland Medical Center
Baltimore, MD

SPEAKERS
Kenneth D. Aldape, MD
The University of Texas MD Anderson Cancer Center
Houston, TX

Susan M. Chang, MD
University of California, San Francisco
San Francisco, CA
LOWER-GRADE GLIOMAS, MOLECULAR MARKERS, AND THERAPEUTIC CHOICES

Treatment of Adult Lower-Grade Glioma in the Era of Genomic


Medicine
Susan M. Chang, MD, Daniel P. Cahill, MD, PhD, Kenneth D. Aldape, MD, and Minesh P. Mehta, MBChB, FASTRO

OVERVIEW

By convention, gliomas are histopathologically classified into four grades by the World Health Organization (WHO) legacy
criteria, in which increasing grade is associated with worse prognosis and grades also are subtyped by presumed cell of origin.
This classification has prognostic value but is limited by wide variability of outcome within each grade, so the classification is
rapidly undergoing dramatic re-evaluation in the context of a superior understanding of the biologic heterogeneity and
molecular make-up of these tumors, such that we now recognize that some low-grade gliomas behave almost like malignant
glioblastoma, whereas other anaplastic gliomas have outcomes comparable to favorable low-grade gliomas. This clinical
spectrum is partly accounted for by the dispersion of several molecular genetic alterations inherent to clinical tumor
behavior. These molecular biomarkers have become important not only as prognostic factors but also, more critically, as
predictive markers to drive therapeutic decision making. Some of these, in the near future, will likely also serve as potential
therapeutic targets. In this article, we summarize the key molecular features of clinical significance for WHO grades II and III
gliomas and underscore how the therapeutic landscape is changing.

S everal challenges limit the efficacy of surgery, radio-


therapy (RT), chemotherapy, and targeted agents for
WHO grades II and III glioma (categorized as lower-grade
and biologically different subgroups that are nested within
each grade category. 1 In addition, the interevaluator
variability that has been demonstrated, especially for
glioma, or LrGG). These include the diffuse and imprecisely mixed histologies, is a major limitation in provision of
defined boundaries of extension, proximity to, and growth uniform and reproducible pathologic diagnoses.2 Recent
into critical cortical and subcortical structures, inability of molecular characterization has identified potential bio-
therapeutic agents to adequately cross the blood-brain markers that can serve to define the framework of different
barrier, active transport mechanisms of drug efflux, high subtypes of these tumors with more uniform prognosis
plasma protein binding of most conventional agents, and and also potentially can serve as indicators for treatment
multiple and redundant drug and radiation resistance selection.3,4
mechanisms. Biologic tumor heterogeneity, with respect to Somatic mutations in the isocitrate dehydrogenase genes
response to treatment and resistance mechanisms, despite IDH1 and IDH2 are of important prognostic value. These
similar grade or histology, is another important challenge. mutations are present in 50% to 80% of WHO grades II and III
The era of genomic medicine holds the promise of gener- astrocytic and oligodendroglial tumors in adults and also
ating personalized medicine treatment paradigms to im- occur with similar frequency in secondary glioblastoma.
prove outcomes. The ability of isocitrate dehydrogenase These mutant enzymes lead to conversion of a-ketoglutarate
(IDH) mutations, loss of heterozygosity of 1p and 19q, and into D-2-hydroxyglutarate, an oncometabolite that drives
methylation of methylguanine methyl transferase (MGMT)
the oncogenic activity of IDH mutations.5 Irrespective of
status to serve as predictive biomarkers is of specific interest
treatment choice, patients with IDH-mutated tumors ex-
in the treatment of LrGG.
perience superior survival compared with patients who
have IDH wild-type gliomas of the same histologic grade; this
MOLECULAR CHARACTERIZATION OF LOWER- difference clearly underscores the prognostic significance of
GRADE GLIOMA this marker. In tumors of oligodendroglial lineage, the most
Clinicopathologic assessment, valuable to distinguish common cytogenetic alteration consists of an unbalanced
grades of glioma, does not address the distinct genetic t1,19(q10;p10) translocation that results in the fusion of the

From the University of California, San Francisco, San Francisco, CA; Harvard Medical School, Boston, MA; Toronto General Hospital/Research Institute, Toronto, Canada; University of
Maryland, Baltimore, MD.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Minesh P. Mehta, MD, FASTRO, Department of Radiation Oncology, University of Maryland, 22 South Greene St., Baltimore, MD 21201; email: mmehta@umm.edu.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 75


CHANG ET AL

chromosomal arms of 1p and 19q, accompanied by the loss 1p19q codeletion, and mutations in CIC and FUBP1, with an
of one hybrid chromosome, which thus results in the loss of absence of mutations in TP53 or ATRX.
heterozygosity of 1p and 19q. These tumors have better
prognoses than histologically identical tumors of the same WHO GRADE III ANAPLASTIC
grade that do not harbor this codeletion. Furthermore, this OLIGODENDROGLIOMA: THE CASE FOR A
biomarker has been proven to be a critical predictive bio- PREDICTIVE BIOMARKER FOR CHEMOTHERAPY
marker of chemotherapy response, and improved survival On the basis of several retrospective series and phase II
after chemoradiotherapy, compared with RT alone. trials that demonstrated the chemosensitivity of oligoden-
Recent reports also have reinforced the redefini- drogliomas, two prospective randomized phase III trials
tion of subgroups of LrGG by incorporating additional evaluated the role of postoperative chemoradiotherapy
biomarkersfor example, activating mutations of the tel- compared with RT alone.6,7 In both studies, the experimental
omerase reverse transcription (TERT) promotor and muta- arm was RT combined with procarbazine, lomustine, and
tions and losses of alpha thalassemia/mental retardation vincristine (PCV). In the Radiation Therapy Oncology Group
syndrome X linked (ATRX) and tumor protein P53 (TP53). By (RTOG) trial RTOG-9402, patients were randomly assigned
using these genetic alterations, the vast majority of adult either to four cycles of intensified PCV followed by RT or to
diffuse gliomas fall into one of three distinct categories that immediate RT without chemotherapy. Mature data from this
more reproducibly predict clinical outcome and guide study showed that the median survival of those with
treatment by creating more homogeneous subsets.3,4 These codeleted tumors who were treated with PCV plus RT was
consist of (1) molecular glioblastomas, which are highly twice that of patients who received only RT (14.7 vs. 7.3
aggressive tumors characterized by the TERT promoter years). The survival of patients with codeleted tumors was
mutation and concomitant deletions of chromosome 10 and better than that of those with non-codeleted tumors re-
gain of chromosome 7 with EGFR amplification, and they are gardless of treatment, which emphasizes the prognostic
notable for the absence of IDH1/2 mutations (i.e., they value of the 1p19q codeletion. The survival was not sta-
are IDH wild type); (2) molecular astrocytomas, which are tistically different for patients with tumors that lacked the 1p
characterized by mutations in IDH1/2, TP53, and ATRX, and and 19q deletion, irrespective of treatment (median survival,
the absence of promoter mutations or codeletion of 1p19q; 2.6 vs. 2.7 years), which suggests that the codeletion also has
and (3) molecular oligodendrogliomas, which are char- predictive value. In another analysis of patients enrolled in
acterized by IDH mutation and TERT promoter mutation, this study, IDH mutation status had predictive value as well;
although patients with codeleted tumors lived longest,
patients with non-codeleted IDH-mutated tumors also lived
longer after chemoradiotherapy compared to RT alone.8 The
KEY POINTS implication here is that, for WHO grade III tumors, con-
ventionally labeled as oligodendroglioma, treatment with
WHO grades II and III gliomas, conventionally chemoradiotherapy imparts a substantial survival advantage
substratified as astrocytomas or oligodendrogliomas (or when the tumors are either codeleted for 1p19q or are non-
mixed), are highly heterogeneous tumors with complex, codeleted tumors only with expression of an IDH mutation.
overlapping, and varied biological behavior. Non-codeleted tumors without IDH mutations have the
In the past decade, significant new therapeutic strides worst prognosis and do not benefit from the addition of
have been made to improve overall survival in almost all chemotherapy.
of these entities, in large measure, as a consequence of
In the European Organization for Research and Treat-
robust, international clinical trial collaborations with
parallel tissue collection for molecular analysis.
ment of Cancer (EORTC) 26951 trial, 368 patients who had
This tissue collection effort, with well-annotated predominantly grade III oligodendroglial tumors received
matching clinical outcomes, has secondarily identified immediate RT only or RT followed by six cycles of PCV.
several key molecular markers that have crucial Patients with tumors that had the 1p and 19q codeletion
prognostic significance, such as IDH1/2 mutations, had better outcomes regardless of therapy (prognostic).
1p19q co-deletions, MGMT methylation, and more. In addition, MGMT promoter methylation was of prog-
These markers are also increasingly identified as crucial nostic value in this cohort. Similar to the results of RTOG-
predictive markers, which thereby influence therapeutic 9402, long-term follow-up showed that the addition of
decision making. PCV to RT significantly increased progression-free survival
Some of these molecular markers, such as the IDH1/2 (PFS; median, 157 vs. 50 months) in patients with the
mutation, are now also emerging as future therapeutic
1p19q codeletion, and there was a trend toward increased
targets. Multivariate analysis of clinical and molecular
markers is identifying subgroups that likely need to be
overall survival (OS; OS not reached in the RT/PCV group
treated with greater therapeutic intensity and others for vs. OS of 112 months in the RT-alone group; hazard
which therapeutic de-intensification could potentially ratio [HR] 0.56; 95% CI, 0.311.03). These two studies
be contemplated, posing novel challenges for clinical confirmed the value of the 1p19q codeletion as an im-
trial design and conduct. portant prognostic and predictive marker and influenced
the design of studies in anaplastic glioma; in ongoing

76 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


LOWER-GRADE GLIOMAS, MOLECULAR MARKERS, AND THERAPEUTIC CHOICES

international studies, patient arms are being stratified on Because of the relatively low proliferative index of grade II
the basis of this biomarker. An international intergroup glioma, these tumors were previously considered chemo-
phase III trial is being conducted in patients who have therapy resistant. Reports of objective responses in patients
newly diagnosed grade 3 glioma with the 1p19q status with LrGG, however, raised interest in the use of chemo-
codeletion.7 Patients are randomly assigned to two arms, therapy in the adjuvant setting. In a small Southwest On-
RT with concomitant and adjuvant temozolomide or RT cology Group trial, there was no survival difference among
with adjuvant PCV. patients with incompletely excised grade II gliomas ran-
domly assigned to RT alone or to the combination of RT and
lomustine.13 The RTOG-9802 study examined the role of
WHO GRADE III ANAPLASTIC ASTROCYTOMA adjuvant PCV chemotherapy for high-risk adults (i.e., those
The role of chemotherapy in anaplastic astrocytoma is not of any age with tumors that had less than total resection, or
well established. Most phase III trials have demonstrated those older than age 40 with any resection) who had grade II
no benefit of chemotherapy compared with radiation glioma. Two hundred fifty-one patients were randomly
alone in this tumor. Carmustine and PCV are associated assigned to RT alone or RT followed by six cycles of PCV. At a
with minimal improvement in survival. The Glioma Meta- median follow-up of 12 years, a significant improvement in
Analysis Trialists group showed a 6% increase in 1- and OS in the RT/PCV group (13.3 years) compared with the RT-
2-year survival rates for patients who received chemo- alone group (7.8 years) was reported.14
therapy (2-year survival, 37% vs. 31%), whereas a large The significance of 1p19q, MGMT, and IDH was unknown at
randomized trial of adjuvant PCV compared with RT the time of study initiation, and several molecular analyses
alone did not show any benefit from PCV.9,10 RTOG-9813 were post hoc, performed on non-mandatorily collected
was a phase III study to compare radiation with bis- tumor blocks, slides, or cores submitted for 117 patients; of
chloroethylnitrosourea (BCNU) or lomustine to radiation with these, 113 (57 of 126 [45%] and 56 of 125 [45%] in the RT
temozolomide, and the results of this study demonstrated alone and RT/PCV arms, respectively) were suitable for IDH-
no difference in survival between the two chemotherapy R132H immunohistochemistry interpretation. IDH1-R132H
arms. Molecular characterization in a subset of patients mutations were detected in 35 (61.4%) of 57 patients and 36
showed that IDH mutation was a strong prognostic factor. (64.3%) of 56 patients in the RT alone and RT/PCV arms,
The number of patients was too small to determine any respectively, and in 77.6%, 53.8%, and 48.0% of patients with
differential benefit of the type of chemotherapy on the oligodendroglioma, oligoastrocytoma, and astrocytoma, re-
basis of IDH mutation status.11 spectively. Tissue sufficient for determination of other IDH
The NOA-04 phase III trial compared the efficacy of RT mutations or of the 1p19q codeletion was available for
followed by chemotherapy at progression to initial che- analysis in 29 and 34 patients in the RT and in the RT/PCV
motherapy followed by RT at progression in newly diagnosed arms, respectively. In a subset analysis of PFS by IDH1-R132H
anaplastic gliomas and did not show any difference in the mutation status, patients with mutations experienced sig-
median time to failure, PFS, or OS among the arms. The study nificantly longer PFS than those without (p , .001), regardless
demonstrated the prognostic value of IDH1 mutations in of treatment, which established the mutation as prognosti-
anaplastic gliomas, with a favorable impact that was more cally significant. In addition, patients with tumoral IDH1-
distinct than that of 1p19q codeletion or MGMT promoter R132H mutations had longer PFS if they received RT/PCV than
methylation.12 if they received RT alone (p = .003), which established the
A large international trial in which patients with newly mutation status as a predictive variable. The number of events
diagnosed non1p19q-codeleted grade III glioma were in patients without the IDH1-R132H mutation was too small to
randomly assigned to radiation with or without concom- determine the association of treatment effect in this subset.
itant and with or without adjuvant temozolomide after In an exploratory subset analysis of OS by IDH1-R132H mu-
RT recently completed accrual, and initial interim analysis tation status, patients with mutations experienced signifi-
showed a significant benefit with adjuvant temozolomide cantly longer OS than those without the mutation, regardless
(p = .0084). Results for concurrent temozolomide and for the of treatment (log rank p = .02). Median survival times with and
overall treatment regimen are not yet available. Tissue without the mutation were 13.1 and 5.1 years, respectively.
acquisition was mandatory for this study and will allow Patients with tumoral IDH1-R132H mutations also experi-
evaluation of the role of concurrent or adjuvant temozo- enced longer OS if they received RT/PCV than if they received
lomide on the basis of molecular subgroups. RT alone (log rank p = .02). The number of events in patients
without the IDH1-R132H mutation was too small to de-
WHO GRADE II GLIOMA termine the association of treatment effect in this subset.15
Data and conclusions pertaining to WHO grade II gliomas RTOG-0424 was a study of concurrent and adjuvant
have largely been derived from patient subsets entered onto temozolomide and RT in a high-risk low-grade glioma
therapeutic trials. As a consequence, these recommen- population, and the 3-year OS rate was 73.1% (95% CI,
dations do not apply to the patients with lowest-risk dis- 65.3%80.8%), which was significantly improved compared
ease, that is, younger patients with completed resected with the prespecified historical control reported by Pignatti
tumors, especially oligodendroglioma. et al (p , .0001).16 Molecular analysis on this trial is pending.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 77


CHANG ET AL

An ongoing intergroup phase III trial is attempting to address patients with newly diagnosed anaplastic gliomas. Three
the role of adjunctive temozolomide in LrGG. hundred eighteen patients, most of whom had grade 3
anaplastic astrocytoma (with very few oligodendroglioma),
ROLE OF RADIOTHERAPY were randomly assigned 2:1:1 (arms A:B1:B2) to receive
Historically, RT trials have established that earlier intervention conventional RT (arm A), PCV (arm B1), or temozolomide
with RT prolongs PFS and reduces symptom burden, especially (arm B2) at diagnosis. At progression or unacceptable tox-
related to seizures, but does not improve survival compared icity, patients in arm A were treated with PCV or temozo-
with deferred RT.17 In addition, a categorical dose effect on lomide (1:1 random assignment), whereas patients in arms
survival is not observed with RT in the 45- to 64-Gy range.18 B1 or B2 received RT. The median PFS (HR 1.0; 95% CI,
Grade III tumors were historically labeled malignant gliomas 0.71.3) and OS (HR 1.2; 95% CI, 0.81.9) were similar for all
and were conglomerated in trials that included grade IV three arms. Hypermethylation of the MGMT promoter (HR
glioblastomas, which made it difficult to discern the precise 0.59; 95% CI, 0.361.0) and mutations of IDH1 gene (HR 0.48;
survival advantage from RT. More contemporary trials in grade 95% CI, 0.290.77) reduced the risk of progression, and IDH
II astrocytoma and oligodendroglioma, as well as in grade III mutations had the largest impact. MGMT promoter meth-
oligodendroglioma, have confimed that chemoradiotherapy ylation also was associated with prolonged PFS in the
yields a survival advantage compared with RT alone. A com- chemotherapy and RT arm. Therefore, initial RT or che-
mon theme to emerge from these trials is that, for the fa- motherapy achieved comparable results in patients with
vorable tumors, especially those with IDH mutations, a anaplastic gliomas.12 This trial has sometimes been used to
dramatic survival advantage emerges with combination che- support the logic of initiating chemotherapy first, followed
moradiotherapy; in the case of grade III tumors, even those in by RT at progression, for grade II gliomas (both astrocytic and
the 1p19q non-codeleted subset, a survival advantage with oligodendroglial) and grade III oligodendroglioma, but it
chemoradiotherapy is observed if the tumor has IDH mutant must be borne in mind that the results may not necessarily
status. An important caveat here is that these patients have extrapolate to these entities.
prolonged survival, so concerns have been expressed about
possible cognitive decline after RT; in practice, some clinicians SURGERY IN LOWER-GRADE GLIOMA
simply treat with chemotherapy alone and reserve RT for The role of surgical resection in adult diffuse gliomas is
salvage therapy. Level-1 data to support this practice do not twofold: diagnostic and therapeutic. Historically, the di-
exist, and, until it is categorically established that RT dose agnostic grading of a glioma has been dependent upon the
reduction or dose elimination is safe, these patients should type of procedure performed and the volume of tissue
continue to be treated with combination chemoradiotherapy. specimen provided to the pathologist for review. Thus, a
The IDH wild-type tumors do not exhibit this survival advantage less-extensive biopsy procedure can undersample a glioma,
with chemoradiotherapy, so it would be reasonable to consider which can result in diagnostic histologic undergrading. For
treating patients who have these tumors with RT alone. example, Glantz et al20 demonstrated that, in 262 patients
The EORTC has completed a large, international, ran- undergoing resection, 214 (82%) were ultimately rendered a
domized trial in 477 patients with WHO grade II tumors final diagnosis of glioblastoma (WHO grade IV) and 48 (18%)
(22033-26033), stratified by 1p deletion, and randomized to were diagnosed with anaplastic astrocytoma (WHO grade
RT alone or temozolomide alone. A difference between the III). In contrast, of 67 patients undergoing stereotactic bi-
two treatment strategies was not observed for PFS, although opsy, 33 (49%) were diagnosed with glioblastoma, whereas
RT was numerically favored. However, molecular tumor 34 (51%) were diagnosed with anaplastic astrocytoma.
characterization may allow the treatment approach to be These observations led to the conclusion that it was likely
personalized and one or the other treatment modality to be that some of the anaplastic astrocytomas diagnosed by
selected. A post hoc analysis of molecular markers was done stereotactic biopsy were undersampled and actually rep-
in 407 patients and was reported recently. IDH1 or IDH2 resented glioblastomas.20
mutations were detected in 85.8% of patients. Codeletions A similar theme was noted by Jackson et al,21 in a study
of 1p19q were identified in 33%. MGMT was methylated in based on biopsy specimens and the subsequent resection
90%, of which the majority, 86%, was IDH mutant. Mutation specimens from the same patient (paired samples), without
of IDH1 or IDH2, regardless of 1p19q codeletion, was a intervening delay or therapeutic intervention. In this study,
positive prognostic factor. Patients with IDH-mutated, non- the authors noted that the diagnoses rendered from the
codeleted tumors had shorter PFS after treatment with biopsy specimen differed from the diagnoses achieved by
temozolomide than after RT (HR 1.86; 95% CI, 1.212.87; log using the more extensive surgical resection specimen in 50
rank p = .0043), whereas no difference was observed be- (49%) of 82 instances. They concluded that stereotactic
tween these treatments for patients with IDH wild-type biopsy is frequently inaccurate in provision of a correct
tumors and IDH-mutated, codeleted tumors.19 diagnosis and thus recommended surgical resection, even
One additional study, the phase III German NOA-04 trial, purely for diagnostic purposes.21
deserves mention, relative to the role of RT in LrGG. The In a more recent analysis, Kim et al22 noted that molecular
NOA-04 trial compared the efficacy of RT followed by testing can be used to resolve these diagnostic discrepancies
chemotherapy at progression with the reverse sequence in that develop because of limited pathologic sampling. Similar

78 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


LOWER-GRADE GLIOMAS, MOLECULAR MARKERS, AND THERAPEUTIC CHOICES

to results by Glantz et al,20 these investigators noted a been proven highly effective in the randomized studies
volume-based discrepancy between diagnoses of grade IV discussed in this article.
and grade III tumors, whereby grade IV diagnoses were more
common in larger resection specimens. Notably, however,
the frequency of IDH1 mutation was constant between the
TARGETED THERAPY IN LOWER-GRADE GLIOMA
Aberrant signaling in pathways, including the PI3K/Akt/
two cohorts, which demonstrated that molecular status was
mTOR network, have been identified in grade II glioma,
volume independent. Importantly, the diagnosis of ana-
and clinical trials are ongoing to target this pathway as a
plastic astrocytoma from a less-extensive resection was
therapeutic approach.27 In addition, ongoing studies are
associated with significantly poorer survival (glioblastoma-
evaluating inhibitors of IDH.28 The ability to image levels of
like). Thus, the investigators concluded that, for smaller
the oncometabolite 2-hydroxyglutarate is an exciting area
surgical specimens, underdiagnosis of glioblastoma may
of research to develop noninvasive robust biomarkers of
occur when the analysis is restricted to histopathology
treatment response and clinical outcome in IDH-mutated
alone.22 Moving forward, the WHO will now integrate
tumors.29 These approaches mandate the selection of pa-
molecular scoring into the diagnostic categorization of these
tients on the basis of specific molecular/cytogenetic char-
lesions.23
acteristics and highlight the future of medical treatment of
With regard to the therapeutic benefit of resection, recent
LrGG in the genomic era.
studies have focused on the benefit of removal of the
nonenhancing tumor that makes up the majority (or en-
tirety) of an LrGG. Most studies of surgery for glioma are CONCLUSION
retrospective and nonrandomized and, therefore, are po- There is increasing evidence that multiple molecular markers
tentially biased because of patient selection. In a molecular are prognostic, and possibly even predictive, in patients with
study, Beiko et al24 found that IDH1 mutation was in- LrGG. The 1p19q codeletion historically has conferred im-
dependently associated with complete resection of en- proved prognosis and recently has been identified as a
hancing disease in grade III and IV astrocytomas and that predictive marker. IDH mutations, similarly, have also been
additional resection of nonenhancing disease was associ- identified as prognostic and, possibly, predictive.5,8,11 By
ated with an additional survival benefit only in the IDH1- using a combination of IDH1/2, 1p19q, and TERT mutations,
mutant cohort.24 For LrGG, Shaw et al25 observed 111 it is possible to subdivide these tumors into three working
patients who were assigned to observation after surgeon- subgroups: molecular glioblastoma, molecular astrocytoma,
determined gross-total resection in the RTOG-9802 study of and molecular oligodendroglioma. Effectively, this stratifies
low-risk, low-grade (WHO grade II) glioma.25 The most LrGGs from the worst prognostic category that has outcomes
favorable-risk cohort shared three factors: tumor diameter comparable to GBM to the best with very long survival rates
less than 4 cm on preoperative MRI scan, oligodendroglioma measured in decades and includes an in between category
histology, and less than 1 cm of residual tumor on post- as well; all categories likely differ in response to therapy.
operative MRI scan, indicative of the survival benefit Such stratification now calls into question both the existing
associated with more extensive resection. In a well- grade-based conventional therapeutic strategies and the
characterized cohort, Smith et al26 demonstrated an asso- value of ongoing clinical trials that do not include these as
ciation between gross-total resection of nonenhancing stratification variables.
tumor and survival in low-grade (WHO grade II) gliomas. The For the worst prognostic group of LrGG, more effective
majority of patients in these studies likely had IDH1-mutant therapies might be necessary, because conventional ther-
tumors. apies are relatively ineffective; patients with these tumors
Thus, the emerging molecular classification forms a could be enrolled in clinical trials of glioblastoma, perhaps
practical guide for surgical decision making in patients with as a separate subgroup. Among those with the best prog-
adult diffuse gliomas. For molecular glioblastomas, the nosis, median survival exceeds a decade, and this subgroup
evidence supports a surgical goal of complete resection of might include patients who can actually be observed in lieu
enhancing disease. There is no evidence yet in this pop- of immediate intervention. These patients might be suited
ulation that surgical resection of nonenhancing disease for treatment de-intensification, but this possibility raises
offers a survival benefit; surgical goals therefore should be the conundrum of how to risk treatment de-intensification,
curtailed for lesions in eloquent cortex with a focus on because even a modest loss in survival would take longer
safety. For molecular astrocytomas, evidence supports a than a decade to detect. One possible approach would be
surgical goal of maximal resection for both enhancing and to de-intensify only those components that have greater
nonenhancing disease. The standard of care for subsequent toxicity; for example, perhaps the indirect data about the
adjuvant treatment of patients with molecular astrocytoma efficacy of temozolomide in LrGGs would be adequate to
awaits guidance from ongoing randomized studies. Finally, consider its use instead of PCV?11,30,31 From an RT per-
for molecular oligodendrogliomas, a surgical goal of maximal spective, perhaps these are the tumors better suited for
safe resection of enhancing and nonenhancing disease again proton therapy, with its inherently lower integral dose.
is warranted but tempered with the knowledge that, for How do we move this genomic information directly to the
these patients, well-established adjuvant therapies have therapeutic realm, specifically in terms of tailored therapies?

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 79


CHANG ET AL

IDH-directed therapies have entered phase I clinical testing, resection, a surgical concept promulgated by Duffau?33
but, with prolonged median survival rates, how exactly does Will metabolic imaging that is based on the IDH-associated
one establish the therapeutic potential of such an agent? Of oncometabolite 2-hydroxyglutartate provide an avenue to
course, testing in the recurrence situation with response monitor residual disease or the effects of various therapies
rates or PFS as endpoints is one initial step, but making the or will it perhaps become a tool for contouring the target
leap to a survival-based trial in the newly diagnosed context for RT?34
will likely prove challenging. For example, one recent report IDH mutations represent the emergence of a new par-
suggests that IDH inhibitors might in fact be radioprotective, adigm; recently described molecular classifications cat-
which potentially negates their clinical value. 32 In the egorically demonstrate a number of less common, but
meantime, data are emerging that these IDH-mutated tu- potentially targetable, alterations, all of which individually
mors might be amenable to more complete resections and, are sufficiently rare that it would be near impossible to
maybe, that these are the patients who, after a gross total mount classic randomized phase III trials for each and
resection, could in fact receive observation only.24,33 Are every one. Perhaps the concept of the National Cancer
these perhaps also the patients best suited for and driv- Institutesponsored MATCH trial could be expanded to
ing the lengthy survival rates associated with supratotal these LrGGs.35

References
1. Louis DN, Ohgaki H, Wiestler OD, et al. The 2007 WHO classification of 14. Buckner JC, Pugh SL, Shaw EG, et al. Phase III study of radiation therapy
tumours of the central nervous system. Acta Neuropathol. 2007;114: (RT) with or without procarbazine, CCNU, and vincristine (PCV) in low-
97-109. grade glioma: RTOG 9802 with Alliance, ECOG, and SWOG. J Clin Oncol.
2. van den Bent MJ. Interobserver variation of the histopathological di- 2014;32:5s (suppl;abstr 2000).
agnosis in clinical trials on glioma: a clinicians perspective. Acta 15. Buckner JC, Shaw EG, Pugh SL, et al. Long-term results of R9802: a Phase
Neuropathol. 2010;120:297-304. III study of radiation therapy (RT) with or without procarbazine, CCNU,
3. Brat DJ, Verhaak RG, Aldape KD, et al; Cancer Genome Atlas Research and vincristine (PCV) in low-grade gliomaRTOG with NCCTG, ECOG,
Network. Comprehensive, integrative genomic analysis of diffuse and SWOG. N Engl J Med. In press.
lower-grade gliomas. N Engl J Med. 2015;372:2481-2498. 16. Pignatti F, van den Bent M, Curran D, et al. Prognostic factors for survival
4. Eckel-Passow JE, Lachance DH, Molinaro AM, et al. Glioma groups based in adult patients with cerebral low-grade glioma. J Clin Oncol. 2002;20:
on 1p/19q, IDH, and TERT promoter mutations in tumors. N Engl J Med. 2076-2084.
2015;372:2499-2508. 17. Karim AB, Afra D, Cornu P, et al. Randomized trial on the efficacy of
5. Yan H, Parsons DW, Jin G, et al. IDH1 and IDH2 mutations in gliomas. radiotherapy for cerebral low-grade glioma in the adult: European
N Engl J Med. 2009;360:765-773. Organization for Research and Treatment of Cancer Study 22845 with
6. Cairncross G, Wang M, Shaw E, et al. Phase III trial of chemo- the Medical Research Council study BRO4an interim analysis. Int J
radiotherapy for anaplastic oligodendroglioma: long-term results of Radiat Oncol Biol Phys. 2002;52:316-324.
RTOG 9402. J Clin Oncol. 2013;31:337-343. 18. Shaw E, Arusell R, Scheithauer B, et al. Prospective randomized trial of
7. van den Bent MJ, Brandes AA, Taphoorn MJ, et al. Adjuvant pro- low- versus high-dose radiation therapy in adults with supratentorial
carbazine, lomustine, and vincristine chemotherapy in newly diagnosed low-grade glioma: initial report of a North Central Cancer Treatment
anaplastic oligodendroglioma: long-term follow-up of EORTC brain Group/Radiation Therapy Oncology Group/Eastern Cooperative On-
tumor group study 26951. J Clin Oncol. 2013;31:344-350. cology Group study. J Clin Oncol. 2002;20:2267-2276.
8. Cairncross JG, Wang M, Jenkins RB, et al. Benefit from procarbazine, 19. Baumert BG, Hegi ME, Mason WP, et al. Radiotherapy in relation to
lomustine, and vincristine in oligodendroglial tumors is associated with temozolomide: subgroup analysis of molecular markers of the ran-
mutation of IDH. J Clin Oncol. 2014;32:783-790. domized phase III study by the EORTC/NCIC-CTG/TROG/MRC-CTU
9. Stewart LA. Chemotherapy in adult high-grade glioma: a systematic (EORTC 22033-26033) in patients with a high-risk low-grade glioma.
review and meta-analysis of individual patient data from 12 randomised J Clin Oncol. 2015;33 (suppl; abstr 2006).
trials. Lancet. 2002;359:1011-1018. 20. Glantz MJ, Burger PC, Herndon JE II, et al. Influence of the type of
10. Medical Research Council Brain Tumor Working Party. Randomized trial surgery on the histologic diagnosis in patients with anaplastic gliomas.
of procarbazine, lomustine, and vincristine in the adjuvant treatment of Neurology. 1991;41:1741-1744.
high-grade astrocytoma: a Medical Research Council trial. J Clin Oncol. 21. Jackson RJ, Fuller GN, Abi-Said D, et al. Limitations of stereotactic biopsy
2001;19:509-518. in the initial management of gliomas. Neuro Oncol. 2001;3:193-200.
11. Chang SM, Zhang P, Cairncross JG, et al. Results of NRG oncology/RTOG 22. Kim BY, Jiang W, Beiko J, et al. Diagnostic discrepancies in malignant
9813: A phase III randomized study of radiation therapy (RT) and astrocytoma due to limited small pathological tumor sample can be
temozolomide (TMZ) versus RT and nitrosourea (NU) therapy for an- overcome by IDH1 testing. J Neurooncol. 2014;118:405-412.
aplastic astrocytoma (AA). J Clin Oncol. 2015;33 (suppl; abstr 2002). 23. Louis DN, Perry A, Burger P, et al; International Society Of
12. Wick W, Hartmann C, Engel C, et al. NOA-04 randomized phase III trial of NeuropathologyHaarlem. International Society Of Neuropathology
sequential radiochemotherapy of anaplastic glioma with procarbazine, Haarlem consensus guidelines for nervous system tumor classification
lomustine, and vincristine or temozolomide. J Clin Oncol. 2009;27: and grading. Brain Pathol. 2014;24:429-435.
5874-5880. 24. Beiko J, Suki D, Hess KR, et al. IDH1 mutant malignant astrocytomas are
13. Shaw EG, Wisoff JH. Prospective clinical trials of intracranial low-grade more amenable to surgical resection and have a survival benefit as-
glioma in adults and children. Neuro Oncol. 2003;5:153-160. sociated with maximal surgical resection. Neuro Oncol. 2014;16:81-91.

80 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


LOWER-GRADE GLIOMAS, MOLECULAR MARKERS, AND THERAPEUTIC CHOICES

25. Shaw EG, Berkey B, Coons SW, et al. Recurrence following neurosurgeon- diagnosed anaplastic oligodendrogliomas and mixed anaplastic oli-
determined gross-total resection of adult supratentorial low-grade goastrocytomas: long-term results of RTOG BR0131. J Neurooncol.
glioma: results of a prospective clinical trial. J Neurosurg. 2008;109: 2015;124:413-420.
835-841. 31. Fisher BJ, Hu C, Macdonald DR, et al. Phase 2 study of temozolomide-
26. Smith JS, Chang EF, Lamborn KR, et al. Role of extent of resection in the based chemoradiation therapy for high-risk low-grade gliomas: pre-
long-term outcome of low-grade hemispheric gliomas. J Clin Oncol. liminary results of Radiation Therapy Oncology Group 0424. Int J Radiat
2008;26:1338-1345. Oncol Biol Phys. 2015;91:497-504.
27. McBride SM, Perez DA, Polley MY, et al. Activation of PI3K/mTOR 32. Molenaar RJ, Botman D, Smits MA, et al. Radioprotection of IDH1-
pathway occurs in most adult low-grade gliomas and predicts patient mutated cancer cells by the IDH1-mutant inhibitor AGI-5198. Cancer
survival. J Neurooncol. 2010;97:33-40. Res. 2015;75:4790-4802.
28. Rohle D, Popovici-Muller J, Palaskas N, et al. An inhibitor of mutant IDH1 33. Duffau H. Long-term outcomes after supratotal resection of diffuse low-
delays growth and promotes differentiation of glioma cells. Science. grade gliomas: a consecutive series with 11-year follow-up. Acta
2013;340:626-630. Neurochir (Wien). 2015;158:51-58.
29. Andronesi OC, Loebel F, Bogner W, et al. Treatment response as- 34. Dang L, White DW, Gross S, et al. Cancer-associated IDH1 mutations
sessment in IDH-mutant glioma patients by non-invasive 3D functional produce 2-hydroxyglutarate. Nature. 2009;462:739-744.
spectroscopic mapping of 2-hydroxyglutarate. Clin Cancer Res. 2016;22: 35. Clinical Trial NCT02465060. NCI-MATCH: targeted therapy directed by
1632-1641. genetic testing in treating patients with advanced refractory solid tu-
30. Vogelbaum MA, Hu C, Peereboom DM, et al. Phase II trial of pre- mors or lymphomas. https://clinicaltrials.gov/ct2/show/NCT02465060.
irradiation and concurrent temozolomide in patients with newly Accessed November 11, 2015.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 81


DEVELOPMENTAL THERAPEUTICS AND
TRANSLATIONAL RESEARCH

Biomarkers, Blood-Based Testing,


and the Heterogeneous Tumor

CHAIR
Charles Swanton, MBPhD, FRCP, FMedSci
The Francis Crick Institute
London, United Kingdom

SPEAKERS
Julia A. Beaver, MD
U.S. Food and Drug Administration
Silver Spring, MD

Melissa Lynne Johnson, MD


Sarah Cannon Research Institute
Nashville, TN
MANAGEMENT OF INTRATUMOR HETEROGENEITY

Tumor Evolutionary Principles: How Intratumor


Heterogeneity Influences Cancer Treatment and Outcome
Subramanian Venkatesan, BSc, and Charles Swanton, MBPhD, FRCP, FMedSci

OVERVIEW

Recent studies have shown that intratumor heterogeneity contributes to drug resistance in advanced disease. Intratumor
heterogeneity may foster the selection of a resistant subclone, sometimes detectable prior to treatment. Next-generation
sequencing is enabling the phylogenetic reconstruction of a cancers life history and has revealed different modes of cancer
evolution. These studies have shown that cancer evolution is not always stochastic and has certain constraints. Consid-
eration of cancer evolution may enable the better design of clinical trials and cancer therapeutics. In this review, we
summarize the different modes of cancer evolution and how this might impact clinical outcomes. Furthermore, we will
discuss several therapeutic strategies for managing emergent intratumor heterogeneity.

rule books of cancer evolution might have important


T he selection of fitter subclones in the context of tumor
evolution was postulated by Nowell in the 1970s.1 The
existence of multiple phenotypes within a single tumor was
implications for designing trials and cancer treatments. In
this review, we will touch upon how intratumor heteroge-
determined 3 decades ago2; however, the emergence of neity may affect clinical outcome and discuss the different
next-generation sequencing has enabled the more detailed modes of cancer evolution. We will also discuss different
study of cancer evolution and intratumor heterogeneity. therapeutic strategies for the management of intratumor
Genome instability is one of the proposed hallmarks of heterogeneity.
cancer and is believed to be acquired early in tumorigenesis,
promoting the acquisition of other cancer hallmarks.3,4 It THE RELATIONSHIP BETWEEN INTRATUMOR
is a collective term, referring to a spectrum of different HETEROGENEITY AND CLINICAL OUTCOME
genetic aberrations ranging from point mutations to chro- Genome instability and intratumor heterogeneity are closely
mosomal rearrangements, gains, and losses.5,6 Genome associated, as genome instability increases the rate of
instability equips cancer with the potential to create new mutation (in the broadest sense referring to nucleotide and
genetic aberrations in daughter cells and to adapt to new to whole chromosome level changes) and the subsequent
environments.3 Once one of these genetic aberrations chance of subclonal (i.e., in a fraction of cancer cells) di-
confers a fitness advantage, genetically distinct subclones versification (reviewed by Burrell et al12). The influence of
may arise within a single cancer. Many cancers show hall- genome instability and intratumor heterogeneity upon clin-
marks of genome instability that support the development ical outcome is becoming clearer. Chromosomal instability
of intratumor heterogeneity, evidenced by elevated rates (CIN) is a specific form of genome instability and refers to the
of point mutations,7-9 chromosomal rearrangements,10 and rate of acquiring whole-chromosome or segmental chro-
somatic copy-number aberrations.10,11 Intratumor hetero- mosomal aneuploidies.13 CIN may contribute to extensive
geneity and cancer evolution contribute to resistance to intratumor heterogeneity in clear cell renal cell carcinomas
therapy and therefore demand investigation. (ccRCC)14; therefore, the identification of CIN-driving events
As the number of studies reporting on the extent of may inform how to manage intratumor heterogeneity. In
intratumor heterogeneity in different cancer types is rising some cases, genome-doubled tumors are thought to pre-
sharply, it is becoming clear that cancer evolution is not cede the acquisition of a CIN phenotype.15 The ability of a
always stochastic. Recurring commonalities observed within diploid cell to tolerate a genome-doubling event sub-
each cancer type suggests there are certain rules and sequently fosters the propagation of aneuploid cells and
constraints dictating how a tumor can evolve. Identifying the progressive evolution of chromosomally unstable tumor

From the UCL Cancer Institute, CRUK Lung Cancer Centre of Excellence, London, United Kingdom; The Francis Crick Institute, London, United Kingdom.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Charles Swanton, MBPhD, FRCP, FMedSci, Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, 44 Lincolns Inn Fields, London WC2A 3LY;
email: charles.swanton@crick.org.uk.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e141


VENKATESAN AND SWANTON

cells.15 Emerging evidence suggests that genome doubling FIGURE 1. The Trade-off Between Genome
itself is a predictor of a poorer relapse-free survival in early- Instability/Intratumor Heterogeneity and Clinical
stage colorectal15 and ovarian cancers.16 Outcome
Interestingly, a mouse model with a heterozygous knockout
of the gene centrosome-associated protein E (Cenp-e) led
to increased aneuploidy and CIN.17 The Cenp-e+/2 mice de-
veloped more spontaneous spleen and lung tumors compared
with the Cenp-e+/+ mice. In contrast, genetically (through
homozygous p19ARF deletion) and chemically (through 7,12-
dimethylbenz[a]anthracene exposure) induced tumorigen-
esis was inhibited in the Cenp-e+/2 mice compared with
wild-type mice. These results suggest that CIN, in some
cases, can both promote and inhibit tumorigenesis.17 This
study supports the mutational meltdown theory postulated
by Lynch et al regarding deleterious mutations in asexual
populations.18 Here, the accumulation of deleterious mu-
tations combined with the inability of sexual recombination might be related to the trade-off between tumor fitness and
contributes to the extinction process, which they termed genome instability as previously discussed (Fig. 1).25 Col-
mutational meltdown. Cancer cells also divide and re- lectively, these studies suggest a range of optimal genome
produce in an asexual manner; therefore, the observations instability favoring the tumors fitness, which at the right
by Weaver et al17 suggested that the mutational meltdown threshold, may negatively affect clinical outcome (Fig. 1).
theory18 may also apply to human cancers. Our group in- However, beyond a certain threshold, excessive genome
vestigated the association between the level of CIN and instability may result in diminished cancer cell survival and
clinical outcome.19,20 We found, in different cancer types, is paradoxically associated with an improvement in cancer
that patients with tumors containing intermediate levels survival outcomes (Fig. 1).
of CIN had the worst prognosis, whereas tumors with The influence of specific somatic alterations on the rate of
high levels of CIN were associated with improved prognosis evolution has been subject to investigation. Genomic ab-
(Fig. 1).19,20 These observations have been recapitulated in errations in cancer may accumulate either gradually such as
recent studies of high-grade serous ovarian cancer,21 es- through point mutations (microevolution), or in a punctu-
trogen receptornegative breast cancer,19,22 and a pan- ated manner through processes such as chromoplexy,26 or
cancer study across 12 cancer types.23 in a single catastrophic event such as through chromo-
Although the number of patients with ovarian cancer thripsis (macroevolution).27 The distinction between these
studied by Schwarz et al and patients with esophageal cancer different magnitudes of genomic changes might be of im-
studied by Murugaesu et al were probably not large enough portance for the rate of disease progression. For example,
to detect the nonlinear relationship between the extent of chromothripsis was identified in a small subset of patients in
intratumor heterogeneity and response to therapy, they still multiple myeloma and was often related to rapid relapse and
demonstrated that patients with more heterogeneous death within 1 to 2 years.28
cancers had poorer response.21,24 In glioblastoma, Kim et al It is of clinical importance to elucidate how cancers tol-
report that TP53 mutations coincide with higher frequencies erate such somatic events and propagate diversity and
of subclonal mutations and better clinical outcome, which branched evolution, as this may inform how cancer prog-
resses and support new therapeutic approaches to limit
these processes. Through deep, multiregion, and longitu-
dinal sequencing studies of the primary tumor and metas-
KEY POINTS tases, it might be possible to order somatic events more
effectively and identify genetic events enabling the toler-
Cancer evolution is not always stochastic and is ance and propagation of genome instability. For example,
subjected to several constraints evidenced by parallel prostate cancer metastases were enriched for TP53 muta-
and convergent evolution. tions, whereas this mutation was subclonal in the primary
There is a trade-off between tumor fitness and genome tumor.29 Conceivably, metastasis may be enhanced by loss-
instability. of-function of TP53, possibly allowing for greater tolerance
Different modes of cancer evolution may be operative
of genome instability and the subsequent gain of a
over the lifetime of a cancer.
metastasis-driving event.
Multiregion sequencing and/or longitudinal monitoring
of cancer subclones enables a better understanding of Unfortunately, the clinical effect of targeted therapeutic
evolutionary life histories. drugs has been limited by the rapid acquisition of drug
Understanding cancer evolution may inform the better resistance in the metastatic setting. There are now multiple
design of clinical trials and cancer treatments. reports suggesting that drug-resistant subclones reside
within the tumor at low frequency prior to therapy in

e142 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


MANAGEMENT OF INTRATUMOR HETEROGENEITY

melanoma,30 prostate cancer,31 colorectal cancer,32,33 ovarian the same patient acquiring (epi)genetic alterations in similar
cancer,21 and medulloblastoma.34 We recently reported in a genes, protein complexes, or signaling pathways (Fig. 2C).49
study of nine cancer types that mutations in many driver Conversely, parallel evolution refers to subclones derived
genes can be subclonal across multiple different tumor types. from the same parental clone acquiring (epi)genetic alter-
These data suggest that targeting founder events present in ations in similar genes, protein complexes, or signaling
every tumor cell may facilitate a more effective drug devel- pathways (Fig. 2D).50,51 Although many tumors follow a
opment process.35 branched evolutionary pattern, presumably many of these
tumors experience common selective pressures from the
microenvironment or constraints inherited from the pa-
MODES OF CANCER EVOLUTION rental founder cell, which force the different subclones to
In the past decade, different modes of cancer evolution have independently converge on similar cellular processes. The
been discovered. Historically, cancer was believed to follow level of convergent and parallel evolution is of emerging
the multistep process of oncogene activation, tumor sup- interest because of developments in the use of multiregion
pressor gene loss, and subsequent clonal sweeps by the and single cell sequencing (Table 1).
fittest clone.1,36 With current advances in next-generation Up to this point, we have described cancer as co-evolving,
sequencing technology, the initial hypothesis has been re- mutually independent subclones; however, reality is more
fined and additional models of macroevolution26,27,37 and complex as these subclones also interact. In an environment
neutral evolution38-40 have been postulated. In this section, with limited space and nutritional resources, there are
we will summarize the current state of knowledge of cancer complex dynamics of clonal cooperation and clonal in-
evolution (Table 1 and Fig. 2). terference (Fig. 2E), which are only now beginning to be
understood. In vitro52-55 and in vivo54-57 data show that
Darwinian Cancer Evolution subclonal populations can protect and/or drive cancer as a
Deep sequencing analysis of hematologic and solid tumors whole. Marusyk et al elegantly demonstrated in a mouse
indicate that cancer is usually of clonal origin and often model that a minor subclone with inferior fitness drove
follows a branched tumor evolutionary pattern.41 Using tumor growth in a noncell-autonomous fashion through
tumor purity estimates, local copy number aberrations, and the secretion of a specific ligand.56 This enabled the tumor to
mutation variant allele frequency, it is possible to calculate maintain growth, clonal diversity, and even metastatic po-
the cancer cell fraction of a mutation and phylogenetically tential. These tumor phenotypes could be abrogated once
reconstruct the life history of cancers.42-45 The trunk of the the driving subclones were omitted from the tumor, high-
phylogenetic tree describes the clonal events where the lighting the importance of examining minor subclones in
founder driver events are present. Trunk driver events are addition to dominant subclones. Similar clonal dynamics
present in all cancer cells derived from the cell of origin. are likely also operative in patients. Clonal interference
As the cancer genome evolves during cancer progres- between a dominant subclone and a minor subclone may
sion, subclonal events are introduced into a subset of the prevent the outgrowth of the minor subclone during tumor
progeny, termed branched events (Fig. 2A). A clonal sweep progression (Fig. 2E). However, the harsh selective pressure
occurs if a branch driver event increases the fitness of a imposed by the tumor microenvironment, the cancer
subclone to the extent that it out-competes all other sub- therapeutics directed toward the dominant clone, or the
clones in the tumor. This mode of cancer evolution has been process of metastasis in general may permit a resistant
termed linear evolution (Fig. 2B). Reports suggest that minor subclone residing in the tumor to emerge by com-
branched evolution is common in cancer, as there may petitive release.58 Gatenby et al hypothesized that a tumor
be one or more subclonal/branched driver events pre- may consist of chemotherapy-sensitive cells that have a
sent within distinct subclones of a tumor. Nevertheless, higher fitness in the absence of chemotherapeutic pressure
in several cancer types, cases of linear evolution have been and suppress the outgrowth of less fit chemotherapy-
documented such as in acute myeloid leukemia, 46,47 resistant subpopulation. They demonstrated that adapting
chronic lymphocytic leukemia, 48 and VHL-associated the chemotherapy dose according to tumor dynamicsthat
ccRCC. 49 is, higher doses upon tumor growth and lower doses upon
We have studied four clonally independent primary kidney tumor reductionwas able to stabilize the tumor and
cancers in the context of a germline VHL mutation.49 Several prevent the resistant subpopulation from reaching lethal
observations were made from this patient: (1) the tumors proportions.58 This therapeutic strategy was appropriately
were polyclonal, (2) the tumors shared no other muta- called adaptive therapy, and, related to this, it was recently
tions apart from the germline VHL mutation highlighting shown to be potentially relevant in the treatment of
independent evolutionary trajectories despite similar mi- BRAFV600E-mutant melanomas (Fig. 2F).59
croenvironments, and (3) the tumors showed signs of A different approach to address an emerging treatment-
evolutionary constraints shown by the absence of branched resistant subclone may include altering the drug instead
evolution and the presence of convergent evolution toward of the drug doses with the aim to eradicate the cancer
the PI3K-Akt-mTOR pathway (Fig. 2C).49 Convergent evo- (i.e., sequential therapy; Fig. 2G). In sequential therapy,
lution refers to the process of independent tumors within the treatment is sequentially adapted according to the

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e143


VENKATESAN AND SWANTON

evolution of new treatment-resistant events. The poten- tumor and the accompanying metastases is enabling a
tial of sequential therapy has been demonstrated in the clearer picture of the metastatic process. Studies by
treatment of drug-sensitive EGFR-driven (often EGFRL585R Gundem et al and Hong et al in prostate cancer show that
or EGFR exon 19 deletion) lung cancer with first-generation the following patterns of metastasis occur frequently
EGFR inhibitors (erlotinib or gefitinib). Tumors often (Fig. 2H): (1) multiple subclones can seed the same
progress as a result of the emergence of an EGFRT790M metastatic site (termed polyclonal seeding), (2) combi-
resistanceconferring mutation.60 However, this mutation nations of shared subclones between these polyclonal
renders cancer cells sensitive to the third-generation EGFR seeds are recurring (hinting toward clonal cooperation),
inhibitors, AZD929161 and rociletinib.62 Nevertheless, se- (3) subclones within a metastasis can spread to another
quential therapy may eventually encounter a resistant metastatic site (termed metastasis-to-metastasis spread
subclone that is not yet amenable to treatment because or metastatic cross-seeding), and (4) tumor cells from
treating tumors that harbor the EGFRT790M variant with the metastasis may return back to the primary tumor
either AZD929163 or rociletinib 64 appears to select for the (termed self-seeding).65 The formation of metastatic sub-
re-emergence of EGFRT790M-negative cells, EGFRC797S clones were long thought to occur through the clonal se-
mutant cells, or EGFRT790M-positive cells with a yet un- lection of the fittest and latest evolving subclone. However,
identified resistance event (Fig. 2G). Clonal interactions are recent studies show that metastatic progression can also
not limited to the primary tumor but also extend to the occur through the early diverged clonal expansion of the
metastatic sites. Phylogenetic reconstruction of the primary trunk clone.21,29

TABLE 1. Different Modes of Cancer Evolution Reported in a Selection of Different Cancer Types
Cancer Type Mode of Evolution
AML Branched evolution80,81
Parallel evolution81: FLT3-ITD
Linear evolution46,47,82
Breast Cancer Branched evolution83
Parallel evolution83: PTEN, FGFR2, TP53, RUNX1
Punctuated and catastrophic evolution7,83: chromoplexy, chromothripsis, kataegis
Colorectal Cancer Catastrophic evolution27: chromothripsis
Neutral evolution39,40
Esophageal Branched evolution24,84,85
Cancer
Catastrophic evolution27: chromothripsis
Parallel evolution24: NOTCH1, GNPTAB
Polyclonal tumorigenesis85
Follicular Branched evolution86,87
Lymphoma
Parallel evolution86,87: chromatin-modifying genes (KMT2D, KMT2C, EZH2, CREBBP, MEF2B, EP300, HIST1H1E,
HIST1H1C, SWI/SNF complex [ARID1A, SMARCA4])
Punctuated evolution7: kataegis
Glioma Branched evolution88,89 in distant relapse90
Catastrophic evolution27: chromothripsis
Linear evolution in local relapse25,90
Neutral evolution38,40
Parallel evolution25,90,91: EGFR, TP53, PTEN, RB1, NF1, CDKN2A/B
Melanoma Branched evolution30,92
Catastrophic evolution27: chromothripsis
Metastasic cross-seeding93
Neutral evolution38,40
Parallel evolution30,92,93: KRAS, BRAF, MEK1, PIK3R2, PHLPP1, CTNNB1
Multiple Branched evolution94-96
Myeloma
Linear evolution94-97
Parallel evolution96,97: NRAS, KRAS, BRAF, NF1, RASA2, FAM46C
Punctuated and catastrophic evolution28,96: kataegis, chromothripsis

Continued

e144 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


MANAGEMENT OF INTRATUMOR HETEROGENEITY

TABLE 1. Different Modes of Cancer Evolution Reported in a Selection of Different Cancer Types (contd)
Cancer Type Mode of Evolution
NSCLC Branched evolution98,99
Neutral evolution38,40
Polyclonal tumorigenesis98
Punctuated and catastrophic evolution7,27,98,100: chromoplexy, chromothripsis, kataegis
Ovarian Cancer Branched evolution21
Metastasic cross-seeding21
Neutral evolution38,40
Prostate Cancer Branched evolution101,102
Punctuated and catastrophic evolution26: chromoplexy
Parallel evolution102: MYC, methylation sites
Metastasic cross-seeding103
Neutral evolution38,40
Polyclonal seeding103
Sporadic ccRCC Branched evolution45,51
Catastrophic evolution27: chromothripsis
Neutral evolution38,40
Parallel evolution45,51,68: mTOR pathway, SETD2, PTEN, KDM5C, PIK3CA, BAP1, PBRM1, SWI/SNF complex
VHL-associated Linear evolution49
ccRCC
Convergent evolution49: mTOR pathway (TSC1, mTOR)
Polyclonal tumorigenesis49
Abbreviations: AML, acute myeloid leukemia; NSCLC, nonsmall cell lung cancer; ccRCC, clear cell renal cell carcinoma.

Neutral Evolution mutations were clonal in individual tumor glands and it is


In addition to the model of Darwinian selection of the improbable that a whole gland would migrate.
fittest subclone, a different model was postulated by This model of neutral cancer evolution states that after
Sottoriva et al termed the big bang model, or neutral malignant transformation, individual subclones grow at similar
cancer evolution (Fig. 2I).39 In this study in colorectal rates, despite showing distinct mutational patterns. In this
cancer, individual tumor glands and two bulk fragments context, the timing of a new mutation occurring is the major
originating from the left and right side of the resected determinant of its prevalence within the tumor, rather than
tumors were subjected to different genomic techniques clonal selection for that mutation. In follow-up studies, the
including whole-exome sequencing and single nucleotide role of neutral evolution across different cancer types was
polymorphism arrays. As expected, they found a pro- investigated.38,40 These studies found that neutral evolution
portion of the aberrations to be clonal, but unexpectedly, may play an important role in tumor development of cervical
the same set of subclonal (or private) mutations could be squamous cell carcinoma and endocervical adenocarcinoma,
found in different tumor glands on opposite sides of the colorectal, stomach, lung, prostate, bladder, and urothelial
resection. These observations suggest that once the initial cancers.38,40 However, once treatment has been initiated,
cell has transformed into a cancer cell, subclonal mutations it is likely that other forms of Darwinian cancer evolution
rapidly accumulate in the primordial tumor. Subsequently, take over and eventually force the selection of treatment-
the daughter cells of specific subclones may migrate small resistant subclones.
distances within the primordial tumor mass. Indeed, this
short-range cellular migration may be a basic feature of
aggressive tumor growth.66 The distance between these APPROACHES TO TRANSLATE KNOWLEDGE OF
daughter cells is presumably amplified by the subsequent INTRATUMOR HETEROGENEITY INTO CANCER
(big banglike) growth, possibly explaining why tumor TREATMENT
glands of the same subclone are scattered throughout the Over the past few years, the cancer research community has
tumor. Other explanations for the finding that exactly the hunted for substantially mutated genes or driver genes. The
same subclonal mutations are found in different parts of the underlying premise was that cancer cells have become
colorectal cancer could include that these subclonal mu- addicted to the driver gene and, consequently, inhibition of
tations are acquired late in tumor evolution with the these genes may halt tumor progression. However, the
subsequent migration of these cancer cells. However, as presence of multiple subclones with different driver and
the authors stressed, this is unlikely because these private resistance mutations in a single tumor is a major hurdle for

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e145


VENKATESAN AND SWANTON

FIGURE 2. Different Modes of Cancer Evolution Contribute to Different Patterns of Intratumor Heterogeneity

e146 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


MANAGEMENT OF INTRATUMOR HETEROGENEITY

targeted therapy. Cancer drug development is undoubtedly subclonal events,71-74 minimal residual disease, and the
made more costly by the ability of a resistant subclone to be acquisition of new somatic alterations associated with drug
selected during treatment in a matter of weeks or months. resistance,31,32,75 and it can be used to monitor re-
There is a need to implement the knowledge gained by sponse73,76 (reviewed in Crowley et al77 and Krebs et al78).
cancer evolutionary models into designing better anticancer In addition to adapting treatment to the adapting cancer, it
therapies. In this section, we will provide a brief summary may be attractive to interfere with the causes of genome
of different strategies incorporating knowledge of cancer instability. Reducing genome instability may reduce the
evolutionary medicine. adaptability of cancer cells to treatments. Conversely, in-
A therapeutic strategy that has been advocated by us and creasing genome instability may tip the cancer cell beyond
others to maximize tumor response is to target trunk driver the levels of tolerance, forcing cell-autonomous lethality. As
events present in every tumor cell.67 In this case, it is im- there are many causes of genome instability (reviewed in
portant to accurately determine the clonal driver events. Aguilera et al6 and Burrell et al12), identifying the mecha-
Parallel evolution is widespread in different cancer types and nistic basis of micro- and macroevolutionary events may
represents the constraints imposed on cancer evolution enable the therapeutic intervention and prevention of major
(Table 1). Therefore, targeting parallel evolutionary events and resistance-forming events from occurring.
exploiting these cancer dependencies may provide a com-
pelling approach to therapy.49,68,69 Indeed, these observations
were initially made in ccRCC, in which rapalogs worked ex- CONCLUSION
ceptionally well in tumors that harbored multiple mutations The number of studies investigating cancer evolution is rising
that converged upon the mTOR pathway,68 suggesting that sharply and has contributed to early insights into cancer evo-
parallel evolution could pose as an Achilles heel of cancer. lutionary rule books. Sequencing of bulk tumor samples in
As cancer evolves during treatment, dynamic therapy projects such as The Cancer Genome Atlas have set the foun-
strategies may allow the clinician to maintain a stable dations for precision medicine; however, intratumor heteroge-
population of treatment-sensitive cells (adaptive therapy),58 neity is still largely underestimated in these datasets. Multiregion
or even force the tumor down a particular evolutionary sequencing and longitudinal monitoring via liquid biopsy together
route resulting in acquired sensitivity to a different drug with postmortem studies is necessary to attain a more accurate
(sequential therapy). The utility of sequential therapy has picture of cancer evolution. TRACERx (TRAcking nonsmall cell
recently been demonstrated in a patient with an ALK- lung Cancer Evolution through therapy [Rx]) is such a prospective
rearranged nonsmall cell lung cancer, who sequentially study, which aims to define the evolutionary trajectories
responded to different generations of ALK inhibitors.70 To of nonsmall cell lung cancer.79
inform the next step in adaptive and sequential therapy, Although the complexities of cancer evolution are just
longitudinal monitoring of the cancer is essential. Liquid beginning to be understood, much has been learned over the
biopsy analysis of circulating tumor DNA or circulating tumor past decade. We anticipate that cancer evolutionary models
cells is a convenient way to monitor cancer genetic changes. will inform clinicians how to design clinical trials and cancer
Furthermore, liquid biopsy has the potential to detect treatments.

References
1. Nowell PC. The clonal evolution of tumor cell populations. Science. 9. Kandoth C, McLellan MD, Vandin F, et al. Mutational landscape and
1976;194:23-28. significance across 12 major cancer types. Nature. 2013;502:333-339.
2. Heppner GH. Tumor heterogeneity. Cancer Res. 1984;44:2259-2265. 10. Yang L, Luquette LJ, Gehlenborg N, et al. Diverse mechanisms of
3. Negrini S, Gorgoulis VG, Halazonetis TD. Genomic instabilityan somatic structural variations in human cancer genomes. Cell. 2013;
evolving hallmark of cancer. Nat Rev Mol Cell Biol. 2010;11:220-228. 153:919-929.
4. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. 11. Zack TI, Schumacher SE, Carter SL, et al. Pan-cancer patterns of so-
Cell. 2011;144:646-674. matic copy number alteration. Nat Genet. 2013;45:1134-1140.
5. Aguilera A, Gomez-Gonz
alez B. Genome instability: a mechanistic view 12. Burrell RA, McGranahan N, Bartek J, et al. The causes and conse-
of its causes and consequences. Nat Rev Genet. 2008;9:204-217. quences of genetic heterogeneity in cancer evolution. Nature. 2013;
6. Aguilera A, Garca-Muse T. Causes of genome instability. Annu Rev 501:338-345.
Genet. 2013;47:1-32. 13. Geigl JB, Obenauf AC, Schwarzbraun T, et al. Defining chromosomal
7. Alexandrov LB, Nik-Zainal S, Wedge DC, et al; Australian Pancreatic instability. Trends Genet. 2008;24:64-69.
Cancer Genome Initiative; ICGC Breast Cancer Consortium; ICGC 14. Martinez P, Birkbak NJ, Gerlinger M, et al. Parallel evolution of tumour
MMML-Seq Consortium; ICGC PedBrain. Signatures of mutational subclones mimics diversity between tumours. J Pathol. 2013;230:
processes in human cancer. Nature. 2013;500:415-421. 356-364.
8. Lawrence MS, Stojanov P, Mermel CH, et al. Discovery and saturation 15. Dewhurst SM, McGranahan N, Burrell RA, et al. Tolerance of whole-
analysis of cancer genes across 21 tumour types. Nature. 2014;505: genome doubling propagates chromosomal instability and acceler-
495-501. ates cancer genome evolution. Cancer Discov. 2014;4:175-185.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e147


VENKATESAN AND SWANTON

16. Carter SL, Cibulskis K, Helman E, et al. Absolute quantification of somatic 38. Sottoriva A, Graham T. A pan-cancer signature of neutral tumor
DNA alterations in human cancer. Nat Biotechnol. 2012;30:413-421. evolution. bioRxiv. 2015. http://biorxiv.org/content/early/2015/02/
17. Weaver BA, Silk AD, Montagna C, et al. Aneuploidy acts both onco- 11/014894. Accessed February 12, 2016.
genically and as a tumor suppressor. Cancer Cell. 2007;11:25-36. 39. Sottoriva A, Kang H, Ma Z, et al. A Big Bang model of human colorectal
18. Lynch M, Burger R, Butcher D, et al. The mutational meltdown in tumor growth. Nat Genet. 2015;47:209-216.
asexual populations. J Hered. 1993;84:339-344. 40. Williams MJ, Werner B, Barnes CP, et al. Identification of neutral
19. Roylance R, Endesfelder D, Gorman P, et al. Relationship of extreme tumor evolution across cancer types. Nat Genet. Epub 2016 Jan 18.
chromosomal instability with long-term survival in a retrospective 41. Greaves M. Evolutionary determinants of cancer. Cancer Discov. 2015;
analysis of primary breast cancer. Cancer Epidemiol Biomarkers Prev. 5:806-820.
2011;20:2183-2194. 42. Campbell PJ, Pleasance ED, Stephens PJ, et al. Subclonal phylogenetic
20. Birkbak NJ, Eklund AC, Li Q, et al. Paradoxical relationship between structures in cancer revealed by ultra-deep sequencing. Proc Natl
chromosomal instability and survival outcome in cancer. Cancer Res. Acad Sci USA. 2008;105:13081-13086.
2011;71:3447-3452. 43. Miller CA, White BS, Dees ND, et al. SciClone: inferring clonal ar-
21. Schwarz RF, Ng CK, Cooke SL, et al. Spatial and temporal heterogeneity chitecture and tracking the spatial and temporal patterns of tumor
in high-grade serous ovarian cancer: a phylogenetic analysis. PLoS evolution. PLOS Comput Biol. 2014;10:e1003665.
Med. 2015;12:e1001789. 44. Roth A, Khattra J, Yap D, et al. PyClone: statistical inference of clonal
22. Jamal-Hanjani M, AHern R, Birkbak NJ, et al. Extreme chromosomal population structure in cancer. Nat Methods. 2014;11:396-398.
instability forecasts improved outcome in ER-negative breast cancer: 45. Gerlinger M, Rowan AJ, Horswell S, et al. Intratumor heterogeneity
a prospective validation cohort study from the TACT trial. Ann Oncol. and branched evolution revealed by multiregion sequencing. N Engl J
2015;26:1340-1346. Med. 2012;366:883-892.
23. Andor N, Graham TA, Jansen M, et al. Pan-cancer analysis of the extent 46. Klco JM, Spencer DH, Miller CA, et al. Functional heterogeneity of
and consequences of intratumor heterogeneity. Nat Med. 2016;22: genetically defined subclones in acute myeloid leukemia. Cancer Cell.
105-113. 2014;25:379-392.
24. Murugaesu N, Wilson GA, Birkbak NJ, et al. Tracking the genomic 47. Garg M, Nagata Y, Kanojia D, et al. Profiling of somatic mutations in
evolution of esophageal adenocarcinoma through neoadjuvant acute myeloid leukemia with FLT3-ITD at diagnosis and relapse. Blood.
chemotherapy. Cancer Discov. 2015;5:821-831. 2015;126:2491-2501.
25. Kim H, Zheng S, Amini SS, et al. Whole-genome and multisector exome 48. Landau DA, Tausch E, Taylor-Weiner AN, et al. Mutations driving CLL and
sequencing of primary and post-treatment glioblastoma reveals their evolution in progression and relapse. Nature. 2015;526:525-530.
patterns of tumor evolution. Genome Res. 2015;25:316-327. 49. Fisher R, Horswell S, Rowan A, et al. Development of synchronous VHL
26. Baca SC, Prandi D, Lawrence MS, et al. Punctuated evolution of syndrome tumors reveals contingencies and constraints to tumor
prostate cancer genomes. Cell. 2013;153:666-677. evolution. Genome Biol. 2014;15:433.
27. Stephens PJ, Greenman CD, Fu B, et al. Massive genomic rear- 50. Juric D, Castel P, Griffith M, et al. Convergent loss of PTEN leads to
rangement acquired in a single catastrophic event during cancer clinical resistance to a PI(3)Ka inhibitor. Nature. 2015;518:240-244.
development. Cell. 2011;144:27-40. 51. Gerlinger M, Horswell S, Larkin J, et al. Genomic architecture and
28. Magrangeas F, Avet-Loiseau H, Munshi NC, et al. Chromothripsis evolution of clear cell renal cell carcinomas defined by multiregion
identifies a rare and aggressive entity among newly diagnosed sequencing. Nat Genet. 2014;46:225-233.
multiple myeloma patients. Blood. 2011;118:675-678. 52. Hobor S, Van Emburgh BO, Crowley E, et al. TGFa and amphiregulin
29. Hong MK, Macintyre G, Wedge DC, et al. Tracking the origins and paracrine network promotes resistance to EGFR blockade in colorectal
drivers of subclonal metastatic expansion in prostate cancer. Nat cancer cells. Clin Cancer Res. 2014;20:6429-6438.
Commun. 2015;6:6605. 53. Liu J, Joha S, Idziorek T, et al. BCR-ABL mutants spread resistance to
30. Kemper K, Krijgsman O, Cornelissen-Steijger P, et al. Intra- and inter- non-mutated cells through a paracrine mechanism. Leukemia. 2008;
tumor heterogeneity in a vemurafenib-resistant melanoma patient 22:791-799.
and derived xenografts. EMBO Mol Med. 2015;7:1104-1118. 54. Zhang M, Tsimelzon A, Chang CH, et al. Intratumoral heterogeneity
31. Romanel A, Tandefelt DG, Conteduca V, et al. Plasma AR and in a Trp53-null mouse model of human breast cancer. Cancer Discov.
abiraterone-resistant prostate cancer. Sci Transl Med. 2015;7:312re10. 2015;5:520-533.
32. Diaz LA Jr, Williams RT, Wu J, et al. The molecular evolution of acquired 55. Inda MM, Bonavia R, Mukasa A, et al. Tumor heterogeneity is an active
resistance to targeted EGFR blockade in colorectal cancers. Nature. process maintained by a mutant EGFR-induced cytokine circuit in
2012;486:537-540. glioblastoma. Genes Dev. 2010;24:1731-1745.
33. Laurent-Puig P, Pekin D, Normand C, et al. Clinical relevance of KRAS- 56. Marusyk A, Tabassum DP, Altrock PM, et al. Non-cell-autonomous
mutated subclones detected with picodroplet digital PCR in advanced driving of tumour growth supports sub-clonal heterogeneity. Nature.
colorectal cancer treated with anti-EGFR therapy. Clin Cancer Res. 2014;514:54-58.
2015;21:1087-1097. 57. Cleary AS, Leonard TL, Gestl SA, et al. Tumour cell heterogeneity
34. Morrissy AS, Garzia L, Shih DJ, et al. Divergent clonal selection dominates maintained by cooperating subclones in Wnt-driven mammary can-
medulloblastoma at recurrence. Nature. 2016;529:351-357. cers. Nature. 2014;508:113-117.
35. McGranahan N, Favero F, de Bruin EC, et al. Clonal status of actionable 58. Gatenby RA, Silva AS, Gillies RJ, et al. Adaptive therapy. Cancer Res.
driver events and the timing of mutational processes in cancer 2009;69:4894-4903.
evolution. Sci Transl Med. 2015;7:283ra54. 59. Sun C, Wang L, Huang S, et al. Reversible and adaptive resistance to
36. Fearon ER, Vogelstein B. A genetic model for colorectal tumorigenesis. BRAF(V600E) inhibition in melanoma. Nature. 2014;508:118-122.
Cell. 1990;61:759-767. 60. Pao W, Miller VA, Politi KA, et al. Acquired resistance of lung ade-
37. Gerlinger M, McGranahan N, Dewhurst SM, et al. Cancer: evolution nocarcinomas to gefitinib or erlotinib is associated with a second
within a lifetime. Annu Rev Genet. 2014;48:215-236. mutation in the EGFR kinase domain. PLoS Med. 2005;2:e73.

e148 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


MANAGEMENT OF INTRATUMOR HETEROGENEITY

61. Cross DA, Ashton SE, Ghiorghiu S, et al. AZD9291, an irreversible EGFR 84. Ross-Innes CS, Becq J, Warren A, et al; Oesophageal Cancer Clinical
TKI, overcomes T790M-mediated resistance to EGFR inhibitors in lung and Molecular Stratification (OCCAMS) Study Group; Oesophageal
cancer. Cancer Discov. 2014;4:1046-1061. Cancer Clinical and Molecular Stratification OCCAMS Study Group.
62. Walter AO, Sjin RT, Haringsma HJ, et al. Discovery of a mutant- Whole-genome sequencing provides new insights into the clonal
selective covalent inhibitor of EGFR that overcomes T790M- architecture of Barretts esophagus and esophageal adenocarcinoma.
mediated resistance in NSCLC. Cancer Discov. 2013;3:1404-1415. Nat Genet. 2015;47:1038-1046.
63. Thress KS, Paweletz CP, Felip E, et al. Acquired EGFR C797S mutation 85. Stachler MD, Taylor-Weiner A, Peng S, et al. Paired exome analysis of
mediates resistance to AZD9291 in non-small cell lung cancer har- Barretts esophagus and adenocarcinoma. Nat Genet. 2015;47:
boring EGFR T790M. Nat Med. 2015;21:560-562. 1047-1055.
64. Piotrowska Z, Niederst MJ, Karlovich CA, et al. Heterogeneity underlies 86. Green MR, Kihira S, Liu CL, et al. Mutations in early follicular lym-
the emergence of EGFRT790 wild-type clones following treatment of phoma progenitors are associated with suppressed antigen pre-
T790M-positive cancers with a third-generation EGFR inhibitor. sentation. Proc Natl Acad Sci USA. 2015;112:E1116-E1125.
Cancer Discov. 2015;5:713-722. 87. Okosun J, Bodor C, Wang J, et al. Integrated genomic analysis
65. Kim MY, Oskarsson T, Acharyya S, et al. Tumor self-seeding by cir- identifies recurrent mutations and evolution patterns driving the
culating cancer cells. Cell. 2009;139:1315-1326. initiation and progression of follicular lymphoma. Nat Genet. 2014;
66. Waclaw B, Bozic I, Pittman ME, et al. A spatial model predicts that 46:176-181.
dispersal and cell turnover limit intratumour heterogeneity. Nature. 88. Sottoriva A, Spiteri I, Piccirillo SG, et al. Intratumor heterogeneity in
2015;525:261-264. human glioblastoma reflects cancer evolutionary dynamics. Proc Natl
67. Yap TA, Gerlinger M, Futreal PA, et al. Intratumor heterogeneity: Acad Sci USA. 2013;110:4009-4014.
seeing the wood for the trees. Sci Transl Med. 2012;4:127ps10. 89. Johnson BE, Mazor T, Hong C, et al. Mutational analysis reveals the
68. Voss MH, Hakimi AA, Pham CG, et al. Tumor genetic analyses of origin and therapy-driven evolution of recurrent glioma. Science.
patients with metastatic renal cell carcinoma and extended benefit 2014;343:189-193.
from mTOR inhibitor therapy. Clin Cancer Res. 2014;20:1955-1964. 90. Kim J, Lee IH, Cho HJ, et al. Spatiotemporal evolution of the primary
69. Wei EY, Hsieh JJ. A river model to map convergent cancer evolution glioblastoma genome. Cancer Cell. 2015;28:318-328.
and guide therapy in RCC. Nat Rev Urol. 2015;12:706-712. 91. Francis JM, Zhang CZ, Maire CL, et al. EGFR variant heterogeneity in
70. Shaw AT, Friboulet L, Leshchiner I, et al. Resensitization to Crizotinib by the glioblastoma resolved through single-nucleus sequencing. Cancer
Lorlatinib ALK Resistance Mutation L1198F. N Engl J Med. 2016;374:54-61. Discov. 2014;4:956-971.
71. Murtaza M, Dawson SJ, Pogrebniak K, et al. Multifocal clonal evolution 92. Shi H, Hugo W, Kong X, et al. Acquired resistance and clonal evolution
characterized using circulating tumour DNA in a case of metastatic in melanoma during BRAF inhibitor therapy. Cancer Discov. 2014;4:
breast cancer. Nat Commun. 2015;6:8760. 80-93.
72. Siravegna G, Mussolin B, Buscarino M, et al. Clonal evolution and 93. Sanborn JZ, Chung J, Purdom E, et al. Phylogenetic analyses of mel-
resistance to EGFR blockade in the blood of colorectal cancer patients. anoma reveal complex patterns of metastatic dissemination. Proc Natl
Nat Med. 2015;21:827. Acad Sci USA. 2015;112:10995-11000.
73. Russo M, Siravegna G, Blaszkowsky LS, et al. Tumor heterogeneity and 94. Keats JJ, Chesi M, Egan JB, et al. Clonal competition with alternating
lesion-specific response to targeted therapy in colorectal cancer. dominance in multiple myeloma. Blood. 2012;120:1067-1076.
Cancer Discov. 2016;6:147-153. 95. Magrangeas F, Avet-Loiseau H, Gouraud W, et al. Minor clone
74. Lohr JG, Adalsteinsson VA, Cibulskis K, et al. Whole-exome sequencing provides a reservoir for relapse in multiple myeloma. Leukemia. 2013;
of circulating tumor cells provides a window into metastatic prostate 27:473-481.
cancer. Nat Biotechnol. 2014;32:479-484. 96. Bolli N, Avet-Loiseau H, Wedge DC, et al. Heterogeneity of genomic
75. Murtaza M, Dawson SJ, Tsui DW, et al. Non-invasive analysis of ac- evolution and mutational profiles in multiple myeloma. Nat Commun.
quired resistance to cancer therapy by sequencing of plasma DNA. 2014;5:2997.
Nature. 2013;497:108-112. 97. Melchor L, Brioli A, Wardell CP, et al. Single-cell genetic analysis
76. Dawson SJ, Tsui DW, Murtaza M, et al. Analysis of circulating tumor DNA reveals the composition of initiating clones and phylogenetic patterns
to monitor metastatic breast cancer. N Engl J Med. 2013;368:1199-1209. of branching and parallel evolution in myeloma. Leukemia. 2014;28:
77. Crowley E, Di Nicolantonio F, Loupakis F, et al. Liquid biopsy: monitoring 1705-1715.
cancer-genetics in the blood. Nat Rev Clin Oncol. 2013;10:472-484. 98. de Bruin EC, McGranahan N, Mitter R, et al. Spatial and temporal
78. Krebs MG, Metcalf RL, Carter L, et al. Molecular analysis of circulating diversity in genomic instability processes defines lung cancer evo-
tumour cells-biology and biomarkers. Nat Rev Clin Oncol. 2014;11:129-144. lution. Science. 2014;346:251-256.
79. Jamal-Hanjani M, Hackshaw A, Ngai Y, et al. Tracking genomic cancer 99. Zhang J, Fujimoto J, Zhang J, et al. Intratumor heterogeneity in lo-
evolution for precision medicine: the lung TRACERx study. PLoS Biol. calized lung adenocarcinomas delineated by multiregion sequencing.
2014;12:e1001906. Science. 2014;346:256-259.
80. Welch JS, Ley TJ, Link DC, et al. The origin and evolution of mutations in 100. George J, Lim JS, Jang SJ, et al. Comprehensive genomic profiles of
acute myeloid leukemia. Cell. 2012;150:264-278. small cell lung cancer. Nature. 2015;524:47-53.
81. Paguirigan AL, Smith J, Meshinchi S, et al. Single-cell genotyping 101. Haffner MC, Mosbruger T, Esopi DM, et al. Tracking the clonal origin of
demonstrates complex clonal diversity in acute myeloid leukemia. Sci lethal prostate cancer. J Clin Invest. 2013;123:4918-4922.
Transl Med. 2015;7:281re2. 102. Brocks D, Assenov Y, Minner S, et al; ICGC Early Onset Prostate
82. Hughes AE, Magrini V, Demeter R, et al. Clonal architecture of sec- Cancer Project. Intratumor DNA methylation heterogeneity reflects
ondary acute myeloid leukemia defined by single-cell sequencing. clonal evolution in aggressive prostate cancer. Cell Reports. 2014;8:
PLoS Genet. 2014;10:e1004462. 798-806.
83. Yates LR, Gerstung M, Knappskog S, et al. Subclonal diversification of 103. Gundem G, Van Loo P, Kremeyer B, et al; ICGC Prostate UK Group. The
primary breast cancer revealed by multiregion sequencing. Nat Med. evolutionary history of lethal metastatic prostate cancer. Nature.
2015;21:751-759. 2015;520:353-357.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e149


DEVELOPMENTAL THERAPEUTICS AND
TRANSLATIONAL RESEARCH

Biosimilars: Here and Now

CHAIR
Steven J. Lemery, MD, MHS
U.S. Food and Drug Administration
Silver Spring, MD

SPEAKERS
Francisco J. Esteva, MD, PhD
New York University Clinical Cancer Center
New York, NY

Martina Weise, MD
Federal Institute for Drugs and Medical Devices
Bonn, Germany
BIOSIMILARS: HERE AND NOW

Biosimilars: Here and Now


Steven J. Lemery, MD, MHS, Francisco J. Esteva, MD, PhD, and Martina Weise, MD

OVERVIEW

Congress passed the Biologics Price Competition and Innovation Act (BPCI Act) as part of the Affordable Care Act on March
23, 2010. The BPCI Act authorized an approval pathway for biosimilar and interchangeable products. It defines biosimilarity
to mean that the biological product is highly similar to the reference product notwithstanding minor differences in clinically
inactive components and that there are no clinically meaningful differences between the biological product and the
reference product in terms of safety, purity, and potency of the product. The biosimilar pathway has the potential to
facilitate access to biologic products through increased competition, in the same manner as biosimilars have done for almost
10 years in Europe. The goal of a biosimilar program is not to independently establish safety and effectiveness for each
condition of use. Rather, the goal is to demonstrate biosimilarity through an extensive analytical characterization and a
targeted clinical program designed to assess for clinically meaningful differences, if they exist. The regulatory approaches in
both the United States and Europe involve a totality-of-the-evidence approach to demonstrate biosimilarity. Importantly,
the biosimilar pathway allows for extrapolation of data across indications so that a sponsor, with adequate scientific
justification, need not conduct clinical studies in each intended condition of use. Without extrapolation, development may
not be feasible for many products, and patients and resources could be diverted from clinical studies of newer agents for
cancer.

C ongress passed the BPCI Act on March 23, 2010, as part


of the Affordable Care Act. The BPCI Act created a new
abbreviated licensure pathway for biologic products that are
should not be considered formal or binding FDA or European
Medicine Agencies (EMA) advice on regulatory or policy
matters related to biosimilar drugs.
shown to be biosimilar to, or interchangeable with, a U.S. The goal of a biosimilar development program differs from
Food and Drug Administration (FDA)-licensed reference traditional drug development. The goal of a traditional drug
product. According to the BPCI Act, a reference product is development program is to demonstrate that the biologic
the single biologic product licensed (i.e., approved) by FDA product is safe and effective for one or more indications. The
that the proposed biosimilar is being compared with in an objective of a biosimilar development program is to dem-
application.1 The statutory definition requires the reference onstrate through a totality-of-the-evidence approach that
product to be licensed under Section 351(a) of the Public the biosimilar product is highly similar to the U.S.-licensed
Health Service Act. The reference product must be an orig- reference product, notwithstanding minor differences in
inator drug approved based on a full dossier of preclinical and clinically inactive components and that there are no clinically
clinical data, and not a biosimilar, which is licensed under meaningful differences between the biosimilar product and
Section 351(k) and not 351(a).1 the reference product in terms of safety, purity, and po-
The abbreviated licensure pathway for biosimilar and in- tency.2 A biosimilar sponsor does not need to independently
terchangeable products is possible because the proposed establish the safety and effectiveness of its product; it es-
product may rely for licensure on, among other things, publicly tablishes this through a demonstration of biosimilarity. The
available information on FDAs prior determination that the biosimilar pathway also allows for extrapolation of data
reference product is safe, pure, and potent. This article will across indications. With adequate scientific justification, a
discuss scientific considerations as understood by the authors sponsor would not need to conduct a clinical study to
regarding the development and approval of biosimilar ther- support approval of a biosimilar for each indication in the
apeutic biologic proteins (e.g., monoclonal antibodies or reference products label.
certain cytokines) in the United States. It also highlights the Although the biosimilar pathway is an abbreviated ap-
experience of biosimilars in Europe. As such, this article proval pathway, the data package required for approval of a

From the U.S. Food and Drug Administration, Silver Spring, MD; New York University Clinical Cancer Center, New York, NY; Federal Institute for Drugs and Medical Devices (BfArM), Bonn,
Germany.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Steven J. Lemery, U.S. Food and Drug Administration, 10903 New Hampshire Ave., Silver Spring, MD 20993; email: steven.lemery@fda.hhs.gov.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e151


LEMERY, ESTEVA, AND WEISE

biosimilar product can be quite extensive. Therefore, the route of administration. They also must be bioequivalent to
development of these products by sponsors and the review the reference listed drug.6
of these products by regulatory authorities, including FDA Unlike small molecule drugs, whose structure usually can be
and EMA, is quite thorough to ensure that the products are completely defined and reproduced, biologic products typically
highly similar and will be safe and effective. Ultimately, FDA are more complex and difficult to characterize. In addition, all
uses a totality-of-the-evidence approach to determine that a biologics display a certain degree of microheterogeneity be-
product is analytically highly similar to the approved ref- cause of the inherent variability of the biologic systems from
erence product and that there are no clinically meaningful which they are derived as well as their manufacturing pro-
differences between the biosimilar product and the refer- cesses.7 The reference product manufactured by an originator
ence product.2 Likewise, EMA considers a totality-of-the- may differ from batch to batch and may change over time
evidence approach when evaluating biosimilars. Under the because of modifications of the manufacturing process after
provisions of the BPCI Act, FDA licensed the first biosimilar licensing. As such, the biologic product that a patient receives
product, Zarxio (filgrastim-sndz), on March 6, 2015.3-5 in clinical practice is unlikely to be identical to the reference
Because oncologists prescribe biologic products, they need product administered to patients in the clinical study or studies
to familiarize themselves with biosimilar productshow they that supported approval. However, product quality is tightly
are approved and how they will be incorporated into clinical controlled within predefined acceptance ranges, and changes
practice. This article describes biosimilar products and their in the manufacturing processes are evaluated and approved by
approval process. Aspects on incorporation of the products the competent regulatory authority to ensure unchanged
into clinical practice may differ by state and are not covered in clinical performance of the product. Since 1996, FDA has
this article. approved many manufacturing process changes for refer-
Biosimilar products are not generic drug products. Fur- ence biologic products based on demonstration of product
thermore, the data package required to support the approval comparabilitybefore and after the process change(s)
of a biosimilar differs from a generic drug. Generic drugs are often without the need for additional nonclinical or clinical
copies of brand-name drugs. They contain the same active data. The EMA also has approved many such changes to
ingredient as the reference listed drug (e.g., the brand-name approved biologic products. These process changes can in-
drug) and are the same in terms of dosage form, strength, and clude changes in suppliers of cell culture media, new purifi-
cation methods, or new manufacturing sites. A 2013 report
from Dr. Schneider from the Danish Health and Medicines
Authority indicated that Remicade (infliximab) has undergone
KEY POINTS over 35 manufacturing process changes since approval and
MabThera (rituximab) has undergone at least five process
Approval in the United States requires a biosimilar changes.8
product to be highly similar to the reference product Both the biosimilar and reference product are essentially
notwithstanding minor differences in clinically inactive different versions of the same active drug substance.19 As
components and that there are no clinically such, scientific principles that underlie the comparability
meaningful differences between the biological product exercise for manufacturing process changes of the originator
and the reference product in terms of safety, purity, and product are also applicable to the analytical similarity exercise
potency of the product. to demonstrate biosimilarity.9 Although the scientific prin-
Although biosimilar products are not identical to the ciples are applicable to both exercises, additional steps to
reference product, the reference product also may differ
justify similarity are usually necessary in biosimilar develop-
from batch to batch and may change over time. As such,
the biologic product a patient receives in clinical practice
ment, either analytically or clinically, because of differences in
is unlikely to be identical to the reference product cell lines and manufacturing processes, among others. Ulti-
administered to patients in the clinical study or studies mately, the sponsors, including the originators, and regulators
that supported approval. need to understand how any differences in the structure of
Comparative clinical studies in biosimilar development the product may affect the function of the product. They also
programs should be directed toward an assessment of need to ensure that these differences are evaluated thor-
clinically meaningful differences, if they exist, and not oughly to ensure safety and effectiveness of the product.
directed toward independently establishing the safety
and effectiveness of the biosimilar product.
The BPCI Act allows for the potential, with adequate APPROACHES TO ASSESS ANALYTICAL
scientific justification, to extrapolate data to support a
demonstration of biosimilarity in more than one
SIMILARITY
indication.
In the United States, a determination of biosimilarity re-
Both the United States and the EMA use a totality-of- quires data demonstrating that the biologic product is highly
the-evidence approach when assessing whether a similar to the reference product, notwithstanding minor
product meets the standards to be designated as differences in clinically inactive components. The FDA gen-
biosimilar. erally expects that the biosimilar product will be composed of
the same amino acid sequence as the reference product.2

e152 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


BIOSIMILARS: HERE AND NOW

Because of the complexity of biologic products, FDA expects particular element is unnecessary in an application to
sponsors to characterize both their product and the reference support a demonstration of biosimilarity.
product. FDA also expects that this characterization will serve Comparative clinical study results (e.g., a clinical study
as the foundation for the demonstration of biosimilarity.2 powered to demonstrate similar pharmacokinetic profiles
Such characterization generally will include analyses of mul- or a clinical study to demonstrate a lack of clinically mean-
tiple attributes including, but not limited to, primary struc- ingful differences in safety and efficacy) cannot be used to
tures (e.g., amino acid sequences), higher order structures, support that a proposed biosimilar product is biosimilar to the
enzymatic post-translational modifications (e.g., glycosylation reference product when the proposed biosimilar product has
patterns), other variants (e.g., deamidation or charge vari- otherwise not been demonstrated to be highly similar
ants), and any other intentional modifications (e.g., addition to the reference product. Highly similar and no clinically
of PEGylation).2 meaningful differences are two separate standards, each of
Comparative functional assays provide additional data which needs to be met to support a demonstration of bio-
to support a determination that the biosimilar product similarity. Clinical data cannot be used to overcome relevant
is highly similar to the reference product. For example, differences in quality attributes.2,11
comparative functional assays may include binding assays, In general, FDA expects an application for a biosimilar
antibody-dependent cell-mediated cytotoxicity assays, and product to include data from a clinical pharmacology study
cell-based bioactivity assays to assess the potential for cy- demonstrating pharmacokinetic similarity between the
tokine release.2 Furthermore, a sponsor should consider other biosimilar product and the reference product.12 If one or
factors such as product expression systems, manufacturing more relevant pharmacodynamic markers exist, demon-
processes, impurities, and stability.2 stration of pharmacodynamic similarity may reduce residual
The growing number of analytical tools allows sponsors uncertainty and permit a more selective and targeted ap-
and regulatory agencies to better understand how attributes proach to additional clinical studies. Important elements
of a protein or biologic product can affect the function of that that FDA considers in the pharmacokinetic and/or pharma-
protein. As such, the comparative analytical characterization codynamic study include: (1) the appropriate (i.e., sensitive)
in a biosimilar program is quite extensive. population to detect differences, if they exist; (2) the appro-
A sponsor can submit analytical data to regulatory priate dose, which may differ from the approved dose and
agencies before clinical development. This allows regulatory ought to be on the steep part of the dose response curve; (3)
authorities to assess residual uncertainty regarding the de- route of administration; and (4) and the assays used to measure
termination of whether the proposed biosimilar product is pharmacokinetic and/or pharmacodynamic markers.12
biosimilar to the reference product, and what additional In some cases, a healthy volunteer population, if ethically
studies are needed to support the determination that the appropriate, may be the most appropriate and sensitive
product is biosimilar. Factors considered in the assessment of population rather than a patient population for the phar-
residual uncertainty may include: (1) which specific attributes macokinetic and/or pharmacodynamic similarity assessment
were tested, understanding that it is necessary to assess for because tumor burden may increase the variability in phar-
differences in any critical quality attributes; (2) the number of macokinetic measurements of certain biologic products. With
attributes tested (in a theoretical example, a more extensive justification, the dose and schedule of the biologic products
characterization with a fingerprint-like analysis10 could re- used in the study could be lower than the approved dose of
duce uncertainty); (3) the number of lots tested for both the the reference product to improve the safety margin and/or
proposed biosimilar product and the reference product; and sensitivity of such a study.
(4) what differences, if any, were observed between products The FDA recommends sponsors to pursue a step-wise
and what impact the differences could have on safety and approach to biosimilar development. For example, FDA
efficacy. can review the analytical data and clinical pharmacokinetic
and/or pharmacodynamic data and determine what residual
uncertainly exists before determining whether additional
CLINICAL STUDIES clinical studies are necessary to address the uncertainty and
As stated, the foundation of a biosimilar development support a demonstration of biosimilarity. If sponsors request
program is data demonstrating that the biosimilar product is FDA advice on the design of clinical studies before FDAs
analytically highly similar to the reference product. In ad- review of analytical data, which does not follow the FDA-
dition, the BPCI Act states that an application for a biosimilar recommended step-wise approach, FDA may assume residual
product must include, among other things, information from uncertainty exists. The FDA may recommend a more extensive
animal studies and a clinical study or studies (including the clinical program than otherwise might have been necessary.
assessment of immunogenicity and pharmacokinetics or Furthermore, the design of additional studies should be
pharmacodynamics) that are sufficient to demonstrate influenced by the type of residual uncertainty. For example,
safety, purity, and potency in one or more conditions of use as a scientific matter, an adequate assessment of immuno-
for which the reference product is licensed and for which genicity is expected in a biosimilar development program.2
licensure is sought for the biosimilar product.1 However, The nature of the data and the design of a study to generate
the BPCI Act provides FDA discretion to determine whether a comparative immunogenicity data may depend on specific

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e153


LEMERY, ESTEVA, AND WEISE

analytical characteristics of the biosimilar compared with the be safe and effective for the proposed indications. The FDA
reference product that could change the risk of an immune understands that because of chance, there may be differences
response (e.g., different excipients or other formulation between the study arms in the incidence of certain adverse
changes) as well as the publicly known risk of immunogenicity events in a comparative clinical study. Therefore, sponsors
(and risk of adverse events if immunogenicity occurs) asso- may discuss proposals with FDA to prioritize the collection and
ciated with the reference product. analysis of certain cardinal or key adverse events of interest.
The objective of a comparative clinical study in a biosimilar For example, anti-VEGF pathway antibodies increase the
development program is to support a demonstration of no risk of hypertension, epistaxis, proteinuria, and dysponia.
clinically meaningful differences between the biosimilar An analysis of these adverse events may be more relevant to
product and the reference product. In these comparative an assessment of no clinically meaningful differences for
clinical studies, this determination is not made by in- anti-VEGF pathway antibodies than analyses of fatigue or
dependently establishing the safety and effectiveness of the pain in a patient population with advanced cancer.
biosimilar product. As such, some endpoints used for the
approval of the reference product in the oncology setting
(e.g., overall survival or progression-free survival) may be less EXTRAPOLATION OF CLINICAL DATA ACROSS
informative. This is because they are not only influenced by INDICATIONS
the performance of the medicinal product but also by factors The FDA has essentially allowed extrapolation of data for
such as the general health status of the patient. Therefore, approved biologic products without additional clinical studies
they are insufficiently sensitive to detect product-specific for years following the introduction of certain manufacturing
differences between the reference product and the bio- process changes. Many biologic drugs used in oncology are
similar product, if they exist. In some cases, if scientifically approved for multiple indications, including nononcology
relevant, pharmacodynamic endpoints, such as objective re- indications. Based on the BPCI Act, if the proposed product
sponse rate, may be more appropriate. These endpoints meets the statutory requirements for licensure as a biosimilar
measure the pharmacological action of the biologic in a less product based on, among other things, data derived from a
confounded way and may be more sensitive to any po- clinical study sufficient to demonstrate safety, purity, and
tential differences in the biologic products. potency in an appropriate condition of use, the potential
If multiple indications are approved for the FDA-licensed exists for the biosimilar product to be licensed for one or more
reference product, and the biosimilar sponsor intends to additional conditions of use for which the reference product
seek licensure for more than one of these indications, FDA is licensed.13 A conclusion of biosimilarity in one or more
may provide advice regarding which indication is more indications potentially allows for extrapolation of data to
sensitive in the assessment of clinically meaningful differ- support a conclusion of biosimilarity for other indications.13
ences. Selection of a sensitive indication may allow a sponsor With adequate scientific justification, a sponsor would not
to request extrapolation of data to other indications without need to conduct a clinical study to support licensure of a
conducting additional clinical studies. biosimilar for each condition of use for which the reference
In general, FDA recommends that sponsors design com- product is licensed.
parative clinical studies to demonstrate that the proposed Sponsors need to provide scientific justification for such
biosimilar product has neither decreased nor increased ac- extrapolation. Justification should include a discussion of
tivity (e.g., efficacy) or has safety concerns (e.g., toxicity) that how the mechanism of action of the drug relates to each
differ from the reference product.2 Unless there is justifica- condition of use; whether pharmacokinetics and/or phar-
tion for a one-sided test, or other design, such a comparative macodynamics may differ in different patient populations;
clinical study generally uses a two-sided test based on a differences in expected toxicities in different patient pop-
prespecified similarity margin.2 The FDA will consider whether ulations; and any other factor that may affect the safety or
the proposed study duration will provide sufficient exposure efficacy in the different patient populations.2,13 It is important
to the proposed biosimilar and the reference product to to note that differences among indications or conditions of
assess for clinically meaningful differences, and whether the use do not preclude extrapolation or necessitate additional
study is adequately designed to assess for potential differ- clinical data. However, any differences need to be adequately
ences in immunogenicity.2 addressed with data and information to support extrapolation.
Because FDA uses a totality-of-the-evidence approach to Ultimately, sponsors should consider conducting clinical
support a demonstration of biosimilarity, it will consider studies that are adequately sensitive to detect clinically
whether observed differences in a clinical study between the meaningful differences between the biosimilar product and
proposed biosimilar and the reference product constitute the comparator product and that would best reduce any
clinically meaningful differences. A comparative evaluation residual uncertainty regarding extrapolation based on the
of multiple data points in a comparative clinical study (e.g., factors described above. For example, choosing to conduct
efficacy and cardinal-safety findings), combined with phar- the comparative clinical study in a relevant indication with a
macokinetic similarity and an extensive analytical comparison consistent and/or large treatment effect may have greater
demonstrating that the products are highly similar, will pro- sensitivity to detect differences between the products, if
vide solid ground to conclude that the biosimilar product will they exist. Also, choosing to conduct a comparative clinical

e154 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


BIOSIMILARS: HERE AND NOW

study in a relevant indication whose mechanism of action policies.16 Currently, none of the European Union (EU) coun-
overlaps with other indications being sought could reduce tries have authorized unrestricted automatic substitution.
the residual uncertainty regarding extrapolation. Some countries (e.g., Belgium, Italy, Norway, and the United
Extrapolation can reduce the time and cost of develop- Kingdom) prohibit pharmacy-level substitution, whereas other
ment by abrogating the need to conduct clinical studies for countries (e.g., France, Germany, and the Netherlands) allow
each indication previously approved for the reference product. substitution in certain instances. France permits substitution
Extrapolation is one of the tenets of the abbreviated approval of a biosimilar for the prescribed (reference) biologic in patients
pathway for biosimilar products. When FDA approves a bio- who are treatment nave.17 The Dutch Medicines Evaluation
similar product, it has determined that the product meets Board accepts substitution of biosimilars under the condition
the requirements for approval and has been demonstrated to that both the treating physician and the pharmacist are in-
have no clinically meaningful differences from the reference volved.18 In Germany, the pharmacist may only substitute a
product in terms of safety, purity, and potency, including any bioidentical product (i.e., a biosimilar from the same
indications that were supported by extrapolation. manufacturer).
In summary, extrapolation can be supported for a product Importantly, lack of designation as interchangeable does
that is highly similar to the U.S.-licensed reference product not imply that a biosimilar product is an inferior product
based on the totality of the evidence in which the biosimilar to a biosimilar that has also been determined to be in-
product would be expected to be safe and effective for each terchangeable with the reference product. By being bio-
indication of use, even without conducting a separate clinical similar, the biosimilar product has been determined to be
study in each indication. Without extrapolation, for products highly similar and has no clinically meaningful differences
with multiple indications, sponsors would likely need to from the reference product for the labeled conditions of use.
conduct comparative clinical studies with thousands to tens Nevertheless, a demonstration of interchangeability might
of thousands, or more, patients depending on the treatment require additional data (e.g., to evaluate the risk of switching
effect and the sensitivity of the population(s). Such a re- or alternating) to address the additional standards, and
quirement could have multiple consequences. These could sponsors need to make a business decision about whether to
include keeping development costs high and rendering the pursue a claim of interchangeability.
pathway infeasible and potentially diverting patients and
resources from clinical studies of newer therapies for cancer.
Despite these considerations, however, licensing of safe and DISCUSSION
effective drugs is the highest priority for regulators and Biosimilar products are an additional treatment option for
extrapolation is only allowed if a sponsor provides adequate health care providers and patients. They may help facilitate
scientific justification. access to treatment and decrease costs for patients and the
health care system in the United States, in the same manner
as these products have done for almost 10 years in Europe.
INTERCHANGEABILITY The FDA licensed the first biosimilar product, Zarxio (filgrastim-
The statutory provisions for interchangeability are unique to sndz), on March 6, 2015.3
the United States. According to the BPCI Act, for FDA to Health care providers in Europe have already gained sub-
determine that a product is interchangeable with a refer- stantial experience with safely prescribing biosimilar prod-
ence product, the product must be biosimilar to the ref- ucts. The legal pathway for authorizing biosimilar medicines
erence product and can be expected to produce the same was established in 2003, and the first biosimilar product,
clinical result in any given patient.14 Additionally, FDA must somatotropin, was approved in 2006.16,19,20 Approved bio-
determine that for a biological product that is administered similar products in Europe include hematopoietic growth
more than once to an individual, the risk in terms of safety or factors, such as filgrastim and epoetin, as well as insulin,
diminished efficacy of alternating or switching between use somatotropin, follitropin, and the monoclonal antibody
of the proposed interchangeable product and the reference infliximab.9,20 The uptake of biosimilars in the EU has in-
product is not greater than the risk of using the reference creased steadily but at different rates in different countries.
product without such alternation or switching.14 Similar to Germany has shown the highest uptake of approximately
U.S. generic drugs, an interchangeable biologic product may 50% volume.21 In certain European countries, adoption of
be substituted for the reference product without the in- such products has been widespread and thousands of pa-
tervention of the health care provider who prescribed the tients have been safely treated with biosimilar drugs.16,22-26
product.15 According to the BPCI Act, only a product des- Concerns about safety and efficacy have been voiced for
ignated as interchangeable may be substituted. Therefore, biosimilar filgrastim and epoetins, especially in extrapolated
the treating health care providerand not the pharmacist indications for which no clinical data with the biosimilar have
can decide whether to prescribe a biosimilar product not been generated.9 Epoetins were among the first biosimilars
designated as interchangeable at any time, including deciding approved in the EU. The major concern with epoetins is the
whether to change their patient from one product to another. rare development of neutralizing, cross-reacting anti-epoetin
Although most biosimilars in Europe are approved cen- antibodies that can cause pure red cell aplasia (PRCA).
trally by EMA, individual member states make substitution Product-related factors, such as aggregation and contaminants

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e155


LEMERY, ESTEVA, AND WEISE

(e.g., leachables from rubber stoppers or tungsten from sy- structural and functional assays using state-of-the art science.
ringes), have been implicated in the development of anti- In the United States, such comparative analytical testing must
epoetin antibodies.27,28 show that the biosimilar product is analytically highly similar
Although regulators and sponsors now better understand to the reference product. Although the specific language of
the factors that contribute to immunogenicity, as a scientific the regulations differs in Europe, the same scientific principles
matter, both FDA and EMA expect an adequate assessment apply with regard to the requirements for demonstrating
of immunogenicity as part of biosimilar development pro- analytical similarity.
grams. The EMA guideline on the development of biosimilar In addition to analytical testing, biosimilar applications in
epoetins requests clinical immunogenicity data with sub- the United States generally will contain comparative data
cutaneous use in patients with renal anemia and, therefore, from one or more clinical studies showing pharmacokinetic
with the route of administration and in the patient population similarity and, if necessary, an additional comparative clinical
at highest risk for developing PRCA.29 As such, immunogenicity study or studies to support a demonstration of no clinically
data from these patients can then be extrapolated to lower-risk meaningful differences. Data in humans further serve to
clinical settings, such as intravenous use or to immunocom- reduce the residual uncertainty on the clinical impact arising
promised patients who have chemotherapy-induced anemia. from any observed analytical differences and support a
The immunogenicity assessment of approved biosimilar demonstration of biosimilarity. Ultimately, FDA and EMA
epoetins in Europe includes both prelicensing studies and, determinations that a product is biosimilar based on a totality-
because of the rarity of PRCA, extensive postmarketing of-the-evidence approach to ensure that these products are
surveillance programs to gain a more precise estimate of its safe and effective for the labeled indications, including for the
frequency. Reassuringly, postmarketing studies and phar- treatment of patients with cancer. Such access will help to
macovigilance surveillance in Europe confirm efficacy and facilitate treatment of patients with cancer in both the United
safety of licensed biosimilars, lending further support to the States and Europe.
adequacy of the respective scientific guidelines and approval
processes.22-25
Biosimilar products are not generics. Requirements for ACKNOWLEDGMENT
approval of a biosimilar product differ in many ways from The authors would like to thank Leah Christl, PhD, Associate
generic drug products. Analytical testing for biosimilar drug Director for Therapeutic Biologics, FDA, for providing
products is much more extensive and encompasses numerous comments and edits to the manuscript.

References
1. Patient Protection and Affordable Care Act, Pub. L No. 111-148, Sections 10. U.S. Food and Drug Administration. Quality Considerations in Dem-
7002(a)(2) and (b)(3); Public Health Service Act, Pub. L No. 78-410, onstrating Biosimilarity of a Therapeutic Protein Product to a Reference
Sections 351(k), 351(i)(2), and 351(i)(4). Product: Guidance for Industry. http://www.fda.gov/downloads/
2. U.S. Food and Drug Administration. Scientific Considerations in drugs/guidancecomplianceregulatoryinformation/guidances/
Demonstrating Biosimilarity to a Reference Product: Guidance for In- ucm291134.pdf. Accessed March 17, 2016.
dustry. http://www.fda.gov/downloads/DrugsGuidanceCompliance 11. European Medicines Agency. Guideline on Similar Biological Medicinal
RegulatoryInformation/Guidances/UCM291128.pdf. Accessed March 17, Products. http://www.ema.europa.eu/docs/en_GB/document_library/
2016. Scientific_guideline/2014/10/WC500176768.pdf. Accessed January 29,
3. U.S. Food and Drug Administration. BLA Approval Letter. http://www. 2016.
accessdata.fda.gov/drugsatfda_docs/appletter/2015/ 12. U.S. Food and Drug Administration. Guidance for Industry: Clinical
125553Orig1s000ltr.pdf. Accessed January 7, 2016. Pharmacology Data to Support a Demonstration of Biosimilarity to a
4. U.S. Food and Drug Administration. FDA Approves First Biosimilar Product Reference Product, Draft Guidance. http://www.fda.gov/downloads/
Zarxio. http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ drugs/guidancecomplianceregulatoryinformation/guidances/
ucm436648.htm. Accessed January 29, 2016. ucm397017.pdf.
5. Christl LD, Deisseroth A. Development and Approval of Biosimilar 13. U.S. Food and Drug Administration. Guidance for Industry on Bio-
Products. The ASCO Post. March 25, 2015;6. similars: Q & As Regarding Implementation of the BPCI Act of 2009:
6. U.S. Food and Drug Administration. Abbreviated New Drug Appli- Questions and Answers Part I. http://www.fda.gov/Drugs/Guidance
cation (ANDA): Generics. http://www.fda.gov/Drugs/DevelopmentApproval ComplianceRegulatoryInformation/Guidances/ucm259809.htm.
Process/HowDrugsareDevelopedandApproved/ApprovalApplications/ Accessed January 8, 2016.
AbbreviatedNewDrugApplicationANDAGenerics/. Accessed January 8, 2016. 14. U.S. Food and Drug Administration. Information for Industry (Bio-
7. Weise M, Bielsky MC, De Smet K, et al. Biosimilars: what clinicians similars). http://www.fda.gov/Drugs/DevelopmentApprovalProcess/
should know. Blood. 2012;120:5111-5117. HowDrugsareDevelopedandApproved/ApprovalApplications/Therapeutic
8. Schneider CK. Biosimilars in rheumatology: the wind of change. Ann BiologicApplications/Biosimilars/ucm241720.htm. Accessed January 8,
Rheum Dis. 2013;72:315-318. 2016.
9. Weise M, Kurki P, Wolff-Holz E, et al. Biosimilars: the science of ex- 15. U.S. Food and Drug Administration. Information for Consumers (Bio-
trapolation. Blood. 2014;124:3191-3196. similars). http://www.fda.gov/Drugs/DevelopmentApprovalProcess/

e156 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


BIOSIMILARS: HERE AND NOW

HowDrugsareDevelopedandApproved/ApprovalApplications/Therapeutic HaEmatology and Oncology: results of the ORHEO observational study.


BiologicApplications/Biosimilars/ucm241718.htm. Accessed January 8, BMC Cancer. 2014;14:503.
2016. 23. Dellanna F, Fluck RJ, Lonnemann G, et al. Results from a safety cohort of
16. Renwick MJ SK, Smolina K, Gladstone EJ, et al. Postmarket policy patients with renal anemia receiving the biosimilar epoetinzeta: The
considerations for biosimilar oncology drugs. Lancet Oncol. 2016;17: PASCO I study. Clin Nephrol. 2015;84:280-288.
e31-e38. 24. Horbrand F, Bramlage P, Fischaleck J, et al. A population-based study
17. Generics and Biosimilars Initiative. France to Allow Biosimilars Sub- comparing biosimilar versus originator erythropoiesis-stimulating agent
stitution. http://gabionline.net/Policies-Legislation/France-to-allow- consumption in 6,117 patients with renal anaemia. Eur J Clin Pharmacol.
biosimilars-substitution. Accessed January 29, 2016. 2013;69:929-936.
18. Medicines Evaluation Board. MEB Stance on Prescribing Biosimilars. http:// 25. Schmitt M, Publicover A, Orchard KH, et al. Biosimilar G-CSF based
english.cbg-meb.nl/latest/news/2015/03/31/meb-stance-on-prescribing-% mobilization of peripheral blood hematopoietic stem cells for autologous
E2%80%9Cbiosimilars%E2%80%9D. Accessed January 29, 2016. and allogeneic stem cell transplantation. Theranostics. 2014;4:280-289.
19. European Medicines Agency. Questions and Answers on Biosimilar 26. Gascon P, Aapro M, Ludwig H, et al. Treatment patterns and outcomes
Medicines (Similar Biological Medicinal Products). http://www.ema. in the prophylaxis of chemotherapy-induced (febrile) neutropenia with
europa.eu/docs/en_GB/document_library/Medicine_QA/2009/12/ biosimilar filgrastim (the MONITOR-GCSF study). Support Care Cancer.
WC500020062.pdf. Accessed January 8, 2016. 2016;24:911-925.
20. European Medicines Agency. European Public Assessment Reports. 27. Boven K, Stryker S, Knight J, et al. The increased incidence of pure red
http://www.ema.europa.eu/ema/index.jsp?curl=pages%2Fmedicines%2 cell aplasia with an Eprex formulation in uncoated rubber stopper
Flanding%2Fepar_search.jsp&mid=WC0b01ac058001d124&searchTab= syringes. Kidney Int. 2005;67:2346-2353.
searchByAuthType&alreadyLoaded=true&isNewQuery=true&status= 28. Seidl A, Hainzl O, Richter M, et al. Tungsten-induced denaturation and
Authorised&keyword=Enter+keywords&searchType=name&taxonomy aggregation of epoetin alfa during primary packaging as a cause of
Path=&treeNumber=&searchGenericType=biosimilars&generics immunogenicity. Pharm Res. 2012;29:1454-1467.
KeywordSearch=Submit. Accessed January 8, 2016. 29. European Medicines Agency. Committee for Medicinal Products for
21. Generics and Biosimilars Initiative. Use of Biosimilars in Europe Differs Human Use: Guideline on non-clinical and clinical development of
Across Countries. http://gabi-journal.net/news/use-of-biosimilars-in- similar biological medicinal products containing recombinant eryth-
europe-differs-across-countries. Accessed January 29, 2016. ropoietins (revision). http://www.ema.europa.eu/docs/en_GB/document_
22. Michallet M, Luporsi E, Soubeyran P, et al; ORHEO study group. BiO- library/Scientific_guideline/2010/04/WC500089474.pdf. Accessed January
similaRs in the management of anaemia secondary to chemotherapy in 29, 2016.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e157


DEVELOPMENTAL THERAPEUTICS AND
TRANSLATIONAL RESEARCH

Clinical Trial Eligibility Criteria:


Tradition Versus Reality

CHAIR
Edward S. Kim, MD, FACP
Levine Cancer Institute, Carolinas HealthCare System
Charlotte, NC

SPEAKERS
Gwynn Ison, MD
U.S. Food and Drug Administration
College Park, MD

Jeffrey R. Infante, MD
Sarah Cannon Research Institute
Nashville, TN
PRACTICAL VERSUS TRADITIONAL ELIGIBILITY CRITERIA

Transforming Clinical Trial Eligibility Criteria to Reflect


Practical Clinical Application
Edward S. Kim, MD, FACP, Jennifer Atlas, MD, Gwynn Ison, MD, and Jennifer L. Ersek, MSPH

OVERVIEW

Historically, oncology clinical trials have focused on comparing a new drugs efficacy to the standard of care. However, as our
understanding of molecular pathways in oncology has evolved, so has our ability to predict how patients will respond to a
particular drug, and thus comparison with a standard therapy has become less important. Biomarkers and corresponding
diagnostic testing are becoming more and more important to drug development but also limit the type of patient who may
benefit from the therapy. Newer clinical trial designs have been developed to assess clinically meaningful endpoints in
biomarker-enriched populations, and the number of modern, molecularly driven clinical trials are steadily increasing. At the
same time, barriers to clinical trial enrollment have also grown. Many barriers contribute to nonenrollment in clinical trials,
including patient, physician, institution, protocol, and regulatory barriers. At the protocol level, eligibility criteria have
become a large roadblock to clinical trial accrual. Over time, eligibility criteria have become more and more restrictive. To
accrue an adequate number of patients to molecularly driven trials, we should consider eligibility criteria carefully and
attempt to reduce restrictive criteria. Reducing restrictive eligibility criteria will allow more patients to be eligible for clinical
trial participation, will likely increase the speed of drug approvals, and will result in clinical trial results that more accurately
reflect treatment of the population in the clinical setting.

C linical trials and their availability for treatment of pa-


tients serve as a longstanding foundation in the prac-
tice of medicine and drug development. Through all phases
could be relaxed. Clinical models and structures that support
therapeutic development are not keeping pace with scientific
innovations.1 We must carefully evaluate the evolution of
of study (I-IV), the risk-benefit ratio has always encompassed clinical trial design and the corresponding eligibility criteria
assessing safety as well as offering opportunity for treatment based on molecular targets and drugs and, in turn, implement
benefit. In the past, the primary focus of early-phase studies these into the conduct of future clinical trials to successfully
was defining safe doses, and late-phase studies served as the incorporate the use of novel therapeutics into oncologic care
final hurdle to regulatory approval. Initially, with only lim- with results that are generalizable to the greater population.
ited treatments available, therapies were tested against best
supportive care. As more therapies were approved, combi-
nation therapies were compared with historical standards,
IMPACT OF BIOMARKERS AND SURROGATE
ENDPOINTS ON CLINICAL TRIALS
leading to the desire for homogenous patient populations.
Molecular medicine is quickly changing the landscape of
Although homogenous populations allow for direct compar- medical oncology. The era of molecular medicine is lead-
ison of study results, they also reduce the number of patients ing to drug approval at an unprecedented rate in oncology,
eligible to participate in the study. From the perspective of with more than 40 drug approvals based on specific tumor
study investigators and sponsors, there may be an interest in biomarkers in the past decade.2 These drugs add to the
keeping study populations tight and homogenous to allow for growing armamentarium of treatment options available to
comparison with standard therapies; however, increasingly patients with cancer. However, it is becoming apparent
strict eligibility criteria lead to increased difficulty in accrual. that phenotypic characteristics are limited in their ability to
With modern clinical trials incorporating biomarkers and predict response. More recently, biomarkers are included as
targeted therapies, there is less need to compare the results critical components in modern clinical trials involving tar-
of these trials to standard therapies, and thus trial eligibility geted agents, shifting the priority to identifying targets

From the Department of Medicine, Hematology and Oncology, Wake Forest School of Medicine, Winston-Salem, NC; Office of Hematology and Oncology Products, Center for Drug
Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD; Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Edward S. Kim, MD, FACP, Solid Tumor Oncology and Investigational Therapeutics, Levine Cancer Institute, Carolinas HealthCare Stystem, 1021 Morehead
Medical Dr., Suite 3100, Charlotte, NC 28204; email: edward.kim@carolinashealthcare.org.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 83


KIM ET AL

for therapeutic development alongside drug development. acceptable toxicity if the drug is found to have clinical ef-
Consideration also must be given to appropriate choice of ficacy. The most important endpoint traditionally in phase I
primary endpoints. Although overall survival remains the trials has been identifying dose-limiting toxicity (DLT). His-
gold standard, progression-free survival, tumor response, torically, phase I trials have not restricted patient eligibility
and duration of tumor response may serve as surrogate on the basis of diagnosis or molecular phenotype; however,
endpoints in early-phase studies and may allow for early this is changing.
approval. Interpretation of emerging data from trials is From a regulatory standpoint, many deficiencies sent to
hindered by a lack of specific reporting standards for bio- sponsors of investigational new drug applications (INDs) do
markers.3 Stronger evidence-based outcomes incorporating not pertain to the eligibility criteria, provided that the first-
biomarkers could be achieved by implementing standard- in-human study is conducted in an appropriately advanced
ized biomarker-reporting systems with molecular selection cancer patient population, but pertain to the definition of
criteria guided by the mutation gene at the variant level.3 DLTs. The stringency applied to the DLT definition stems
As an example, listing specific variant mutations (e.g., del19, from the interpretability of the study results, as potential
T790) instead of listing a mutation generically as an EGFR signals of toxicity should be captured as early in develop-
mutation would be more descriptive. ment as possible. Unfortunately, the inflexible nature of the
commonly used 3+3 design of first-in-human trials may lead
to the premature discontinuation of dose escalation because
GOAL OF CLINICAL TRIALS of toxicities that are either unrelated to the drug or are not
Clinical trials are fundamental to understanding the risks and dose-dependent. To minimize the potential for premature
benefits of treatment in a specific intended population. discontinuation of dose escalation, strict eligibility criteria
Clinical trials provide a formalized process of integrating regarding organ function, life expectancy, comorbidities,
scientific findings into clinical practice in a safe and system- performance status, HIV status, and laboratory parameters
atic manner. With increasing response rates noted in many are instituted.
molecularly driven trials, small incremental gains may no In phase II clinical trials, the primary objective is to de-
longer be adequate for drug approval. There needs to be a termine whether the therapy is effective. The primary
focus on utilizing molecular medicine to achieve clinically endpoints in these studies have historically been tumor
meaningful endpoints. response and survival. Because of inconsistent factors, in-
From a regulatory perspective, the U.S. Food and Drug cluding eligibility criteria and protocol design, phase II trials
Administrations (FDA) primary objective in evaluating trial can be difficult to interpret in comparison with one another.
eligibility criteria, from a first-in-human trial to a trial in- Historically, phase III clinical trials have evaluated an ex-
tended to support a marketing application, is the safety of perimental therapy in comparison with standard-of-care
the patient.4 Eligibility criteria that encourage clinical trial therapy. Randomized clinical trials remain the gold stan-
accrual, permit patient access to investigational drugs, and dard. Following trials in these phases, sponsors present data
create clinical trial results that are generalizable to the from their studies and file applications for approval. Federal
broader U.S. population are directly in line with FDAs regulations govern aspects of IND applications and New
mission. Drug Applications and Biologic License Applications. 5,6
Although there are no specific regulations regarding eligi-
TRADITIONAL CLINICAL TRIAL ELIGIBILITY bility criteria, deciding whom to include or exclude from a
CRITERIA AND DESIGN trial may determine the difference between a successful trial
Traditional clinical trials have been categorized on the basis and a failed one.
of phases of drug development. In phase I clinical trials, a Phase IV clinical trials, often referred to as postmarketing
small number of patients are enrolled, and the focus is on surveillance trials, are completed after a drug has received
safety. The goal of phase I clinical trials is primarily to es- approval. Common reasons for conducting these trials in-
tablish the maximum tolerated dose that would have clude monitoring a drugs long-term efficacy, observing
the long-term toxicities and impact on quality of life, and
comparisons between a novel drug and other therapies that
are available on the market. The strict criteria developed
KEY POINTS in the early-phase setting are not relaxed for later-phase
studies, as there is a legitimate interest to avoid confounding
Discuss the impact of molecular medicine and of the interpretation of the results of trials by factors related
biomarkers in oncology clinical trials.
to the disease condition, rather than the therapeutic product.
Review traditional versus modern clinical trial design.
Review of barriers to protocol enrollment.
The unintended consequence of this approach is that the
Discuss issues related to traditional versus modern results of these studies may not be applicable to the patients
clinical trial eligibility. who eventually will receive the drug in the postmarketing
Discuss regulatory issues related to clinical trial setting. Although postmarketing studies may capture some
eligibility. of this information, they are not systematically per-
formed, resulting in off-label use without appropriate safety

84 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


PRACTICAL VERSUS TRADITIONAL ELIGIBILITY CRITERIA

information. Studying subgroups with broader eligibility validity and integrity of the trial. Adaptive clinical trial
earlier in development may provide information that is more designs are becoming more and more popular, as statisti-
useful to prescribers and patients in the postapproval set- cians can preplan adjustments to the study design on the
ting, where comorbidities are more frequent, and may aid in basis of data from the study itself. Multiple variations of
making the benefit-risk calculation for a new agent with the adaptive design exist, and combinations can be used
more relevant data. simultaneously (multiple adaptive design). In the phase I
setting, adaptive dose-finding designs are used to establish
minimum and maximum dose levels and may be used in
MODERN/PRACTICAL CLINICAL TRIALS conjunction with the group sequential design. The group
Modern trial designs, such as adaptive, umbrella, and basket
sequential design allows preplanned examination of the
trials, have allowed the FDA to develop and implement
study data to determine if the trial can be stopped early
accelerated approval programs in some cases, based on
because of efficacy or futility. In clinical trials involving the
surrogate endpoints. A growing number of targeted ther-
use of biomarkers, biomarker-adaptive designs are used to
apies have been adopted into the FDA programs that assist
allow for changes to the study design on the basis of bio-
with the accelerated development and approval of prom-
marker response.15,16
ising drugs (Fast Track designation, Breakthrough Therapy
Phase I trials have increasingly been restricted to specific
designation, and Priority Review designation).7 The FDA
tumor types known to harbor molecular targets or to tu-
granted accelerated approval to 35 oncology products for
mors that demonstrate the presence of a specific molecular
47 new indications from December 11, 1992, to July 1, 2010,
target.17 The use of contemporary designs in dose esca-
with clinical benefit confirmed in postmarketing trials for
lation studies that allow for more flexibility with regard to
26 of the 47 new indications, with subsequent conversion to
dose selection could permit the broadening of the eligi-
regular approval.8 The FDA has given breakthrough therapy
bility criteria. An adaptive design that allows for dose es-
designation and accelerated approval to several targeted
calation, dose de-escalation, and dose re-escalation may
drugs to be used in cancer therapy in an effort to speed
filter out potential signals of toxicity stemming from the drug
up the availability of promising drugs based on surrogate
product, as opposed to the disease process or underlying
endpoints felt to be reasonably likely to predict clinical
comorbidities.
benefit in diseases with unmet medical needs or drugs that
have demonstrated a substantial improvement over current
available therapy. The median time between accelerated Umbrella Trials
approval and regular approval of oncology products was Umbrella trials involve the testing of a variety of drugs tar-
3.9 years (range, 0.8-12.6 years), and the mean time was geting different mutations in a single cancer subtype. They
4.7 years.8 Reduction in approval time clearly shows the allow a large number of patients to be screened for multiple
benefit of these FDA accelerated approval programs, with biomarkers, which is particularly beneficial for low-prevalence
the implication that appropriate postmarketing trials should markers. These trials involve a group of two or more en-
be completed to confirm efficacy results. Examples of tar- richment designs, connected through a central infrastructure
geted therapies developed in conjunction with biomarkers that oversees screening and identification of patients. The
that received approval under expedited programs include rationale for the umbrella trial design is to facilitate screening
combination trametinib and dabrafenib for unresectable and accrual to trials, with the goal of identifying a patient
or metastatic melanoma with BRAF V600E or V600K mu- population with the best chance for improved outcomes
tations,9 osimertinib for metastatic EGFR T790M mutation- with therapy. The BATTLE trial (Biomarker-Based Approaches
positive nonsmall cell lung cancer (NSCLC),10 and erlotinib of Targeted Therapy for Lung Cancer Elimination) in 2005 is
and gefitinib for NSCLC with EGFR exon 19 deletions or an example of an umbrella trial. In the study, patients with
exon 21 (L858R) substitution mutations.11,12 There continues previously diagnosed and treated lung cancer underwent
to be a growing interest in developing tumor biomarkers to repeat biopsies to assess biomarkers, which were used to
assess the effectiveness of therapy, especially in relation to select treatment recommendations with one of several bi-
targeted therapy, and in using surrogate endpoints as a basis ologic agents.16 Other examples of umbrella trials include
for accelerated approval by the FDA. We must continue to ALCHEMIST, Lung-MAP, and I-SPY 2.18-20
strive to overcome the design and regulatory barriers that
exist so as not to impede approval of promising drugs.
Basket Trials
Basket trials are genotype-focused designs involving the
Adaptive Trials testing of a single drug on a specific mutation or mutations
In 2004, the FDA published a critical path initiative docu- in a variety of cancer types. Therefore, these trials have the
ment highlighting the need for new, innovative clinical trial ability to include rare cancers that would be difficult to
designs as well as integration of biomarkers.13 Adaptive study in randomized controlled trials.21 The basket trial
designs, as defined by Vladimir Dragalin,14 are a multistage approach allows flexibility to continually open and close
study design that uses accumulating data to decide how treatment arms, which makes it possible to screen the
to modify aspects of the study without undermining the impact of many drugs over several different tumors under

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 85


KIM ET AL

one study. Patients are matched to the drug based solely on There are several obstacles to trial enrollment associated
the genetic abnormality, not on the type of cancer. One with patient characteristics. Because of the more extensive
example of a basket trial is vemurafenib in V600-mutated need for tissue and blood samples in molecularly driven
nonmelanomas.22 In this trial, patients were enrolled into studies, some patients decline clinical trial enrollment because
prespecified cohorts of multiple tumor types. Cohorts were of the fear that additional testing will delay the initiation of
closed or combined for tumor types with low accrual. Re- critical antineoplastic therapy or out of fear of undergoing
sponse rate and progression-free survival were evaluated, additional procedures. Socioeconomic issues, including dis-
and the authors concluded that vemurafenib is active in tance from centers with trials and frequency of follow-up
some, but not all, tumor types. The National Cancer In- visits, may influence the populations enrolled on protocols.
stitutes (NCI) MATCH is a basket/master trial, meaning that Patients who lack insurance may present with more advanced
new drugs can be added to the trial at any time. In 2015 the stages of cancer and symptomatic disease, precluding trial
NCI launched the NCI-MATCH trial (Molecular Analysis for enrollment. Because of socioeconomic differences, there are
Therapy Choice), with plans to screen up to 3,000 patients, subsets of patients with poor enrollment on clinical trials due
with an enrollment of at least 1,000 individuals to a targeted to lack of understanding of the rationale for the clinical studies
drug combination and independent of tumor histology in and/or mistrust of physicians. Christian et al27 reported that
patients with advanced solid tumors (gastrointestinal stro- although the enrollment of minority patients on clinical trials in
mal tumor, NSCLC, breast, gastric, melanoma, and thyroid) the United States has remained relatively stable, because the
and lymphomas.23 The primary objective of this trial is to overall number of individuals enrolled in trials has increased
determine if the efficacy of targeted therapy is better than over time, the minority percentage has therefore decreased. A
that of standard therapy. Another example of the basket trial decreased number of minority patients represented in clinical
approach includes the NCI M-PACT trial (Molecular Profiling- trials could lead to potentially important biologic differences
Based Assignment of Cancer Therapy), which will randomly being overlooked.
assign patients with a known mutation in a specific genetic Physician-driven barriers to protocol enrollment exist as
pathway to either pathway-driven targeted therapies or a well. These include the complexity of protocol design and
treatment not known to be pathway-specific.24 These types eligibility criteria, lack of awareness of an available protocol,
of trials will help answer questions about what types of absence of an available protocol for an individual patient, or
patients, tumors, or aberrations have the best outcomes lack of consideration for clinical trial enrollment of patients
with molecularly targeted therapy compared with standard with decreased performance status. Sohal et al28 reported
chemotherapy.25 that the lack of clinical trial access and a decline in functional
status or death were the most commonly cited reasons
patients were not enrolled on molecularly driven protocols
Registry Trials
by their physicians. The inability of physicians to keep up to
Clinical studies are investigational in nature, whereas the
date with the research literature of molecular compounds
focus of registry studies is on observation. Registry studies
and markers is overwhelming and will limit discussions with
allow observational conclusions to be drawn from the
patients about clinical trials.
evaluation of multiple drugs directed at multiple biologic
Additionally, there are funding and regulatory barriers to
targets in several cancers. As an example, the American
clinical trial enrollment that directly affect the protocol-
Society of Clinical Oncologys (ASCOs) TAPUR (Targeted
level barriers. With the evolution of trial design and safety
Agent and Profiling Utilization Registry Study) will enroll
regulations, there is a growing number of mandated re-
patients with advanced solid tumors, B-cell non-Hodgkin
quirements in an effort to standardize protocols. Typically,
lymphoma, and multiple myeloma from three health care
criteria mandated for exclusion of certain patients perceived
systems, including the Carolinas HealthCare Systems Levine
as being potentially at higher risk to receive investigational
Cancer Institute, with the goal of matching tumor profiles
therapy. These patients at higher risk may be defined as
with available agents. A molecular tumor board will review
having a prior history of malignancy, active brain metas-
the proposed drug-target match and report to the physician
tases, suboptimal hepatic or renal function, or HIV positivity.
on potential treatments either on or off the study. Safety and
However, as the field becomes more comfortable with early
efficacy outcomes will be recorded; however, the primary
integration of available interdisciplinary data during the
endpoint is objective response rate.26
course of drug development,1 the FDAs Office of Hematology
and Oncology Products would encourage sponsors to broaden
BARRIERS TO CLINICAL TRIAL ENROLLMENT these criteria, allowing for the best examination of real world
Many barriers to clinical trial enrollment exist and are experience as well as providing patients with life-threatening
noted in the literature. These can be broken into several diseases access to investigational agents. From a funding
broad categories: patient-level barriers (including socio- perspective, increasing complexity of trials and eligibility
demographic and disease-related barriers), physician-level criteria has led to increased costs to conduct such studies.
barriers, institutional-level barriers, and regulatory and With decreasing funding available, studies must be carefully
protocol-level barriers, including restrictive eligibility designed, as meaningful clinical benefits will likely be ex-
criteria. amined more closely in the future.

86 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


PRACTICAL VERSUS TRADITIONAL ELIGIBILITY CRITERIA

IMPORTANCE OF CLINICAL TRIAL ELIGIBILITY effect profiles are quite different. As more clinical in-
Although eligibility criteria are a necessary aspect of clinical formation about the toxicity profile of a particular drug is
protocols, their purpose is to define the characteristics of obtained, the eligibility criteria for subsequent trials may be
the intended patient population to receive the study drug. tailored to account for this information. For example, if it is
However, as trial designs adapt to incorporate molecular observed that an agent causes hepatic toxicity during early
medicine, so must eligibility criteria. From a design per- development, then the eligibility criteria for the subsequent
spective, restrictions on eligibility are effective only if the trials may be amended either to include more intensive
restrictions lead to increased statistical power of the study.29 evaluation of hepatic injury and function and/or to exclude
In clinical practice, eligibility criteria employed on clinical patients with a certain degree of baseline hepatic impair-
trials are rarely adhered to. ment. Dedicated studies in patients with hepatic dysfunc-
Traditionally, we have relied on the decision-making ca- tion would also be informative, especially in diseases that
pacity of the individual physician, with consent of the pa- have a propensity to metastasize to the liver. However,
tient, to determine eligibility for trial enrollment; however, other criteria, such as a minimum age requirement or comor-
the paradigm has shifted over time so that all aspects of bidities (e.g., underlying HIV positivity), could be relaxed or
the trial, including determination of patient eligibility, dose amended to allow for less stringent requirements as the de-
modifications, and allowed supportive measures, are ex- velopment progresses into later phases.17 Safety concerns are a
plicitly included in the protocol.29 Reevaluation of clinical commonly cited cause for restrictive eligibility criteria. If a drug
trial eligibility criteria, especially those related to the sci- has a toxicity profile known to be harmful in a particular subset
entific objective and generalizability of the results to of patients, then these patients should be excluded. One ex-
other populations, throughout the entire drug development ample of this would be giving bevacizumab to patients with lung
process should be considered, while keeping the safety of cancer with squamous cell carcinoma histology. Ideally, safety
the patient at the forefront. criteria should be limited to those in which no medical judgment
is allowed. Increased complexity of protocol design and in-
structions on dose modifications do not replace a clinicians
RESTRICTIVE ELIGIBILITY CRITERIA: A judgment concerning an individual patients suitability for
ROADBLOCK TO TRIAL ENROLLMENT clinical trial enrollment.
Although eligibility criteria remain an essential component Patients with brain metastases are a patient subset
of clinical trials, the growing number of eligibility criteria and typically excluded from clinical trials. Over time, the
complexity of trial designs has increasingly become a barrier number of patients with brain metastases are likely to
to trial enrollment. Eligibility criteria have generally been increase in frequency, based on the natural history of
categorized as inclusion (e.g., must have a specific molecular many tumor types and the sites of frequent brain me-
aberration) versus exclusion criteria (e.g., evidence of brain tastases. In addition, as adjuvant chemotherapy continues
metastases). We assessed barriers to enrollment on trials, to improve outcomes, it is likely that we will continue to
including patient-related barriers, physician-driven barriers, see more disease relapses in the brain given the limited
and funding and regulatory barriers.17 Protocol eligibility number of antineoplastic therapies that can penetrate the
criteria represent a modifiable barrier to increasing clinical blood-brain barrier. Carden et al30 advocate that we
trial availability to a larger subset of patients. Table 1 lists a should rethink exclusion of patients with asymptomatic
comparison of traditional versus potential modern clinical brain metastases from clinical trials. Because of the de-
trial eligibility criteria. velopment of more-sensitive brain-imaging techniques,
Certain diagnostic restrictions are required to ensure that metastatic brain lesions are often detected earlier and
patients enrolled on clinical protocols involving targeted may be asymptomatic at the time of detection. In many
drugs would have a reasonable chance of clinical benefit. trials, brain imaging is a part of the screening workup for
Protocol barriers, such as tissue availability and specific trial eligibility. Traditionally, patients with brain metas-
laboratory testing, are increasingly frequent requirements of tases have been excluded from trial participation because
trials involving molecularly targeted agents. George29 de- of the shortened life expectancy associated with symp-
scribed the types of problems restrictive eligibility criteria tomatic brain metastases and a concern that the patient
create in phase III clinical trials in cancer with regard to would not receive a sufficient duration of the study drug;
scientific interpretation, medical applicability, complexity, therefore, clinical benefit would not be expected and may
costs, and patient accrual. In molecularly driven trials, eli- confound study results. Although safety is a priority,
gibility criteria related to patient characteristics, including universally precluding patients with brain metastases
restrictions on age, performance status, laboratory findings from clinical trials limits their access to potentially ef-
addressing organ function, and history of previous cancer fective therapy, making it difficult to extrapolate the use
diagnosis, previous antineoplastic therapy, or evidence of of these drugs to the general population.
metastatic disease to the brain, have become cumbersome. The more restrictive the eligibility criteria, the more
Many restrictive criteria could be reevaluated when limited the heterogeneity of the study population, limiting
considering the effects of traditional cytotoxic therapy the ability to generalize trial results to the population who
versus modern targeted therapy because these drug side- will be treated off-study. We must be concerned that the

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 87


KIM ET AL

TABLE 1. Comparison of Traditional Versus Potential Modern Clinical Trial Eligibility Criteria
Traditional Modern
Trial Methodology
Preclinical Data Less requirements, usually some diagnostic restrictions More diagnostic restrictions; usually require biopsy/tissue and
specific laboratory testing to identify actionable target
Clinical Trial Design Dose escalation Dose escalation with expansion cohorts
(Early Phase)
Clinical Trial Design Randomized registries Randomized in biomarker enriched population
(Late Phase)
Clinical Trial Maximum tolerated dose Response rate; however, FDA policies in place for Breakthrough
Endpoints Therapies and Accelerated Approval. Greater use of surrogate
(Early Phase) endpoints.
Clinical Trial OS, PFS OS
Endpoints
(Late Phase)
Patient Characteristics
Target Population Unselected Molecularly targeted subset
Age Typically restricted Typically restricted, but could advocate for broadening age re-
strictions given different side-effect profiles of these drugs
Performance Typically limited to ECOG PS 0-1 Could include EGOG PS 2
Status
Life Expectancy Typically, patients must be expected to live several months Could possibly be relaxed given different side-effect profiles
Organ Function/ Required; some patients may be excluded because of abnormal Required; some patients may be excluded because of abnormal
Laboratory laboratory values laboratory values. May require reevaluation as a result of
Abnormalities different side-effect profiles.
Medical History Required; some conditions are excluded Required; some conditions are excluded
Previous Therapy May exclude from trials looking at front-line therapy May exclude from trials looking at front-line therapy
Medications/ Less likely to exclude Increased concern for drug-drug interactions
Steroid Use
Comorbidities More stringent exclusion based on comorbidities (e.g., cardiac Some comorbidities traditionally restricting enrollment could be
comorbidities) relaxed
QOL/Depression Sometimes required that patients self-reported QOL/depression Could be relaxed; including patients with low self-reported QOL
scores are high enough for inclusion could be important for trials where efficacy is similar, but one
drug is better tolerated
Disease Characteristics
Confirmed Required Somewhat important, but druggable target is main driver
Diagnosis/Tumor
Histology
Target Lesion Required Required
Nontarget Lesion Usually not required May be required
Brain Metastases Traditionally excluded Traditionally excluded; possibly needs to be modified, especially
for patients with asymptomatic brain metastases
Metastatic Disease Usually allowable Usually allowable
Other Than Brain
Prior/Concurrent Usually excluded Usually excluded
Malignancy
Prior Progression Sometimes restricted in traditional trials Could be relaxed in trials of molecularly targeted drug
Prior Clinical Usually allowable Usually allowable
Response
Molecular Analysis
Biopsy/Specimen Not necessarily required Required
Study Therapy
Treated Previously Traditionally excluded Could be included if subsequent biopsies have target abnormality
With Study Drug
Known Sensitivity Not necessarily required Usually required
Other Investiga- Traditionally excluded Usually excluded
tional Drug
Continued

88 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


PRACTICAL VERSUS TRADITIONAL ELIGIBILITY CRITERIA

TABLE 1. Comparison of Traditional Versus Potential Modern Clinical Trial Eligibility Criteria (contd)
Traditional Modern
Toxicity More likely to exclude from trial or lead to patient coming off trial; Different side-effect profile than traditional cytotoxic therapy;
likely more grade 3 or 4 toxicity seen with traditional cytotoxic generally less grade 3 or 4 toxicity seen with targeted therapy
therapy
Investigator Traditionally more investigator judgment incorporated; drug or Less investigator opinion, shift to explicit instructions on all
Opinion combination therapy compared with physician preference protocol aspects
Regulatory
Consent/ Required Required
Compliance
Additional Not often required Often required for specimen collection/molecular analysis
Optional Consent
Abbreviations: FDA, U.S. Food and Drug Administration; OS, overall survival; PFS, progression-free survival; ECOG, Eastern Cooperative Oncology Group; PS, performance status; QOL,
quality of life.

study population is not accurately reflecting associated utilization of information obtained from ongoing adaptive
toxicities seen with these novel agents if only the youn- clinical trials to determine the appropriate treatment for
gest and healthiest patients qualify for study enrollment. each patient. There must be a concentrated effort to
Unger et al31 found that clinical trial participation was limit restrictive eligibility criteria while allowing continued
associated with better survival in the first year of en- modification of eligibility criteria as a drug moves through
rollment, which was postulated to be related to exclusion the research process. ASCO initiatives support the evolution
of patients having a poor prognosis with comorbidities of more practical, modernized, clinical trial design.1,17 At
and decreased functional status. the center of this movement for eligibility criteria reform
Restrictive, complex eligibility criteria likely result in is the implementation of trials that involve targeted
decreased accrual rates. Slow accrual to clinical trials therapies with molecular biomarkers. The higher the
means delays in answers to important clinical questions. strength of a drug and a specific molecular target mea-
The negative impact of low accrual rates extends to pro- sured on the tumor, the higher the priority of identifying
longed trial duration, early closure of studies with poor this patient population for treatment. In these settings,
accrual, and delays in analysis of trial results.32 If eligibility an active drug-to-target (e.g., high response rate) should
criteria are broadened, this may result in a faster accrual supersede other potentially restrictive eligibility criteria.
rate and observation of a wider array of clinical toxicities Because many markers have low incidence rates, greater
that more accurately reflect the general intended pop- collaboration will be needed to effectively complete
ulation. However, expanding clinical trial eligibility would meaningful studies. We must continue to prioritize the
perhaps allow for trial conclusions to more accurately development of modernized, practical clinical proto-
mimic general clinical practice and allow for extrapolation cols and encourage enrollment in these innovative trials.
to patients who were not treated on-study. We must increase access and awareness of molecularly
driven protocols to include more patients as we answer
FUTURE DIRECTIONS these important clinical questions and continue to in-
Personalized, precision medicine will require the in- tegrate new targeted drugs and biomarkers into clinical
corporation of clinical exams, molecular testing, and practice.

References
1. Meropol NJ, Kris MG, Winer EP. The American Society of Clinical 7. U.S. Department of Health and Human Services. U.S. Food and Drug
Oncologys blueprint for transforming clinical and translational cancer Administration. Guidance for Industry Expedited Programs for Serious
research. J Clin Oncol. 2012;30:690-691. ConditionsDrugs and Biologics. http://www.fda.gov/downloads/Drugs/
2. Masters GA, Krilov L, Bailey HH, et al. Clinical cancer advances 2015: GuidanceComplianceRegulatoryInformation/Guidances/UCM358301.pdf.
annual report on progress against cancer from the American Society of Accessed January 28, 2016.
Clinical Oncology. J Clin Oncol. 2015;33:786-809. 8. Johnson JR, Ning YM, Farrell A, et al. Accelerated approval of oncology
3. Mockus SM, Patterson SE, Statz C, et al. Clinical trials in precision products: the food and drug administration experience. J Natl Cancer
oncology. Clin Chem. 2016;62(3):442-448. Epub 2015 Nov 27. Inst. 2011;103:636-644.
4. Code of Federal Regulations. General principles of the IND submission, 9. U.S. Food and Drug Administration. Trametinib/Dabrafenib. http://
21 CFR Sect. 312. 22; 2014. www.fda.gov/Drugs/InformationOnDrugs/ApprovedDrugs/ucm476837.
5. Code of Federal Regulations. IND content and format, 21 CFR Sect. 312. htm. Accessed January 28, 2016.
23; 2014. 10. U.S. Food and Drug Administration. Osimertinib. http://www.fda.gov/
6. Code of Federal Regulations. Applications for FDA approval to market a Drugs/InformationOnDrugs/ApprovedDrugs/ucm472565.htm. Accessed
new drug, 21 CFR Sect. 314; 2014. January 28, 2016.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 89


KIM ET AL

11. U.S. Food and Drug Administration. Gefitinib. http://www.fda.gov/ 23. National Cancer Institute. NCI Molecular Analysis for Therapy
Drugs/InformationOnDrugs/ApprovedDrugs/ucm454692.htm. Accessed Choice Program (MATCH) & Pediatric MATCH. http://www.cancer.gov/
January 28, 2016. clinicaltrials/noteworthy-trials/match. Accessed February 2, 2015.
12. U.S. Food and Drug Administration. Erlotinib. http://www.fda.gov/ 24. Do K, OSullivan Coyne G, Chen AP. An overview of the NCI precision
Drugs/InformationOnDrugs/ApprovedDrugs/ucm352317.htm. Accessed medicine trials-NCI MATCH and MPACT. Chin Clin Oncol. 2015;4:
January 28, 2016. 31-38.
13. U.S. Food and Drug Administration. Innovation or stagnation: challenge 25. Kim ES. The future of molecular medicine: biomarkers, BATTLEs, and big
and opportunity on the critical path to new medical products. Wash- data. Am Soc Clin Oncol Educ Book. 2015;22-27.
ington, DC. 2004. 26. Andrews A. ASCO and NCI launch largest precision medicine trials using
14. Dragalin V. Adaptive designs: terminology and classification. Drug Inf J. real-world evidence. Am Health Drug Benefits. 2015;8:37.
2006;40(4):425-435. 27. Christian MC, Trimble EL. Increasing participation of physicians and
15. Bowalekar S. Adaptive designs in clinical trials. Perspect Clin Res. 2011;2: patients from underrepresented racial and ethnic groups in National
23-27. Cancer Institute-sponsored clinical trials. Cancer Epidemiol Biomarkers
16. Kim ES, Herbst RS, Wistuba II, et al. The BATTLE trial: personalizing Prev. 2003;12:277s-283s.
therapy for lung cancer. Cancer Discov. 2011;1:44-53. 28. Sohal DP, Rini BI, Khorana AA, et al. Prospective clinical study of
17. Kim ES, Bernstein D, Hilsenbeck SG, et al. Modernizing eligibility criteria precision oncology in solid tumors. J Natl Cancer Inst. 2015;108:
for molecularly driven trials. J Clin Oncol. 2015;33:2815-2820. djv332.
18. National Cancer Institute. ALCHEMIST (The Adjuvant Lung Cancer 29. George SL. Reducing patient eligibility criteria in cancer clinical trials.
Enrichment Marker Identifcation and Sequencing Trials): Question J Clin Oncol. 1996;14:1364-1370.
and Answers. http://www.cancer.gov/newscenter/newsfromnci/2014/ 30. Carden CP, Agarwal R, Saran F, et al. Eligibility of patients with brain
ALCHEMISTlaunchQandA. Accessed February 2, 2015. metastases for phase I trials: time for a rethink? Lancet Oncol. 2008;9:
19. Lung-MAP. http://www.lung-map.org. Accessed January 28, 2016. 1012-1017.
20. I-SPY 2 Trial. http://www.ispy2trial.org. Accessed January 28, 2016. 31. Unger JM, Barlow WE, Martin DP, et al. Comparison of survival out-
21. Siu LL, Conley BA, Boerner S, et al. Next-generation sequencing to guide comes among cancer patients treated in and out of clinical trials. J Natl
clinical trials. Clin Cancer Res. 2015;21:4536-4544. Cancer Inst. 2014;106:dju002.
22. Hyman DM, Puzanov I, Subbiah V, et al. Vemurafenib in multiple 32. Lara PN Jr, Higdon R, Lim N, et al. Prospective evaluation of cancer
nonmelanoma cancers with BRAF V600 mutations. N Engl J Med. 2015; clinical trial accrual patterns: identifying potential barriers to enroll-
373:726-736. ment. J Clin Oncol. 2001;19:1728-1733.

90 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


DEVELOPMENTAL THERAPEUTICS AND
TRANSLATIONAL RESEARCH

Immunotherapy: Beyond
Checkpoint Inhibitors

CHAIR
Gregory L. Beatty, MD, PhD
Hospital of the University of Pennsylvania
Philadelphia, PA

SPEAKERS
George E. Peoples, MD
Cancer Vaccine Development Program
San Antonio, TX

Marcela Valderrama Maus, MD, PhD


Abramson Cancer Center of the University of Pennsylvania
Philadelphia, PA
ARMING THE IMMUNE SYSTEM TO PREVENT CANCER RECURRENCE

Arming the Immune System Through Vaccination to Prevent


Cancer Recurrence
Diane F. Hale, MD, Timothy J. Vreeland, MD, and George E. Peoples, MD

OVERVIEW

Cancer vaccines have the potential to provide a nontoxic treatment for the prevention of cancer recurrence in the adjuvant
setting. Many cancer vaccines have been tested in multiple phase III trials with minimal success. However, through these
failed clinical trials, we have learned that the ideal setting for vaccine therapy is the adjuvant setting. Also, we have learned
important lessons about patient selection to maximize the probability of success. This article will highlight some of the
successes, our trial results in the adjuvant setting, and future directions.

T he creation of a vaccine to prevent or cure cancer is a very


alluring goal, but the pursuit of this goal has been fraught
with difficulty. To create a cancer vaccine, the first key is to
analyses from multiple trials of vaccines given to patients
with advanced cancers have shown that, although vaccines
produced disappointing results overall, they perform much
find the perfect target: a tumor-associated antigen (TAA) or better in patients with minimal or no residual disease. Taken
antigens expressed on a high percentage of tumor cells but as a whole, these results indicate that cancer vaccines per-
not on normal cells. Once the target is identified, the goal is form better when used in patients with less aggressive dis-
to use an efficient, safe, and easily reproducible method to ease, whether by biology or by disease burden.
stimulate the immune system to eliminate targeted tumor Building on these lessons, cancer vaccines have been used
cells. These goals have been approached in a number of ways to treat patients with the absolute minimal disease burden
in the saga of cancer vaccine creation, but with few clinically after they are rendered disease-free by standard-of-care
meaningful successes. To date, the majority of cancer vaccines therapies. Priming the immune system to create an active
have been tested in the metastatic setting with disappointing immune response to prevent recurrence rather than trying
results. The high disease burden, immune escape mechanisms, to eradicate advanced, established, and aggressive disease is
and immune suppression of the tumor micro-environment very appealing. First, after patients complete and recover
encountered in the setting of metastatic or aggressive disease from standard-of-care therapy (generally immunosuppres-
is more than the immune system can overcome. This may not sive chemotherapy), they have improved immune function,
be the ideal setting for cancer vaccines to become clinically allowing a full development of antitumor immunity.1 Second,
relevant, at least not as monotherapy. in a disease-free state, there is theoretically less tumor-related
There are, however, exceptions to the overall discouraging immune suppression, fewer immune escape mechanisms, and
results of vaccines in the metastatic setting, and important no tumor microenvironment.1 With the bulk of tumor removed
lessons can be learned from these exceptions. The most or eradicated, the immune system can then be used to eliminate
notable exception, and the only vaccine granted U.S. Food the small number of tumor cells left after therapy, which is a
and Drug Administrationapproval, is sipuleucel-T, an autol- more realistic goal for this mechanism. Finally, the long-term
ogous dendritic cell vaccine directed against the TAA prostatic immune memory created by a successful cancer vaccine offers
acid phosphate. This vaccine is approved for use in metastatic, continued protection against dormant circulating or dissemi-
asymptomatic or minimally symptomatic castration-resistant nated tumor cells that may still be present or may re-emerge in a
(hormone-refractory) prostate cancer. The success of sipuleucel- disease-free host; this characteristic and distinct advantage
T in the metastatic setting is likely due to the indolent nature of distinguishes active specific immunotherapy from traditional
prostate cancer, which makes it a more realistic target for a cancer therapies. Consequently, cancer vaccines offer the unique
cancer vaccine, even in advanced disease. Similarly, subset promise of long-term protection against cancer recurrence.

From the Department of Surgery, San Antonio Military Medical Center, Fort Sam Houston, TX; Department of Surgery, Womack Army Medical Center, Fort Bragg, NC; Cancer Vaccine
Development Program, San Antonio, TX; Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: George E. Peoples, MD, Cancer Vaccine Development Program, c/o Metis Foundation, 300 Convent St., San Antonio, TX 78205; email: georgepeoples2@
hotmail.com.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e159


HALE, VREELAND, AND PEOPLES

Although using the vaccine platform to prevent recurrence for the prevention of solid tumor recurrence. There have
in the adjuvant setting is quite attractive, this setting has been many vaccines that found clinical improvement in
unique challenges. There is less tolerance for toxicity when phase II evaluation but failed to find clinical significance in
patients are disease-free; therefore, any therapy being phase III. The lessons learned from previous trials have
added in the adjuvant setting must come with a very low assisted in creating improved trial designs and, most im-
toxicity profile or else compliance is expected to be poor. portantly, selecting patients who will derive the most benefit
Also, proving a clinically substantial decrease in recurrence from a cancer vaccine. Highlighted below are a few key
can be quite challenging when evaluating patients with less phase III trials that demonstrate important points for future
aggressive disease. The attractive aspect of studying a new trial design.
therapy in patients with advanced disease is the relatively OncoVax is an autologous tumor cell vaccine combined
short time period needed to show a reduction in tumor with bacillus Calmette-Guerin that has been studied
burden or improvement in progression-free survival. Con- extensively in colon cancer. There have been three phase
versely, in the adjuvant setting, the time period required to III trials using this vaccine in the adjuvant setting after
reach a noteworthy number of recurrences may be quite resection of stage II/III colon and rectal cancer, enrolling a
lengthy. Depending on the tumor biology and documented total of 764 patients.2-4 The initial phase III trial enrolled
recurrence rates, this may necessitate a trial spanning patients with both colon and rectal cancer. Overall, there
multiple years and with large treatment groupsultimately was no significant differences in the treatment groups, but
resulting in a very expensive trial. If the vaccine strategy is when breaking down the results by tumor type (colon vs.
truly more suitable in the adjuvant setting, these limitations rectal) and focusing only on those patients with colon
lead to a difficult first step to prove efficacy and likely explain cancer, there was a significant improvement in both overall
why this strategy has been around for so long but has so few survival (OS) and disease-free survival (DFS) for the vacci-
successes. As a result, much of the current evidence showing nated versus control patients (OS 83% vs. 52%, hazard ratio
benefit of vaccines comes from subset analyses of larger [HR] 3.97; 95% CI, 1.2412.72; p = .020; DFS 67% vs. 44%, HR
trials focusing on patients with minimal disease burden. 2.67; 95% CI, 1.056.76; p = .039).2 This led to the second
We will review the evidence of vaccine success in patients phase III trial, which focused on patients with surgically
with minimal disease burden and current immunologic resected stage II/III colon cancer. Again, the overall results of
vaccine therapies being studied in the adjuvant setting, and this trial failed to show a significant difference with treatment,
we will lay out the best available data supporting this theory but within the subset of stage II disease, vaccinated patients did
with current successes. We will also discuss future plans for show an improved recurrence free interval (p = .011) and 4-year
combination therapies. recurrence-free survival (88% vs. 74%, p = .032).3 The largest of
the phase III OncoVax trials enrolled 412 patients with surgically
resected stage II/III colon cancer and had a 7.6-year median
PROOF OF CONCEPT IN PHASE III TRIALS follow-up.4 This trial did not show a significant improvement in
Although developing and funding an adjuvant study may be OS, DFS, or time-to-recurrence with vaccination.4 Based on the
difficult, it has been attempted in several cancer types with a combined results of these trials, the next phase III OncoVax trial
variety of vaccine platforms. Table 1 summarizes the pre- is focused on patients with minimal disease, where subset
vious and current phase III trials of vaccines given in the analysis has shown the most promise, enrolling only patients
adjuvant setting (with no or minimal evidence of disease), with resected stage II colon cancer (NCT02448173). Overall, the
work with this vaccine nicely demonstrates identification of
patients with no evidence of disease with low disease burden
KEY POINTS as the most suitable population for cancer vaccines.
A similar finding of improved outcomes in vaccinated
Cancer vaccines represent a nontoxic therapeutic patients with lower disease burden was found in a trial using
modality with great antitumor specificity. vitespen for renal cell carcinoma. Vitespen (HSPPC-96) is an
Peptide vaccines effectively stimulate a specific immune autologous, tumor-derived, heat shock protein vaccine. In
response and are simple, easily produced, and the phase III evaluation, it was given to patients in the
efficiently exported to the community. adjuvant setting that were stage I-IV, surgically resected and
The adjuvant setting is an appropriate clinical setting to with no evidence of disease; with 1.9-year median follow-
test cancer vaccines as it optimizes the chances of an up, there were no overall differences in recurrence events
effective immune response when the tumor burden is (p = .506). However, on subset analysis of patients with
the lowest.
stage I/II disease, there was a decrease in recurrence for
Combinations of other immunotherapies with vaccines
may be even more clinically effective.
patients who were treated with the vaccine versus patients
In the metastatic setting, combining vaccines as in the control group (15% vs. 27%, p = .056).5
T-celleliciting agents with checkpoint inhibitors may Patient selection in trials using an adjuvant vaccine to
synergistically enhance the benefits of each modality prevent recurrence is a key to demonstrate clinical efficacy.
and improve long-term clinical results. Beyond a low disease burden, it is also important to un-
derstand the specific mechanism of action of each vaccine to

e160 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


TABLE 1. Adjuvant Phase III Solid Tumor Vaccine Trials
Solid Tumor Reference or
Type Vaccine Strategy Vaccine Type Immunoadjuvant Study Type No. Patients Study Name Results NCT Number
DENDRTIC CELL VACCINES
Melanoma DCaT-RNA DC Autologous tumor RNA loaded Adjuvant, NED, resected, monosomy 3 200* Currently enrolling, study start date June 2014, esti- NCT01983748
(Uveal) onto autologous DC mated primary completion June 2020
Glioblastoma DC Vax-L DC Autologous DC pulsed with au- Adjuvant, NED, new diagnosis in addition to 348* Active (not enrolling), study start date December 2006, NCT00045968
tologous whole tumor lysate standard of care, must be resected estimated primary completion September 2016
disease
Glioblastoma ICT-107 DC Autologous DC pulsed with 6 Adjuvant, evidence of disease after standard 414* Currently enrolling, study start date December 2015, NCT02546102
peptides (MAGE-1, HER2, AIM- of care , 1 cm3 of disease may be present estimated primary completion December 2019
2, TRP-2, GP100, IL-13Ra2)
PEPTIDE VACCINES
Breast E75 Peptide HER2/neu peptide vaccine GM-CSF Adjuvant, NED, high-risk for recurrence 700* PRESENT Active (not enrolling), study start date November 2011, NCT01479244
estimated primary completion date April 2018
Melanoma POL-103A Multiple Shed peptides from 3 allogenic Alum Adjuvant, NED, surgically resected, stage IIb, 1,059* MAVIS Currently enrolling, study start date April 2012, esti- NCT01546571
peptides melanoma cell lines IIc, III mated primary completion date July 2016
Melanoma Tyrosinase, gp100, Multiple GM-CSF Adjuvant, NED, surgically resected, stage IV 815 OS (p = .528), RFS (p = .131) NCT01989572
MART-1 Peptides
Melanoma GM2-KLH Protein GM2 antigen combined with KLH QS-21 Adjuvant, NED, stage II 1,314 EORTC 18961 Terminated after second interim analysis: 1.8 years Eggermont
to improve immunogenicity median follow-up, detrimental OS (HR 1.66; p = .02) et al19
and futile RFS (HR 1.00; p = .99). After 4-year follow-
up, vaccinated patients RFS 1.2% (HR 1.03) and OS
rate 2.1% (HR 1.16)
Melanoma MAGE-A3 Protein recMAGE-A3 AS15 Adjuvant, NED, stage IIIB-C 1,351* DERMA Terminated after showing lack of efficacy NCT00796445
Melanoma MART-1, NA17-A, Multiple Adjuvant, NED 13 Terminated for low accrual NCT00036816
(Ocular) gp100, peptides
tyrosinase
NSCLC MAGE-A3 Protein recMAGE-A3 AS15 Adjuvant, NED, MAGE-A3 positive, resected, 2,272 MAGRIT 38.8-month median follow-up, median DFS 60.5 Vansteenkiste
stage IB, II, IIIA months vaccine vs. 57.9 months control (p = .74) et al20
Glioblastoma Rindopepimut Peptide 14-mer peptide of EGFRvIII with GM-CSF Adjuvant, must have attempted surgical 700* ACT IV Active (not enrolling), study start date November 2011, NCT01480479
(CDX-110) KLH resection and chemoradiation, may have estimated primary completion date November 2016
evidence of disease, must have stable
disease from postoperative to
postchemoradiation
RCC HSPPC-96 Protein Autologous, tumor-derived, heat Adjuvant, NED, surgically resected, stage I-IV 728 1.9-year median follow-up, no difference in overall Wood et al5
(vitespen) shock protein recurrence events (37.7% vaccine vs. 39.8% control; p
= .506), slightly improved in stage I/II (15.2% vaccine
vs. 27% control; p = .056)
TUMOR CELL VACCINES
Pancreas Algenpantucel-L Tumor cell Alpha-1,3-glactosyltransferase Adjuvant, NED, resected pancreatic cancer 722* IMPRESS Active (not enrolling), study start date April 2010, NCT01072981
(hyperacute) expressing allogeneic pancre- stage I/II estimated primary completion date June 2016
atic tumor cell vaccine
NSCLC Belagenpumatecel- Tumor cell Allogenic tumor cell with anti- Adjuvant (maintenance after first-line che- 532 STOP Trial No difference OS (p = .594), no difference PFS (p = .947); Giaccone
L (lucanix) sense plasmid motherapy), evidence of disease allowed regression analysis found improved survival with et al21
to have SD (CR or PR) randomization within 12 weeks of completion of
chemotherapy (p = .002) and prior radiation im-
proved survival (p = .032)
Melanoma Polyvalent cultured Tumor cell Allogeneic melanoma vaccine BCG Adjuvant, NED, stage III/IV 1,656 MMAIT Terminated after interim analysis for futility: 5-year Morton et al22
allogenic mela- estimated survival 42.3% for stage IV and 63.4% for

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK


noma vaccine stage III
(canvaxin)

e161
ARMING THE IMMUNE SYSTEM TO PREVENT CANCER RECURRENCE

Continued
TABLE 1. Adjuvant Phase III Solid Tumor Vaccine Trials (cont'd)

e162
Solid Tumor Reference or
Type Vaccine Strategy Vaccine Type Immunoadjuvant Study Type No. Patients Study Name Results NCT Number
Melanoma Polyvalent tumor- Tumor cell Allogeneic melanoma vaccine DETOX (detoxi- Adjuvant, NED, stage II/III 689 S9035 12.1-year median follow-up, ITT no difference in RFS Carson et al6
cell lysate fied Freund (p = .58) or OS (p = .61), subset analysis vaccinated
(melacine) adjuvant) and HLA-A2/Cw3+ 10-year RFS 66% vs. controls 54%
(p = .02)
Colon and Active specific Tumor cell Autologous tumor cells BCG Adjuvant, NED, stage II/III colon or rectal 98 93-month median follow-up, OS trend toward favoring Hoover et al2
HALE, VREELAND, AND PEOPLES

Rectal immunotherapy vaccine (p = .088), cohort analysis: colon cancer


(OncoVax) significant OS (p = .02) and DFS (p = .039) for vaccine,
no benefit in rectal
Colon Active specific Tumor cell Autologous tumor cells BCG Adjuvant, NED, stage II/III colon 254 8701 5.3-year median follow-up, recurrence free interval Vermorken et
immunotherapy (p = .023) improved in vaccine with larger impact on al3
(OncoVax) RFS (p = .032) in stage II vaccine
Colon Active specific Tumor cell Autologous tumor cells BCG Adjuvant, NED, stage II/III colon 412 ECOG E5283 7.6-year median follow-up, ITT no difference OS, DFS, or Harris et al4
immunotherapy time to recurrence, cohort analysis vaccinated pa-
(OncoVax) tient with treatment compliance signaled a trend in
DFS (p = .078) and OS (p = .12)
Colon Active specific Tumor cell Autologous tumor cells BCG Adjuvant, NED, stage II colon 550* Currently enrolling, study start date May 2015, esti- NCT02448173
immunotherapy mated primary completion date July 2020
(OncoVax)
Colon and Newcastle disease, Tumor cell Viral vector of autologous tumor Adjuvant, NED, after resection (to include 51 116.1-month follow-up for vaccine, 112.4-month Schulze et al23
Rectal virus-infected cells from resected liver liver metastasis resection) follow-up for control, no difference in OS, DFS, or
autologous tu- metastasis metastases-free survival. Subgroup analysis: advan-

2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


mor cell vaccine tage for vaccinated colon OR (p = .042) and
(ATV-; NDV) metastases-free survival (p = .047)

*Anticipated number.
Abbreviations: DC, dendritic cell; NED, no evidence of disease; GM-CSF, granulocyte macrophage colony-stimulating factor; OS, overall survival; DFS, disease-free survival; NSCLC, nonsmall cell lung cancer; RCC, renal cell carcinoma; SD, stable
disease; CR, complete response; PR, partial response; BCG, bacillus Calmette-Guerin; ITT, intention-to-treat.
ARMING THE IMMUNE SYSTEM TO PREVENT CANCER RECURRENCE

identify the patients who will have the best clinical benefit. minimal toxicity with the majority of local and systemic
The Southwest Oncology Groups phase III trial of melacine toxicities being grade 1. It was also able to produce effective
demonstrates the importance of human leukocyte antigen immunity to HER2. With a median follow-up of 60 months,
(HLA) characteristics in patient selection and understanding the vaccinated patients in this trial showed a trend toward
how the vaccine stimulates the immune system. Melacine improved DFS with a 48% reduction in relative risk of re-
is a vaccine created from an allogeneic tumor cell line that currence (RRR; p = .08).7 Given the dose escalation nature of
contains numerous melanoma-associated TAAs. The phase III the first stage of the trial, some patients did not receive the
trial randomly selected 689 patients with stage II/III resected optimal dose of the vaccine. When we examined patients
melanoma to receive the vaccine or to be in the control group. based on dosing, we found a significant increase in DFS in
At a median follow-up of 12.1 years, the trial had an overall patients who received the optimal dosing regimen com-
negative result, but subset analysis demonstrated a significant pared with suboptimal dosed and observational controls
recurrence-free survival (RFS) and OS advantage in the pa- (94.6% vs. 87.1% vs. 80.2%, respectively; p = .05).7 During
tients who received the vaccine compared with the control this trial, it was discovered that, after completing the pri-
group when limiting analysis to patients with HLA-A2+ and/or mary vaccination series, patients demonstrated waning
HLA-Cw3+ expression (5-year RFS, 78% vs. 65%; 10-year RFS, immunity to the targeted peptide. Therefore, a voluntary
66% vs. 54%; p = .02; 5-year OS, 90% vs. 76%; 10-year OS, 75% booster program was instituted and patients received ad-
vs. 63%; p = .01).6 These findings demonstrate the importance ditional booster inoculations every 6 months. When ex-
of identifying the appropriate target population for a vaccine amining patients who were optimally dosed and boosted, we
and selecting patients with the appropriate biologic features. found an improvement numerically in DFS versus all other
vaccinated patients versus the control arm (95.2% vs. 88.4%
vs. 80.2%; p = .11).7 Another important patient character-
PROOF OF EFFICACY istic, which had been previously demonstrated and was also
As previously discussed, demonstrating efficacy of a new identified here, was the effect of HER2 expression on response
therapy in the adjuvant setting is inherently difficult, but it has to the vaccine.8 For patients with non-overexpression of HER2
been done. We will highlight the work our group has completed (immunohistochemistry [IHC] 1+ or 2+), there was a trend
with cancer vaccines in the adjuvant setting; these trials are toward improvement in 5-year DFS in vaccinated patients
summarized in Table 2. We have extensively researched three versus the control (88% vs. 78%; p = .16).7 There were also
HER2-based peptide vaccines (Fig. 1). These peptides (E75, GP2, important limitations of this trial; there was no blinded,
and AE37) were paired with an immunoadjuvant, granulocyte placebo-treated control group receiving GM-CSF, which led to
macrophage colony-stimulating factor (GM-CSF) and given in questions about the effects of this immunoadjuvant therapy.
the adjuvant setting to disease-free patients with breast cancer Thus, this feature was incorporated into our future trials.
at high risk of recurrence. Two of the vaccines are HLA-A2 and Integrating the lessons learned from this initial trial, a phase
HLA-A3restricted (E75 and GP2) and directly stimulate III trial was designed that included a GM-CSFtreated control
CD8+ T cells to generate immunity. The third, AE37, is a longer arm and optimal dosing of the vaccine to include a booster
peptide vaccine, is not HLA-restricted, and stimulates CD4+ series, and it enrolled only patients with HER2 1-2+ expression.
T cells to create HER2-directed immunity. Similar to the HER2- The phase III PRESENT (Prevention of Recurrence in Early Stage
based peptide vaccines, we have also targeted folate binding node-positive breast cancer with low to intermediate HER2
protein (FBP)-expressing tumors with a peptide vaccine (E39 Expression with NeuVax Treatment) trial was initiated in the
and GM-CSF) in ovarian, endometrial, and breast cancers. adjuvant setting for patients with high-risk HLA-A2+ or HLA-A3+
Lastly, we have recently expanded into an autologous dendritic breast cancers who were deemed disease free after standard-
cell vaccine platform using autologous tumor lysate particle- of-care therapy. The patients were randomly selected to re-
loaded dendritic cells in the adjuvant setting, targeting patients ceive E75 with GM-CSF or GM-CSF only. This trial has com-
with surgically resected melanoma. pleted enrollment and is monitoring for the primary endpoint
of 3-year DFS.
E75 (Nelipepimut-S, NeuVax)
E75 (nelipepimut-S or NeuVax) is a peptide (KIFGSLAFL, HER2 AE37 and GP2
aa: 369-377) derived from the extracellular domain of HER2 Our second large phase II peptide vaccine trial in the ad-
(Fig. 1) and given in combination with GM-CSF. Its immu- juvant setting, which is nearing completion, is a dual trial
nologic activity is mainly restricted to specific HLA types HLA- testing GP2 and AE37. GP2 (IISAVVGIL, HER2 aa: 654-675) is a
A2 and HLA-A3, but with the possibility of binding several peptide derived from the transmembrane domain of HER2
other less common alleles HLA-A24 and HLA-A26. E75 was (Fig. 1). GP2 is a major histocompatibility complex (MHC)
evaluated in one of the largest adjuvant breast cancer class I peptide vaccine that stimulates antigen-specific CD8+
vaccine phase I/II trials to date, with 195 enrolled and 187 T cells in patients positive for HLA-A2 or HLA-A3. AE37
evaluable patients.7 Patients who were positive for HLA-A2/ (GVGSPYVSRLLGICL, HER2 aa: 776-790) is the Ii-Key (LRMK)
A3 were placed in the treatment arm, and patients who were hybrid of the HER2-derived MHC class II peptide, AE36, from
negative for HLA-A2/A3 were followed prospectively in an the intracellular domain of HER2 (Fig. 1) and is not HLA-
observational control group. This vaccine was found to have restricted. The trial design selected patients by HLA status,

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e163


HALE, VREELAND, AND PEOPLES

TABLE 2. Current and Future Trials in the Adjuvant NED Setting to Prevent Recurrence
Solid
Tumor Vaccine No. Reference or
Type Vaccine Strategy Type Immunoadjuvant Phase Study Type Patients Result NCT Number
PEPTIDE
Breast E75 (NeuVax, Peptide HER2 (aa: GM-CSF II Adjuvant, NED, 195 Median follow-up 60 months; 5-year Mittendorf
nelipepi- 369-377) high-risk DFS ITT: 90% vaccinated vs. 80% et al7
mut-S) recurrence control (p = .08); subset populations
NCT00841399
with most benefit optimally dose 95%
vaccine vs. 80% control (p = .05) and NCT00854789
both optimally dosed and boosted:
95% vaccinated vs. 80% control (p =
.11)
Breast E75 (NeuVax, Peptide HER2 (aa: GM-CSF II Adjuvant, NED, 108* Anticipated start date March 2016, es- NCT02636582
(DCIS) nelipepi- 369-377) DCIS timated primary completion Sep-
mut-S) tember 2019
Breast AE37 Peptide HER2 (aa: GM-CSF II Adjuvant, NED, 298 Median follow-up 25 months; 5-year DFS Mittendorf
776-790 high-risk IIT: 81% vaccine vs. 80% controls (p = et al10
+LRMK) recurrence .70); subset populations with improved
NCT00524277
benefit HER2 1-2+ expressing tumors:
77% vaccine vs. 66% controls (p = .21)
and TNBC 78% vaccine vs. 49% controls
(p = .12)
Breast GP2 Peptide HER2 (aa: GM-CSF II Adjuvant, NED, 180 Median follow-up 34 months; 5-year Schneble
654-675) high-risk DFS IIT: 88% vaccine vs. 81% controls et al9
recurrence (p = .43); subset population with im-
NCT00524277
proved benefit HER3+ expressing tu-
mors: 94% vaccine vs. 89% controls (p
= .86)
Prostate E75 (NeuVax, Peptide HER2 (aa: GM-CSF I/IIA Adjuvant, NED 40 Median follow-up 58 months; PSA re- Gates et al24
nelipepi- 369-377) s/p resection, currence rate: 29% vaccine vs. 26%
mut-S) high-risk control (p = .9); DFS: 41.3 vs. 37.9
recurrence months (p = .6)
Ovarian E39 Peptide Folate bind- GM-CSF I/IIA Adjuvant, NED 51 Median follow-up 12 months; 5-year Greene
ing pro- DFS ITT: 43% vaccine vs. 34% control et al15
tein (aa: (p = .36); subset population with im-
NCT01580696
191-199) proved benefit optimally dosed
group: 86% vaccine vs. 34% control (p
= .03)
DENDRITIC CELL
Melanoma Dendritoma DC Autologous IL-2 I/IIA Adjuvant, NED 25 Median OS 16 months; overall 5-year Greene et al17
DC fused and ED, stage DFS: 29%; OS improvements in NED
with au- IV patients patients: 80% vs. ED 14% (p = .004)
tologous enrolled
tumor
lysate
Melanoma TLPLDC DC Autologous GM-CSF IIB Adjuvant, NED 120* Actively enrolling, estimated primary NCT02301611
DC loaded completion June 2018
with au-
tologous
tumor
lysate
COMBINATION
Breast E75 (NeuVax, Peptide and HER2 (aa: GM-CSF II Adjuvant, NED, 300* Actively enrolling, estimated primary NCT01570036
Nelipepi- monoclo- 369-377) high-risk recur- completion December 2016
mut-S) and nal and HER2 rence with
trastuzu- antibody monoclo- HER2 1-2+
mab nal expressing
(herceptin) antibody tumors
Breast E75 (NeuVax, Peptide and HER2 (aa: GM-CSF II Adjuvant, NED, 100* Actively enrolling, estimated primary NCT02297698
Nelipepi- monoclo- 369-377) high-risk recur- completion October 2016
mut-S) and nal and HER2 rence with
trastuzu- antibody monoclo- HER2 3+
mab nal expressing
(herceptin) antibody tumors
*Anticipated number.
Abbreviations: DC, dendritic cell; NED, no evidence of disease; GM-CSF, granulocyte macrophage colony-stimulating factor; OS, overall survival; DFS, disease-free survival; NSCLC,
nonsmall cell lung cancer; RCC, renal cell carcinoma; SD, stable disease; CR, complete response; PR, partial response; BCG, bacillus Calmette-Guerin; ITT, intention-to-treat; DCIS,
ductal carcinoma in situ; TNBC, triple-negative breast cancer; TLPLDC, tumor lysate particle-loaded dendritic cell.

e164 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


ARMING THE IMMUNE SYSTEM TO PREVENT CANCER RECURRENCE

FIGURE 1. HER2 Peptide Vaccines (E75, GP2, AE37)

Abbreviations: MHC, major histocompatibility complex; HLA, human leukocyte antigen.

and placed the patients who were positive for HLA-A2/A3 controls (77% vs. 66%; p = .21) and a 32% reduction in RRR.
into the GP2 trial and patients who were negative for HLA- This was even more pronounced in patients with triple-
A2/A3 into the AE37 trial. Patients in both trials were then negative breast cancer, who experienced a 56% reduction in
randomly selected to receive the respective vaccine or the RRR and had a trend toward improved 5-year DFS with the
control, GM-CSF alone. This dual trial has also completed vaccination (78% vs. 50%; p = .12). Patients with triple-
enrollment, with 298 patients enrolled in the AE37 trial and negative breast cancer currently have limited options for
180 patients in the GP2 trial.9,10 From the analysis of these long-term recurrence prevention, making the AE37 vaccine
trials, we have established that the minimal toxicity in all an interesting option for future study.10
patients was primarily related to the GM-CSF given similar Overall, through these phase II studies, a potential benefit
toxicity levels between the vaccine and control arms. of peptide vaccine therapy in the adjuvant setting was
The primary analysis of the GP2 trial revealed a 37% reduction identified in patient populations depending on biologic char-
in RRR for the vaccinated arm with a 34-month median follow- acteristics of the tumor and synergy with current standard-
up. Vaccinated patients showed a nonsignificant increase in of-care therapy, specifically passive immunotherapy.
estimated 5-year DFS (88% vs. 81%; p = .43). Interestingly,
subset analysis by HER2 expression in this trial showed that E39
the best DFS was found in patients with HER2 overexpression Similar to HER2 expression in breast cancer, FBP has also
(IHC 3+ or FISH+) who received the vaccine after completing been found to be overexpressed in breast, lung, endome-
trastuzumab. Although intention-to-treat (ITT) analysis in this trial, and ovarian cancers and has been linked to a poor
subgroup showed a 94% estimated 5-year DFS in vaccinated prognosis.11-14 E39 (EIWTHSYKV, FBP aa: 191-199,) is an HLA-
patients versus 89% in the control arms (p = .86), the per- A2restricted FBP peptide vaccine, also given with GM-CSF.
treatment analysis (excluding patients who experienced re- Our group has completed an initial phase I/IIA trial of the E39
currence during the primary vaccination series and therefore with GM-CSF vaccine given in the adjuvant setting to pa-
did not complete the series) of HER2 overexpression showed tients with ovarian and endometrial cancers after they re-
improved 5-year DFS in the vaccinated patients versus the ceived standard-of-care therapies. Similar to previous
control (100% vs. 89%; p = .08). Although it is difficult to show a peptide-based vaccine trials, there was minimal toxicity
statistically significant improvement over the already impres- with the treatment. Interim analysis after median follow-up
sive DFS of patients receiving standard-of-care trastuzumab, of 12 months revealed overall ITT 5-year estimated DFS was
based on these results, the GP2 vaccine appears to have 43% for vaccinated patients versus 34% for controls (p = .36).
synergism with the HER2-directed trastuzumab.9 Like the initial E75 trial, given the phase I/IIA design of this
The primary analysis of the AE37 arms revealed no sig- trial, not all patients received what was deemed to be the
nificant difference in 5-year DFS for vaccinated versus optimal dose of the vaccine. In subset analysis based on
control patients (83.5% vs. 82.1%; p = .7). Again, planned dosing, optimally dosed patients had a statistically signifi-
subset analysis revealed the patient populations with the cant increase in survival over suboptimally dosed and control
greatest benefit. First, patients with HER2 1-2+ expression patients (86% vs. 34% vs. 21%, respectively; p = .03). Because
showed a trend of separation in the Kaplan Meier plots, with of the aggressive nature of ovarian and endometrial cancers
5-year DFS enhanced in patients who were vaccinated versus as opposed to breast cancer, this trial was able to show a

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e165


HALE, VREELAND, AND PEOPLES

survival difference between groups in a relatively short breast cancer with HER2 overexpression changed based on
time.15 the approval of trastuzumab. During these trials, we were
able to compare patients who received a combination of
Tumor Lysate Particle-Loaded Dendritic Cell Vaccine peptide vaccine and trastuzumab with patients in the control
We are currently evaluating a novel dendritic cell vaccine arms, who received trastuzumab without a vaccine. From
technology. The new technology revolutionizes the ability to these trials, there have been 89 patients with HER2 over-
efficiently produce a personalized dendritic cell vaccine. The expression who received trastuzumab, with 55 of these
vaccine is produced by loading autologous tumor lysate into patients also receiving one of our peptide vaccines. With a
prepared yeast cell wall particles, which are naturally and median follow-up of 36 months, patients receiving the
efficiently phagocytized by isolated and immature dendritic combination of vaccine and trastuzumab had a 100% DFS
cells. Along with the immunoadjuvant CpG, the loading versus only 84% in those receiving trastuzumab alone
process matures the dendritic cells resulting in the final (p = .012; unpublished data from project narrative). To
product referred to as the tumor lysate particle-loaded further elucidate the potential for synergy and verify the
dendritic cell (TLPLDC) vaccine. The use of autologous tu- possible improved clinical benefit, we are currently enroll-
mor lystate allows for the inclusion of the full range of ing two separate trials combining E75 and trastuzumab
tumor antigens from a given patients tumor, making it in the adjuvant setting. The first trial is focused on the
widely applicable to any patient and any tumor type. patients with HER2 overexpression (HER2 3+ or FISH+;
TLPLDC can be created in 48 hours and requires only a small NCT01570036), and the second trial is enrolling patients
amount of tumor (1 cm3). In comparison with previous with HER2 non-overexpression (HER2 1-2+; NCT02297698).
dendritic cell vaccine technologies, TLPLDC costs less and In both trials, patients are randomly selected to receive
requires fewer dendritic cells. This technology affords the trastuzumab with E75 and GM-CSF versus trastuzumab and
ability to create a personalized vaccine for any solid tumor GM-CSF alone. Patients with HER2 1-2+ expression cur-
in an expeditious manner. rently have limited options in the adjuvant setting after
The TLPLDC vaccine technology builds from the previous completion of chemotherapy, as they do not qualify to
technology of dendritoma (fusion of dendritic cells with receive trastuzumab. Our trial, along with the NSABP-47
autologous tumor), which is a relatively cumbersome trial, seeks to address the use of trastuzumab in this
technique.16 We have completed a phase I/IIA trial of the population. Additionally, we seek to assess the vaccine in
dendritoma vaccine in 25 patients with stage IV melanoma. this population as well as the toxicity associated with
With a median follow-up of 16 months, the overall 5-year trastuzumab alone or in combination with the vaccine in
DFS was 29%. Following the theme of previous trials, the these patients in the adjuvant setting.
patients with no evidence of disease prior to initiation of Though beyond the scope of this article, vaccines may
therapy experienced a significant benefit compared with now, largely for the first time, be successfully applied to the
those with evidence of disease (80% vs. 14%; p = .004).17 nonadjuvant setting. Given the successes of checkpoint
Although these results are encouraging, the dendritoma inhibitors (anti-CTLA4 and PD-1) to protect and promote
technology is too cumbersome and expensive to be evaluated endogenous antitumor T cells, there is now rationale for
in a larger trial. The TLPLDC vaccine technology accomplishes combining tumor-reactive T-celleliciting vaccines with
the same goal as the dendritoma technologythe introduction clinically proven checkpoint inhibitors for patients with
of the entire antigenic repertoire from the autologous tumor larger tumor burdens. Furthermore, the knowledge gained
into the cytoplasm of the dendritic cell. The efficacy of the from the checkpoint inhibitorrelated studies underscores
TLPLDC vaccine has been confirmed in an ongoing basket trial of why so many vaccines have previously failed to demon-
multiple solid tumors.18 Current results from this ongoing phase strate benefit in patients who have metastatic disease
I/IIA trial indicate that the autologous TLPLDC vaccination is with established tumors capable of suppressing/evading
well-tolerated and safe. The TLPLDC vaccine had clinical benefit the vaccine-induced immune response. To assess the likely
in 60% of patients, with 30% objective response including synergism between vaccines and checkpoint inhibitors,
complete response in metastatic melanoma.18 Therefore, we have initiated a new trial for patients with metastatic
building on the results of the dendritoma trial in melanoma and melanoma receiving standard-of-care checkpoint inhibitors
utilizing the improved TLPLDC technology (confirmed to be but who have stable or slowly progressive disease. For
effective in melanoma18), we have initiated a prospective, these patients, we will add a personalized TLPLDC vaccine
randomized, blinded, placebo-controlled phase IIB trial in pa- in combination with their checkpoint inhibitors. The end-
tients with fully resected stage III and IV melanoma to prevent points of the trial are safety and tumor response.
recurrence (NCT02301611), with estimated completion of the
primary endpoint in 2018.
CONCLUSION
Combination Immunotherapy Cancer vaccines represent a nontoxic therapeutic modality
Exploring combination therapy of passive and active im- with great antitumor specificity to prime the immune sys-
munotherapies is a growing area of interest. During the tem, but they have yet to be proven and/or gain acceptance
enrollment of the E75 and GP2 trials, the standard of care for in clinical practice. Based on lessons learned from previous

e166 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


ARMING THE IMMUNE SYSTEM TO PREVENT CANCER RECURRENCE

trials, the most promising use of this therapy is to prevent patients with metastatic disease if given in combination with
recurrence in disease-free patients. Our group has made checkpoint inhibitors.
strides toward showing the efficacy of this approach with
multiple vaccines/strategies applied to several tumor types. DISCLAIMER
Additionally, there is potential synergistic clinical benefit of The view(s) expressed herein are those of the author(s) and
combination therapy with monoclonal antibodies, which do not reflect the official policy or position of San Antonio
needs to be further evaluated. Finally, with a better under- Military Medical Center, Womack Army Medical Center, the
standing of the tumor microenvironment and the tools to U.S. Army Medical Department, the U.S. Army Office of the
protect tumor-reactive T cells in this hostile setting, cancer Surgeon General, the Department of the Army, Department
vaccines may now, largely for the first time, be effective in of Defense, or the U.S. Government.

References

1. Hale DF, Clifton GT, Sears AK, et al. Cancer vaccines: should we be molecular identification as a folate-binding protein. Am J Pathol. 1993;
targeting patients with less aggressive disease? Expert Rev Vaccines. 142:557-567.
2012;11:721-731. 14. Li PY, Del Vecchio S, Fonti R, et al. Local concentration of folate bind-
2. Hoover HC Jr, Brandhorst JS, Peters LC, et al. Adjuvant active specific im- ing protein GP38 in sections of human ovarian carcinoma by in vitro
munotherapy for human colorectal cancer: 6.5-year median follow-up of a quantitative autoradiography. J Nucl Med. 1996;37:665-672.
phase III prospectively randomized trial. J Clin Oncol. 1993;11:390-399. 15. Greene J, Schneble E, Berry J, et al. Preliminary results of the phase I/IIa
3. Vermorken JB, Claessen AM, van Tinteren H, et al. Active specific dose finding trial of a folate binding protein vaccine (E39+GM-CSF) in
immunotherapy for stage II and stage III human colon cancer: a rand- ovarian and endometrial cancer patients to prevent recurrence. J Clin
omised trial. Lancet. 1999;353:345-350. Oncol. 2015;33 (suppl; abstr e14031).
4. Harris JE, Ryan L, Hoover HC Jr, et al. Adjuvant active specific immu- 16. Holmes LM, Li J, Sticca RP, et al. A rapid, novel strategy to induce tumor
notherapy for stage II and III colon cancer with an autologous tumor cell cell-specific cytotoxic T lymphocyte responses using instant den-
vaccine: Eastern Cooperative Oncology Group Study E5283. J Clin Oncol. tritomas. J Immunother. 2001;24:122-129.
2000;18:148-157. 17. Greene JM, Schneble EJ, Jackson DO, et al. A phase I/IIa clinical trial in
5. Wood C, Srivastava P, Bukowski R, et al; C-100-12 RCC Study Group. An stage IV melanoma of an autologous tumor-dendritic cell fusion
adjuvant autologous therapeutic vaccine (HSPPC-96; vitespen) versus (dendritoma) vaccine with low dose interleukin-2. Cancer Immunol
observation alone for patients at high risk of recurrence after ne- Immunother. Epub 2016 Feb 19.
phrectomy for renal cell carcinoma: a multicentre, open-label, rand- 18. Greene J, Hale D, Schneble E, et al. Initial phase I/IIa trial results of an
omised phase III trial. Lancet. 2008;372:145-154. autologous tumor lysate + yeast cell wall particles + dendritic cells
6. Carson WE, III, Unger JM, Sosman JA, et al. Adjuvant vaccine immu- vaccine (TLPLDC) in patients with solid tumors. Cancer Immunol Res.
notherapy of resected, clinically node-negative melanoma: long-term 2016;4(suppl, abstr A044).
outcome and impact of HLA class I antigen expression on overall 19. Eggermont AM, Suciu S, Rutkowski P, et al. Adjuvant ganglioside GM2-KLH/
survival. Cancer Immunol Res. 2014;2:981-987. QS-21 vaccination versus observation after resection of primary tumor
7. Mittendorf EA, Clifton GT, Holmes JP, et al. Final report of the phase I/II . 1.5 mm in patients with stage II melanoma: results of the EORTC 18961
clinical trial of the E75 (nelipepimut-S) vaccine with booster in- randomized phase III trial. J Clin Oncol. 2013;31:3831-3837.
oculations to prevent disease recurrence in high-risk breast cancer 20. Vansteenkiste J, Cho B, Vanakesa T, et al. MAGRIT, a double-blinded,
patients. Ann Oncol. 2014;25(9):1735-1742. randomized, placebo-controlled phase III study to assess the efficacy
8. Benavides LC, Gates JD, Carmichael MG, et al. The impact of HER2/neu of the recMAGE-A3 + AS15 cancer immunotherapeutic as adjuvant
expression level on response to the E75 vaccine: from U.S. Military therapy in patients with resected MAGE-A3-positive non-small cell lung
Cancer Institute Clinical Trials Group Study I-01 and I-02. Clin Cancer cancer (NSCLC). Ann Oncol. 2014;25(suppl 4):iv409-iv16.
Res. 2009;15:2895-2904. 21. Giaccone G, Bazhenova LA, Nemunaitis J, et al. A phase III study of
9. Schneble E, Perez S, Murray J, et al. Primary analysis of the prospective, belagenpumatucel-L, an allogeneic tumour cell vaccine, as mainte-
randomized, phase II trial of GP2+GM-CSF vaccine versus GM-CSF alone nance therapy for non-small cell lung cancer. Eur J Cancer. 2015;51:
administered in the adjuvant setting to high-risk breast cancer patients. 2321-2329.
J Clin Oncol. 2014;32 (suppl 26; abstr 134). 22. Morton D, Mozzillo N, Thompson J, et al. An international, randomized,
10. Mittendorf EA, Schneble EJ, Perez SA, et al. Primary analysis of the phase III trial of bacillus Calmette-Guerin (BCG) plus allogeneic mela-
prospective, randomized, single-blinded phase II trial of AE37 vaccine noma vaccine (MCV) or placebo after complete resection of melanoma
versus GM-CSF alone administered in the adjuvant setting to high-risk metastatic to regional or distant site. J Clin Oncol. 2007;25:8508.
breast cancer patients. J Clin Oncol. 2014;32:5s (suppl; abstr 638). 23. Schulze T, Kemmner W, Weitz J, et al. Efficiency of adjuvant active
11. Peoples GE, Anderson BW, Lee TV, et al. Vaccine implications of folate specific immunization with Newcastle disease virus modified tumor
binding protein, a novel cytotoxic T lymphocyte-recognized antigen cells in colorectal cancer patients following resection of liver metas-
system in epithelial cancers. Clin Cancer Res. 1999;5:4214-4223. tases: results of a prospective randomized trial. Cancer Immunol
12. Peoples GE, Anderson BW, Fisk B, et al. Ovarian cancer-associated Immunother. 2009;58:61-69.
lymphocyte recognition of folate binding protein peptides. Ann Surg 24. Gates JD, Carmichael MG, Benavides LC, et al. Longterm followup as-
Oncol. 1998;5:743-750. sessment of a HER2/neu peptide (E75) vaccine for prevention of re-
13. Garin-Chesa P, Campbell I, Saigo PE, et al. Trophoblast and ovarian currence in high-risk prostate cancer patients. J Am Coll Surg. 2009;208:
cancer antigen LK26. Sensitivity and specificity in immunopathology and 193-201.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e167


GREGORY L. BEATTY

Overcoming Therapeutic Resistance by Targeting Cancer


Inflammation
Gregory L. Beatty, MD, PhD

OVERVIEW

Tumor-infiltrating myeloid cells are a prominent feature of most solid malignancies. This inflammatory immune response,
driven by tumor-intrinsic signaling pathways, is a major checkpoint to therapeutic efficacy achieved with immunotherapy
and standard cytotoxic therapies. To overcome therapeutic resistance mediated by cancer inflammation, ongoing clinical
trials are evaluating strategies that (1) deplete myeloid cells from tumors, (2) inhibit tumor-promoting properties of myeloid
cells, and (3) redirect myeloid cells with tumor-inhibitory activity.

outgrowth of nascent transformed cells,8,9 it has become


T he microenvironment that surrounds solid tumors is
fundamental to tumor biology and a critical barrier to
therapeutic efficacy. A defining feature of this microenvi-
increasingly clear that the immune system, in addition to its
potential to be harnessed for antitumor activity, can be a
ronment is a leukocyte infiltrate that can vary widely in major proponent of tumor development and a barrier to
its complexity.1 For example, some tumors demonstrate a therapeutic efficacy.5,10 This paradoxic role of the immune
robust infiltration of T lymphocytes, whereas others show a system in cancer can be explained by the plasticity of the
near absence or a spatial restriction of T lymphocytes to the immune system, which can acquire either pro- or antitumor
tumor margin.2-4 In contrast, innate immune cells composed properties. For productive immunosurveillance to eliminate
of macrophages, granulocytes, and immature myeloid cells cancer, components of innate and adaptive immunity must
are universally seen within tumors, although to varying be aligned or else peripheral tolerance will ensue.11 In many
degrees.5 In some tumors, this innate immune inflammatory settings, alignment is not achieved because leukocytes
response can be robust, even representing the majority of recruited to tumors most often orchestrate an immuno-
the cellular mass of a tumor lesion. suppressive, rather than an immunostimulatory, microen-
Although inflammation is a well-recognized proponent of vironment that acts to spoil the antitumor potential of
cancer, the phenotype of tumor-infiltrating inflammatory the immune system.12 In preclinical models, elimination
cells is pliable and strongly dependent on cues received of this myeloid response by inhibiting ICAM-1mediated
from the surrounding microenvironment.6 As such, tumor- macrophage recruitment to tumors early during cancer
infiltrating leukocytes can acquire either pro- or antitumor initiation can block tumor development.13 Similarly,
properties.7 With a rapidly advancing understanding of this blockade of colony-stimulating factor 1/receptor colony-
biology regulating the phenotype and recruitment of in- stimulating factor 1 (CSF-1/CSF-1R) signaling, a key signaling
flammatory cells to tumors, novel therapeutic targets have pathway for macrophages, can improve the response to
been identified and are being investigated in early-phase immunotherapy with PD-1 and CTLA-4 antagonists.14
clinical trials for their potential to enhance current standard Tumor-infiltrating inflammatory cells, including macro-
therapies (e.g., radiation and chemotherapy) and to derail phages, dendritic cells, granulocytes, and immature mye-
resistance mechanisms to the success of T celldependent loid cells, are commonly seen across a vast majority of solid
immunotherapies. tumor types. These myeloid cell subsets are recruited to
tumors by a variety of chemoattractants released by both
INFLAMMATION: A HALLMARK OF CANCER malignant cells and nonmalignant cells residing within the
Although the immunosurveillance hypothesis proposed tumor microenvironment. Myeloid cells that infiltrate tu-
by Thomas and Burnet in 1957 postulated a critical role for mors are primarily thought to originate from progenitors in
the immune system in controlling the development and the bone marrow and are recruited to tumor tissue via the

From the Abramson Cancer Center of the University of Pennsylvania, Philadelphia, PA; Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine,
University of Pennsylvania, Philadelphia, PA.

Disclosures of potential conflicts of interest provided by the author are available with the online article at asco.org/edbook.

Corresponding author: Gregory L. Beatty, MD, PhD, Abramson Cancer Center of the University of Pennsylvania, Smilow Center for Translational Research, Room 8-112, Bldg 421, 3400
Civic Center Blvd., Philadelphia, PA 19104-5156; email: gregory.beatty@uphs.upenn.edu.

2016 by American Society of Clinical Oncology.

e168 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


OVERCOMING THERAPEUTIC RESISTANCE BY TARGETING CANCER INFLAMMATION

bloodstream.15 However, the spleen may also be an abundant recently being unraveled. For example, activation of Kras in
source of tumor-infiltrating myeloid cells.16 In contrast, the pancreatic cancer induces the secretion of chemokines, such
contribution to the tumor microenvironment by tissue- as interleukin (IL)-8,21 and myeloid growth factors, including
resident myeloid cells, which are derived from yolk-sac granulocyte-macrophage colony stimulating factor,22 which
progenitors, remains ill defined.17,18 can promote the recruitment and differentiation of myeloid
The recruitment of myeloid cells to the tumor microen- cells with protumor activities.23 PTEN loss in preclinical
vironment is dependent on chemoattractants, such as models of pancreatic cancer has also been associated
chemokine:chemokine receptor interactions.19 For example, with a robust infiltration of inflammatory cells.24 In ad-
the chemokine CCL2 regulates myeloid trafficking through dition, PTEN loss in melanoma has been found to correlate
interaction with chemokine receptor CCR2, which is with T-cell exclusiona finding that was associated with an
expressed at high levels on a subset of monocytes, com- increase in proinflammatory cytokines including CCL2 and
monly referred to as inflammatory or classical mono- vascular endothelial growth factor.25 In this study, neu-
cytes. In some cases, a single chemokine may bind to tralization of vascular endothelial growth factor enhanced
multiple chemokine receptors. For example, CCL5 has T-cell infiltration into tumors, and pharmacologic inhibition
binding affinity for chemokine receptors CCR1, CCR3, and of the PI3K pathway improved the effectiveness of antiPD1
CCR5. Similarly, a single chemokine receptor may also bind immunotherapy. Similar findings have been reported with
multiple chemokines. For example, chemokine receptor activation of the b-catenin pathway, which was shown to
CCR5 can bind both CCL3 and CCL5. Together, this com- regulate the expression of tumor-derived chemokines,
plexity associated with expression of chemokine receptors specifically CCL4.26 Here, CCL4 was important for the re-
on distinct myeloid subsets and the interaction of chemo- cruitment of dendritic cells involved in T-cell priming. This
kine receptors with select chemokines determines, at least emerging role for tumor-intrinsic signaling pathways in
in part, the heterogeneity of the myeloid infiltration into regulating the recruitment of inflammatory cells has also
tumors. been observed in models of prostate adenocarcinoma,
Both malignant and nonmalignant cells can release che- where loss of Smad4 leads to hyperactivation of Hippo-YAP
mokines within the tumor microenvironment that act to signaling, which induces CXCL5 upregulation in cancer cells
regulate the infiltration of myeloid cells. For example, in and subsequent recruitment of immunosuppressive CXCR2+
preclinical models of breast cancer, CCL2 released by CD11b+ Gr-1+ myeloid cells.27 Together, these studies sug-
both malignant and nonmalignant cells is required for gest the importance of tumor genetics in shaping the in-
recruitment of CCR2+ monocytes to promote cancer flammatory immune response to cancer.
metastasis.20 However, the importance of tumor genetics The phenotype of myeloid cells within tumors can be quite
in defining the chemokine milieu within tumors is only variable and probably best described as a spectrum rang-
ing from pro- to antitumor based on functional properties
(e.g., T cellsuppressive activity, cytokine production, tumor-
icidal activity, pro- versus antiangiogenic properties, etc.).7
KEY POINTS However, a diverse array of factors (e.g., extracellular matrix,
hypoxia, cytokines, and other soluble factors) can influence
Tumor-infiltrating myeloid cells, including macrophages, the phenotype of a myeloid cell. As a result, the complexity
dendritic cells, granulocytes, and immature myeloid of the myeloid response to cancer has remained a challenge
cells, are a hallmark of solid malignancies. to describe in vivo. Many attempts have been made to
Tumor-intrinsic oncogenic signaling pathways (e.g.,
categorize tumor-infiltrating myeloid cells based on protein
Kras, PTEN/Akt/PI3K, WNT/b-catenin) shape the
composition, phenotype, and function of tumor-
and molecular signatures determined under in vitro culture
infiltrating myeloid cells with implications for defining conditions. For example, macrophages studied under dis-
tumor immunogenicity. tinct in vitro culture conditions with interferon gamma and
The pro- versus antitumor biology of myeloid cell lipopolysaccharide or IL-4 and IL-13 can be classified as M1
subsets within solid tumors is pliable and determined by or M2 macrophages, respectively.28 However, almost in-
microenvironmental signals released by malignant and variably when macrophages are obtained from in vivo
nonmalignant cells. settings, they more commonly display a mixture of an M1
Strategies to intervene on the myeloid response to and M2 phenotype, thereby diluting the potential relevance
cancer include therapeutic approaches designed to (1) of this simplistic approach.29
deplete myeloid cells from the tumor In cancer, the myeloid cell-differentiation program be-
microenvironment, (2) inhibit the tumor-promoting
comes altered from the earliest stages of myelopoiesis,30
properties of myeloid cells, and (3) redirect tumor-
infiltrating myeloid cells with tumor-inhibitory
occurring within the bone marrow, through myeloid cell
properties. transit in the peripheral blood to infiltration into the tumor
Therapeutic manipulation of tumor-infiltrating myeloid microenvironment.31 At each step along this lifecycle of a
cells holds promise for enhancing the efficacy of T-cell tumor-infiltrating myeloid cell, soluble factors released in
immunotherapy and standard cytotoxic therapies. response to cancer have been identified that shape myeloid
cell biology. For example, in pancreatic cancer, mobilization

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e169


GREGORY L. BEATTY

of CCR2+ monocytes from the bone marrow into the pe- Blocking Recruitment
ripheral blood is enhanced and correlates with elevated Strategies to block inflammatory cell recruitment to tumors
serum levels of CCL2.32 Within the peripheral blood, human involve the neutralization of chemokines with antibodies,
monocytes are then exposed to serum-soluble factors that the delivery of small-molecule inhibitors to block chemokine
can modulate monocyte biology. For example, in patients receptor signaling, and selective depletion of myeloid
with renal cell carcinoma, human blood monocytes were subsets. For example, neutralizing antibodies against CCL2
found to be induced with pro-angiogenic properties by the (e.g., carlumab) and antagonists of CCR2 (e.g., PF-04136309
IL-1IL-1R cytokine axis even before entry into tumor tis- and CCX872) are being evaluated in early phase 1 stud-
sue.33 Once in the tumor microenvironment, tumor-derived ies in patients with advanced solid malignancies to block
soluble factors then further shape the ultimate fate of monocyte recruitment to tumors. Similarly, antagonists of
tumor-infiltrating myeloid cells. However, because the CXCR1/2 (e.g., AZD5069 and reparixin) are being studied in
secretome of malignant cells can vary between tumor-type, early-phase clinical trials to inhibit granulocyte recruitment to
the cellular fate of myeloid cells is tumor-specific and likely tumors. To deplete myeloid cell populations, ongoing clinical
differentially skewed between cancers and perhaps even investigations are testing antagonists of CSF1R (PLX3397,
between lesions within the same patient.34,35 ARRY-382, IMC-CS4, FPA008, AMG820, and RO5509554)
and a chemotherapeutic agent, trabectedin, which was
found to selectively induce capsase-8dependent apoptosis
TARGETING INFLAMMATION FOR THERAPEUTIC in monocytes via signaling through tumor necrosis factor
BENEFIT IN CANCER related apoptosis-inducing ligand receptor 2.40
Changes in inflammatory cell biology that occur in cancer can
affect the efficacy of standard treatments such as chemo-
therapy. In response to cytotoxic stress, tumor cells secrete Inhibiting Activity
chemokines that can recruit myeloid cells, which may then Tumor-infiltrating myeloid cells are active components of
suppress the efficacy of chemotherapy and/or inhibit the the tumor microenvironment. Within tumors, myeloid cells
development of antitumor T celldependent immunity.10,36 shape the supply of nutrients important for T-cell activity
Tumor-infiltrating inflammatory cells can also directly sup- (e.g., tryptophan and arginine) and release soluble factors
press T-cell antitumor immunity by producing immuno- (e.g., IL-6, IL-10, and transforming growth factor beta) that
suppressive cytokines and depleting critical amino acids, can be immunosuppressive and supportive of tumor
including tryptophan and arginine, which are necessary for survival.37,41-43 Ongoing studies are evaluating strategies to
normal T-cell activity.37-39 For these reasons, strategies to inhibit indoleamine 2,3-dioxygenase (IDO), a key enzyme
intervene on cancer-induced inflammation are actively expressed by myeloid cells and important for tryptophan
being investigated in early-phase clinical trials. metabolism in tumors. In preclinical models, IDO inhibition
There are three potential approaches (Table 1) to altering was shown to promote antitumor T-cell activity,37 and in
the inflammatory response to cancer including: clinical trials, IDO inhibition has demonstrated safety and
1. blocking inflammatory cell recruitment to tumors, tolerability.44,45 Similarly, early-phase clinical trials are un-
2. inhibiting tumor-promoting activities of myeloid cells, derway to evaluate the safety and preliminary efficacy of
and inhibiting distinct signaling pathways in myeloid cells, such
3. redirecting tumor-infiltrating myeloid cells with anti- as JAK/STAT signaling, which is important for the biologic
tumor activity. activity of cytokines, including IL-6 and IL-10 (Table 1).

TABLE 1. Therapeutic Strategies to Cancer Inflammation in Solid Malignancies

Approach to Cancer Inflammation Target Agents


Block Infiltration or Deplete Myeloid Subsets CCL2/CCR2 Carlumab, PF-04136309, CCX872
CXCR1/CXCR2 AZD5069, reparixin
CSF-1/CSF-1R PLX3397, ARRY-382, IMC-CS4, FPA008, AMG820, RO5509554
TRAIL-R2 Trabectedin
Inhibit Protumor Activity IDO Epacadostat/INCB024360, indoximod, NLG919, 1-methyl-D-tryptophan
JAK Ruxolitinib, pacritinib, momelotinib, INCB039110
Redirect With Antitumor Activity CD40 RO7007789/CP-870,893, APX005M, ADC-1013, Chi Lob 7/4, SEA-CD40
CSF-1R PLX3397, ARRY-382
CD47-Sirpa CC-90002, TTI-621
Abbreviations: CCL2, chemokine (C-C motif) ligand 2; CCR2, chemokine (C-C motif) receptor 2; CXCR1, chemokine (C-X-C motif) receptor 1; CXCR2, chemokine (C-X-C motif) receptor 2;
CSF-1, colony-stimulating factor 1; CSF-1R, colony-stimulating factor 1 receptor; TRAIL-R2, tumor necrosis factorrelated apoptosis-inducing ligand receptor 2; IDO, indoleamine 2,3
dioxygenase; JAK, Janus kinase; Sirpa, signal regulatory protein alpha.

e170 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


OVERCOMING THERAPEUTIC RESISTANCE BY TARGETING CANCER INFLAMMATION

TABLE 2. Targeting Cancer Inflammation in Combination With Checkpoint Immunotherapy


Therapeutic Approach Targets Agents
Block Infiltration or Deplete Myeloid Subsets in Combination With Checkpoint Inhibition CXCR2, PD-L1 AZD5069, MEDI4736
CSF-1R, PD-1 FPA008, nivolumab
CSF-1R, PD-L1 RO5509554, MPDL3280A
Inhibit Inflammatory Signaling Pathways in Combination With Checkpoint Blockade IDO, PD-1 INCB024360, nivolumab
IDO, PD-1 INCB024360, pembrolizumab
IDO, PD-L1 INCB024360, MEDI4736
IDO, CTLA4 INCB024360, ipilimumab
JAK, PD-1 INCB039110, pembolizumab
Redirect Inflammation in Combination With Checkpoint Blockade CD40, PD-L1 RO7007789, MPDL3280A
CSF1R, PD-1 PLX3397, pembrolizumab
Abbreviations: CXCR2, chemokine (C-X-C motif) receptor 2; CSF-1, colony-stimulating factor 1; CSF-1R, colony-stimulating factor 1 receptor; IDO, indoleamine 2,3 dioxygenase; CTLA4,
cytotoxic T-lymphocyteassociated protein 4; JAK, Janus kinase.

Redirecting Activity these approaches hold promise in combination with cytotoxic


The biology of tumor-infiltrating myeloid cells is pliable therapies and immunotherapy. To this end, multiple clinical
and therefore may potentially be redirected from pro- to trials are underway investigating modulation of cancer in-
antitumor or, alternatively, from immunosuppressive to flammation in combination with immunotherapy (e.g., PD-1/
immunostimulatory. This premise forms the rationale for PD-L1 blockade; Table 2), chemotherapy, and radiation. These
therapeutic approaches designed to unleash the antitumor studies will help to inform whether distinct strategies designed
potential of tumor-infiltrating myeloid cells. For example, to manipulate the myeloid cell response to cancer can com-
ongoing clinical studies are evaluating antagonists of the plement existing therapeutic approaches.
CD47-Sirpa pathway, which delivers inhibitory signals to
macrophages and blocks macrophage phagocytosis of tumor
cells. In preclinical studies, CD47 antagonists have demon- CONCLUSION
strated the capacity to induce macrophage-dependent an- Tumor-infiltrating myeloid cells are a prominent feature
titumor activity46 and promote dendritic cellmediated of most solid malignancies and most often portend a poor
induction of T-cell antitumor immunity. 47 An alternative prognosis. However, the impact of myeloid cells on tumor
approach to redirecting myeloid cells with antitumor development, progression, and growth can vary sub-
properties is to block signaling pathways (e.g., CSF-1), which stantially, which is explained by their inherent cellular
can instill macrophages with protumor functions.48 For plasticity and dependence on local microenvironmental
example, although antibody-based strategies that target cues, which determine their ultimate cellular fate. Tumor-
CSF-1R (e.g., IMC-CS4, FPA008, AMG820, RO5509554) may intrinsic oncogenic signaling pathways have emerged as a
deplete myeloid cells via antibody-dependent cellular major determinant of the complexity of the myeloid cell
cytotoxicity,49 small-molecule inhibitors of CSF-1R (e.g., infiltrate in tumorsa finding that has strong implications
PLX3397, ARRY-382) have the potential for shifting the in understanding resistance mechanisms to the efficacy of
polarity of myeloid cells from pro- to antitumor.48 Finally, cytotoxic and immunotherapeutic strategies. Based on
CD40 agonists have demonstrated the capacity to redirect strong preclinical rationale, ongoing clinical trials are
tumor-infiltrating myeloid cells with both tumoricidal and investigating strategies designed to deplete, inhibit, and
antifibrotic activity.50,51 In recent years, several CD40 ago- redirect myeloid cells in cancer for therapeutic benefit.
nists have been developed and are now undergoing clinical
investigation (Table 1).
Each of the aforementioned strategies for modulating cancer ACKNOWLEDGMENT
inflammation is being evaluated in clinical trials. Although This work was supported by a National Institutes of Health
merely manipulating the myeloid response to cancer is unlikely grant K08 CA138907 and by grant 2013107 from the Doris
by itself, to produce long-lasting and major tumor regressions, Duke Charitable Foundation.

References
1. Coussens LM, Zitvogel L, Palucka AK. Neutralizing tumor-promoting 3. Galon J, Costes A, Sanchez-Cabo F, et al. Type, density, and location of
chronic inflammation: a magic bullet? Science. 2013;339:286-291. immune cells within human colorectal tumors predict clinical outcome.
2. Llosa NJ, Cruise M, Tam A, et al. The vigorous immune microenvi- Science. 2006;313:1960-1964.
ronment of microsatellite instable colon cancer is balanced by multiple 4. Gajewski TF, Schreiber H, Fu YX. Innate and adaptive immune cells in the
counter-inhibitory checkpoints. Cancer Discov. 2015;5:43-51. tumor microenvironment. Nat Immunol. 2013;14:1014-1022.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e171


GREGORY L. BEATTY

5. Qian BZ, Pollard JW. Macrophage diversity enhances tumor progression 28. Mantovani A, Sica A, Locati M. Macrophage polarization comes of age.
and metastasis. Cell. 2010;141:39-51. Immunity. 2005;23:344-346.
6. Lavin Y, Winter D, Blecher-Gonen R, et al. Tissue-resident macrophage 29. Murray PJ, Allen JE, Biswas SK, et al. Macrophage activation and po-
enhancer landscapes are shaped by the local microenvironment. Cell. larization: nomenclature and experimental guidelines. Immunity. 2014;
2014;159:1312-1326. 41:14-20.
7. Long KB, Beatty GL. Harnessing the antitumor potential of macrophages 30. Redente EF, Orlicky DJ, Bouchard RJ, et al. Tumor signaling to the bone
for cancer immunotherapy. OncoImmunology. 2013;2:e26860. marrow changes the phenotype of monocytes and pulmonary mac-
8. Burnet FM. The concept of immunological surveillance. Prog Exp Tumor rophages during urethane-induced primary lung tumorigenesis in A/J
Res. 1970;13:1-27. mice. Am J Pathol. 2007;170:693-708.
9. Thomas L. On immunosurveillance in human cancer. Yale J Biol Med. 31. Sica A, Bronte V. Altered macrophage differentiation and immune
1982;55:329-333. dysfunction in tumor development. J Clin Invest. 2007;117:1155-1166.
10. DeNardo DG, Brennan DJ, Rexhepaj E, et al. Leukocyte complexity 32. Sanford DE, Belt BA, Panni RZ. M, et al. Inflammatory monocyte mo-
predicts breast cancer survival and functionally regulates response to bilization decreases patient survival in pancreatic cancer: a role for
chemotherapy. Cancer Discov. 2011;1:54-67. targeting the CCL2/CCR2 axis. Clin Cancer Res. 2013;19:3404-3415.
11. Mellor AL, Munn DH. Creating immune privilege: active local sup- 33. Chittezhath M, Dhillon MK, Lim JY, et al. Molecular profiling reveals a
pression that benefits friends, but protects foes. Nat Rev Immunol. tumor-promoting phenotype of monocytes and macrophages in human
2008;8:74-80. cancer progression. Immunity. 2014;41:815-829.
12. Beatty GL, Gladney WL. Immune escape mechanisms as a guide for 34. Lundholm M, Hagglof C, Wikberg ML, et al. Secreted Factors from
cancer immunotherapy. Clin Cancer Res. 2015;21:687-692. Colorectal and Prostate Cancer Cells Skew the Immune Response in
13. Liou GY, Doppler H, Necela B, et al. Mutant KRAS-induced expression of Opposite Directions. Sci Rep. 2015;5:15651.
ICAM-1 in pancreatic acinar cells causes attraction of macrophages to 35. Bogels M, Braster R, Nijland PG, et al. Carcinoma origin dictates dif-
expedite the formation of precancerous lesions. Cancer Discov. 2015;5: ferential skewing of monocyte function. OncoImmunology. 2012;1:
52-63. 798-809.
14. Zhu Y, Knolhoff BL, Meyer MA, et al. CSF1/CSF1R blockade reprograms 36. Qian DZ, Rademacher BL, Pittsenbarger J, et al. CCL2 is induced by
tumor-infiltrating macrophages and improves response to T-cell chemotherapy and protects prostate cancer cells from docetaxel-
checkpoint immunotherapy in pancreatic cancer models. Cancer Res. induced cytotoxicity. Prostate. 2010;70:433-442.
2014;74:5057-5069. 37. Liu X, Shin N, Koblish HK, et al. Selective inhibition of IDO1 effectively
15. Franklin RA, Liao W, Sarkar A, et al. The cellular and molecular origin of regulates mediators of antitumor immunity. Blood. 2010;115:
tumor-associated macrophages. Science. 2014;344:921-925. 3520-3530.
16. Cortez-Retamozo V, Etzrodt M, Newton A, et al. Origins of tumor- 38. Bronte V, Zanovello P. Regulation of immune responses by L-arginine
associated macrophages and neutrophils. Proc Natl Acad Sci USA. 2012; metabolism. Nat Rev Immunol. 2005;5:641-654.
109:2491-2496. 39. Kortylewski M, Kujawski M, Wang T, et al. Inhibiting Stat3 signaling in
17. Ostuni R, Kratochvill F, Murray PJ, et al. Macrophages and cancer: from the hematopoietic system elicits multicomponent antitumor immunity.
mechanisms to therapeutic implications. Trends Immunol. 2015;36: Nat Med. 2005;11:1314-1321.
229-239. 40. Germano G, Frapolli R, Belgiovine C, et al. Role of macrophage targeting
18. Gomez Perdiguero E, Klapproth K, Schulz C, et al. Tissue-resident in the antitumor activity of trabectedin. Cancer Cell. 2013;23:249-262.
macrophages originate from yolk-sac-derived erythro-myeloid pro- 41. Lesina M, Kurkowski MU, Ludes K, et al. Stat3/Socs3 activation by IL-6
genitors. Nature. 2015;518:547-551. transsignaling promotes progression of pancreatic intraepithelial
19. Bonecchi R, Locati M, Mantovani A. Chemokines and cancer: a fatal neoplasia and development of pancreatic cancer. Cancer Cell. 2011;19:
attraction. Cancer Cell. 2011;19:434-435. 456-469.
20. Qian BZ, Li J, Zhang H, et al. CCL2 recruits inflammatory monocytes to 42. Spranger S, Spaapen RM, Zha Y, et al. Up-regulation of PD-L1, IDO, and T
facilitate breast-tumour metastasis. Nature. 2011;475:222-225. (regs) in the melanoma tumor microenvironment is driven by CD8(+)
21. Sparmann A, Bar-Sagi D. Ras-induced interleukin-8 expression plays a T cells. Sci Transl Med. 2013;5:200ra116.
critical role in tumor growth and angiogenesis. Cancer Cell. 2004;6: 43. Uyttenhove C, Pilotte L, Theate I, et al. Evidence for a tumoral immune
447-458. resistance mechanism based on tryptophan degradation by indole-
22. Pylayeva-Gupta Y, Lee KE, Hajdu CH, et al. Oncogenic Kras-induced GM- amine 2,3-dioxygenase. Nat Med. 2003;9:1269-1274.
CSF production promotes the development of pancreatic neoplasia. 44. Soliman HH, Jackson E, Neuger T, et al. A first in man phase I trial of the
Cancer Cell. 2012;21:836-847. oral immunomodulator, indoximod, combined with docetaxel in pa-
23. Bayne LJ, Beatty GL, Jhala N, et al. Tumor-derived granulocyte- tients with metastatic solid tumors. Oncotarget. 2014;5:8136-8146.
macrophage colony-stimulating factor regulates myeloid in- 45. Beatty GL, ODwyer PJ, Clark J, et al. Phase I study of the safety,
flammation and T cell immunity in pancreatic cancer. Cancer Cell. 2012; pharmacokinetics (PK), and pharmacodynamics (PD) of the oral in-
21:822-835. hibitor of indoleamine 2,3-dioxygenase (IDO1) INCB024360 in patients
24. Ying H, Elpek KG, Vinjamoori A, et al. PTEN is a major tumor suppressor (pts) with advanced malignancies. J Clin Oncol. 2013;31(suppl; abstr
in pancreatic ductal adenocarcinoma and regulates an NF-kB-cytokine 3025).
network. Cancer Discov. 2011;1:158-169. 46. Weiskopf K, Ring AM, Ho CC, et al. Engineered SIRPa variants as im-
25. Peng W, Chen JQ, Liu C, et al. Loss of PTEN Promotes Resistance to T Cell- munotherapeutic adjuvants to anticancer antibodies. Science. 2013;
Mediated Immunotherapy. Cancer Discov. 2016;6:202-216. 341:88-91.
26. Spranger S, Bao R, Gajewski TF. Melanoma-intrinsic b-catenin 47. Liu X, Pu Y, Cron K, et al. CD47 blockade triggers T cell-mediated de-
signalling prevents anti-tumour immunity. Nature. 2015;523: struction of immunogenic tumors. Nat Med. 2015;21:1209-1215.
231-235. 48. Pyonteck SM, Akkari L, Schuhmacher AJ, et al. CSF-1R inhibition alters
27. Wang G, Lu X, Dey P, et al. Targeting YAP-Dependent MDSC Infiltration macrophage polarization and blocks glioma progression. Nat Med.
Impairs Tumor Progression. Cancer Discov. 2016;6:80-95. 2013;19:1264-1272.

e172 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


OVERCOMING THERAPEUTIC RESISTANCE BY TARGETING CANCER INFLAMMATION

49. Ries CH, Cannarile MA, Hoves S, et al. Targeting tumor-associated in patients with advanced pancreatic ductal adenocarcinoma. Clin Cancer
macrophages with anti-CSF-1R antibody reveals a strategy for cancer Res. 2013;19:6286-6295.
therapy. Cancer Cell. 2014;25:846-859. 51. Beatty GL, Chiorean EG, Fishman MP, et al. CD40 agonists alter tumor
50. Beatty GL, Torigian DA, Chiorean EG, et al. A phase I study of an agonist stroma and show efficacy against pancreatic carcinoma in mice and
CD40 monoclonal antibody (CP-870,893) in combination with gemcitabine humans. Science. 2011;331:1612-1616.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e173


DEVELOPMENTAL THERAPEUTICS AND
TRANSLATIONAL RESEARCH

Pharmacokinetics, Dynamics,
and Genomics in the Era of
Immunotherapy and Small
Molecules

CHAIR
Emiliano Calvo, MD, PhD
START Madrid, Centro Integral Oncologico
Clara Campal
Madrid, Spain

SPEAKERS
Christine Walko, PharmD, BCOP
Moffitt Cancer Center
Tampa, FL

E. Claire Dees, MD
The University of North Carolina at Chapel Hill
Chapel Hill, NC
PHARMACOGENOMICS, PHARMACOKINETICS, AND PHARMACODYNAMICS OF TARGETED THERAPIES

Pharmacogenomics, Pharmacokinetics, and


Pharmacodynamics in the Era of Targeted Therapies
Emiliano Calvo, MD, PhD, Christine Walko, PharmD, FCCP, BCOP, E. Claire Dees, MD, and Belen Valenzuela,
PharmD, PhD

OVERVIEW

The complex nature of the pharmacologic aspects of cancer therapeutics has become more apparent in the past several years
with the arrival of a cascade of target-based agents and the difficult challenge of bringing individualized precision medicine to
oncology. Interpatient variability in drug action, singularly in novel agents, is in part caused by pharmacogenomic (PG),
pharmacokinetic, and pharmacodynamic (PD) factors, and drug selection and dosing should take this into consideration to
optimize the benefit for our patients in terms of antitumor activity and treatment tolerance. In this regard, somatic genetic
evaluation of tumors is useful in not only predicting response to initial targeted therapies but also in anticipating and guiding
therapy after the development of acquired resistance; therapeutic drug monitoring of novel small molecules and
monoclonal antibodies must be incorporated in our day-to-day practice to minimize the negative effect on clinical outcome
of interindividual variability on pharmacokinetic processes of these drugs for all patients, but especially for fragile patient
populations and those with organ dysfunction or comorbidities. For these populations, incorporating frailty assessment
tools into trials of newer agents and validating frailty-based dose adjustment should be an important part of further drug
development.

majority of patients. In fact, most failures in phase III1 trials


O ver the past several years, we have witnessed a dra-
matic and encouraging burst of newly approved anti-
cancer therapeutics. The treatment of cancer has continued
are related to a lack of efficacy that can be attributed to
underdosing situations, because interindividual variability
to shift from traditional cytotoxic therapies aimed at inhibiting in pharmacokinetic processes plays a critical role in drug
the growth of fast-dividing cells toward targeting a particular exposure variability and, consequently, in the clinical outcome
cellular pathway that appears to be activated in a patients observed. In addition, dosing anticancer chemotherapy in
particular cancer. The concept of individualized therapy elderly individuals and frail patients has long been a problem in
embraces the idea that each patient and his or her cancer oncology. Because the pivotal clinical trials of most anticancer
both have unique properties, including how the drug will be chemotherapeutics included only patients with normal organ
handled in the body and whether it will be effective against function and excellent performance status, we have limited
the patients cancer. Many of these new drugs are targeted information in dosing a more real-world population.
agents, small molecules, and monoclonal antibodies with Therefore, we review some of the data available on the vari-
unique biologic effects and challenges in optimizing their clinical ability in drug action, particularly in newer agents, caused by
use include variability in pharmacogenomics, pharmacokinetics, genomic, kinetic, and dynamic factors and how dosing should be
and pharmacodynamics among patients with cancer. approached in the context of individualized precision medicine.
Pharmacogenomic analysis of tumor tissue is an important
tool in identifying therapies that will be effective against
certain cancers while also preventing patients for whom ROLE OF PHARMACOGENOMICS IN THE
these therapies are unlikely to be effective from suffering MOLECULAR ERA
the physical and economic toxicities inherent to each The process of dissecting the heterogeneity of an individual
treatment. Tyrosine kinase inhibitors (TKIs) and monoclonal patients cancer begins as soon as a sample of the malignancy
antibodies are administered at fixed-flat or weight-based is able to be assessed and subjected to a gamut of staining,
doses, and frequently, they are not the right dose for the immunohistochemistry, fluorescence in situ hybridization,

From the DeBartolo Family Personalized Medicine Institute, H. Lee Moffitt Cancer Center, Tampa, FL; UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC; Platform of
Oncology, Hospital Quiron,
Torrevieja, Alicante, Spain; START Madrid, Early Clinical Drug Development Program, Centro Integral Oncologico
Clara Campal, Madrid, Spain.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Emiliano Calvo, MD, PhD, START Madrid, Early Clinical Drug Development Program, Centro Integral Oncologico
~a, 10, 28050 Madrid,
Clara Campal, Calle On
Spain; email: emiliano.calvo@start.stoh.com.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e175


CALVO ET AL

and cytogenetics, as well as genetic and methylation as- or panitumumab will be useful in the management of the
says in some instances. All of these provide both prog- tumor.4 This emphasizes not only finding the right biomarker
nostic and predictive information used to begin to narrow for each agent but also ensuring that the selected patient
down specific treatment options and expected outcomes. sample will assess the value of this biomarker. Ideal predictive
The discovery of specific biomarkers has been essential for biomarker trials should include two comparison treatment
drug development. For example, the initial trial of pan- groups and should demonstrate that the outcome is different
itumumab and best supportive care (BSC) compared with BSC for patients with biomarker-positive results compared with
alone among patients with metastatic colorectal cancer that those with biomarker-negative results.5
had progressed after standard therapy demonstrated The role of somatic mutation testing has continued to
an improvement in median progression-free survival (mPFS) evolve past the initial diagnosis of tumors to help prioritize
from 7.3 weeks to 8 weeks in the BSC and panitumumab and guide additional treatment options. Improved under-
groups, respectively. In comparison, mean progression-free standing of cancer biology and drug pharmacology has
survival (PFS) was 8.5 weeks and 13.8 weeks for the BSC allowed for expanded investigation into novel therapy
and panitumumab groups, respectively, suggesting the ex- options. Greater understanding of the role of a mutated pro-
istence of a subset of patients who responded better to the tein in a process essential to cancer survival, such as DNA
panitumumab.2 Unlike the story of the HER2-inhibitor tras- repair, has supported the interrogation of alterations in genes
tuzumab, overexpression of EGFR was not the predictive involved in this process in terms of response to DNA-repair
biomarker for the EGFR inhibitor. Rather, mutations in the inhibitors like the PARP inhibitor, olaparib. PARP inhibitors
Kirsten rat sarcoma (KRAS) oncogene downstream of EGFR were initially developed with the hope of taking advantage
were predictive of responses to these inhibitors in colorectal of synthetic lethality for patients deficient in the tumor sup-
cancer, as evidenced by a secondary analysis of the same trial pressor genes BRCA1 and BRCA2. PARP is required for base-
showing KRAS mutations in 43% of the patients and corre- excision repair of damaged DNA, whereas BRCA is involved
lation with mPFS. For patients with wild-type KRAS, pan- in homologous repair. In a BRCA1- or BRCA2-deficient tumor,
itumumab resulted in mPFS of 12.3 weeks compared with PARP is able to compensate for this loss of homologous repair;
7.3 weeks with BSC, whereas patients with KRAS mutations thus, inhibition of PARP would ultimately result in cell death.6
had similar mPFS regardless of treatment (7.4 weeks com- The PARP inhibitor olaparib was initially approved by the U.S.
pared with 7.3 weeks in the panitumumab and BSC groups, Food and Drug Administration (FDA) in December 2014 for
respectively).3 Based on these and additional findings, a newly patients with ovarian cancer with defective BRCA genes. Nu-
diagnosed metastatic colorectal cancer is standardly assessed merous other mutations can occur in genes involved in DNA
for the alterations in KRAS and NRAS and this helps to de- repair and are common in cancers, including prostate cancer.
termine whether EGFR-directed therapies such as cetuximab The clinical efficacy of olaparib in metastatic castration-
resistant prostate cancer was assessed among 16 patients
with homologous deletions, deleterious mutations, or both in
DNA-repair genes. These genes included ATM, FANCA, CHEK2,
KEY POINTS PALB2, and others, including BRCA1 and BRCA2. A response
was seen for 88% of patients, including four of five patients
Precision medicine in oncology must take into account with ATM alterations.7 Ongoing trials with olaparib and inves-
interpatient variability in drug action, particularly in tigational PARP inhibitors are underway for patients with a
newer agents, caused by genomic, kinetic, and dynamic variety of tumor types with DNA-repair gene alterations.
factors.
As our knowledge about cancer biology grows, so too
Pharmacogenomics of somatic tumors, through genetic
analyses of solid samples and liquid biopsies of the
does the cancers ability to overcome these novel therapies.
disease, relates not only to initial selection of therapy Somatic genetic evaluation of tumors is useful in not only
but also to sequencing of therapy options. predicting response to initial targeted therapies but also
Interindividual variability on pharmacokinetic processes in anticipating and directing therapy after the development
is a definite source of imprecision in the use of targeted of acquired resistance. EGFR-activating mutations were
drugs, with profound clinical consequences, especially in identified in 2004 and are found for approximately 10%
frail patients, that can be decreased through therapeutic of patients with nonsmall cell lung cancer (NSCLC) with
drug monitoring. adenocarcinoma histology. The majority of these EGFR-
Measuring frailty might become an important focus of activating mutations occur in exons 18 to 21 and predict
oncology research, because some of these tools response to EGFR TKIs including erlotinib, gefitinib, and
accurately predict toxicity and may prompt
afatinib. Median PFS with these agents is approximately
management change.
To continue to establish genetic biomarkers and
9 to 14 months when resistance develops. The EGFR
therapeutic drug monitoring of novel therapies, we T790M mutation is responsible for approximately 60% of the
must evolve the designs of our clinical trials, make them acquired resistance and prevents TKI binding through steric
as inclusive as possible, and facilitate access to patients hindrance and increased EGFR affinity for ATP.8,9 Novel anti-
willing to enroll in them. EGFR TKIs are being developed to overcome this specific
mutation and include the recently approved osimertinib,

e176 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


PHARMACOGENOMICS, PHARMACOKINETICS, AND PHARMACODYNAMICS OF TARGETED THERAPIES

which has affinity for both activating mutations in EGFR and PHARMACOKINETICS VARIABILITY AND
the T790M. The use of serial sequencing is necessary for THERAPEUTIC DRUG MONITORING OF
depiction of this mutational landscape. The value of this NEWER AGENTS
sequential genetic analysis and integration with correlative Oral Targeted Therapies
science is illustrated by a case report of a patient with NSCLC TKIs are the main oral targeted therapies and comprise a
harboring an anaplastic lymphoma kinase (ALK) rearrange- growing group of drugs for the personalized treatment of
ment. The patient was initially started on crizotinib and both hematologic and solid tumors. To date, more than 15
developed resistance to the agent after 18 months. Genetic TKIs have been approved and many others are at different
analysis at this time showed an ALK C1156Y mutation known stages of their clinical development.
to confer resistance to crizotinib. The patient was then treated Regarding the absorption process, limited solubility of TKIs
with ceritinib and other non-ALK directed therapies until she in the gastrointestinal tract, gastrointestinal surgery, and
was ultimately enrolled in a clinical trial with a novel ALK in- first-pass metabolism by enterocytes and the liver are the
hibitor, lorlatinib. The patient initially had a 41% decrease in main factors limiting their oral absorption. Moreover, con-
her tumor and a response that lasted for 8 months. After comitant administration with food significantly modifies
progressive disease, a subsequent genetic analysis of the tu- plasma levels for some TKIs. For example, lapatinib and nilo-
mor showed the C1156Y mutation but also a new L1198F tinib exposure is increased more than 50% with high-fat meals,
mutation. This later mutation results in resistance to lorlatinib whereas no influence of food has been reported for imatinib,
but paradoxically enhances response to the original crizotinib, dasatinib, sunitinib, or sorafenib absorption. In the opposite
ultimately negating the C1156Y mutation.10 This was elegantly case, drug exposure of axitinib and pazopanib is increased by
demonstrated with correlative in vitro studies and crystal 49% and twofold, respectively, under fasting conditions.1,14,15
structures of the mutations and drug therapies, underscoring Finally, adherence to treatment is another factor that deter-
the importance of team science approaches to interrogating mines variability in drug exposure. Thus, poor imatinib compli-
extraordinary responding patients. Exploring and determining ance results in very low trough plasma concentrations, reduces
the clinical effect a mutation can have on a particular protein cytogenetic responses, and contributes to treatment failure.16,17
can help develop the library of known mutations and further There are a number of important host-related variables
explore a variant of nearly known significance. These var- that are correlated with variability in toxic and therapeutic
iants are found in clinically relevant genes in areas of the gene drug response. For example, TKIs bind to plasma proteins,
that are known to related to clinical function but the exact mainly albumin and alpha-1-acid glycoprotein (AGP). Be-
variant itself has not been functionally characterized. En- cause the only unbound (free) drugs are pharmacologically
couraging collaborative science exploration of these variants active, hypoalbuminemia secondary to cachexia or liver
using functional assays and pharmacologic insight will help to metastases can increase the amount of free drug, leading to
elucidate the clinical value of these types of variants. increased toxicity. In fact, AGP levels have been correlated
Finally, it may not be the specific mutation itself but rather with imatinib and erlotinib clearance. Sarcopenia and low
a number of mutations that may help to predict benefit from body mass index have been also shown to be predictors of
therapy. The search for a reproducible biomarker to predict dose-limiting toxicity of sunitinib and sorafenib.18,19 The
the response to novel immunotherapies, including the PD-1 major route of elimination of TKIs is metabolism by CYP3A4
inhibitors nivolumab and pembrolizumab as well as the CTLA-4 enzymes. Hepatic expression on these enzymes may vary
inhibitor ipilimumab, has yielded limited results. A promising more than 20-fold among patients and may lead to vari-
finding demonstrated across tumor types and immunother- ability in plasma levels. Moreover, genetic polymorphisms
apies is the correlation between the number of mutations and influence TKI metabolism, as has been determined for
clinical response to treatment. Whole-exome sequencing of CYP2D6 and gefitinib or UGT1A9 and sorafenib.16 It is also
34 patients with NSCLC treated with pembrolizumab dem- important to take into account the potential inhibition or
onstrated longer PFS in tumors with a higher number of induction of metabolic enzymes as a result of drugdrug
nonsynonymous mutations (hazard ratio 0.19; 95% CI, interactions because patients with cancer are frequently
0.080.47, p = .0004). The median number of nonsynonymous receiving more than one drug treatment. Finally, chronic ad-
mutations among patients with a durable clinical response ministration may also induce drug elimination, as occurred
and those without was 302 and 148, respectively.11 This was with imatinib among patients with a gastrointestinal stro-
also shown for patients with melanoma who were receiving mal tumor, in which clearance was increased by 33% after
ipilimumab, in which those with more than 100 nonsyn- 3 months of treatment.20
onymous mutations as well as a specific neoantigen land- There is accumulating evidence for potential benefits of
scape demonstrated improved survival.12 Finally, tumors therapeutic drug monitoring (TDM) in the treatment of
with mismatch repair deficiency, which is associated with TKIs.21 Relationships between exposure and response
a higher number of mutations, were also associated with (efficacy/toxicity) have been established for most TKIs. Area
improved mPFS from pembrolizumab.13 The specific under the plasma concentration-time curve (AUC) and
mutation cut point is dependent on the analysis and will trough plasma concentration (Cmin) are the pharmacokinetic
need further prospective validation trials to confirm this parameters often correlated with clinical outcomes and/or
biomarker. toxicity effects of TKIs in several types of cancer.22 For

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e177


CALVO ET AL

imatinib, the pharmacokinetic target has been retrospec- been totally elucidated and several mechanisms have been
tively established in both patients with chronic myeloid proposed, including proteolysis by the liver and re-
leukemia (CML) and those with gastrointestinal stromal ticuloendothelial system, target-mediated elimination, and
tumor. For imatinib, a Cmin value below 1,000 ng/mL is nonspecific endocytosis.29 Because the target-mediated
associated with lower clinical benefit and shorter time to route of elimination depends on the amount of the target
progression. To validate this pharmacokinetic target and expression and the affinity to binding, it leads to nonlinear
include TDM as a clinical routine tool for patients receiving and time-dependent changes in clearance.
imatinib treatment, two prospective randomized controlled Although TDM studies for monoclonal antibodies are
trials were performed.22,23 The final results of the ran- limited, exposure-response relationships have been de-
domized OPTIM imatinib study of 133 patients with chronic scribed. Strong exposure response has determined for
phase CML were recently communicated,24 which showed trastuzumab. Thus, the results of a case-matched control
that only one-third of patients were correctly treated with comparison study showed that patients with the lowest
the standard fixed dose (i.e., these patients achieved the quartile of trastuzumab exposure at the end of the cycle 1
Cmin threshold of 1,000 ng/mL) and two-thirds of patients (Cmin , 11.8 mg/L on day 21) had a median overall survival
were not exposed enough to imatinib. The imatinib dose was 8 months shorter than in the other quartiles, whereas no
increased guided by TDM to obtain the Cmin target and relationship was found between exposure and toxicity.30
resulted in a higher major molecular response rate at Based on these results, the FDA review team recommended
12 months (63% vs. 37%). Moreover, among 493 patients performing a prospective trial of trastuzumab TDM-dose
with imatinib-resistant or imatinib-intolerant CML treated intensification among patients who were underexposed.28
with nilotinib, it was recently demonstrated that those who An exposure-response relationship for trastuzumab-emtansine
had a nilotinib Cmin below 500 ng/mL had a significantly was also found among patients with HER2-positive meta-
longer time to achieve complete cytogenetic response and static breast cancer. After adjusting for baseline risk factors,
molecular response. In addition, nilotinib Cmin was also a higher trough concentration at day 21 (the end of cycle 1)
correlated with elevations in total bilirubin and lipase was associated with improved efficacy (measured as overall
levels.24 For dasatinib, results of the prospective OPTIM survival, PFS, and objective response rates).31 These results
dasatinib trial among patients newly diagnosed with justify that trastuzumab-emtansine TDM-guided dosing
chronic-phase-CML demonstrated that a dasatinib maxi- is a necessary tool to improve patient exposure and avoid
mum serum concentration (Cmax) above 50 ng/mL was as- inadequate exposure.
sociated with clinical response and a Cmin below 2.5 ng/mL For patients with metastatic colorectal cancer treated with
prevented fluid retention and pleural effusion.25 These re- cetuximab, a relationship between trough serum concen-
sults provide a strong rationale to use TDM as a new strategy tration and clinical benefit was also found. Thereby, patients
for optimizing the drug dosage to maximize the clinical with a Cmin below the median value of 40 mg/L at day 14 had
benefit (faster and more pronounced clinical responses) for significantly shorter mPFS than patients with a Cmin above
patients with CML and to promote another prospective this value (3.3 vs. 7.8 months).32
clinical trials to clarify the pharmacokinetic targets for more Similar results to those seen in solid tumors had been
TKIs. reported as well for hematologic malignancies. Thus, Cmin
values of alemtuzumab,27 rituximab,27 and obinutuzumab33
were higher in responders versus nonresponders and were
Monoclonal Antibodies related to better clinical outcomes.
The pharmacokinetics of monoclonal antibodies are complex
and different from others anticancer drugs but their in-
terindividual variability on pharmacokinetic processes is DOSING PATIENTS WHO ARE ELDERLY OR FRAIL:
similar to that observed for TKIs (for example, trastuzumab PHARMACODYNAMIC CHALLENGES
clearance varies up to 40%). Monoclonal antibodies are Although people older than age 65 comprise approximately
dosed by body weight or at fixed doses but high variability in 60% of all patients with newly diagnosed cancer, only 36% of
mAb exposure has been observed after their administration patients enrolled in clinical trials are age 65 or older.34 This
at the labeled dose, supporting the need to individualize becomes a more important disparity as the population ages
dosing to account for variability and ensure efficacy.26,27 and as the population of patients with cancer ages. Many
Absorption and elimination are the main sources of phar- oncologists empirically dose reduce older patients, often
macokinetic variability. Absorption after subcutaneous or without much data to guide them. There are growing efforts
intramuscular administrations is slow, incomplete, and among several collaborative and cooperative groups to
variable. The Cmax is reached between 2 and 8 days after evaluate anticancer therapeutics specifically in elderly
administration. Factors that have been identified as po- populations. In addition, techniques for measuring frailty,
tential sources to limit mAb absorption are loss through independent of age, have been developed, are being in-
presystemic catabolism, flow of blood and lymph, age, body corporated into clinical trials, and may help guide dosing.
weight (especially in obese patients), and injection site.28 Because marrow reserve and organ function often decline
The elimination routes of monoclonal antibodies have not with age, efforts to evaluate therapies specifically for patients

e178 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


PHARMACOGENOMICS, PHARMACOKINETICS, AND PHARMACODYNAMICS OF TARGETED THERAPIES

with organ dysfunction are particularly relevant to the ap- effective than the standard regimens. Thus, it also was not
propriate treatment of older adults with cancer. recommended as an acceptable alternative for elderly
Several commonly used cytotoxic chemotherapy regimens patients.43
have been evaluated for older patients and those with The taxanes are one of the most widely used classes of
impaired organ function. Yet there is variability in regard to chemotherapy for breast cancer as well as for other tumor
which traditional anticancer chemotherapy agents need types. The toxicity of this class of drugs has been specifically
dose reductions in these populations. If we look at breast evaluated in elderly patients in several trials. Docetaxel was
cancer chemotherapy as an example, there appears to be no evaluated in the above-described trial.42 The relationship
age-related difference in toxicity in the common breast between age and paclitaxel pharmacokinetics and toxicity
cancer adjuvant therapy regimens, doxorubicin and cyclo- was evaluated in a CALGB trial, which enrolled 155 patients
phosphamide (AC) or cyclophosphamide, epirubicin, and ranging in age from 55 to 86. This study found age-related
fluorouracil (CEF). One small study evaluated AC across a decline in paclitaxel clearance and in severity of neutropenia,
range of ages and found no age-related differences in which did not appear to result in adverse clinical sequelae
neutropenia complications, cardiac dysfunction, or quality of such as hospitalization or infection.44 In a subsequent pooled
life.35 Another small study evaluated CEF and similarly found analysis of two other cooperative group trials, these same
no age-related difference in leukopenia or other toxicity investigators again demonstrated an age-related increase in
among patients older than age 70 compared with those leukopenia as well as anorexia, hyperbilirubinemia, and
younger than age 70.36 In a large cooperative group trial neuropathy. However, no age-related difference in pacli-
evaluating doses of AC, there was no difference in grade 4 taxel efficacy was noted.45 Finally, Hurria et al46 recently
toxicity across age strata.37 By contrast, several investigators evaluated nab-paclitaxel among older patients across an age
have found that the adjuvant regimen cyclophosphamide, range. Although these investigators found an age-related
methotrexate, and fluorouracil (CMF) is less well tolerated increase in AUC (borderline significance), there was no re-
by older patients than by their younger counterparts.38,39 lationship between age and dose reduction, dose omissions,
However, this disparity in toxicity may be partly explained by or grade 3 toxicity. However, the chemotherapy toxicity risk
age-related renal dysfunction. At least one study has shown score did predict increased toxicity. This score (which included
that apparent age-related differences in toxicity with CMF age and variables such as CrCl, anemia, hearing impairment,
were abrogated by dosing according to creatinine clearance falls, and need for assistance with instrumental activities of
(CrCl).40 daily living) is an attempt to capture frailty, which may be
The Cancer and Leukemia Group B (CALGB; now ALLIANCE) more important than chronological age in toxicity prediction
evaluated age-related toxicity in three of their trials of and treatment and dosing decisions.46
adjuvant chemotherapy in node-positive breast cancer. The Measuring frailty has become an important focus of ge-
regimens were three schedules of CAF, AC with paclitaxel, or riatric oncology research. Several tools have been de-
AC without paclitaxel and two schedules of AC followed by veloped, including the chemotherapy toxicity prediction tool
paclitaxel. Interestingly, even in these relatively modern described above and used by the Cancer and Aging Research
trials (accrued from 1985 to 1999), only 7% of patients Group (CARG)47 and the Chemotherapy Risk Assessment
enrolled were age 65 or older. Nevertheless, this pooled Scale for High-Age Patients (CRASH) score.48 The types and
retrospective analysis was able to show that patients age 65 utility of geriatric assessments have recently been the
or older are 66% more likely to have grade 4 hematologic subject of a consensus paper from the International Society
toxicity than younger patients, discontinue therapy early of Geriatric Oncology.49An intriguing new imaging tool for
more commonly, and have a higher incidence of treatment- measuring frailty by assessing sarcopenia (or skeletal muscle
related death.41 wasting) on CT scans has also been shown to predict che-
More recently, several large randomized trials tested al- motherapy toxicity and outcomes (reviewed in Kazemi-
ternate adjuvant breast cancer regimens in older popu- Bajestani et al50).
lations. For example, in the Italian ELDA trial, women age 65 Although it is clear from multiple studies that some of these
to 79 were randomly assigned to receive adjuvant CMF or tools measuring frailty accurately predict toxicity and may
weekly docetaxel. No difference in disease-free or overall prompt management change, studies validating that dosing
survival was seen. The pattern of toxicity was notable for based on risk scores yield better outcomes still are needed.
worse hematologic toxicity in the CMF arm and worse Similar to our conundrum with the more traditional
nonhematologic toxicity in the docetaxel arm. Furthermore, chemotherapy drugs, we have little data to guide us in
docetaxel resulted in worse quality of life for participants. treating elderly individuals, frail patients, or patients with
Thus, docetaxel was not recommended as an acceptable organ dysfunction with novel targeted agents. Table 1 reviews
alternate for elderly patients.42 CALGB (now ALLIANCE) many of the agents approved in 2014 to 2015, the age range
conducted a randomized trial of adjuvant breast cancer of patients enrolled in the pivotal trials, and data in the
therapy for older patients comparing oral capecitabine to package insert regarding dosing in geriatric patients and
either CMF or AC. This randomized study enrolled 633 those with organ dysfunction.
patients, 65% of whom were older than age 70. Although It is encouraging to note that the majority of the pivotal
capecitabine was less toxic, it was found to be less trials for these newer targeted therapies enrolled a higher

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e179


CALVO ET AL

TABLE 1. Recently Approved Anticancer Therapeutics


Median Package Insert Guidelines
Patient Age in
Mechanism Pivotal Trial, Geriatric Renal Hepatic
Drug Name Indication of Action Years (Range) Use Impairment Impairment Reference
Alectinib ALK plus NSCLC ALK inhibitor 54 (2979); No data No data in severe No data in Shaw et al55
18% age moderate
65 and older to severe
52 (2279); Ou et al56
10% age
65 and older
Belinostat Peripheral HDAC inhibitor 64 (2881); No need No data in CrCl , No data in OConnor et al57
T cell 48% age for dose 39 mL/min moderate
lymphoma 65 and older reduction to severe
Blinatumomab Ph- preB- Bispecific Ab 32 (1877); Patients older No data No data Topp et al58
cell ALL to CD19/CD3 13% age than age 65
65 and older had higher
neuro- and
infectious
toxicity
Ceritinib ALK plus ALK inhibitor 53 (2280); No difference in Dose reduce for No data in Shaw et al59
NSCLC 16% age toxicity or CrCl , 30 moderate
65 and older efficacy mL/min to severe
Cobimetinib BRAF MEK inhibitor 55 (2388) No data No data in severe No data in Larkin et al60
mutation moderate
melanoma to severe
Daratumumab MM Anti-CD38 64 (4476); No differences No dose No data in Lokhorst et al61
45% age in safety or adjustment moderate
65 and older efficacy needed to severe
Elotuzumab MM Anti-SLAMF7 67 (3788); No comment No data No data Lonial et al62
57% age
65 and older
Ibrutinib Mantle BTK inhibitor 67 (5678); Toxicity more No dose Dose reduce in Dreyling et al63
cell, CLL 62% age frequent adjustment mild. Avoid in
65 and older among for CrCl . 25 moderate
patients mL/min. No to severe
older than data for CrCl
age 65 , 25 mL/min
67 (3086) Byrd et al64
61% older
than age 65
Idelalisib CLL, B-cell PI3K-delta 71 (4890); Higher No dose Increased Furman et al65
NHL, SLL inhibitor 78% age incidence modification AUC if
65 and older of SAEs for CrCl . 15 LFTs . ULN
and death mL/min
Ixazomib MM Proteasome 55% age 65 No difference Dose reduce for Dose reduce Moureau et al66
inhibitor and older in safety or CrCl , 30 for total
efficacy mL/min bilirubin .
1.53 ULN
Levatinib Thyroid Kinase inhibitor 64 (NR); No difference Dose reduce for Dose reduce Schlumberger
cancer 45% age in safety or CrCl , 30 for Child- et al67
65 and older efficacy mL/min Pugh C
Necitumumab Squamous EGFR antagonist 62 (3284); Higher incidence No data No data Thatcher et al68
NSCLC 39% age of VTE among
65 and older patients older
than age 70
Nivolumab Melanoma AntiPD-1 59 (2388); No difference No need for No dose Weber et al69
squamous 35% age in safety or dose reduction reduction
NSCLC 65 and older efficacy in mild. No
data in
moderate
to severe

Continued

e180 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


PHARMACOGENOMICS, PHARMACOKINETICS, AND PHARMACODYNAMICS OF TARGETED THERAPIES

TABLE 1. Recently Approved Anticancer Therapeutics (Contd)


Median Package Insert Guidelines
Patient Age in
Mechanism Pivotal Trial, Geriatric Renal Hepatic
Drug Name Indication of Action Years (Range) Use Impairment Impairment Reference
61 (3784); Borghaei et al70
37% age
65 and older
Olaparib BRCA PARP inhibitor 57 (2979); Grade 3 AEs No data if CrCl No data Kaufman et al71
mutation 20% age more , 50 mL/min
ovarian 65 and older frequent
cancer among
patients
older than
age 65
Osimertinib EGFR EGFR inhibitor 45% older Grade 3 and No data if No data in Ramalingam
mutation than age 65 4 AEs more CrCl , 30 moderate et al72
NSCLC frequent mL/min to severe
among
patients
older than
age 65
Palbociclib ER+ HER22 CDK4/6 inhibitor 63 (5471); No difference No dose No dose Finn et al73
breast 44% age in safety or reduction reduction
cancer 65 and older efficacy required for in mild; no
CrCl . 30 data in
mL/min. No moderate
data in severe to severe
Panobinostat MM Pan-deacetylase 63 (5659) More GI, No dose Dose San-Miguel
inhibitor 42% age cardiac, and reduction modification et al74
65 and older hematologic needed for mild
AEs in older and moderate
patients
Pembrolizumab Melanoma AntiPD-1 60 (2594) No difference No dose No dose Hamid et al75
39% age in safety or adjustment adjustment
65 and older efficacy needed needed in
mild. No
data in
moderate
to severe
Sonidegib Basal cell Smoothened 65 (2493) No difference in No dose No dose Migden et al76
carcinoma receptor 52% age effectiveness; adjustment adjustment
antagonist 65 and older higher incidence needed needed in
of grade 3 or mild. No data
4 AEs in older in moderate
patients to severe
Talimogene Melanoma Oncolytic virus 63 (2294) No difference No data No data Andtbacka et al77
Laherparepvec 48% age in safety or
65 and older efficacy
Abbreviations: ALK, anaplastic lymphoma kinase; NSCLC, nonsmall cell lung cancer; ALL, acute lymphoblastic leukemia; MM, multiple myeloma; CLL, chronic lymphocytic
leukemia; NHL, non-Hodgkin lymphoma; SLL, small lymphocytic lymphoma; HDAC, histone deacetylase; Ab, antibody; MEK, mitogen-activated protein kinase; BTK, Brutons
tyrosine kinase; PI3K, phosphoinositide 3-kinase; CDK, cyclin-dependent kinase; SAE, serious adverse event; VTE, venous thromboembolism; AE, adverse event; GI,
gastrointestinal; CrCl, creatinine clearance; AUC, area under the plasma concentration-time curve; LFT, liver function test; ULN, upper limit of normal; Ph, Philadelphia
chromosome.

proportion of older patients than the historical trials sum- to lessen confidence. These new targeted therapies and
marized above. More than 40% patients were age 65 or older immunotherapies also vary in need for dose reduction for
in 12 of the 20 trials highlighted. The relationship between renal or hepatic dysfunction (Table 1).
age and toxicity appears to vary between drugs. Some Few targeted therapies have been specifically evaluated to
studies have noted increased toxicity in older patients date in populations who have organ dysfunction or are el-
(ibrutinib, idelalisib, necitumumab, osimertinib, panobino- derly or frail. Several examples of treatments that have been
stat, sonidegib, blinatumumab, olaparib). Others report no assessed in special populations include the CARG study of
age-related increase in toxicity (belinostat, ceritinib, dar- bevacizumab among elderly patients51 and clinical trials of
atumumab, ixazomib, lenvatinib, nivolumab, palbociclib), pazopanib52 and sorafenib53 among patients with organ
although most comment that sample sizes are small enough dysfunction.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e181


CALVO ET AL

Some of the newer drugs shown in Table 1 have in fact tumor acquisition as well as heterogeneity of metastatic
been evaluated in populations who are older than age 65 cancers. Cell-free DNA and liquid biopsies are currently being
and are more representative of the disease population. For assessed as predictors of response and more experience
example, in the pivotal study of idelalisib and rituximab in with these assays will help to determine their value and
relapsed chronic lymphocytic leukemia,54 78% of patients en- place in standard clinical management. Pharmacogenomic
rolled were age 65 or older, 40% had a CrCl below 60 mL/min, analysis of tumor tissue has evolved from focused in-
and 85% had a Cumulative Illness Rating Scale score greater terrogation of select genes to guide targeted therapies to
than 6. Although patients older than age 65 benefited from multigene panels being used in conjunction with molecular
idelalisib similarly to their younger counterparts, the package tumor boards to help direct optimal therapy for patients.
insert reveals that there were higher rates or serious adverse The encouraging results for pharmacokinetic optimization
events, treatment discontinuation, and death among patients of TKIs and monoclonal antibodies justify the need to in-
older than age 65 in this trial as well as the pivotal trials in crease knowledge about the exposure-response relationship
non-Hodgkin lymphoma and small lymphocytic lymphoma. for other anticancer targeted agents to maximize the
In conclusion, pivotal studies of recently approved drugs therapeutic benefit of these expensive therapies. As Gao
generally have had higher representation of patients older et al14 said, Perhaps the time is ripe for us to quit guessing
than age 65. This yields more information on toxicity in the and start measuring. The implementation of TDM in routine
older population with these therapies. Unfortunately, it clinical practice would require (1) simple, rapid, and re-
appears that toxicity of many of these new targeted drugs is producible analytical methods to quantify plasma or serum
higher in older patients. However, package inserts do not drug concentrations; (2) a pharmacokinetic target related to
give guidance on alternate dosing or schedules in older and efficacy or toxicity; (3) population pharmacokinetic models
frail patients. In this special population, TDM or validated previously developed and a Bayesian approach to determine
frailty assessments could be useful tools to characterize the the individual pharmacokinetic parameters taking into ac-
real exposure and manage possible dose reduction accord- count the interindividual variability in all of the pharma-
ingly, minimizing the risk of greater reductions than those cokinetic processes; and (4) highly qualified professionals
that are really needed. (within multidisciplinary teams) trained to translate the TDM
results and to propose new doses adjusted to individual
FUTURE DIRECTIONS OF PERSONALIZED needs of each patient. This is a feasible way of optimizing our
THERAPY ACCORDING TO PHARMACOGENOMIC, current and future therapeutic armamentarium and it
PHARMACOKINETIC, AND PHARMACODYNAMIC should be encouraged.
PARAMETERS Finally, considering that with these agents, as with the
The role of somatic tumor genetics relates not only to initial older drugs, chronological age is not as important as
selection of therapy but also to sequencing of therapy op- physiologic age/frailty, incorporating frailty assessment
tions, and it must be optimized in standard clinical practice. tools into trials of newer agents and validating frailty-based
To continue to establish genetic biomarkers of novel ther- dose adjustment will be an important part of further drug
apies, we need clinical trials and patients willing to enroll in development. Ongoing efforts to assess approved antican-
them. Enthusiasm behind basket trials must continue to fuel cer therapeutics for older patient populations and those
histology agnostic, biomarker-driven trials that are as in- with organ dysfunction are an important effort for the
clusive as possible for the tumor types in which they will be oncology clinical research community.
most useful. In addition, novel methods of obtaining sam- All together, these efforts will allow us to push precision
ples for genetic analysis are needed to address challenges of medicine in oncology to the next level for our patients.

References
1. Klumpen HJ, Samer CF, Mathijssen RH, et al. Moving towards dose American Society of Clinical Oncology Provisional Clinical Opinion
individualization of tyrosine kinase inhibitors. Cancer Treat Rev. 2011; Update 2015. J Clin Oncol. 2016;34:179-185.
37:251-260. 5. Ballman KV. Biomarker: predictive or prognostic? J Clin Oncol. 2015;33:
2. Van Cutsem E, Peeters M, Siena S, et al. Open-label phase III trial of 3968-3971.
panitumumab plus best supportive care compared with best supportive 6. Iglehart JD, Silver DP. Synthetic lethalitya new direction in cancer-drug
care alone in patients with chemotherapy-refractory metastatic co- development. N Engl J Med. 2009;361:189-191.
lorectal cancer. J Clin Oncol. 2007;25:1658-1664. 7. Mateo J, Carreira S, Sandhu S, et al. DNA-repair defects and olaparib in
3. Amado RG, Wolf M, Peeters M, et al. Wild-type KRAS is required for metastatic prostate cancer. N Engl J Med. 2015;373:1697-1708.
panitumumab efficacy in patients with metastatic colorectal cancer. 8. Thomas A, Liu SV, Subramaniam DS, et al. Refining the treatment of
J Clin Oncol. 2008;26:1626-1634. NSCLC according to histological and molecular subtypes. Nat Rev Clin
4. Allegra CJ, Rumble RB, Hamilton SR, et al. Extended RAS gene mutation Oncol. 2015;12:511-526.
testing in metastatic colorectal carcinoma to predict response to anti- 9. Oxnard GR, Arcila ME, Sima CS, et al. Acquired resistance to EGFR
epidermal growth factor receptor monoclonal antibody therapy: tyrosine kinase inhibitors in EGFR-mutant lung cancer: distinct natural

e182 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


PHARMACOGENOMICS, PHARMACOKINETICS, AND PHARMACODYNAMICS OF TARGETED THERAPIES

history of patients with tumors harboring the T790M mutation. Clin 27. Mould DR, Dubinsky MC. Dashboard systems: pharmacokinetic/
Cancer Res. 2011;17:1616-1622. pharmacodynamic mediated dose optimization for monoclonal anti-
10. Shaw AT, Friboulet L, Leshchiner I, et al. Resensitization to crizotinib by bodies. J Clin Pharmacol. 2015;55(Suppl 3):S51-S59.
the lorlatinib ALK resistance mutation L1198F. N Engl J Med. 2016;374: 28. Wang W, Chen N, Shen X, et al. Lymphatic transport and catabolism of
54-61. therapeutic proteins after subcutaneous administration to rats and
11. Rizvi NA, Hellmann MD, Snyder A, et al. Cancer immunology. Mutational dogs. Drug Metab Dispos. 2012;40:952-962.
landscape determines sensitivity to PD-1 blockade in non-small cell lung 29. Keizer RJ, Huitema AD, Schellens JH, et al. Clinical pharmacokinetics of
cancer. Science. 2015;348:124-128. therapeutic monoclonal antibodies. Clin Pharmacokinet. 2010;49:493-507.
12. Snyder A, Makarov V, Merghoub T, et al. Genetic basis for clinical 30. Yang J, Zhao H, Garnett C, et al. The combination of exposure-response
response to CTLA-4 blockade in melanoma. N Engl J Med. 2014;371: and case-control analyses in regulatory decision making. J Clin Phar-
2189-2199. macol. 2013;53:160-166.
13. Le DT, Uram JN, Wang H, et al. PD-1 blockade in tumors with mismatch- 31. Wang J, Song P, Schrieber S, et al. Exposure-response relationship of T-
repair deficiency. N Engl J Med. 2015;372:2509-2520. DM1: insight into dose optimization for patients with HER2-positive
14. Gao B, Yeap S, Clements A, et al. Evidence for therapeutic drug metastatic breast cancer. Clin Pharmacol Ther. 2014;95:558-564.
monitoring of targeted anticancer therapies. J Clin Oncol. 2012;30: 32. Azzopardi N, Lecomte T, Ternant D, et al. Cetuximab pharmacokinetics
4017-4025. influences progression-free survival of metastatic colorectal cancer
15. Widmer N, Bardin C, Chatelut E, et al. Review of therapeutic drug patients. Clin Cancer Res. 2011;17:6329-6337.
monitoring of anticancer drugs part twotargeted therapies. Eur J 33. Gibiansky E, Gibiansky L, Carlile DJ, et al. Population pharmacokinetics
Cancer. 2014;50:2020-2036. of obinutuzumab (GA101) in chronic lymphocytic leukemia (CLL) and
16. Ibrahim AR, Eliasson L, Apperley JF, et al. Poor adherence is the main non-Hodgkins lymphoma and exposure-response in CLL. CPT Phar-
reason for loss of CCyR and imatinib failure for chronic myeloid leu- macometrics Syst Pharmacol. 2014;3:e144.
kemia patients on long-term therapy. Blood. 2011;117:3733-3736. 34. Talarico L, Chen G, Pazdur R. Enrollment of elderly patients in clinical
17. Bui BN, Italiano A, Miranova A, et al. Trough imatinib plasma levels in trials for cancer drug registration: a 7-year experience by the US Food
patients treated for advanced gastrointestinal stromal tumors evidence and Drug Administration. J Clin Oncol. 2004;22:4626-4631.
of large interpatient variations under treatment with standard doses. 35. Dees EC, OReilly S, Goodman SN, et al. A prospective pharmacologic
J Clin Oncol. 2008;26:15s (suppl; abstr 10564). evaluation of age-related toxicity of adjuvant chemotherapy in women
18. Huillard O, Mir O, Peyromaure M, et al. Sarcopenia and body mass index with breast cancer. Cancer Invest. 2000;18:521-529.
predict sunitinib-induced early dose-limiting toxicities in renal cancer 36. Cascinu S, Del Ferro E, Catalano G. Toxicity and therapeutic response to
patients. Br J Cancer. 2013;108:1034-1041. chemotherapy in patients aged 70 years or older with advanced cancer.
19. Mir O, Coriat R, Blanchet B, et al. Sarcopenia predicts early dose-limiting Am J Clin Oncol. 1996;19:371-374.
toxicities and pharmacokinetics of sorafenib in patients with hepato- 37. Fisher B, Anderson S, Wickerham DL, et al. Increased intensification and
cellular carcinoma. PLoS One. 2012;7:e37563. total dose of cyclophosphamide in a doxorubicin-cyclophosphamide
20. Eechoute K, Fransson MN, Reyners AK, et al. A long-term prospective regimen for the treatment of primary breast cancer: findings from
population pharmacokinetic study on imatinib plasma concentrations National Surgical Adjuvant Breast and Bowel Project B-22. J Clin Oncol.
in GIST patients. Clin Cancer Res. 2012;18:5780-5787. 1997;15:1858-1869.
21. Yu H, Steeghs N, Nijenhuis CM, et al. Practical guidelines for therapeutic 38. Crivellari D, Bonetti M, Castiglione-Gertsch M, et al. Burdens and
drug monitoring of anticancer tyrosine kinase inhibitors: focus on the benefits of adjuvant cyclophosphamide, methotrexate, and fluorouracil
pharmacokinetic targets. Clin Pharmacokinet. 2014;53:305-325. and tamoxifen for elderly patients with breast cancer: the International
22. Gotta V, Widmer N, Decosterd LA, et al. Clinical usefulness of thera- Breast Cancer Study Group Trial VII. J Clin Oncol. 2000;18:1412-1422.
peutic concentration monitoring for imatinib dosage individualization: 39. Hurria A, Brogan K, Panageas KS, et al. Patterns of toxicity in older
results from a randomized controlled trial. Cancer Chemother Phar- patients with breast cancer receiving adjuvant chemotherapy. Breast
macol. 2014;74:1307-1319. Cancer Res Treat. 2005;92:151-156.
23. Rousselot P, Johnson-Ansah H, Huguet F, et al. 133 personalized daily 40. Gelman RS, Taylor SG 4th. Cyclophosphamide, methotrexate, and 5-
doses of imatinib by therapeutic drug monitoring increase the rates of fluorouracil chemotherapy in women more than 65 years old with ad-
molecular responses in patients with chronic myeloid leukemia. Final vanced breast cancer: the elimination of age trends in toxicity by using
results of the randomized OPTIM imatinib study. Paper presented at: doses based on creatinine clearance. J Clin Oncol. 1984;2:1404-1413.
57th Annual Meeting of the American Society of Hematology; De- 41. Muss HB, Berry DA, Cirrincione C, et al; Cancer and Leukemia Group B
cember 2015; Orlando, FL. Experience. Toxicity of older and younger patients treated with adju-
24. Giles FJ, Yin OQ, Sallas WM, et al. Nilotinib population pharmacokinetics vant chemotherapy for node-positive breast cancer: the Cancer and
and exposure-response analysis in patients with imatinib-resistant or Leukemia Group B Experience. J Clin Oncol. 2007;25:3699-3704.
-intolerant chronic myeloid leukemia. Eur J Clin Pharmacol. 2013;69: 42. Perrone F, Nuzzo F, Di Rella F, et al. Weekly docetaxel versus CMF as
813-823. adjuvant chemotherapy for older women with early breast cancer: final
25. Rousselot P, Mollica L, Guerci-Bresler A, et al. Dasatinib daily dose results of the randomized phase III ELDA trial. Ann Oncol. 2015;26:
optimization based on residual drug levels resulted in reduced risk of 675-682.
pleural effusions and high molecular response rates: final results of the 43. Muss HB, Berry DA, Cirrincione CT, et al; CALGB Investigators. Adjuvant
randomized OPTIM dasatinib trial. Paper presented at: 19th Congress of chemotherapy in older women with early-stage breast cancer. N Engl J
the European Hematology Association; June 2014; Milan, Italy. Med. 2009;360:2055-2065.
26. Oude Munnink TH, Henstra MJ, Segerink LI, et al. Therapeutic drug 44. Lichtman SM, Hollis D, Miller AA, et al; Cancer and Leukemia Group B
monitoring of monoclonal antibodies in inflammatory and malignant (CALGB 9762). Prospective evaluation of the relationship of patient age
diseasetranslating TNF-a experience to oncology. Clin Pharmacol and paclitaxel clinical pharmacology: Cancer and Leukemia Group B
Ther. Epub 2015 Aug 11. (CALGB 9762). J Clin Oncol. 2006;24:1846-1851.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e183


CALVO ET AL

45. Lichtman SM, Hurria A, Cirrincione CT, et al; Cancer and Leukemia Group B. 63. Dreyling M, Jurczak W, Jerkeman M, et al. Ibrutinib versus temsirolimus
Paclitaxel efficacy and toxicity in older women with metastatic breast cancer: in patients with relapsed or refractory mantle-cell lymphoma: an in-
combined analysis of CALGB 9342 and 9840. Ann Oncol. 2012;23:632-638. ternational, randomised, open-label, phase 3 study. Lancet. Epub 2015
46. Hurria A, Blanchard MS, Synold TW, et al. Age-related changes in Dec 4.
nanoparticle albumin-bound paclitaxel pharmacokinetics and phar- 64. Byrd JC, Brown JR, OBrien S, et al. Ibrutinib versus ofatumumab in previously
macodynamics: influence of chronological versus functional age. On- treated chronic lymphoid leukemia. N Engl J Med. 2014;371:213-223.
cologist. 2015;20:37-44. 65. Furman RR, Sharman JP, Coutre SE, et al. Idelalisib and rituximab in
47. Hurria A, Togawa K, Mohile SG, et al. Predicting chemotherapy toxicity relapsed chronic lymphocytic leukemia. N Engl J Med. 2014;370:
in older adults with cancer: a prospective multicenter study. J Clin 997-1007.
Oncol. 2011;29:3457-3465. 66. Moreau P, Masszi T, Grzasko N, et al. Ixazomib, an investigational oral
48. Extermann M, Boler I, Reich RR, et al. Predicting the risk of chemotherapy proteasome inhibitor (PI), in combination with lenalidomide and
toxicity in older patients: the Chemotherapy Risk Assessment Scale for dexamethasone (IRd), significantly extends progression-free survival
High-Age Patients (CRASH) score. Cancer. 2012;118:3377-3386. (PFS) for patients (Pts) with relapsed and/or refractory multiple my-
49. Wildiers H, Heeren P, Puts M, et al. International Society of Geriatric eloma (RRMM): the phase 3 Tourmaline-MM1 Study (NCT01564537).
Oncology consensus on geriatric assessment in older patients with Blood. 2015;126 (abstr 727).
cancer. J Clin Oncol. 2014;32:2595-2603. 67. Schlumberger M, Tahara M, Wirth LJ, et al. Lenvatinib versus placebo in
50. Kazemi-Bajestani SM, Mazurak VC, Baracos V. Computed tomography- radioiodine-refractory thyroid cancer. N Engl J Med. 2015;372:621-630.
defined muscle and fat wasting are associated with cancer clinical 68. Thatcher N, Hirsch FR, Luft AV, et al. Necitumumab plus gemcitabine
outcomes. Semin Cell Dev Biol. Epub 2015 Sep 3. and cisplatin versus gemcitabine and cisplatin alone as first-line therapy
51. Mohile SG, Hardt M, Tew W, et al; Cancer and Aging Research Group. in patients with stage IV squamous non-small-cell lung cancer (SQUIRE):
Toxicity of bevacizumab in combination with chemotherapy in older an open-label, randomised, controlled phase 3 trial. Lancet Oncol. 2015;
patients. Oncologist. 2013;18:408-414. 16:763-774.
52. Shibata SI, Chung V, Synold TW, et al. Phase I study of pazopanib in 69. Weber JS, DAngelo SP, Minor D, et al. Nivolumab versus chemotherapy
patients with advanced solid tumors and hepatic dysfunction: a Na- in patients with advanced melanoma who progressed after anti-CTLA-4
tional Cancer Institute Organ Dysfunction Working Group study. Clin treatment (CheckMate 037): a randomised, controlled, open-label,
Cancer Res. 2013;19:3631-3639. phase 3 trial. Lancet Oncol. 2015;16:375-384.
53. Miller AA, Murry DJ, Owzar K, et al. Phase I and pharmacokinetic study 70. Borghaei H, Paz-Ares L, Horn L, et al. Nivolumab versus docetaxel in
of sorafenib in patients with hepatic or renal dysfunction: CALGB 60301. advanced nonsquamous non-small-cell lung cancer. N Engl J Med. 2015;
J Clin Oncol. 2009;27:1800-1805. 373:1627-1639.
54. Furman RR, Sharman JP, Coutre SE, et al. Idelalisib and rituximab in 71. Kaufman B, Shapira-Frommer R, Schmutzler RK, et al. Olaparib mon-
relapsed chronic lymphocytic leukemia. N Engl J Med. 2014;370: otherapy in patients with advanced cancer and a germline BRCA1/2
997-1007. mutation. J Clin Oncol. 2015;33:244-250.
55. Shaw AT, Gandhi L, Gadgeel S, et al. Alectinib in ALK-positive, crizotinib- 72. cSS, Chih-Hsin Yang J, Lee CK, et al. AZD9291, a mutant-selective EGFR
resistant, non-small-cell lung cancer: a single-group, multicentre, phase inhibitor, as first-line treatment for EGFR mutation-positive advanced
2 trial. Lancet Oncol. 2016;17:234-242. non-small cell lung cancer (NSCLC): results from a phase 1 expansion
56. Ou SI, Ahn JS, De Petris L, et al. Alectinib in crizotinib-refractory ALK- cohort. J Clin Oncol. 2015;33 (suppl; abstr 8000).
rearranged non-small-cell lung cancer: a phase II global study. J Clin 73. Finn RS, Crown JP, Lang I, et al. The cyclin-dependent kinase 4/6 in-
Oncol. Epub 2015 Nov 23. hibitor palbociclib in combination with letrozole versus letrozole alone
57. OConnor OA, Horwitz S, Masszi T, et al. Belinostat in patients with as first-line treatment of oestrogen receptor-positive, HER2-negative,
relapsed or refractory peripheral T-cell lymphoma: results of the pivotal advanced breast cancer (PALOMA-1/TRIO-18): a randomised phase 2
phase II BELIEF (CLN-19) study. J Clin Oncol. Epub 2015 Jun 22. study. Lancet Oncol. 2015;16:25-35.
58. Topp MS, Gokbuget N, Stein AS, et al. Safety and activity of blinatumomab 74. San-Miguel JF, Hungria VT, Yoon SS, et al. Panobinostat plus bortezomib
for adult patients with relapsed or refractory B-precursor acute lym- and dexamethasone versus placebo plus bortezomib and dexameth-
phoblastic leukaemia: a multicentre, single-arm, phase 2 study. Lancet asone in patients with relapsed or relapsed and refractory multiple
Oncol. 2015;16:57-66. myeloma: a multicentre, randomised, double-blind phase 3 trial. Lancet
59. Shaw AT, Ou SH, Bang YJ, et al. Crizotinib in ROS1-rearranged non-small- Oncol. 2014;15:1195-1206.
cell lung cancer. N Engl J Med. 2014;371:1963-1971. 75. Hamid O, Robert C, Daud A, et al. Safety and tumor responses with
60. Larkin J, Ascierto PA, Dreno B, et al. Combined vemurafenib and lambrolizumab (anti-PD-1) in melanoma. N Engl J Med. 2013;369:
cobimetinib in BRAF-mutated melanoma. N Engl J Med. 2014;371: 134-144.
1867-1876. 76. Migden MR, Guminski A, Gutzmer R, et al. Treatment with two different
61. Lokhorst HM, Plesner T, Laubach JP, et al. Targeting CD38 with dar- doses of sonidegib in patients with locally advanced or metastatic basal
atumumab monotherapy in multiple myeloma. N Engl J Med. 2015;373: cell carcinoma (BOLT): a multicentre, randomised, double-blind phase 2
1207-1219. trial. Lancet Oncol. 2015;16:716-728.
62. Lonial S, Weiss BM, Usmani SZ, et al. Daratumumab monotherapy in 77. Andtbacka RH, Kaufman HL, Collichio F, et al. Talimogene laherparepvec
patients with treatment-refractory multiple myeloma (SIRIUS): an improves durable response rate in patients with advanced melanoma.
open-label, randomised, phase 2 trial. Lancet. Epub 2016 Jan 6. J Clin Oncol. 2015;33:2780-2788.

e184 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


GASTROINTESTINAL (COLORECTAL) CANCER

Potentially Resectable Metastatic


Colorectal Cancer: A
Multidisciplinary Discussion

CHAIR
Gunnar Folprecht, MD
University Hospital Carl Gustav Carus
Dresden, Germany

SPEAKERS
David M. Liu, MD
University of British Columbia
Vancouver, BC, Canada

Laura A. Dawson, MD
Princess Margaret Cancer Centre
Toronto, ON, Canada

Steven J. Nurkin, MD
Roswell Park Cancer Institute
Buffalo, NY
GUNNAR FOLPRECT

Liver Metastases in Colorectal Cancer


Gunnar Folprecht, MD

OVERVIEW

Resection of colorectal liver metastases is a treatment standard because patients experience long-term disease-free survival
or are even cured after undergoing this procedure. Improved surgical techniques for liver resection in combination with
downsizing liver metastases by chemotherapy, interventions to induce liver hypertrophy before resection, and the use of
ablative techniques have allowed us to expand the indications for liver surgery and local treatment in situations with limited
metastatic colorectal cancer. Resectability and identification of patients who might benefit from liver surgery and local
ablative techniques are key factors for the treatment of patients with colorectal cancer. Despite the wide acceptance of liver
surgery and ablative techniques, there are many open questions on the management of limited metastatic disease, such as
which patients benefit from an aggressive surgical approach, what the indications for ablative and other local techniques are,
and what the role of chemotherapy is for patients with resectable or resected disease. Unfortunately, results of randomized
trials are only available for a limited number of these questions.

R esection of colorectal liver metastases is a treatment


standard because patients experience long-term disease-
free survival or are even cured after undergoing this pro-
In daily clinical practice, technical resectability and prog-
nostic factors contribute to the decision making on whether
to resect liver metastases. A higher number of metastases, a
cedure. Improved surgical techniques for liver resection and short disease-free interval before diagnosis of the metastases,
downsizing liver metastases by chemotherapy, interventions the presence of extrahepatic metastases, larger metasta-
to induce liver hypertrophy before resection, and the use of ses, an advanced primary tumor stage, and elevated tumor
ablative techniques have allowed us to widen the indications marker levels (carcinoembryonic antigen [CEA] or CA 19-9)
for liver surgery and local treatment in situations with limited
are important risk factors for the prognosis of patients.3 The
metastatic colorectal cancer.
Memorial Sloan Kettering Cancer Center score4 is one of the
Resectability and identification of patients who might benefit
from liver surgery and local ablative techniques are key factors most widely used scores and is easily calculated. There is no
for the treatment of patients with colorectal cancer. Despite the fixed upper limit for the number of liver metastases defining
wide acceptance of liver surgery and ablative techniques, there nonresectability or a lower limit for the disease-free interval
are many open questions on the management of limited before the (re)occurrence of liver metastases, however,
metastatic disease, such as which patients benefit from an these two factors have often have an important influence in
aggressive surgical approach, what the indications for ablative daily treatment decisions. The lack of clear definitions on
and other local techniques are, and what the role of chemo- prognostic factors limiting the value of resection contributes
therapy is for patients with resectable or resected disease. to the high subjectivity of decisions and might explain the
Unfortunately, results of randomized trials are only available high variability of treatment decisions observed, i.e., in surgical
for a limited number of these questions. reviews of CT scans the CELIM trial.2 Multidisciplinary care
team discussions can only partly overcome the lack of studies
RESECTABILITY defining limits for prognostic resectability.
Technical resectability is a necessary condition for the re- In addition to technical resectability and tumor-related
section of metastases. For liver metastases, resectability is prognostic factors, treatment decisions are driven by patient-
determined by 25% to 30% of functional liver tissue with related comorbidity, which limits surgical treatment options
sufficient inflow (portal vein/liver artery) and outflow (liver and chemotherapy tolerance (Fig. 1A). Systematic research
veins). Conversion chemotherapy1,2 and several surgical for comorbidity and multimodal treatment of liver metastases
techniques can improve technical resectability (Fig. 1). is rare and is therefore not included in this review.

From the University Hospital Carl Gustav Carus, University Cancer Center, Medical Department I, Dresden, Germany.

Disclosures of potential conflicts of interest provided by the author are available with the online article at asco.org/edbook.

Corresponding author: Gunnar Folprecht, MD, Universitatsklinikum Carl Gustav Carus Dresden, Fetscherstrae 74, 01307, Dresden, Germany; email: gunnar.folprecht@
uniklinikum-dresden.de.

2016 by American Society of Clinical Oncology.

e186 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


LIVER METASTASES IN COLORECTAL CANCER

Retrospective analyses have demonstrated that patients a Basingstoke risk score higher than 20 experience a median
who underwent resection for their metastases experienced survival of less than 1.5 years following liver resection.10 Al-
improved survival compared with patients with medical though patients were treated in the era of fluorouracil (5-FU),
treatment alone. This knowledge is based on retrospective survival for the highest risk group is probably not markedly
cohorts1,4 and patients resected for liver and lung metas- longer than in a general patient population with systemic
tases in prospective trials.5 Unfortunately, these reports are treatment alone.11,12 It is important to note that current
influenced by several biases.6 Resections are offered to prognostic scores are not prospectively evaluated to be pre-
patients whose disease responded to chemotherapy and dictive for the benefit of resections. In addition, re-resections of
who have a better prognosis, and the resection cohort further metastases can be beneficial, and patients with ob-
excludes patients who have an early disease progression, a served indications for re-resection have similar prognoses as
worsened performance status, or died before the decision those with initial resections.13-15 Furthermore, surgery/ablative
for resection (i.e., after conversion chemotherapy) could be techniques have the potential to contribute to therapy as a
made. further line of palliative treatment, but prospective trials among
The EORTC CLOCC trial7 was the first prospective ran- patients with technically resectable, poor prognostic disease
domized trial to demonstrate a survival benefit of resection/ are needed to determine which patients benefit most from
ablation versus systemic treatment alone. Patients with up resections.
to 10 liver metastases were selected to receive medical
treatment alone or medical treatment with radiofrequency
ablation. For the vast majority of patients, radiofrequency IMPROVING RESECTABILITY
ablation was performed as an open procedure and com- Among patients whose disease is not regarded as suitable for
bined with resection. In the combined treatment arm, the up-front resection due to the location of the metastases
overall survival was significantly improved compared (technical resectability) or poor prognostic factors, che-
with systemic treatment alone (8-year survival, 36% vs. 9%, motherapy is mostly the first treatment step, but other
respectively; hazard ratio [HR] 0.58; 95% CI, 0.380.88; interventions also can improve resectability.
p , .01).8 This trial demonstrated that resection/ablation Preoperative portal vein embolization of the liver seg-
has a major impact on overall survival. Among patients with ments planned for resection, which induces a hypertrophy of
up to 10 liver metastases without extrahepatic spread, the not-embolized liver segments, enlarges the remaining
approximately 30% were cured by the combined treat- functional liver tissue.16 This procedure is most frequently
ment. Furthermore, progression-free survival increased applied before an extended right hemihepatectomy if the
from 9.9 months (chemotherapy alone) to 16.8 months left segments (2/3) are relatively small and often combined
(combined modality treatment),8 allowing treatment-free with a two-step resection, i.e., resection of metastases in the
intervals. left segments before the right hemihepatectomy.17,18 The
Unfortunately, overall and disease-free survival rates after relatively aggressive concept of a combination of a portal
resection decrease for patients with poor prognostic factors. vein ligation with an intraoperative in-situ splitting of the
In the CELIM study, the median disease-free survival after liver (associating liver partition and portal vein ligation for
conversion therapy and resection was 16.8 months among staged hepatectomy; ALPPS) induces a more rapid liver
patients with less than five metastases, 8.2 months among hypertrophy and is followed by a second resection.19 In a
patients with 5 to 10 metastases, and 2.5 months among pa- recently published register of 320 patients, second re-
tients with more than 10 metastases.9 Similarly, patients with section was performed 14 days after the first interven-
tion (median). During those 2 weeks, the functional
liver remnant increased from 21% to 40%.20 This tech-
nique increases the options for resection of liver me-
KEY POINTS tastases and primary hepatobiliary cancers but was
associated with a 90-day mortality of 8.8% among pa-
Resection of (liver) metastases is part of standard tients with colorectal liver metastases of 5%.20 The com-
treatment in metastatic colorectal cancer. bination of ablation with resection of metastases provides a
Further trials are needed to better define limits of more parenchymal-sparing approach for extensive liver
resectability, especially among patients with a poor metastases.21
prognosis. These techniques increase the level of treatment com-
Resectability can be improved by conversion plexity, and additional treatment steps need additional time
chemotherapy and surgical/interventional methods
for procedures and/or patient recovery, especially in mul-
requiring close multidisciplinary cooperation.
FOLFOXIRI-based regimens and EGFR combinations
tistep resections for liver metastases or sequential re-
(for RAS wild type disease) have demonstrated higher sections, i.e., of liver and lung metastases plus resection of
response and resection rates in randomized trials the primary. In the COIN, AIO 0207, and CAIRO3 trials in-
Perioperative FOLFOX therapy should be considered for vestigating treatment-free intervals versus maintenance
patients with higher risk factors and resectable disease. therapy, the median time from end of chemotherapy
treatment until progression without chemotherapy was 3.0

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e187


GUNNAR FOLPRECT

FIGURE 1. (A) Dimensions of Resectability and (B) Heatmap Combining Prognostic and Technical Factors

to 4.1 months.22-24 This interval is shorter than the time when prognostic factors, such as a high number of metas-
needed for two or three resections. In several centers, tases, limit resection (Fig. 2). In the latter situation, che-
chemotherapy is administered between treatment cycles, motherapy is often used to observe the course of disease
but the number of required interventions may limit the and to resect disease with less aggressive tumor growth.
feasibility of resection at all metastatic sites. With current regimens achieving response rates of 60%
65%, liver resections were reported in approximately 40% of
patients enrolled in trials focusing on liver-limited disease.26
CHEMOTHERAPY IN NONRESECTABLE DISEASE In randomized trials, FOLFOXIRI (5-FU, leucovorin, ox-
There is a strong correlation between the response rate with aliplatin, and irinotecan) improved response27,28 and resection
chemotherapy and the resection rate when different studies rates28 compared with infusion chemotherapy doublets, as
are compared.25 Conceptually, the effect of shrinking larger, did cetuximab/FOLFIRI (5-FU, leucovorin, and irinotecan)
technically nonresectable metastases is more obvious than compared with FOLFIRI alone.29 Bevacizumab improved

e188 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


LIVER METASTASES IN COLORECTAL CANCER

response rates in studies with IFL (irinotecan, 5-FU, leuco- preoperative treatment, it might be reasonable to orientate
vorin) or fluoropyrimidine monotherapy30,31 but not when on the treatment durations for patients with resectable
combined with FOLFOX in first-line treatment.32 disease and to continue the preoperative therapy to
When EGFR antibodies and bevacizumab are directly 3 months and total chemotherapy duration to 6 months,
compared, higher response rates were reported in the although on treatment durations are missing.
CALGB 80405 study,33 and nonsignificant trends were re-
ported in the FIRE3 and PEAK trials.34,35 Furthermore, dif-
ferences were observed for the non-RECIST evaluation of the RESECTABLE LIVER METASTASES:
deepness of response favoring anti-EGFR antibodies.36 PERIOPERATIVE TREATMENT
A literature-based meta-analysis of all three trials di- Several studies on systemic or liver-directed chemotherapy
rectly comparing anti-EGFR antibodies with bevacizumab have investigated adjuvant or perioperative treatment
described a significantly higher response rate with anti-EGFR of resectable liver metastases. A meta-analysis of two
antibodies (odds ratio 1.3; 95% CI, 1.11.6).37 Similarly, in studiesboth closed early due to poor recruitment
the CALGB study, 82 patients (14.2%) underwent resection demonstrated a strong trend towards improved overall
after treatment with cetuximab, and 50 patients (8.9%) survival with adjuvant bolus 5-FU compared with surgery
underwent resection after receiving bevacizumab-based alone (HR 1.32; 95% CI, 0.951.82; p = .095).39 The EORTC
therapy.38 This difference is statistically significant (p , .01). trial 40983 (EPOC) randomly selected patients with up to
No differences in resection rates were reported in the FIRE3 four metastases to receive perioperative treatment with
and PEAK trials.34,35 Based on these results, FOLFOXIRI (or FOLFOX or liver surgery alone and showed a trend toward
EGFR-based therapies among patients with RAS wild-type improved disease-free survival with perioperative chemo-
disease) might be optimal if conversion therapy is the primary therapy (HR 0.81; 95% CI, 0.641.02; p = .068). The overall
treatment aim. survival was not significantly different between both arms,
During conversion chemotherapy, patients will have re- and the absolute difference in overall survival was relatively
sectable disease after a median treatment duration of small (3.4%; 95% CI, 7.113.8).40 A subgroup analysis of the
4 months. If resectability is not achieved within 6 months, it EPOC trial has shown interaction between CEA level, per-
is very unlikely that further chemotherapy will facilitate later formance status, and obesity on the chemotherapy effect;
resections.2 Therefore, multidisciplinary discussion of the the HR for patients with normal CEA, patients with a per-
treatment strategy might be scheduled every 2 months (or formance status of 1 or higher, and obese patients (body
after 3 and 6 months) during conversion chemotherapy. No mass index $ 30) was greater than 1.0, suggesting harm
trial has been performed to determine the optimal duration from perioperative treatment. The HRs for the opposite
of neoadjuvant therapy when resectability is achieved. groups (elevated CEA, performance status 0, and not obese)
When resectability was achieved within 3 to 6 months after were 0.60 or lower.41 Based on these results, it can be

FIGURE 2. (A) Conversion Chemotherapy in Technical Nonresectable Metastases (B) Effect of Chemotherapy for
Patients With a High Number of Metastases

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e189


GUNNAR FOLPRECT

speculated that perioperative therapy might be indicated for INTRA-ARTERIAL THERAPIES


patients with higher risk for recurrence and excellent per- High response rates in phase II trials using intra-arterial
formance status. chemotherapy have revived the discussion on this treat-
This is especially important because the patients in the ment. In the French OPTILIV study, pretreated patients were
trials mentioned above had relatively favorable risk treated with systemic cetuximab and intra-arterial FOLFOXIRI.
factors: In the EORTC EPOC trial,40 the majority of pa- The response rate was 41%, and 31% of patients were able to
tients had a single liver metastasis and had metachro-
undergo resection for their liver metastases.49 A protocol at
nous metastases. In resectable metastases, there is no
the Memorial Sloan Kettering Cancer Center used systemic
evidence for adding any antibodies to perioperative or
oxaliplatin and irinotecan combined with intra-arterial FUDR
adjuvant chemotherapy.
The British New EPOC trial randomly selected patients in 49 pretreated patients, achieving response rate of 76%. In
with liver metastases to receive perioperative chemotherapy total, 47% of patients were able to undergo resection in this
with or without the EGFR-antibody cetuximab. Progression- trial.50 These intra-arterial strategies are options if resection is
free and overall survival were worse with the addition of the treatment aim in pretreated patients without extrahe-
cetuximab.42 The background for these findings remained patic disease.
unclear and was the subject of long discussions, i.e., whether Selective internal radiation therapy (SIRT) adds another
less extensive surgery and higher rates of positive margins or treatment modality. In the SIRFLOX trial, patients with liver-
unknown biologic aspects explain the unexpected outcome.43 limited or liver-dominant metastases were randomly se-
With the missing benefit of adding any antibody in adjuvant lected to FOLFOX plus SIRT or FOLFOX alone. Progression-
treatment in stage III colon cancer44,45 and the lack of positive free survival (primary endpoint) was not different between
results in perioperative treatment of resectable metastatic the treatment arms (HR 0.93; 95% CI, 0.771.12; p = .43), and
disease, FOLFOX remains the regimen with the best evidence the overall response rates were similar (76% vs. 68%; p = .11).
of potential benefit for patients with resectable disease and Progression-free survival for the liver lesions was longer in
might be indicated especially in the presence of higher risk the SIRT group (HR 0.69; 95% CI, 0.550.90), but overall
factors.
survival data are not yet available.51 Currently, it remains
unclear whether the later progression in the liver translates
CHEMOTHERAPY AND LIVER TOXICITY to a longer overall survival.
Liver-related toxicity was described for irinotecan (stea-
tohepatitis, 20%) and oxaliplatin (sinusoidal obstruction,
19%).46 Patients with steatohepatitis experience increased CONCLUSION
mortality. In the EORTC 40983 study (investigating patients Because multimodal treatment is associated with an im-
with resectable disease), one of the 160 (0.6%) patients did proved prognosis, identifying patients who benefit from
not undergo resection because of oxaliplatin-related liver multimodal treatment is crucial to the treatment of pa-
toxicity.47 Although chemotherapy-induced liver toxicity
tients with metastatic colorectal cancer. Conversion
was intensively discussed after these publications, this risk
chemotherapy and ablative, interventional, and surgical
has to be balanced in the context of patients with meta-
static, nonresectable (or poorly resectable) disease. Be- techniques can improve resectability and require a close
cause operative morbidity increases with the number of multidisciplinary cooperation. Sequencing of the treat-
chemotherapy cycles,48 it is recommended to not further ment steps, the selection of patients who benefit from
extend preoperative chemotherapy if metastases become resection or local treatment, and improved therapies to
resectable. Furthermore, surgical strategies for patients who treat remaining micrometastases are emergent open
have been heavily pretreated must be adapted, i.e., by questions in the treatment of patients with limited me-
parenchymal-saving resections.21 tastatic colorectal cancer.

References
1. Adam R, Delvart V, Pascal G, et al. Rescue surgery for unresectable 4. Fong Y, Fortner J, Sun RL, et al. Clinical score for predicting recurrence
colorectal liver metastases downstaged by chemotherapy. Ann Surg. after hepatic resection for metastatic colorectal cancer: analysis of
2004;240:644-657. 1001 consecutive cases. Ann Surg. 1999;230:309-318, discussion
2. Folprecht G, Gruenberger T, Bechstein WO, et al. Tumour response and 318-321.
secondary resectability of colorectal liver metastases following neo- 5. Masi G, Loupakis F, Pollina L, et al. Long-term outcome of ini-
adjuvant chemotherapy with cetuximab: the CELIM randomised phase tially unresectable metastatic colorectal cancer patients treated with
2 trial. Lancet Oncol. 2010;11:38-47. 5-fluorouracil/leucovorin, oxaliplatin, and irinotecan (FOLFOXIRI) fol-
3. Spelt L, Andersson B, Nilsson J, et al. Prognostic models for outcome lowed by radical surgery of metastases. Ann Surg. 2009;249:420-425.
following liver resection for colorectal cancer metastases: a systematic 6. Palma D a, Salama JK, Lo SS, et al. The oligometastatic state-separating
review. Eur J Surg Oncol. 2012;38:16-24. truth from wishful thinking. Nat Rev Clin Oncol. 2014;11: 549-557.

e190 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


LIVER METASTASES IN COLORECTAL CANCER

7. Ruers T, Punt C, Van Coevorden F, et al; EORTC Gastro-Intestinal Tract combination chemotherapy for first-line treatment of advanced
Cancer Group, Arbeitsgruppe Lebermetastasen undtumoren in der colorectal cancer: results of the randomised phase 3 MRC COIN trial.
Chirurgischen Arbeitsgemeinschaft Onkologie, and the National Cancer Lancet Oncol. 2011;12:642-653.
Research Institute Colorectal Clinical Study Group. Radiofrequency 23. Hegewisch-Becker S, Graeven U, Lerchenmuller CA, et al. Maintenance
ablation combined with systemic treatment versus systemic treatment strategies after first-line oxaliplatin plus fluoropyrimidine plus bev-
alone in patients with non-resectable colorectal liver metastases: acizumab for patients with metastatic colorectal cancer (AIO 0207):
a randomized EORTC Intergroup phase II study (EORTC 40004). Ann a randomised, non-inferiority, open-label, phase 3 trial. Lancet Oncol.
Oncol. 2012;23:2619-2626. 2015;16:1355-1369.
8. Ruers T, Punt CJA, van Coevorden F, et al. Radiofrequency ablation (RFA) 24. Simkens LHJ, van Tinteren H, May A, et al. Maintenance treatment with
combined with chemotherapy for unresectable colorectal liver me- capecitabine and bevacizumab in metastatic colorectal cancer
tastases (CRC LM): long-term survival results of a randomized phase II (CAIRO3): a phase 3 randomised controlled trial of the Dutch Colorectal
study of the EORTC-NCRI CCSG-ALM Intergroup 40004 (CLOCC). J Clin Cancer Group. Lancet. 2015;385:1843-1852.
Oncol. 2015;33;(suppl; abstr 3501). 25. Folprecht G, Grothey A, Alberts S, et al. Neoadjuvant treatment of
9. Folprecht G, Gruenberger T, Bechstein W, et al. Survival of patients with unresectable colorectal liver metastases: correlation between tumour
initially unresectable colorectal liver metastases treated with FOLFOX/ response and resection rates. Ann Oncol. 2005;16:1311-1319.
cetuximab or FOLFIRI/cetuximab in a multidisciplinary concept (CELIM 26. Jones RP, Hamann S, Malik HZ, et al. Defined criteria for resectability
study). Ann Oncol. 2014;25:1018-1025. improves rates of secondary resection after systemic therapy for liver
10. Rees M, Tekkis PP, Welsh FK, et al. Evaluation of long-term survival after limited metastatic colorectal cancer. Eur J Cancer. 2014;50:1590-1601.
hepatic resection for metastatic colorectal cancer: a multifactorial 27. Loupakis F, Cremolini C, Masi G, et al. Initial therapy with FOLFOXIRI and
model of 929 patients. Ann Surg. 2008;247:125-135. bevacizumab for metastatic colorectal cancer. N Engl J Med. 2014;371:
11. Kohne CH, van Cutsem E, Wils J, et al; European Organisation for Re- 1609-1618.
search and Treatment of Cancer Gastrointestinal Group. Phase III study 28. Falcone A, Ricci S, Brunetti I, et al; Gruppo Oncologico Nord Ovest. Phase
of weekly high-dose infusional fluorouracil plus folinic acid with or III trial of infusional fluorouracil, leucovorin, oxaliplatin, and irinotecan
without irinotecan in patients with metastatic colorectal cancer: Eu- (FOLFOXIRI) compared with infusional fluorouracil, leucovorin, and
ropean Organisation for Research and Treatment of Cancer Gastroin- irinotecan (FOLFIRI) as first-line treatment for metastatic colorectal
testinal Group Study 40986. J Clin Oncol. 2005;23:4856-4865. cancer: the Gruppo Oncologico Nord Ovest. J Clin Oncol. 2007;25:
12. de Gramont A, Bosset JF, Milan C, et al. Randomized trial comparing 1670-1676.
monthly low-dose leucovorin and fluorouracil bolus with bimonthly 29. Van Cutsem E, Kohne C-H, Hitre E, et al. Cetuximab and chemotherapy
high-dose leucovorin and fluorouracil bolus plus continuous infusion for as initial treatment for metastatic colorectal cancer. N Engl J Med. 2009;
advanced colorectal cancer: a French intergroup study. J Clin Oncol. 360:1408-1417.
1997;15:808-815. 30. Hurwitz H, Fehrenbacher L, Novotny W, et al. Bevacizumab plus iri-
13. Takahashi S, Inoue K, Konishi M, et al. Prognostic factors for poor notecan, fluorouracil, and leucovorin for metastatic colorectal cancer.
survival after repeat hepatectomy in patients with colorectal liver N Engl J Med. 2004;350:2335-2342.
metastases. Surgery. 2003;133:627-634. 31. Kabbinavar FF, Hambleton J, Mass RD, et al. Combined analysis of
14. Shaw IM, Rees M, Welsh FKS, et al. Repeat hepatic resection for re- efficacy: the addition of bevacizumab to fluorouracil/leucovorin im-
current colorectal liver metastases is associated with favourable long- proves survival for patients with metastatic colorectal cancer. J Clin
term survival. Br J Surg. 2006;93:457-464. Oncol. 2005;23:3706-3712.
15. Lam VWT, Pang T, Laurence JM, et al. A systematic review of repeat 32. Saltz LB, Clarke S, Daz-Rubio E, et al. Bevacizumab in combination with
hepatectomy for recurrent colorectal liver metastases. J Gastrointest oxaliplatin-based chemotherapy as first-line therapy in metastatic
Surg. 2013;17:1312-1321. colorectal cancer: a randomized phase III study. J Clin Oncol. 2008;26:
16. Makuuchi M, Thai BL, Takayasu K, et al. Preoperative portal emboli- 2013-2019.
zation to increase safety of major hepatectomy for hilar bile duct 33. Venook AP, Niedzwiecki D, Lenz H-J, et al. CALGB/SWOG 80405: phase III
carcinoma: a preliminary report. Surgery. 1990;107:521-527. trial of irinotecan/5-FU/leucovorin (FOLFIRI) or oxaliplatin/5-FU/
17. Brouquet A, Abdalla EK, Kopetz S, et al. High survival rate after two- leucovorin (mFOLFOX6) with bevacizumab (BV) or cetuximab (CET)
stage resection of advanced colorectal liver metastases: response- for patients (pts) with KRAS wild-type (wt) untreated metastatic ad-
based selection and complete resection define outcome. J Clin enocarcinoma of the colon or rectum (MCRC). J Clin Oncol. 2014;32:5s
Oncol. 2011;29:1083-1090. (suppl; abstr LBA3).
18. Lam VWT, Laurence JM, Johnston E, et al. A systematic review of two- 34. Heinemann V, von Weikersthal LF, Decker T, et al. FOLFIRI plus
stage hepatectomy in patients with initially unresectable colorectal liver cetuximab versus FOLFIRI plus bevacizumab as first-line treatment for
metastases. HPB (Oxford). 2013;15:483-491. patients with metastatic colorectal cancer (FIRE-3): a randomised,
19. Schnitzbauer AA, Lang SA, Goessmann H, et al. Right portal vein ligation open-label, phase 3 trial. Lancet Oncol. 2014;15:1065-1075.
combined with in situ splitting induces rapid left lateral liver lobe 35. Schwartzberg LS, Rivera F, Karthaus M, et al. PEAK: a randomized,
hypertrophy enabling 2-staged extended right hepatic resection in multicenter phase II study of panitumumab plus modified fluorouracil,
small-for-size settings. Ann Surg. 2012;255:405-414. leucovorin, and oxaliplatin (mFOLFOX6) or bevacizumab plus mFOL-
20. Schadde E, Raptis DA, Schnitzbauer AA, et al. Prediction of mortality FOX6 in patients with previously untreated, unresectable, wild-type
after ALPPS stage-1: an analysis of 320 patients from the international KRAS exon 2 metastatic colorectal cancer. J Clin Oncol. 2014;32:
ALPPS registry. Ann Surg. 2015;262:780-785, discussion 785-786. 2240-2247.
21. Evrard S, Poston G, Kissmeyer-Nielsen P, et al. Combined ablation and 36. Heinemann V, Stintzing S, Modest DP, et al. Early tumour shrinkage
resection (CARe) as an effective parenchymal sparing treatment for (ETS) and depth of response (DpR) in the treatment of patients with
extensive colorectal liver metastases. PLoS One. 2014;9:e114404. metastatic colorectal cancer (mCRC). Eur J Cancer. 2015;51:1927-1936.
22. Adams RA, Meade AM, Seymour MT, et al; MRC COIN Trial Investigators. 37. Khattak MA, Martin H, Davidson A, et al. Role of first-line anti-epidermal
Intermittent versus continuous oxaliplatin and fluoropyrimidine growth factor receptor therapy compared with anti-vascular

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e191


GUNNAR FOLPRECT

endothelial growth factor therapy in advanced colorectal cancer: patients with resected stage III colon cancer (PETACC-8): an open-label,
a meta-analysis of randomized clinical trials. Clin Colorectal randomised phase 3 trial. Lancet Oncol. 2014;15:862-873.
Cancer. 2015;14:81-90. 45. Allegra CJ, Yothers G, OConnell MJ, et al. Phase III trial assessing
38. Venook A, Niedzwiecki D, Lenz H, et al. CALGB/SWOG 80405: analysis of bevacizumab in stages II and III carcinoma of the colon: results of NSABP
patients undergoing surgery as part of treatment strategy. Ann Oncol. protocol C-08. J Clin Oncol. 2011;29:11-16.
2014;25:(suppl 4; abstr LBA10). 46. Vauthey JN, Pawlik TM, Ribero D, et al. Chemotherapy regimen predicts
39. Mitry E, Fields ALA, Bleiberg H, et al. Adjuvant chemotherapy after po- steatohepatitis and an increase in 90-day mortality after surgery for
tentially curative resection of metastases from colorectal cancer: a pooled hepatic colorectal metastases. J Clin Oncol. 2006;24:2065-2072.
analysis of two randomized trials. J Clin Oncol. 2008;26:4906-4911. 47. Nordlinger B, Sorbye H, Glimelius B, et al; EORTC Gastro-Intestinal Tract
40. Nordlinger B, Sorbye H, Glimelius B, et al; EORTC Gastro-Intestinal Tract Cancer Group; Cancer Research UK; Arbeitsgruppe Lebermetastasen
Cancer Group; Cancer Research UK; Arbeitsgruppe Lebermetastasen und-tumoren in der Chirurgischen Arbeitsgemeinschaft Onkologie;
undtumoren in der Chirurgischen Arbeitsgemeinschaft Onkologie; Australasian Gastro-Intestinal Trials Group; Fede ration Francophone de
Australasian Gastro-Intestinal Trials Group; Federation Francophone de Cancerologie Digestive. Perioperative chemotherapy with FOLFOX4 and
Cancerologie Digestive. Perioperative FOLFOX4 chemotherapy and surgery versus surgery alone for resectable liver metastases from co-
surgery versus surgery alone for resectable liver metastases from co- lorectal cancer (EORTC Intergroup trial 40983): a randomised controlled
lorectal cancer (EORTC 40983): long-term results of a randomised, trial. Lancet. 2008;371:1007-1016.
controlled, phase 3 trial. Lancet Oncol. 2013;14:1208-1215. 48. Karoui M, Penna C, Amin-Hashem M, et al. Influence of preoperative
41. Sorbye H, Mauer M, Gruenberger T, et al; EORTC Gastro-Intestinal Tract chemotherapy on the risk of major hepatectomy for colorectal liver
Cancer Group; Cancer Research UK; Arbeitsgruppe Lebermetastasen metastases. Ann Surg. 2006;243:1-7.
und-tumoren in der Chirurgischen Arbeitsgemeinschaft Onkologie; 49. Levi FA, Boige V, Hebbar M, et al; Association Internationale pour la
Australasian Gastro-Intestinal Trials Group; Federation Francophone de Recherche sur le Temps Biologique et la Chronotherapie. Conversion to
Cancerologie Digestive. Predictive factors for the benefit of peri- resection of liver metastases from colorectal cancer with hepatic artery
operative FOLFOX for resectable liver metastasis in colorectal cancer infusion of combined chemotherapy and systemic cetuximab in mul-
patients (EORTC Intergroup Trial 40983). Ann Surg. 2012;255:534-539. ticenter trial OPTILIV. Ann Oncol. 2015;27:267-274.
42. Primrose J, Falk S, Finch-Jones M, et al. Systemic chemotherapy with or 50. DAngelica MI, Correa-Gallego C, Paty PB, et al. Phase II trial of hepatic
without cetuximab in patients with resectable colorectal liver metastasis: artery infusional and systemic chemotherapy for patients with unre-
the New EPOC randomised controlled trial. Lancet Oncol. 2014;15:601-611. sectable hepatic metastases from colorectal cancer: conversion to
43. Nordlinger B, Poston GJ, Goldberg RM. Should the results of the new resection and long-term outcomes. Ann Surg. 2015;261:353-360.
EPOC trial change practice in the management of patients with re- 51. van Hazel GA, Heinemann V, Sharma NK, et al. SIRFLOX: randomized
sectable metastatic colorectal cancer confined to the liver? J Clin Oncol. phase III trial comparing first-line mFOLFOX6 (plus or minus bev-
2014;33:241-243. acizumab) versus mFOLFOX6 (plus or minus bevacizumab) plus selective
44. Taieb J, Tabernero J, Mini E, et al; PETACC-8 Study Investigators. internal radiation therapy in patients with metastatic colorectal cancer.
Oxaliplatin, fluorouracil, and leucovorin with or without cetuximab in J Clin Oncol. Epub 2016 Feb 22.

e192 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


GASTROINTESTINAL (COLORECTAL) CANCER

Questions on Treatment of Locally


Advanced Rectal Cancer

CHAIR
Julio Garcia-Aguilar, MD, PhD
Memorial Sloan Kettering Cancer Center
New York, NY

SPEAKERS
Deborah Schrag, MD, MPH
Dana-Farber Cancer Institute
Boston, MA

Rob Glynne-Jones, MD, FRCP


Mount Vernon Cancer Centre
Middlesex, United Kingdom
GARCIA-AGUILAR, GLYNNE-JONES, AND SCHRAG

Multimodal Rectal Cancer Treatment: In Some Cases, Less


May Be More
Julio Garcia-Aguilar, MD, Rob Glynne-Jones, MBBS, FRCR, FRCP, and Deborah Schrag, MD, MPH

OVERVIEW

A series of clinical trials in the last several decades has resulted in the development of multimodality treatment of locally
advanced rectal cancer that includes neoadjuvant (preoperative) chemoradiotherapy, total mesorectal excision, and
postoperative adjuvant chemoradiotherapy. Owing to this regimen, patients with locally advanced rectal cancer have better
survival rates than patients with colon cancer, but at the cost of substantial morbidity and reduced quality of life. The
challenge is to identify treatment approaches that maintain or even improve oncologic outcomes while preserving quality of
life. We have identified different tumor characteristics that are associated with recurrence and probability of survival for
locally advanced rectal cancer. This risk stratification, based on baseline clinical staging and tumor response to chemo-
radiotherapy, has led us to question whether all patients with locally advanced rectal cancer require every component of the
multimodal regimen. In this article, we will review recent evidence that some patients with locally advanced rectal cancer
can be spared one or more treatment modalities without compromising long-term oncologic outcomes and while preserving
quality of life.

S urgical excision of the rectum and its mesorectal enve-


lope has been the mainstay of treatment for rectal cancer
for over a century.1 Despite advances in surgical technique
credited with the suggestion that patients with rectal cancer
who experience a clinically complete response (cCR) to
neoadjuvant chemoradiotherapy could achieve long-term
and perioperative care, total mesorectal excision remains local tumor control without surgery.4
an operation associated with some mortality, substantial These ideas were initially received with disbelief, but
morbidity, and sequelae that permanently impair quality reports from other institutions have confirmed that surgery
of life.2 can be avoided for select patients with rectal cancer who
received treatment with chemoradiotherapy. However, the
evidence supporting this treatment approach is based on
CAN SURGERY BE AVOIDED FOR SELECTED small institutional series of heterogenous groups of patients
PATIENTS? who were staged using different imaging modalities, treated
Some patients with locally advanced rectal cancer have a according to diverse radiation and chemotherapy regimens,
pathologic complete response (pCR) to neoadjuvant che- evaluated at different times after completion of the neo-
moradiotherapy. Patients with pCR experience lower local adjuvant therapy, selected for observation using different
recurrence and improved survival rates compared with criteria, and followed for relatively short periods of time. In
patients who do not experience pCR, raising the question of spite of these limitations, clinicians are starting to accept a
whether the former need surgery.3 Given that the mortality, paradigm shift for this selected group of patients with rectal
morbidity, and long-term sequelae from multimodal therapy cancer with a cCR after neoadjuvant chemoradiation. Ac-
are related to excision of the rectum, avoiding total mes- ceptance has often been accelerated by patients motivated
orectal excision for selective patients who obtain a sustained to avoid the consequences of a low colorectal anastomosis
response to chemoradiotherapy will improve their quality or a permanent colostomy. The treatment plan after neo-
of life. adjuvant therapy that consists of close active surveillance,
Although the evidence suggesting that some rectal cancers rather than surgery, is called watch-and-wait, wait-and-
can be treated with radiation alone is almost a century old, see, or nonoperative management. We use the term
it is Angelita Habr-Gama from Sao Paulo who should be nonoperative management in this article.

From the Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Medical Oncology, Mount Vernon Centre for Cancer Treatment, London, United
Kingdom; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: email: Julio Garcia-Aguilar, MD, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Ave., New York, NY 10065;
email: garciaaj@mskcc.org.

2016 by American Society of Clinical Oncology.

92 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


RECTAL CANCER TREATMENT: LESS MAY BE MORE

Evidence Supporting Nonoperative Management A group in the Netherlands reported their experience with
Most studies that have reported nonoperative management nonoperative management for 21 patients with cCR among
outcomes in locally advanced rectal cancer have compared a total of 192 patients treated with chemoradiotherapy
recurrence and survival rates between patients with cCR between 2004 and 2010. After a mean follow-up of
entered in a nonoperative management protocol and pa- 25 months, local relapse developed in one patient who then
tients who had similar neoadjuvant treatment followed by underwent curative salvage surgery; the other 20 patients
total mesorectal excision and subsequent pCR. are alive without disease. Outcomes of patients treated with
In Dr. Habr-Gamas protocol, clinical tumor response is the nonoperative management protocol who experienced
assessed 8 weeks after chemoradiotherapy.5 Patients with CR were similar to the outcomes of patients with pCR after
persistent tumor undergo total mesorectal excision; those total mesorectal excision.6
with cCR undergo monthly evaluations including digital rectal Our group at the Memorial Sloan Kettering Cancer Center
examination, proctoscopy, carcinoembryonic antigen (CEA) reported the results of 32 patients with cCR treated with the
blood testing, and biopsy of suspicious lesions. Patients found nonoperative management protocol who started neo-
to have tumor relapse are directed to surgery, whereas pa- adjuvant therapy between 2006 and 2010 and were fol-
tients with a sustained cCR after 1 year continue surveillance lowed for a median of 23 months. We compared these
every 3 months for an additional year and every 6 months results with 57 patients treated during the same period who
thereafter. Of the patients treated according to this protocol, underwent total mesorectal excision and had a pCR. The
27% experienced cCR after 1 year and were spared from total total mesorectal excision group had slightly more proximal
mesorectal excision. Local relapse during follow-up beyond tumors and received adjuvant treatment more often, and
1 year developed in 10% of patients treated with the also had more advanced tumors compared with the patients
nonoperative management protocol, and all proceeded treated with the nonoperative management protcol. Six of
to curative total mesorectal excision. The oncologic results in 32 patients (21%) experienced disease relapse and un-
this nonoperative management group were equivalent to derwent curative salvage surgery; distant metastasis de-
those of patients who had a pCR after total mesorectal veloped in three of these patients. The 3-year disease-free
excision. survival (DFS) and overall survival (OS) rates were not different
between groups.7 In an update to this series, we compared the
results of 73 patients with cCR treated with the nonoperative
management protocol and 72 patients treated with total
mesorectal excision who had pCR between 2006 and 2014.
KEY POINTS
Although relapse occurred among 19 of the 73 patients (26%)
Treatment counseling should convey the risks and treated with nonoperative management, 18 patients received
benefits by using quantitative estimates expressed in salvage therapy, and DFS and OS were equivalent between the
absolutenot relativeterms to empower informed groups. In total, 56 of 73 patients (77%) who were treated
patient decision making. using the nonoperative management protocol preserved the
Multimodality therapy provides excellent local tumor rectum after a median of 4 years of follow-up.8
control and long-term survival for patients with locally Araujo et al reported a retrospective comparison of 42 pa-
advanced rectal cancer. tients with rectal cancer entered into a nonoperative man-
Phased-array MRI can define features associated with a agement protocol with 69 patients treated with neoadjuvant
high risk of metastases and a high likelihood of local therapy and total mesorectal excision with subsequent pCR.
recurrence.
Nonoperative management was not an elective alternative and
Evidence supporting the use or non-use of adjuvant
chemotherapy in patients with rectal cancer is not
was only considered for patients who refused surgery. Relapse
available, and so omission of this treatment can be occurred in a total of 12 (28%) patients in the nonoperative
considered. management group; local recurrence developed in five pa-
Select patients with rectal cancer who experience tients, distant metastasis developed in three patients, and both
complete clinical response after neoadjuvant therapy local recurrence and distant metastasis occurred in four pa-
can avoid total mesorectal excision, but the evidence is tients at a median of 2 years of follow-up. Four of the five
still preliminary. patients with isolated local recurrence had successful salvage
A selective approach to radiotherapy provision is surgery. There was no difference in OS rates, but DFS was
feasible with the provision that we use high-quality MRI reduced in the nonoperative management group compared
and optimal total mesorectal excision surgery. with the surgery group (61% vs. 83%). This study had several
Fluorouracil is best administered using an infusional
unfortunate limitations including pretreatment tumor staging
rather than a bolus schedule. Capecitabine is a
reasonable alternative to infusional 5-FU.
performed by digital examination and a nonstandardized
When FOLFOX is administered, it is essential to avoid definition of clinical response, and the groups were not
long-term toxicity by dose-reducing or discontinuing matched for important clinicopathologic variables such as
oxaliplatin before peripheral neuropathy becomes tumor distance from the anal verge.9
severe or persistent. Li et al reported results from a cohort of 122 patients with
cCR among a total of 900 patients with stage II and III rectal

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 93


GARCIA-AGUILAR, GLYNNE-JONES, AND SCHRAG

cancer treated with chemoradiotherapy. Of the 122 pa- The proportion of patients who respond to chemo-
tients, 92 had rectal resection and 30 entered a non- radiotherapy seems small, and the optimal time to assess
operative management protocol. Both groups of patients clinical response is unknown. Tumor response depends on
were well-matched for clinical characteristics and received radiation dose, but doses beyond 54 Gy are rarely used in
similar treatment. Recurrence and DFS rates were not sig- these patients. Adding other drugs that are effective in colon
nificantly different between groups.10 cancer as radiosensitizers beyond fluoropyrimidines has
A group from the United Kingdom recently reported a been found to be ineffective or prohibitively toxic.12-15
multi-institutional experience with a nonoperative man- Tumor response to chemoradiotherapy is closely associated
agement approach versus surgical resection for patients with time, and, for patients undergoing total mesorectal
with rectal cancer treated with chemoradiotherapy. In excision after chemoradiotherapy, the proportion of tumors
contrast to the previous series, this study compared with pCR increases with the time interval between che-
the outcomes of 129 patients with cCR with 228 patients moradiotherapy and surgery.16 As prolonging the interval to
who had surgical resection after chemoradiotherapy, in- surgery and postoperative systemic chemotherapy may be
dependent of the pathologic stage. Neoadjuvant therapy unsafe for patients at risk for distant metastasis, attempts
was similar between groups. After a median follow-up of have been made to deliver systemic chemotherapy imme-
33 months from start of chemoradiotherapy, 44 (34%) diately before or after chemoradiation.17,18 Delivering sys-
patients with cCR had local regrowth, corresponding to temic chemotherapy before surgery, rather than after, has
an actuarial 3-year local regrowth rate of 38%. Similar to been shown to increase tumor response without delaying
previous findings, most local regrowths were on the bowel the treatment of potential micrometastatic disease. In these
wall, and most underwent successful salvage treatment. The patients, the assessment of clinical response and the rec-
authors developed one-to-one paired cohorts (109 patients ommendation of nonoperative management or surgery are
in each group) using propensity-score matching for the key performed at the completion of both chemoradiotherapy
confounders. The 3-year nonregrowth DFS rate (defined as and systemic chemotherapy. This approach has resulted in
time until death, local recurrence, or distant metastasis, not pCR rates as high as 38% for patients with clinical stage II and
including local regrowths) was 88% for the nonoperative III disease, and it has the added advantage of increasing
management group and 78% for the surgical group (log rank; compliance with adjuvant systemic chemotherapy and
p = .22). The colostomy-free survival rates were 74% and shortening ileostomy time for patients with locally advanced
47%, respectively. The authors concluded that nonoperative rectal cancer.17,18 The experimental arm of the RAPIDO trial
management is safe in a multi-institutional setting, sup- uses short-term radiation and consolidation chemotherapy;
porting the standard adoption of this protocol. However, the the trials results are forthcoming.19
results of this study should be interpreted with caution, as The lack of a reliable and uniform method of distinguishing
tumors in patients treated with nonoperative management post-treatment scar from residual tumor in the bowel wall or
had an earlier pretreatment tumor stage, were less likely to regional lymph nodes is the main obstacle to nonoperative
have nodal involvement, rarely had unfavorable histologic management for patients treated with neoadjuvant therapy.
features, and were more likely to have normal CEA levels. In Most authors agree that digital rectal examination, endos-
addition, comparing patients with and without cCR, in- copy, and imaging studies should be used. A flat white scar
dependent of the pathologic stage, introduces substantial with or without telangiectasia and a normal digital exami-
bias, as tumor response is associated with improved out- nation are good predictors of pCR, whereas the presence of
come compared with nonresponders.11 superficial ulceration or a palpable modularity upon digital
In summary, despite the relatively low-quality evidence, rectal examination indicate an incomplete response.20,21
these studies all suggest that most patients with cCR after Although clinical assessment tends to underestimate tu-
neoadjuvant chemotherapy can achieve prolonged local mor response, there is always a possibility that tumors
tumor control without surgery. Tumor regrowth occurs in are concealed in or behind apparently normal scar tissue
some patients, but most can be effectively treated with in the rectal wall. 22 Endorectal ultrasound, CT, and
18
surgery. As the follow-up period in most of these series F-fludeoxyglucose PET provide an estimate of tumor
is relatively short, long-term survival rates are still regression but are not sensitive enough to identify pCR.23
unknown. Conventional MRI morphologic sequences (e.g., T2- and
T1-weighted images) cannot differentiate residual tumor
from surrounding fibrosis, but diffusion-weighted MRI
Challenges to Nonoperative Management of Rectal sequences may improve the diagnostic performance of
Cancer morphologic MRI sequences for differentiating pCR from
Although the above-mentioned studies all suggest that most residual tumor. 24
patients with cCR after neoadjuvant chemotherapy can The definition of response undoubtedly influences clinical
achieve prolonged local tumor control without surgery, a outcomes: a strict definition reduces the proportion eligible
number of questions must be answered before nonoperative but increases the chance of success with nonoperative
management can be considered a standard option for pa- management, whereas less-strict criteria increase the
tients with locally advanced rectal cancer. number of eligible patients but also risk of local tumor

94 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


RECTAL CANCER TREATMENT: LESS MAY BE MORE

regrowth and distant metastasis. Although there are cur- recurrence observed among patients with rectal cancer
rently no validated criteria defining clinical and radiologic treated with total mesorectal excision alone challenged the
tumor response, a new set of criteria categorizing response benefit of neoadjuvant therapy for patients with low-risk
in a three-tier system is currently being tested in a pro- stage II rectal tumors30-32 and called for a more selective
spective clinical trial.25 approach, particularly for upper rectal cancer.33-35 In ad-
A number of patients with apparent cCR develop tumor dition, for patients with a high risk of metastatic disease,
regrowth during follow-up. As most regrowth occurs in the the integration of more active chemotherapy into the
bowel wall, repeated endoscopic examinations are essential. preoperative setting has become more attractive either in
Any suspicious changes in the scar should be biopsied. MRI addition to or as an alternative for chemoradiotherapy. The
should also be performed regularly to detect nodal clinical and imaging features and histopathologic analysis
regrowth. Changes in the size, contour, heterogeneity, or of tumor determine the inherent risks of both local re-
restriction of diffusion should raise the possibility of relapse. currence and distant metastasis.36,37 Variations in practice
Repeated examinations and continuous monitoring are of- and the different uptake of these modalities in rectal cancer
ten necessary to confirm recurrence. are common, as judgments are made on the balance of risk
Ultimately, finding reliable predictors of response to and toxicity.
neoadjuvant therapy would help identify patients who Surgery and radiotherapy often have permanent sequelae
would most likely to benefit from nonoperative manage- resulting in an adverse effect on quality of life. Preoperative
ment as well as reduce toxicity for patients whose disease chemoradiotherapy enhances local control and can be cu-
will likely have a poor response. Tumor size and stage seem rative on its own for some patients, but it worsens the
to predict response, with smaller early-stage tumors being constellation of changes in bowel function experienced by
more likely to develop pCR. The search for molecular pre- many patients after sphincter-saving rectal cancer sur-
dictors of tumor response has not resulted in breakthrough gery, known as low anterior resection syndrome. Many
findings so far. We have previously shown that rectal tumors patients complain of chronic pain.38 Despite potentially
with a KRAS mutation are less likely to respond to neo- confounding factors such as older age or the effects of
adjuvant therapy.26 However, these findings must be vali- prior surgical procedures in the pelvis, these symptoms
dated in large independent cohorts. are worsened by the addition of short-course pre-
In summary, the nonoperative management approach is operative chemoradiotherapy (SCPRT) or conventional
an attractive alternative for patients with rectal cancer who chemoradiotherapy.39 In the Dutch total mesorectal ex-
experience cCR after neoadjuvant therapy. However, evi- cision trial, 62% of patients who received SCPRT before
dence supporting this approach is still relatively scarce and total mesorectal excision reported fecal incontinence,
too many questions about its safety remain to recommend compared with 38% in the total mesorectal excision alone
this approach outside of a clinical trial. Although a well- group.40 Hence, a paradox: as we are advocating less che-
designed phase III randomized trial comparing surgery moradiotherapy because surgery is getting better, we are
versus nonoperative management for patients with cCR simultaneously advocating chemoradiotherapy to avoid
after neoadjuvant therapy may never be palatable to pro- radical surgery.
spective patients, a number of prospective registries and
phase II trials are currently open to accrual.
Indications for Chemoradiotherapy
There are four main indications for delivering preopera-
CAN WE AVOID NEOADJUVANT THERAPY OR tive radiotherapy or chemoradiotherapy. These comprise
RADIOTHERAPY IN SELECTED PATIENTS? unresectable or borderline resectable cancers; resectable
Historically, surgeons reported a high pelvic recurrence rate cancers with a high risk of local recurrence; early rectal
after radical surgery alone, resulting in a range of symp- cancers in older or frail patients as an alternative to
toms, including profuse mucinous discharge, bleeding, and surgery; and treatment with palliative intent. The radia-
intractable pelvic pain.27 A landmark German study sub- tion oncologist should keep in mind that postoperative
sequently established preoperative chemoradiotherapy chemoradiotherapy is associated with substantially more
as the standard of care.28 Although the rates of local re- acute and long-term morbidity compared with preopera-
currence reported before total mesorectal excision/ tive utilization.28 Hence, postoperative chemoradiotherapy
chemoradiotherapy came into widespread use would is not usually part of the planned management and usually
not be acceptable today, these historical local recurrence reflects poor preoperative decisions, unexpected surgical
rates continue to dominate decision making.29 Many ad- or histopathologic findings, or both.
vocate the use of chemoradiotherapy for all patients staged Tumors are considered unresectable or borderline re-
as cT3N0 or cT3N1 regardless of tumor site (low, middle, or sectable per initial MRI either because the primary tumor or
upper rectum), location (anterior or posterior), depth of involved lymph nodes abut or breach the mesorectal fascia
invasion, or proximity to other vital structures, despite the or there is tumor outside the mesorectal fascia with local
ability of modern MRI to predict the risk of both local extension to pelvic side-wall and sacrum or nodal involve-
recurrence and distant metastasis. The low rate of local ment in the lateral pelvic lymph nodes. Lateral pelvic lymph

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 95


GARCIA-AGUILAR, GLYNNE-JONES, AND SCHRAG

node involvement may reach as high as 15% when tumors or chemoradiotherapy with a brachytherapy boost may be
are low and bulky with multiple visible and presumed in- preferable to surgery for patients at high risk due to age or
volved mesorectal nodes. These lateral pelvic lymph nodes, multiple comorbidities.
although considered regional by the American Joint Com-
mittee on Cancer classification, are not routinely resected
MRI Is the Key to Selection
during a standard total mesorectal excision operation. In
Modern phased-array coil MRI using various sequences and
these circumstances, a degree of response to chemo-
multiple planes provides high-resolution tissue imaging and
radiotherapy is considered essential prior to surgery.41
excellent anatomic depiction of the rectum and other pelvic
Borderline resectable tumors are usually in the low or
structures relative to tumor location. MRI can predict in-
midrectum. They demonstrate various adverse features
volvement of the mesorectal fascia with a sensitivity of 77%
such as extension beyond the muscularis propria by 10 mm
(95% CI, 57%90%) and specificity of 94% (95% CI, 88%
to 15 mm, multiple nodal metastases in the mesorectum
97%).48 The MERCURY trial, a prospective observational
(N2), or the presence of gross extramural vascular invasion.
study that assessed the accuracy of MRI in predicting a
These features are associated with a higher risk of local
curative resection in rectal cancer, reported 92% specificity
recurrence and a high risk of metastatic disease. There are
in predicting a negative circumferential resection margin.49
sometimes less-evident clinical features that may alert the
On multivariate analysis, MRI involvement of the circum-
surgeon to the potential difficulty of the operation and the
ferential resection margin was the only preoperative staging
risk of not achieving an R0 resection. In particular, an an-
parameter that remained predictive for local recurrence and
teriorly placed tumor at or below the level of the prostate,
survival.50
in a large male with a narrow pelvis, is predictive of a
Therefore, MRI is essential to stratify patients into risk
technically challenging dissection. The risk of a positive
categories for local recurrence according to the proximity of
circumferential resection margin in an anteriorly located
the tumor to the circumferential resection margin. These risk
distal rectal cancer is as high as 30% even when treated with
categories are used to separate and select low-risk patients
an abdominoperineal excision. In these circumstances,
who are unlikely to benefit from radiotherapy. MRI can also
chemoradiotherapy to downsize the tumor will reduce the
define a group of patients with a high risk of recurrence
risk of a circumferential resection margin or a defect in the
and metastases, typically suggested by extramural spread,
mesorectum.
extramural vascular invasion, peritoneal reflection involve-
Although the literature suggests that radiotherapy may
ment, and tumor proximity to the levator muscle and
not fully compensate for a positive circumferential resection
sphincter. High risk of metastatic disease means that the
margin, it may reduce the risk of local recurrence for patients
use of chemotherapy at systemically effective doses would
with a suboptimal total mesorectal excision surgery.42
seem essential to improve survival rates for patients with
Surgeons in the United Kingdom who are coming out of
locally advanced rectal cancer. This raises the question of
training may have little experience and certainly face
whether chemoradiotherapy is required if neoadjuvant
a learning curve. As a radiation oncologist, without the
chemotherapy is effective in downstaging the tumor.
availability of records with multiple photographic images of
the surgical specimens to determine the quality of the
mesorectal excision in previous cases, I would likely rec- When Can Radiation Be Omitted?
ommend preoperative chemoradiotherapy. Recent data43 The risk of local recurrence for patients with locally advanced
suggest that many patients with rectal cancer in the United rectal cancer is dependent on tumor stage, the distance of
States are treated by surgeons who perform fewer than six the tumor from the anal verge, and the proximity of the
procedures per year. The circumferential resection margin tumor to the mesorectal fascia.50
(when reported) in such cases has been documented as Tumors located in the upper rectum, distant from the
positive in 17.2% of patients. Although SCPRT or chemo- mesorectal fascia, have a low risk of local recurrence when
radiotherapy do not completely compensate for inadequate treated with total mesorectal excision. The added benefit of
surgery, there is sufficient historical data27,28,44,45 to argue radiation therapy in these patients has been questioned, as
that surgeons entering the learning curve or practicing rectal radiation is associated with substantial toxicity including
cancer surgery in a low-volume environment should offer bowel obstruction, hip fractures, sexual and urinary dys-
their patients neoadjuvant chemoradiotherapy or SCPRT, function, and proctitis.51-53 A growing body of evidence
even if the features mentioned in the preceding paragraph suggests that radiation therapy could be safely avoided for
are not necessarily present. patients with intermediate-risk rectal cancer, such as rectal
Preoperative radiation or chemoradiotherapy followed by cancers located between 5 cm to 12 cm from the anal verge
local excision is also an alternative to total mesorectal ex- that do not threaten the mesorectal fascia on MRI.54,55
cision for patients with early stage rectal cancers treated In a pilot phase II trial conducted at Memorial Sloan
with curative intent.46,47 Chemoradiotherapy and local ex- Kettering Cancer Center, 32 patients with resectable clini-
cision can also be used as a compromise treatment of pa- cally staged II to III rectal cancer were treated with pre-
tients with more advanced rectal cancers who refuse an operative FOLFOX (oxaliplatin, 5-fluorouracil [5-FU], and
abdominoperineal excision of the rectum. Finally, radiation folinic acid) plus antivascular endothelial growth factor and

96 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


RECTAL CANCER TREATMENT: LESS MAY BE MORE

selective chemoradiotherapy based on tumor response.56 chemoradiotherapy dates to 2004, when the German rectal
The 30 patients who completed preoperative chemotherapy cancer trial demonstrated better local control with pre-
experienced tumor regression and underwent total meso- operative chemoradiotherapy with OS identical for pre- and
rectal excision without preoperative chemoradiotherapy. postoperative treatment.28,60 5-FUbased postoperative
No local recurrences were noted at 4 years, and the DFS was chemotherapy was included in both arms of the German
84%.56 These data were used as proof-of-concept for the trial. Also in 2004, the MOSAIC study in stage II and III colon
design of the phase II/III PROSPECT trial, which is now ac- cancer compared 12 cycles of infusional 5-FU to 12 cycles
cruing worldwide. The overarching goal of this randomized of FOLFOX and demonstrated superior survival with
trial is to determine whether pelvic radiation therapy can be FOLFOX.61,62 The benefits were not manifest in the stage II
used selectively for patients with locally advanced rectal subgroup. Although patients with rectal cancer were not
cancer.55 The trial utilizes selective rather than reflexive included, many clinicians have interpreted the results of
chemoradiotherapy to individualize treatment based on this study as pertinent to rectal cancer under the as-
each patients response to neoadjuvant FOLFOX. The hope is sumption that if FOLFOX is preferable for stage III colon
that a trial tailoring therapy more precisely, based on clinical cancer, then logically it should be preferred for stage II and
subgroups and tumor response to treatment, will help III rectal cancer as well.
eliminate the over- or undertreatment noted in previous The persistent controversy regarding adjuvant treatment
trials.54 of rectal cancer can be traced to varying interpretations of
In summary, chemoradiotherapy before total mesorectal these foundational studies. One school of thought considers
excision is currently the treatment of choice for patients with the clear benefit from the original trials conducted in the late
high-risk locally advanced rectal cancer, including those with 1980s as compelling evidence for postoperative 5-FU as
involvement of the pelvic sidewall or mesorectal fascia. In part of the package. Many also accept the legitimacy of
more straightforward cases, there is a question of whether extrapolating from colon to rectal cancer and therefore
chemoradiotherapy alone or surgery alone should be con- including oxaliplatin with 5-FU. Adherents of the opposing
sidered given the long-term effects of both treatments. The viewpoint correctly point out that there is a dearth of
risks presented by either course should be estimated and high-powered studies examining postoperative systemic
discussed with the patient. The argument for any of these chemotherapy among patients treated with chemo-
approaches rests on the provision of using good quality MRI, radiotherapy and that there is compelling evidence that
good quality radiotherapy, and the surgeons record of good patients with stage II tumors and/or cPR have very favorable
performance of total mesorectal excision surgery within the prognoses, particularly in the setting of high-quality pre-
mesorectal plane. operative imaging and optimal surgical technique, even in
the absence of systemic treatment. Posthoc analyses of
complete responders to chemoradiotherapy treated with
ADJUVANT SYSTEMIC CHEMOTHERAPY FOR and without chemotherapy have failed to show the com-
LOCALLY ADVANCED (CLINICAL STAGE II AND III) pelling advantage of the former approach.60,63,64
RECTAL CANCER
The controversy regarding adjuvant systemic chemotherapy
for rectal cancer is best understood in its historical con- Does Systemic Chemotherapy Improve Rectal Cancer
text. In 1990, an American consensus conference57 rec- Outcomes After Preoperative Chemoradiotherapy
ommended postoperative chemoradiotherapy and systemic and Surgical Resection?
5-FU on the basis of two randomized trials that demon- Trials that have specifically addressed the benefit of systemic
strated better OS and reduced local and distant re- chemotherapy compared with observation alone following
currence rates compared with surgery alone or to neoadjuvant chemoradiotherapy are few and have faced
surgery with radiation. 58,59 These trials did not distin- considerable challenges. The EORTC 2991 study, which now
guish between postoperative chemoradiotherapy and has 10 years of follow-up, used a two-by-two factorial design
postoperative chemotherapy, and therefore the benefits to evaluate the inclusion of systemic chemotherapy in both
realized were bundled as a multicomponent package. Since preoperative treatment and postoperative therapy.65 With
then, adjuvant systemic treatment has been an entrenched 500 patients treated with adjuvant therapy and 500 without,
ingredient of curative-intent treatment for locally ad- the power to detect small differences was necessarily lim-
vanced rectal cancer and, indeed, has been adopted as a ited. Although both DFS and OS rates favored the adjuvant
measure of care quality. Unfortunately, the evidence un- treatment arms, the differences were small: 3.3% for DFS
derpinning this treatment recommendation is suboptimal, and 3.4% for OS at 10 years (Table 1). The primary im-
particularly for patients with stage II locally advanced rectal pediment to interpretation of this study is that it used a
cancer. much older systemic treatment regimen, Mayo Clinicstyle
The prevailing approach to curative-intent treatment of bolus 5-FU, which is now recognized to have less efficacy and
rectal cancer for reasonably fit patients in the United States greater toxicity than infusional regimens.
is chemoradiotherapy followed by total mesorectal excision The Dutch Colorectal Cancer Group planned the PROCTOR/
and eight cycles of FOLFOX. The shift to neoadjuvant SCRIPT trial with 840 patients to detect a difference between

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 97


GARCIA-AGUILAR, GLYNNE-JONES, AND SCHRAG

TABLE 1. Outcomes of Patients With Rectal Cancer After Receiving Neoadjuvant Therapy and Subsequent
Treatment With Nonoperative Management or Total Mesorectal Excision

Interval to Salvage After Overall Survival Disease-Free


cCRs pCRs Regrowth in Regrowth in Regrowth in NOM vs. OM Survival NOM vs. Permanent Stoma Evidence
Reference (NOM) (OM) NOM (%) NOM (months) NOM (%)* (p value) OM (p value) NOM vs. OM Level**
Habr-Gama 71 22 2 (3) 60 2 (100) 5-year; 100% vs. 5-year; 92% vs. 0% vs. 41% 3B
et al 20045 88% (p = .01) 83% (p = .09)
Maas et al 21 20 1 (5) 22 1 (100) 2-year; 100% vs. 2-year; 89% vs. 0% vs. 45% 3B
20116 93% (p = .23) 91% (p = .77)
Smith et al 32 57 6 (19) 11 6 (100) 2-year; 96% vs. 2-year; 88% vs. NS 3B
20127 100% (p = .56) 98% (p = .27)
Araujo et al 42 69 5 (12) 48 4 (80) 5-year; 72% vs. 5-year; 61% vs. 7% vs. 19% 4
20159 90% (p = .32) 83% (p = .04)
Li et al 201510 30 92* 2 (7) 22 2 (100) 5-year; 100% vs. 5-year; 90% vs. 0% vs. 43% 4
96% (p = .26) 94% (p = .51)
*Patients with cCR subjected to surgery.
**Level according to Oxford Centre for Evidence-Based Medicine levels of evidence and grades of recommendations for therapeutic interventions.
Abbreviations: NOM, nonoperative management; TME, total mesorectal excision; cCR, clinically complete response; pCR, pathologic complete response; OM, operative management;
NS, not significant.

60% to 70% in OS.66 Unlike the EORTC 2291 trial, patients and was closed prematurely. As a result, clinicians who treat
were randomly assigned to receive 5-FU postoperatively rectal cancer do not have an answer to this fundamental
and after completing preoperative chemoradiotherapy. question. However, the low accrual rates in both the United
PROCTOR/SCRIPT accrued poorly. It was amended to Kingdom and the Netherlands, attributed to lack of equipoise,
substitute eight cycles of capecitabine for the original bolus make it unlikely that trials with an observation arm will ever
5-FU regimens, but it still did not accrue well and closed be conducted. Instead, focus has shifted to intensive efforts to
after a total of 437 patients had been assigned. With 5 years characterize the phenotypes and genotypic subsets of rectal
of follow-up, there is a 7% increase in 5-year DFS in the cancer that are associated with poor prognosis and/or greater
adjuvant therapy arm. This difference did not reach signif- responsiveness to systemic therapy and to tailor treatment
icance. Moreover, OS is nearly identical between the arms. thresholds to risk. Indeed, most recent rectal cancer clinical
An Italian trial reported by Cionini et al randomly assigned trials examine strategies for tailoring treatment to risk profile
635 patients with stage II/III rectal cancer to receive six to strike a balance between either under- or overtreatment.
cycles of bolus 5-FU and leucovorin or observation, and it
found no difference in OS (Table 2).67 Does the Addition of Oxaliplatin to 5-FU Improve
Overall, trials that have directly addressed the question of Outcomes Compared With 5-FU Alone?
the benefit of postoperative adjuvant systemic therapy for More recent trials have assumed that systemic therapy is
patients with rectal cancer treated with neoadjuvant che- beneficial and have focused on addressing the benefit of
moradiotherapy have not demonstrated the superiority of adding oxaliplatin to 5-FU in postoperative adjuvant regi-
treatment to observation. However, it is also fair to say that mens for rectal cancer. In the wake of MOSAIC, oncologists
these studies have not demonstrated the absence of benefit. have accepted the transitive principle and presumed that if
These trials had trouble accruing, were underpowered, and adding oxaliplatin is better than adjuvant infusional 5-FU/
used antiquated bolus 5-FU/leucovorin regimens. The only leucovorin alone in colon cancer, the same must be true in
study that included oxaliplatin68 failed to complete accrual rectal cancer. Typically, the 12 cycles used in MOSAIC are

TABLE 2. Randomized Clinical Trials Comparing Postoperative Systemic Chemotherapy With Observation of
Patients With Rectal Cancer Treated with Preoperative Pelvic Radiotherapy
Disease-Free Survival Overall Survival
Study (Maturity) No. Patients Adjuvant Treatment Rx+ vs. Rx- Difference Rx+ vs. Rx- Difference Reference
EORTC2291 (10-year) 1,101 4 cycles of bolus 5-FU/LV every 47 vs. 43.7 3.3% 51.8 vs. 48.4 3.4% Bossett et al44,65
3 weeks
PROCTOR/SCRIPT 437; closed early 8 cycles of capecitabine or bolus 62.7 vs. 55.4 7.3% 80.4 79.2% Breugom et al66
(5-year) 5-FU/LV
CHRONICLE (3-year) 113; closed early 6 cycles of CaPOx 77.5 vs. 71.3 6.2% 87.8 vs. 88.8 21% Glynne-Jones et al68
Cionini 635 6 cycles of bolus 5-FU/LV every N/A N/A 68 vs. 64 4% Cionini et al67
4 weeks
Abbreviations: 5-FU, 5-fluorouracil; LV, leucovorin; Rx+, with treatment; RX-, without treatment; CaPOx, capecitabine and oxaliplatin; N/A, not applicable.

98 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


RECTAL CANCER TREATMENT: LESS MAY BE MORE

reduced to eight cycles. Somewhat arbitrarily, credit for prefer to administer infusional 5-FU without oxaliplatin
four cycles is given for chemoradiotherapy. rather than no adjuvant systemic therapy. Conversely, for
Notwithstanding the paucity of direct evidence that any patients with substantial comorbidities that suggest life
postoperative chemotherapy is superior to observation in expectancy of less than 5 years, omitting treatment seems
locally advanced rectal cancer, several randomized trials most appropriate.
have directly compared oxaliplatin with nonoxaliplatin- The challenge arises in the treatment of patients with cT3
containing regimens for postoperative adjuvant treatment node-negative tumors with complete or favorable patho-
of rectal cancer. Specifically, the ECOG E320169 study sought logic responses to neoadjuvant chemoradiotherapy in-
to compare eight cycles of postoperative 5-FU and leuco- cluding cT3N0 and yT0N0, T1N0 or T2N0. In this context,
vorin with two to eight cycles of FOLFOX after standard there is reluctance to administer postoperative FOLFOX
neoadjuvant chemoradiotherapy and surgery. The study given its toxicity and the imperfect evidence supporting its
accrued fewer than 100 patients, with surveys indicating a use. The focus of recent and current randomized trials is to
perceived lack of equipoise. evaluate alternative preoperative management strategies
Fortunately, three non-U.S.based studies were more and completion of systemic chemotherapy prior to surgery,
successful. The PETACC-6 study randomly assigned 1,069 so it is possible that this conundrum will eventually have less
patients to receive either six cycles of adjuvant capecitabine salience.
and oxaliplatin (CAPOX) or to capecitabine alone. Although Nevertheless, how can todays clinicians arrive at informed
median follow-up was only 31 months, 3-year DFS rates were decisions in the face of imperfect information? There are
nearly identical in both arms.70 The most recent study from two strategies that may be helpful. The first is careful
the German Rectal Cancer Group randomly assigned 1,265 consideration of the most appropriate treatment prior to
patients to receive either four cycles of Mayo Clinicstyle initiation of any treatment. Although not infallible, MRI
bolus 5-FU or eight cycles of postoperative FOLFOX. With staging helps to identify relatively small rectal cancers with
50 months of follow-up, 3-year DFS was 71.2% for 5-FU and low likelihood of nodal involvement. For rectal tumors that
75.9% for FOLFOX, a difference of 4.7%.15 Mature data and a are cT2N0 or cT3N0, it makes sense to consider up-front
final report have not yet been published. In contrast, the surgical resection. Many of these patients will have node-
recent randomized ADORE trial from South Korea revealed negative disease and can reasonably avoid systemic therapy.
an advantage for FOLFOX compared with Mayo Clinicstyle Patients with node-positive disease require both post-
bolus 5-FU/leucovorin. This study randomly assigned 321 operative chemoradiotherapy and systemic chemotherapy.
patients and reported a 71.6% 3-year DFS rate in the FOLFOX Although less well-tolerated than preoperative treatment, it
group compared with 62.9% in the 5-FU/leucovorin group.71 is important to recall that the German trial showed similar
This difference was significant with a hazard ratio (HR) of OS irrespective of treatment timing.28,60
0.66 (95% CI, 0.430.99; p = .047). In the FOLFOX arm, the The second approach to adjuvant treatment recommen-
3-year OS was 95% versus 85.7% in the control arm, a dif- dations in clinical stage II rectal cancer or in the setting of
ference of 9.3% with a HR of 0.46 (95% CI, 0.220.97). This pCR to neoadjuvant chemoradiation is to scrutinize the
study has been critiqued for its small sample size and the phenotypic and genotypic tumor characteristics for high-risk
different 5-FU backbones in the two arms. As a result, it is features. Recently, Dalerba et al identified the transcription
unclear whether the better outcomes in the intervention factor CDX2 as a negative prognostic factor in stage II and III
arm were attributable to the superiority of infusional versus colon cancer.72 They found that 4.1% of colon cancers lack
bolus 5-FU or to the addition of oxaliplatin. CDX2 expression. In patients with stage II disease, 5-year DFS
was approximately 50% for the subset lacking CDX2 ex-
pression and over 80% for the majority with CDX2 expres-
Treatment Decision Making in the Absence of Perfect sion. Moreover, the 5-year DFS was 91% for the 23 patients
Information with CDX2-positive stage II disease treated with adjuvant
For patients treated with preoperative chemoradiotherapy, chemotherapy and 56% for the 25 patients with CDX2-
decisions about postoperative systemic therapy must be positive disease who did not receive adjuvant treatment.
tailored to the risk of metastatic disease based on pathology. Though information on the prevalence of CDX2 in patients
They must also take into account the patients life expec- with rectal cancer is not yet available, administration of
tancy independent of the cancer, as well as the patients adjuvant systemic treatment to patients with rectal cancer
preferences. In some contexts, decision making is easy. For lacking CDX2 expression is justified even in the setting of
example, extrapolating from the MOSAIC trial, patients with node-negative disease or pCR, given the magnitude of this
node-positive rectal cancer are often recommended eight finding. This study highlights the value of including tissue
cycles of FOLFOX. For patients with no evidence of nodal correlatives alongside clinical trials and using bioinformatics
involvement and excellent response, chemoradiotherapy to identify heterogeneity of risk and treatment re-
may be omitted. In this context, it is important to attenuate sponsiveness. Further discoveries like these practice-
or remove oxaliplatin altogether before peripheral neu- changing results are anticipated and will help us to tailor
ropathy becomes severe (grade 3). Among patients who are adjuvant treatment recommendations to individual risk
frail or do not tolerate oxaliplatin, some oncologists still profiles.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 99


GARCIA-AGUILAR, GLYNNE-JONES, AND SCHRAG

Practice Guidelines numeracy. Clinical trial participation should always be


A considerable body of evidence demonstrates favorable prioritized.
outcomes among patients with average-risk stage II rectal Total mesorectal excision, although effective in the
cancer treated with chemoradiotherapy who have complete or majority of locally advanced rectal cancer cases and still
near-complete pathologic response without adjuvant systemic the mainstay of curative-intent treatment of rectal can-
treatment. European and U.S. practice guidelines recommend cer, harbors the risks of substantial mortality, morbidity,
treatment decisions tailored to individual patient preferences and long-term quality-of-life issues. There is a growing
but interpret the evidence base somewhat differently. In the body of evidence that supports nonoperative manage-
United States, the 2015 National Comprehensive Cancer ment for patients whose disease demonstrates a cCR to
Network guideline recommends postoperative adjuvant sys- neoadjuvant chemoradiotherapy. However, this approach
temic therapy for all patients treated with neoadjuvant che- must be paired with frequent and thorough follow-up,
moradiotherapy irrespective of response.73 The inclusion of especially in the first year after completion of chemo-
oxaliplatin is suggested. In Europe, the European Society of radiotherapy, and patients must be counseled about the
Medical Oncology guideline advises postoperative chemo- risks both of the nonoperative management approach and
therapy in stage III and high-risk stage II cancers.74 However, in the possibility that surgery may be necessary if recurrence
some countries, treatment is not consistently recommended in is detected.
the setting of complete pathologic response and a recent Chemoradiotherapy should be offered to patients with
European Society of Medical Oncology consensus statement unresectable or borderline unresectable tumors and re-
acknowledges persistent uncertainty.75 sectable tumors with high-risk features. Because of its
In summary, there is still much work to be done in elu- proven ability to eradicate some tumors and its less onerous
cidating the circumstances in which adjuvant systemic safety profile, chemoradiotherapy alone may be the most
chemotherapy can be safely omitted from treatment for appropriate choice for selected patients, particularly those
locally advanced rectal cancer. Although there is some ev- unfit or unwilling to undergo surgery.
idence to suggest the safety of omitting this treatment for For some patients with no evidence of clinical nodal in-
some patients with stage II locally advanced rectal cancer, volvement, no high-risk features, and excellent response to
this treatment is indispensable in many situations, notably chemoradiotherapy, the omission of postoperative che-
for patients with post-chemoradiotherapy invasive or node- motherapy may be reasonable provided that there has been
involved tumors and tumors lacking CDX2 expression. explicit discussion about the imperfect evidence supporting
this decision. However, when patients with clinical stage II
rectal cancer or patients with a good response to chemo-
CONCLUSION radiotherapy decide to receive postoperative systemic
All patients embarking on curative-intent therapy for rectal therapy, it is critical to dose-reduce and/or discontinue
cancer should be informed of the risks, benefits, and un- oxaliplatin altogether in the setting of incipient peripheral
certainties of surgery, chemoradiotherapy, and adjuvant neuropathy to prevent long-term adverse effects; in some
systemic therapy, the three pillars of treatment. Modern patients, oxaliplatin may be avoided entirely. Patients must
MRI technology is essential to informed decision making, but understand that when neoadjuvant chemoradiotherapy is
patients comorbidities, natural life expectancy, and pref- administered, postoperative systemic treatment is part of
erences must be considered. The conversations about the package, because it is not always possible to know
treatment recommendations must convey risks and benefits whether the tumor did or did not involve lymph nodes at
in simple language that takes into account patients the outset.

References
1. Heald RJ, Smedh RK, Kald A, et al. Abdominoperineal excision of the therapy: long-term results. Ann Surg. 2004;240:711-717, discussion 717-
rectuman endangered operation. Norman Nigro Lectureship. Dis 718.
Colon Rectum. 1997;40:747-751. 6. Maas M, Beets-Tan RG, Lambregts DM, et al. Wait-and-see policy for
2. Hoerske C, Weber K, Goehl J, et al. Long-term outcomes and quality of clinical complete responders after chemoradiation for rectal cancer.
life after rectal carcinoma surgery. Br J Surg. 2010;97:1295-1303. J Clin Oncol. 2011;29:4633-4640.
3. Maas M, Nelemans PJ, Valentini V, et al. Long-term outcome in patients with a 7. Smith JD RJ, Ruby JA, Goodman KA, et al. Nonoperative management of
pathological complete response after chemoradiation for rectal cancer: rectal cancer with complete clinical response after neoadjuvant ther-
a pooled analysis of individual patient data. Lancet Oncol. 2010;11:835-844. apy. Ann Surg. 2012;256:965-972.
4. Habr-Gama A, de Souza PM, Ribeiro U Jr, et al. Low rectal cancer: impact 8. Smith JJ, Chow OS, Eaton A, et al. Organ preservation in patients with
of radiation and chemotherapy on surgical treatment. Dis Colon Rec- rectal cancer with clinical complete response after neoadjuvant ther-
tum. 1998;41:1087-1096. apy. J Clin Oncol. 2015;33 (suppl 3; abstr 509).
5. Habr-Gama A, Perez RO, Nadalin W, et al. Operative versus nonoperative 9. Araujo RO, Valad~ao M, Borges D, et al. Nonoperative management of
treatment for stage 0 distal rectal cancer following chemoradiation rectal cancer after chemoradiation opposed to resection after complete

100 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


RECTAL CANCER TREATMENT: LESS MAY BE MORE

clinical response. A comparative study. Eur J Surg Oncol. 2015;41: locally advanced rectal cancer treated with chemoradiation plus in-
1456-1463. duction or consolidation chemotherapy, and total mesorectal excision
10. Li J, Liu H, Yin J, et al. Wait-and-see or radical surgery for rectal cancer or nonoperative management. BMC Cancer. 2015;15:767.
patients with a clinical complete response after neoadjuvant chemo- 26. Garcia-Aguilar J, Chen Z, Smith DD, et al. Identification of a biomarker
radiotherapy: a cohort study. Oncotarget. 2015;6:42354-42361. profile associated with resistance to neoadjuvant chemoradiation
11. Renehan AG, Malcomson L, Emsley R, et al. Watch-and-wait approach therapy in rectal cancer. Ann Surg. 2011;254:486-492, discussion 492-
versus surgical resection after chemoradiotherapy for patients with 493.
rectal cancer (the OnCoRe project): a propensity-score matched cohort 27. Swedish Rectal Cancer Trial. Improved survival with preoperative ra-
analysis. Lancet Oncol. 2015;17:174-183. diotherapy in resectable rectal cancer. N Engl J Med. 1997;336:980-987.
12. OConnell MJ CL, Colangelo LH, Beart RW, et al. Capecitabine and 28. Sauer R, Becker H, Hohenberger W, et al; German Rectal Cancer Study
oxaliplatin in the preoperative multimodality treatment of rectal Group. Preoperative versus postoperative chemoradiotherapy for
cancer: surgical end points from National Surgical Adjuvant Breast and rectal cancer. N Engl J Med. 2004;351:1731-1740.
Bowel Project trial R-04. J Clin Oncol. 2014;32:1927-1934. 29. Quirke P, Steele R, Monson J, et al; MRC CR07/NCIC-CTG CO16 Trial
13. Aschele C, Cionini L, Lonardi S, et al. Primary tumor response to pre- Investigators; NCRI Colorectal Cancer Study Group. Effect of the plane
operative chemoradiation with or without oxaliplatin in locally ad- of surgery achieved on local recurrence in patients with operable rectal
vanced rectal cancer: pathologic results of the STAR-01 randomized cancer: a prospective study using data from the MRC CR07 and NCIC-
phase III trial. J Clin Oncol. 2011;29:2773-2780. CTG CO16 randomised clinical trial. Lancet. 2009;373:821-828.
14. Gerard JP, Azria D, Gourgou-Bourgade S, et al. Comparison of two 30. Law WL, Ho JW, Chan R, et al. Outcome of anterior resection for stage II
neoadjuvant chemoradiotherapy regimens for locally advanced rectal rectal cancer without radiation: the role of adjuvant chemotherapy. Dis
cancer: results of the phase III trial ACCORD 12/0405-Prodige 2. J Clin Colon Rectum. 2005;48:218-226.
Oncol. 2010;28:1638-1644. 31. Frasson M, Garcia-Granero E, Roda D, et al. Preoperative chemo-
15. Rodel C, Liersch T, Becker H, et al; German Rectal Cancer Study Group. radiation may not always be needed for patients with T3 and T2N+
Preoperative chemoradiotherapy and postoperative chemotherapy rectal cancer. Cancer. 2011;117:3118-3125.
with fluorouracil and oxaliplatin versus fluorouracil alone in locally 32. Mathis KL, Larson DW, Dozois EJ, et al. Outcomes following surgery
advanced rectal cancer: initial results of the German CAO/ARO/AIO-04 without radiotherapy for rectal cancer. Br J Surg. 2012;99:137-143.
randomised phase 3 trial. Lancet Oncol. 2012;13:679-687. 33. Popek S, Tsikitis VL, Hazard L, et al. Preoperative radiation therapy for
16. Probst CP, Becerra AZ, Aquina CT, et al; Consortium for Optimizing the upper rectal cancer T3,T4/Nx: selectivity essential. Clin Colorectal
Surgical Treatment of Rectal Cancer (OSTRiCh). Extended intervals after Cancer. 2012;11:88-92.
neoadjuvant therapy in locally advanced rectal cancer: the key to 34. Kreis ME, Junginger T, Rodel C, et al. The optimult study concept -
improved tumor response and potential organ preservation. J Am Coll selective neoadjuvant chemoradiation therapy based on preoperative
Surg. 2015;221:430-440. MRI. Zentralbl Chir. 2010;135:302-306.
17. Garcia-Aguilar J, Renfro LA, Chow OS, et al. Organ preservation for 35. Strassburg J, Ruppert R, Ptok H, et al. MRI-based indications for neo-
clinical T2N0 distal rectal cancer using neoadjuvant chemoradiotherapy adjuvant radiochemotherapy in rectal carcinoma: interim results of a
and local excision (ACOSOG Z6041): results of an open-label, single-arm, prospective multicenter observational study. Ann Surg Oncol. 2011;18:
multi-institutional, phase 2 trial. Lancet Oncol. 2015;16:1537-1546. 2790-2799.
18. Cercek A, Goodman KA, Hajj C, et al. Neoadjuvant chemotherapy first, 36. Syk E, Glimelius B, Nilsson PJ. Factors influencing local failure in rectal
followed by chemoradiation and then surgery, in the management of cancer: analysis of 2315 patients from a population-based series. Dis
locally advanced rectal cancer. J Natl Compr Canc Netw. 2014;12: Colon Rectum. 2010;53:744-752.
513-519. 37. Kusters M, Marijnen CA, van de Velde CJ, et al. Patterns of local re-
19. Nilsson PJ, van Etten B, Hospers GA, et al. Short-course radiotherapy currence in rectal cancer; a study of the Dutch total mesorectal excision
followed by neo-adjuvant chemotherapy in locally advanced rectal trial. Eur J Surg Oncol. 2010;36:470-476.
cancerthe RAPIDO trial. BMC Cancer. 2013;13:279. 38. Feddern ML, Jensen TS, Laurberg S. Chronic pain in the pelvic area or
20. Habr-Gama A, Perez RO, Wynn G, et al. Complete clinical response after lower extremities after rectal cancer treatment and its impact on quality
neoadjuvant chemoradiation therapy for distal rectal cancer: charac- of life: a population-based cross-sectional study. Pain. 2015;156:
terization of clinical and endoscopic findings for standardization. Dis 1765-1771.
Colon Rectum. 2010;53:1692-1698. 39. Bregendahl S, Emmertsen KJ, Lous J, et al. Bowel dysfunction after low
21. Smith FM, Wiland H, Mace A, et al. Clinical criteria underestimate anterior resection with and without neoadjuvant therapy for rectal
complete pathological response in rectal cancer treated with neo- cancer: a population-based cross-sectional study. Colorectal Dis. 2013;
adjuvant chemoradiotherapy. Dis Colon Rectum. 2014;57:311-315. 15:1130-1139.
22. Duldulao MP, Lee W, Streja L, et al. Distribution of residual cancer cells 40. Peeters KC, Tollenaar RA, Marijnen CA, et al; Dutch Colorectal Cancer
in the bowel wall after neoadjuvant chemoradiation in patients with Group. Risk factors for anastomotic failure after total mesorectal ex-
rectal cancer. Dis Colon Rectum. 2013;56:142-149. cision of rectal cancer. Br J Surg. 2005;92:211-216.
23. Samdani T, Garcia-Aguilar J. Imaging in rectal cancer: magnetic reso- 41. Oh HK, Kang SB, Lee SM, et al. Neoadjuvant chemoradiotherapy affects
nance imaging versus endorectal ultrasonography. Surg Oncol Clin N the indications for lateral pelvic node dissection in mid/low rectal
Am. 2014;23:59-77. cancer with clinically suspected lateral node involvement: a multicenter
24. Lambregts DM, Lahaye MJ, Heijnen LA, et al. MRI and diffusion- retrospective cohort study. Ann Surg Oncol. 2014;21:2280-2287.
weighted MRI to diagnose a local tumour regrowth during long-term 42. Gosens MJEM, Klaassen RA, Tan-Go I, et al. Circumferential margin
follow-up of rectal cancer patients treated with organ preservation involvement is the crucial prognostic factor after multimodality
after chemoradiotherapy. Eur Radiol. Epub 2015 Oct 30. treatment in patients with locally advanced rectal carcinoma. Clin
25. Smith JJ, Chow OS, Gollub MJ, et al; Rectal Cancer Consortium. Organ Cancer Res. 2007;13:6617-6623.
preservation in rectal adenocarcinoma: a phase II randomized con- 43. Rickles AS, Dietz DW, Chang GJ, et al; Consortium for Optimizing the
trolled trial evaluating 3-year disease-free survival in patients with Treatment of Rectal Cancer (OSTRiCh). High rate of positive

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 101


GARCIA-AGUILAR, GLYNNE-JONES, AND SCHRAG

circumferential resection margins following rectal cancer surgery: a call of Colon Cancer (MOSAIC) Investigators. Oxaliplatin, fluorouracil, and
to action. Ann Surg. 2015;262:891-898. leucovorin as adjuvant treatment for colon cancer. N Engl J Med. 2004;
44. Kapiteijn E, Marijnen CA, Nagtegaal ID, et al; Dutch Colorectal Cancer 350:2343-2351.
Group. Preoperative radiotherapy combined with total mesorectal 62. Andre T, Boni C, Navarro M, et al. Improved overall survival with
excision for resectable rectal cancer. N Engl J Med. 2001;345:638-646. oxaliplatin, fluorouracil, and leucovorin as adjuvant treatment in stage II
45. Bosset JF, Collette L, Calais G, et al; EORTC Radiotherapy Group Trial or III colon cancer in the MOSAIC trial. J Clin Oncol. 2009;27:3109-3116.
22921. Chemotherapy with preoperative radiotherapy in rectal cancer. 63. Garca-Albeniz X, Gallego R, Hofheinz RD, et al. Adjuvant therapy sparing
N Engl J Med. 2006;355:1114-1123. in rectal cancer achieving complete response after chemoradiation.
46. Garcia-Aguilar J, Renfro LA, Chow OS, et al. Organ preservation for World J Gastroenterol. 2014;20:15820-15829.
clinical T2N0 distal rectal cancer using neoadjuvant chemoradiotherapy 64. Bujko K, Glynne-Jones R, Bujko M. Does adjuvant fluoropyrimidine-
and local excision (ACOSOG Z6041): results of an open-label, single-arm, based chemotherapy provide a benefit for patients with resected rectal
multi-institutional, phase 2 trial. Lancet Oncol. 2015;16:1537-1546. cancer who have already received neoadjuvant radiochemotherapy?
47. Verseveld M, de Graaf EJ, Verhoef C, et al. Chemoradiation therapy for A systematic review of randomised trials. Ann Oncol. 2010;21:
rectal cancer in the distal rectum followed by organ-sparing transanal 1743-1750.
endoscopic microsurgery (CARTS study). Br J Surg. 2015;102:853-860. 65. Bosset JF CG, Calais G, Mineur L, et al; EORTC Radiation Oncology Group.
48. Al-Sukhni E, Milot L, Fruitman M, et al. Diagnostic accuracy of MRI for Fluorouracil-based adjuvant chemotherapy after preoperative che-
assessment of T category, lymph node metastases, and circumferential moradiotherapy in rectal cancer: long-term results of the EORTC 22921
resection margin involvement in patients with rectal cancer: a sys- randomised study. Lancet Oncol. 2014;15:184-190.
tematic review and meta-analysis. Ann Surg Oncol. 2012;19:2212-2223. 66. Breugom AJ, van Gijn W, Muller EW, et al; Cooperative Investigators of
49. MERCURY Study Group. Diagnostic accuracy of preoperative magnetic Dutch Colorectal Cancer Group and Nordic Gastrointestinal Tumour
resonance imaging in predicting curative resection of rectal cancer: Adjuvant Therapy Group. Adjuvant chemotherapy for rectal cancer
prospective observational study. BMJ. 2006;333:779. patients treated with preoperative (chemo)radiotherapy and total
50. Taylor FG, Quirke P, Heald RJ, et al; MERCURY study group. Preoperative mesorectal excision: a Dutch Colorectal Cancer Group (DCCG) ran-
high-resolution magnetic resonance imaging can identify good prognosis domized phase III trial. Ann Oncol. 2015;26:696-701.
stage I, II, and III rectal cancer best managed by surgery alone: a pro- 67. Cionini L, Sainato A, De Paoli A, et al. Final results of randomized trial on
spective, multicenter, European study. Ann Surg. 2011;253:711-719. adjuvant chemotherapy after preoperative chemoradiation in rectal
51. Peeters KC, van de Velde CJ, Leer JW, et al. Late side effects of short-course cancer. Radiother Oncol. 2010;96:S113-S114.
preoperative radiotherapy combined with total mesorectal excision for 68. Glynne-Jones R, Counsell N, Quirke P, et al. Chronicle: results of a
rectal cancer: increased bowel dysfunction in irradiated patientsa Dutch randomised phase III trial in locally advanced rectal cancer after
colorectal cancer group study. J Clin Oncol. 2005;23:6199-6206. neoadjuvant chemoradiation randomising postoperative adjuvant
52. Birgisson H, Pahlman L, Gunnarsson U, et al; Swedish Rectal Cancer Trial capecitabine plus oxaliplatin (XELOX) versus control. Ann Oncol. 2014;
Group. Adverse effects of preoperative radiation therapy for rectal 25:1356-1362.
cancer: long-term follow-up of the Swedish Rectal Cancer Trial. J Clin 69. Benson AB, Catalan P, Meropol NJ, et al. ECOG E3201: Intergroup
Oncol. 2005;23:8697-8705. randomized phase III study of postoperative irinotecan, 5-fluorouracil
53. Joye I, Haustermans K. Early and late toxicity of radiotherapy for rectal (FU), leucovorin (LV) (FOLFIRI) vs oxaliplatin, FU/LV (FOLFOX) vs FU/LV
cancer. Recent Results Cancer Res. 2014;203:189-201. for patients (pts) with stage II/III rectal cancer receiving either pre or
54. Gunderson LL, Sargent DJ, Tepper JE, et al. Impact of T and N stage and postoperative radiation (RT)/FU. J Clin Oncol. 2006;24 (suppl; abstr
treatment on survival and relapse in adjuvant rectal cancer: a pooled 3526).
analysis. J Clin Oncol. 2004;22:1785-1796. 70. Schmoll HJ, Haustermans K, Price TJ, et al. Preoperative chemo-
55. Schrag D. Evolving role of neoadjuvant therapy in rectal cancer. Curr radiotherapy and postoperative chemotherapy with capecitabine and
Treat Options Oncol. 2013;14:350-364. oxaliplatin versus capecitabine alone in locally advanced rectal cancer:
56. Schrag D, Weiser M, Goodman KA, et al. Neoadjuvant chemotherapy Disease-free survival results at interim analysis. J Clin Oncol. 2014;32:5s
without routine use of radiation therapy for patients with locally ad- (suppl; abstr 3501).
vanced rectal cancer: a pilot trial. J Clin Oncol. 2014;32:513-518. 71. Hong YS, Nam BH, Kim KP, et al. Oxaliplatin, fluorouracil, and leucovorin
57. NIH consensus conference. Adjuvant therapy for patients with colon versus fluorouracil and leucovorin as adjuvant chemotherapy for locally
and rectal cancer. JAMA. 1990;264:1444-1450. advanced rectal cancer after preoperative chemoradiotherapy
58. Fisher B, Wolmark N, Rockette H, et al. Postoperative adjuvant che- (ADORE): an open-label, multicentre, phase 2, randomised controlled
motherapy or radiation therapy for rectal cancer: results from NSABP trial. Lancet Oncol. 2014;15:1245-1253.
protocol R-01. J Natl Cancer Inst. 1988;80:21-29. 72. Dalerba P, Sahoo D, Paik S, et al. CDX2 as a prognostic biomarker in stage
59. Gastrointestinal Tumor Study Group. Prolongation of the disease-free II and stage III colon cancer. N Engl J Med. 2016;374:211-222.
interval in surgically treated rectal carcinoma. N Engl J Med. 1985;312: 73. Benson AB III, Venook AP, Bekaii-Saab T, et al. Rectal cancer, version
1465-1472. 2.2015. J Natl Compr Canc Netw. 2015;13:719-728, quiz 728.
60. Sauer R, Liersch T, Merkel S, et al. Preoperative versus postoperative 74. Glimelius B, Tiret E, Cervantes A, et al; ESMO Guidelines Working Group.
chemoradiotherapy for locally advanced rectal cancer: results of the Rectal cancer: ESMO Clinical Practice Guidelines for diagnosis, treat-
German CAO/ARO/AIO-94 randomized phase III trial after a median ment and follow-up. Ann Oncol. 2013;24:vi81-vi88.
follow-up of 11 years. J Clin Oncol. 2012;30:1926-1933. 75. Schmoll HJ, Van Cutsem E, Stein A, et al. ESMO Consensus Guidelines for
61. Andre T, Boni C, Mounedji-Boudiaf L, et al; Multicenter International management of patients with colon and rectal cancer. a personalized
Study of Oxaliplatin/5-Fluorouracil/Leucovorin in the Adjuvant Treatment approach to clinical decision making. Ann Oncol. 2012;23:2479-2516.

102 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


GASTROINTESTINAL (NONCOLORECTAL) CANCER

Biliary Tract Cancer: The New and


the Old

CHAIR
Mary F. Mulcahy, MD
Robert H. Lurie Comprehensive Cancer Center of Northwestern University
Chicago, IL

SPEAKERS
John A. Bridgewater, MD, PhD
University College London Cancer Institute
London, United Kingdom

Karyn A. Goodman, MD
University of Colorado School of Medicine
Aurora, CO
BRIDGEWATER ET AL

Biliary Tract Cancer: Epidemiology, Radiotherapy, and


Molecular Profiling
John A. Bridgewater, MD, PhD, Karyn A. Goodman, MD, Aparna Kalyan, MD, and Mary F. Mulcahy, MD

OVERVIEW

Biliary tract cancer, or cholangiocarcinoma, arises from the biliary epithelium of the small ducts in the periphery of the liver
(intrahepatic) and the main ducts of the hilum (extrahepatic), extending into the gallbladder. The incidence and epidemiology
of biliary tract cancer are fluid and complex. It is shown that intrahepatic cholangiocarcinoma is on the rise in the Western
world, and gallbladder cancer is on the decline. Radiation therapy has emerged as an important component of adjuvant therapy
for resected disease and definitive therapy for locally advanced disease. The emerging sophisticated techniques of imaging
tumors and conformal dose delivery are expanding the indications for radiotherapy in the management of bile duct tumors. As
we understand more about the molecular pathways driving biliary tract cancers, targeted therapies are at the forefront of new
therapeutic combinations. Understanding the gene expression profile and mutational burden in biliary tract cancer allows us to
better discern the pathogenesis and identify promising new developmental therapeutic targets.

B iliary tract cancer, or cholangiocarcinoma, arises from


the biliary epithelium of the small ducts in the periphery
of the liver (intrahepatic) and the main ducts of the hilum
topography. This coding has recently changed three times (ICD-
0-1 to ICD-0-2 in 1993 and ICD-0-3 in 2001) and has been
adopted at different times in different countries. In ICD-0-2,
(extrahepatic). Extrahepatic biliary tract cancers include hilar cholangiocarcinoma could be classified pathologically us-
gallbladder cancer, ampullary cancer, and cancer of the ing a histology code, rather than an anatomic code, but was
pancreatic biliary ducts. Although extrahepatic cancers arise cross-referenced to the anatomic code for intrahepatic chol-
from similar epithelia, their etiology can be very different angiocarcinoma rather than extrahepatic cholangiocarcinoma.
because of their anatomy. Furthermore, hilar cholangiocarcinoma could also be correctly
The incidence of cholangiocarcinoma is modest in the western reported as extrahepatic cholangiocarcinoma by using other
world, between 0.35 to 2 per 100,000 annually; however, in histology codes.12 Additionally, there have been local changes in
China and Thailand, the incidence can be up to 40 times the rate data collection, such as the loss of independent verification of
observed in the United Kingdom and, thus, poses significant cause of death in the United Kingdom in 1993. The terminology
public health questions (Fig. 1).1,2 The incidence of gallbladder may also be difficult in certain locations such as in England and
cancer tends to be closely associated with its primary etiology, Wales, where only 1% of cholangiocarcinomas were classified
cholelithiasis. As such, the incidence is uniform for most of the as intrahepatic.5
Western world, however, disease clusters are found in northern This can be considered as an abrupt step change in the
India, Japan, and the Andes region (Fig. 2).3 recorded incidence, such as the step change in incidence in
The incidence of intrahepatic cholangiocarcinoma in the 2001 for intrahepatic cholangiocarcinoma.12 Despite these
Western world is rising.1 Data from the United Kingdom, noted step changes, the overall pattern is a rise in inci-
United States, and other countries show a consistent and dence demonstrated across multiple international data sets.
steady rise in the incidence of intrahepatic cholangiocarcinoma Changes in technology, such as improved stenting, would
from 0.1 to 0.6 per 100,000 over the last 30 years (Figs. 3 similarly present as step changes, although these are not
and 4).4-10 Several reasons have been cited for this change. discernable in the data. Additionally, the incidence appears
The International Classification of Diseases (ICD) for chol- consistent across different tumor sizes and mortality. The
angiocarcinoma has been confusing.11 There are multiple overall picture presents an increase in incidence that ap-
codes for cholangiocarcinoma depending on histology and pears to override short-term changes.

From the UCL Cancer Institute, London, United Kingdom; Department of Radiation Oncology, University of Colorado School of Medicine, Aurora, CO; Northwestern University, Chicago,
IL; Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Mary F. Mulcahy, MD, Robert H. Lurie Comprehensive Cancer Center of Northwestern University, 676 North St., Clair Suite 850, Chicago, IL 60611;
email: mmulcahy@nm.org.

2016 by American Society of Clinical Oncology.

e194 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


BILIARY TRACT CANCER

FIGURE 1. Incidence of Cholangiocarcinoma

One refuted hypothesis to explain the increase in intra- intrahepatic cholangiocarcinoma and carcinoma of unknown
hepatic cholangiocarcinoma is the increase in diagnoses origin.13
of intrahepatic cholangiocarcinomas that were previously The incidence of cholangiocarcinoma in Thailand dem-
thought to be metastases from a carcinoma of unknown onstrates the impact of chronic infection with liver fluke.
origin. Again, if true, this is unlikely to have a substantial Liver cancers rival HIV/AIDS, road traffic accidents, and
impact on incidence, as the numbers are modest. A number stroke as a primary cause of death in Thai males.2 Endemic
of criteria have been proposed to help differentiate between liver fluke infection (Opisthorchis viverrini) is related to
eating raw or poorly cooked fish over 20 years or longer. The
picture in China and Korea is similar, although the de-
scription of data is less detailed, and the culprit organism is
Clonorchis sinensis.14 Fluke-related cancer has a distinctive
microRNA and mutational profile compared with chol-
KEY POINTS
angiocarcinoma of the Western world. The relationship to
The incidence of intrahepatic cholangiocarcinoma in the malignancy appears to be related to the generation of radical
Western world is rising, and the incidence of gallbladder nitrogen species demonstrated in a murine model.
cancer is on the decline. In the Western world, cholangiocarcinoma is associated
The tendency for cholangiocarcinomas to recur locally with chronic inflammation of the biliary tree and hepatic
provides a rationale for additional local therapy after parenchyma. The lifetime incidence of cholangiocarcinoma
definitive surgery. in hepatitis C is modest at 3.5% at 10 years. The data on
A recent meta-analysis of 10 retrospective studies primary sclerosing cholangitis (PSC) suggest a lifetime risk of
evaluating adjuvant radiotherapy after curative 7% and rising, corresponding with the increasing incidence
resection for extrahepatic cholangiocarcinomas of PSC. As for PSC, this increase is primarily among men age 40
demonstrated a significant benefit in overall survival for
or younger and appears to spare the small ducts.
patients receiving adjuvant therapy.
The landscape of molecular mutations in biliary tract
Gallbladder cancer has a different pathophysiology and is
cancers has demonstrated multiple new targetable related primarily, but not entirely, to cholecystitis. The in-
mutations. cidence varies from England and Wales (0.1%) to the Czech
The majority of biliary tract cancer mediators seem to Republic (3.7%6.5%) and La Paz in Bolivia (7.5%13.2% per
affect the epigenetics and transcription. 100,000). The risk factors and demographics for gallbladder
cancer and gallstone disease closely parallel each other. As

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e195


BRIDGEWATER ET AL

FIGURE 2. Incidence of Gallbladder Cancer

such, there is a predominance of women and known risk factors determined by the location of the tumor and the extent of
for gallstone disease (age, obesity, multiple pregnancies, family invasion into adjacent structures. Biliary tract tumors are
history of gallstones, and low levels of physical activity). Chronic generally divided into intrahepatic cholangiocarcinomas,
infection is also implicated with the relative risk consequent on perihilar and extrahepatic cholangiocarcinomas, and gall-
Salmonella (typhi and paratyphi) and Helicobacter (bilis and bladder tumors. For intrahepatic cholangiocarcinomas, which
pylori) isolation, between 2.6 and 7.5. Interestingly, only 1% of are less common, a liver resection based on the anatomic
patients with gallstones develop gallbladder cancer, suggesting involvement of the liver is performed, whereas for extrahe-
that any screening program should be targeted to higher-risk patic cholangiocarcinoma, a pancreaticoduodenectomy may
groups. be required depending on the level of disease within the
The incidence of gallbladder cancer is decreasing in the biliary tree. Gallbladder cancers are often incidentally found
Western world and is related to the increase in routine after a nononcologic surgery, and a repeat oncologic resection
cholecystectomy. This is not the case in Chile, where public to obtain adequate margins at the liver parenchyma may be
health programs have diverted funding for cholecystectomy necessary.
to maternity services, resulting in a slow but substantial in- Even with a complete resection, the 5-year overall sur-
crease in the rate of gallbladder cancer. Nevertheless, pro- vival rates range from 21% to 63% for intrahepatic chol-
phylactic cholecystectomy programs have been proposed. angiocarcinoma, 30% to 40% for perihilar lesions, and
Primary sclerosing cholangitis and natural killer cell poly- 20% to 54% for distal cholangiocarcinomas managed by
morphisms have been associated with gallbladder cancer. pancreaticoduodenectomy.15-17 The recurrence patterns
Choledochal cysts have a 1% to 15% lifetime risk of de- differ by primary site in the biliary tract. Investigators at the
veloping into gallbladder cancer and cholangiocarcinoma. The Memorial Sloan Kettering Cancer Center reviewed the
risk of finding malignancy is higher when the diagnosis is made patterns of recurrence among 156 patients with extrahe-
in adults, but recurrence is rare if it is resected. patic cholangiocarcinoma and gallbladder tumors who un-
derwent definitive resection. Two-thirds of the patients in
ADJUVANT AND DEFINITIVE RADIOTHERAPY each group developed disease recurrence, with an isolated
FOR RESECTABLE AND LOCALLY ADVANCED locoregional disease as the first site of failure in 15% of
BILIARY TRACT CANCER patients with gallbladder cancer compared with 59% of
Role of Adjuvant Therapy for Biliary Tract Tumors patients with extrahepatic cholangiocarcinoma (p , .001).
Complete resection remains the gold standard of treat- By contrast, 85% of gallbladder cancers had distant spread as
ment and offers the best chance of cure for patients with the first site of failure compared with 41% of extrahepatic
biliary tract cancers. The type and extent of resection are cholangiocarcinomas (p , .001).18

e196 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


BILIARY TRACT CANCER

FIGURE 3. Age-Standardized Mortality Rates per FIGURE 4. Age-Adjusted (1970 U.S. Standard)
100,000 in England and Wales5 Incidence Rates of Primary Intrahepatic
Cholangiocarcinoma in the United States, 197319971

Reprinted with permission. 2001 American Association for the Study of Liver
Diseases. All rights reserved. Patel T: Hepatology. 2001;33:1353-1357.

series suggest a benefit, phase III data are lacking.19-22


Moreover, the indications for postoperative radiation
therapy are not yet clear. The most important prognosti-
cator for survival is a complete (R0) resection23,24; none-
theless, the available data suggest isolated locoregional
recurrence rates of up to 60% after curative resection.25-27
A recent meta-analysis of 10 retrospective studies eval-
uating adjuvant radiotherapy after curative resection for
extrahepatic cholangiocarcinomas demonstrated a sig-
nificant benefit in overall survival for patients receiv-
ing adjuvant therapy.28 The pooled hazard ratio (HR) for
overall survival for the addition of adjuvant radiation
therapy versus surgery alone was 0.62 (95% CI, 0.480.78),
despite the fact that the radiotherapy cohorts more fre-
quently had positive margins (69% vs. 31%; p , 0.001).
The authors concluded that adjuvant radiotherapy ap-
peared beneficial, however, this meta-analysis ulti-
mately only included eight studies on extrahepatic
cholangiocarcinomas. Conversely, a Surveillance, Epidemi-
ology, and End Results (SEER) analysis of adjuvant radio-
therapy failed to show a survival benefit, but the authors
caution that key data, including margin status and the use
of combined chemotherapy that might impact on overall
Reprinted by permission from Macmillan Publishers Ltd: Gut. Taylor-Robinson SD outcomes for these patients, were not available through the
et al. Increase in mortality rates from intrahepatic cholangiocarcinoma in England and
Wales 1968-1998. Gut. 2001;48:816-820. 2001.
SEER database.29
The recently reported Intergroup phase II study, led
by the Southwest Oncology Group (SWOG), evaluated
The tendency for cholangiocarcinomas to recur locally the use of adjuvant chemotherapy (gemcitabine and
provides a rationale for additional local therapy after de- capecitabine) followed by chemoradiation (concurrent
finitive surgery. Although several phase II and retrospective radiotherapy and capecitabine) for resected extrahepatic

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e197


BRIDGEWATER ET AL

cholangiocarcinoma or gallbladder tumors. There were a Brachytherapy


total of 79 evaluable patients, 21 (32%) of whom un- Brachytherapy has been used at numerous institutions.
derwent an R1 resection and 49 (62%) of whom had ex- Overall, there appears to be a small trend toward improved
trahepatic cholangiocarcinoma. Two-year overall survival survival with its use as a boost.42,43 Dose distributions can be
was 65% for all patients (67% and 60% in R0 and R1, re- quite satisfactory with the use of Ir-192 catheters. Ideally, an
spectively). Median overall survival was 35 months, and external beam dose designed to eradicate microscopic
only 14 patients developed a local recurrence.30 Grade 3 disease (i.e., 45 to 50 Gy) is given first, followed by seed
and 4 adverse events were observed for 53% and 11% of placement designed to give an additional high dose to the
patients. These promising data demonstrate the feasibility site of gross or highly suspect disease. Long-term compli-
of adjuvant chemotherapy followed by chemoradiation for cations secondary to the high doses and use of stents can
resected extrahepatic cholangiocarcinoma and gallbladder include stricture, bowel obstruction, and bleeding.
tumors.
Adjuvant radiotherapy may also benefit patients with
Bridge to Transplant
intrahepatic cholangiocarcinoma due to the high risk of
The role of liver transplantation for unresectable hilar
positive resection margins.15 An analysis of intrahepatic
cholangiocarcinoma has been established by the excellent
cholangiocarcinoma patients from the SEER database dem-
results from the Mayo Clinic using 4,500 cGy neoadjuvant
onstrated an improvement in median survival for adju-
chemoradiation and concurrent fluorouracil and a brachy-
vant radiotherapy versus surgery alone (11 vs. 6 months,
therapy boost followed by liver transplantation.44 The 5-year
respectively).31
survival rate was 82%, which compared favorably to another
group of patients who underwent resection with a 5-year
survival rate of only 21%. It must be emphasized that
Definitive Radiotherapy for Biliary Tract Tumors
transplantation is reserved for highly selected patients, and
In cases of unresectable nonmetastatic cholangiocarcinoma,
selection criteria are quite stringent.
the use of radiotherapy may improve pain control and bil-
iary decompression. A recent retrospective review of 37 pa-
tients with unresectable extrahepatic cholangiocarcinomas Advances in Radiotherapy Techniques
treated with radiotherapy at Duke University reported 2-year The advancement of radiotherapy over the last 2 decades
actuarial overall survival and local control rates of 22% and has been a remarkable achievement that has improved
71%, respectively. Two patients were alive without evidence delivery, outcomes, and quality of care for patients with
of recurrence at the time of analysis, thus demonstrating long- primary biliary tumors. Historically, radiotherapy for ab-
term survival in a small subset of patients.32 For intrahepatic dominal tumors has been limited by the tolerance of the
cholangiocarcinoma, a dosimetric analysis of patients treated surrounding normal organs to even relatively low doses of
at The University of Texas MD Anderson Cancer Center radiation. With the emergence of more conformal radio-
suggests that a biologically effective dose of greater than therapy techniques and sophisticated treatment-planning
80.5 Gy is associated with improved local control and long- software, more focal treatment fields are now possible.
term survival.33 Classic approaches to portals and fractionation have been
Several small, single-institution, prospective studies have abandoned in favor of more focal techniques of irradiating
evaluated the use of stereotactic body radiotherapy for liver tumors, such as intensity-modulated radiotherapy,
hilar and intrahepatic cholangiocarcinomas, suggesting image-guided radiotherapy, and the use of motion manage-
that the higher dose per fraction is feasible and results in ment. When planning treatment using intensity-modulated
encouraging local control rates for both peripheral and radiotherapy, the radiation dose is prescribed to the target
central biliary tumors.34-40 Stereotactic body radiotherapy volume and strict dose constraints are placed on normal
allows for highly focal dose distributions and sparing of the tissues. A computer-optimized algorithm generates a plan to
surrounding normal tissue; however, given the close deliver radiation of varying intensity to the target volume via
proximity of hilar masses to highly radiosensitive organs multiple beams or even via multiple arcs to meet the re-
such as the small bowel and stomach, stereotactic body quirements for the target volume coverage and normal
radiotherapy for hilar cholangiocarcinomas should be used tissue constraints. The overall result is a highly conformal
with caution and preferably on a clinical trial. A recent dose distribution that is customized to the shape of the
multi-institutional phase II study of hypofractionated tumor. The delivery of conformal intensity-modulated ra-
proton therapy (median dose 58 Gy) for intrahepatic diotherapy plans is predicated on the ability to accurately
cholangiocarcinoma demonstrated a local control rate of target the tumor during treatment, which has led to the
94% and 2-year overall survival of 46%.41 Short-term development of image-guided radiotherapy to reduce the
morbidity associated with radiotherapy includes nausea, uncertainties of tumor positioning. Diagnostic-quality im-
vomiting, malaise, fatigue, or weight loss and intermittent aging can now be performed on the linear accelerator to
fevers due to cholangitis. Long-term complications fol- allow real-time assessment of tumor positioning while
lowing radiotherapy are usually related to duodenal bleeding the patient is on the table prior to treatment delivery.
or stricture. Abdominal tumors can move with respiration, and, thus,

e198 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


BILIARY TRACT CANCER

adaptations to account for motion or reduce motion have PIK3CA/mTOR pathway aberrations were more common in
been developed to allow accurate delivery of the focal ra- extrahepatic cholangiocarcinomas and gallbladder tumors.
diotherapy fields while patients are breathing. These motion Mutations in TP53 have been long established in bili-
management techniques include using an abdominal com- ary tract cancers, with incidence ranging around 44% to
pression belt to minimize the excursion of the diaphragm 47%. Incidence is seen more frequently in extrahepatic
during respiration to reduce motion or to use respiratory cholangiocarcinoma compared with intrahepatic chol-
gating where the linear accelerator turn on and off with the angiocarcinoma, with rates as high as 17.5% and 8.6%,
respiratory cycle. In summary, the emerging sophisticated respectively.48 Multivariate analysis identified that TP53 and
techniques of imaging tumors and conformal dose delivery KRAS are independent predictors of survival.48 Similar fre-
are expanding the indications for radiotherapy in the quency and mutational burden were seen in a study by Churi
management of bile duct tumors. et al in which NGS was performed using a panel of 46 cancer-
related genes.49 In addition to corroborating the findings by
Ross et al, Churi et al found that genetic aberrations in
MOLECULAR TARGETS IN BILIARY TRACT chromatin-modulating genes, BAP1 and PBRM1, were as-
CANCERS sociated with worse overall survival and higher frequency of
A majority of biliary tract cancers (. 90%) are well- bony metastases for patients with extrahepatic chol-
differentiated, mucin-producing adenocarcinomas, whereas angiocarcinoma. KRAS mutations have been shown to
a very small proportion are squamous or small cell in occur at a higher rate in extrahepatic cholangiocarcinoma
origin.45,46 (42%47%) compared with intrahepatic cholangiocarcinoma
(15.7%22%) and gallbladder cancer (11%19.2%), con-
firmed by whole-exome sequencing studies and genomic
Patterns of Molecular Drivers in Biliary Tract Cancer profiling studies (Table 1).47-50
As we understand more about the molecular pathways in- Chromatin-remodeling genes BAP1 (encoding a nu-
volved in biliary tract cancers, targeted therapies are at the clear deubiquitinase), ARID1A (encoding a subunit of the
forefront of new therapeutic combinations. The landscape SWI/SNF chromatin-remodeling complexes), and PBRM1
of molecular mutations in biliary tract cancers has dem- (encoding a subunit of the ATP-dependent SWI/SNF chromatin-
onstrated multiple new targetable mutations. To date, Ross remodeling complexes) have been observed in biliary tract
et al has performed the most comprehensive review of the cancer. The incidence of ARID1A ranges from 11% to 20% in
molecular profile of biliary tract cancers.47 In this study, DNA intrahepatic cholangiocarcinoma specimens, although its
from 412 intrahepatic cholangiocarcinomas, 57 extrahe- incidence is 12% in extrahepatic cholangiocarcinoma and
patic cholangiocarcinomas, and 85 gallbladder cancers were 11% to 13% in gallbladder cancer.47-49,51 Similarly, BAP1 is
extracted and next-generation sequencing (NGS) was per- seen in 9% to 25% of intrahepatic cholangiocarcinomas
formed on these specimens. Genomic profiling encom- and 4% to 13% of gallbladder cancers. The incidence of
passed 182 cancer-related genes plus 37 introns from 14 PBRM1 was 11% to 17% in intrahepatic cholangiocarcinoma,
genes frequently rearranged in cancer. These results dem- 7.7% in gallbladder cancer, and 3.5% to 5% in extrahepatic
onstrate that biliary tract cancers all share genomic cholangiocarcinoma. The frequency of these chromatin-
aberrations in cell cycle regulators (specifically CDKN2B) remodeling genes indicates that not only is chromatin
and chromatin remodeling (ARID1A). Intrahepatic chol- remodeling a key component in cholangiocarcinoma, but
angiocarcinomas feature FGFR fusions, IDH1/2 sub- these mutations may lend themselves to be sensitive to
stitutions, BRAF substitutions, and MET amplifications with a therapeutic agents targeting the chromatin-remodeling
low KRAS mutational frequency.48 ERBB2 amplification and genes.47-49,51

TABLE 1. Incidence of Molecular Mutations in Biliary Tract Cancer as Determined by Genomic Sequencing

Mutation Intrahepatic Cholangiocarcinoma Extrahepatic Cholangiocarcinoma Gallbladder Cancer


ERBB2 Amplification 3% 11% 16%
BRAF Substitution 5% 3% 1%
KRAS 15%22% 42%47% 11%19.2%
PI3KCA Substitution 5% 7% 14%
FGFR1-3 Fusion 11%12.5% 0 3%
CDKN2A/B Loss 18% 17% 19%
IDH1/2 Substitution 15%23% 3%4% 0
ARID1A Alteration 11%20% 12% 11%13%
MET 4% 0 0
BAP1 9%25% 0 4%13%

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e199


BRIDGEWATER ET AL

Molecular Targets gemcitabine, capecitabine, and bevacizumab had a median


EGFR. The EGFR pathway is commonly expressed in biliary progression-free survival of 8.1 months and overall survival
tract cancer. According to one study, it is expressed in 100% of 11.3 months. These results are similar to those ob-
of intrahepatic cholangiocarcinomas, 52.6% of extrahepatic served with traditional chemotherapy. Other antiangiogenic
cholangiocarcinomas, and 38.5% of gallbladder cancers.52 agents, including sorafenib and sunitinib, have been eval-
Erlotinib, a selective and reversible inhibitor of EGFR, has been uated in advanced biliary tract cancers, with response rates
studied in the management of advanced biliary tract cancer in less than 10% when given as either as monotherapy or as
several phase II studies. In a small phase II study evaluating combination therapy.59-61
erlotinib for 42 patients, overall survival was 7.5 months and
median progression-free survival was 2.6 months.52 However, PIK3/mTOR. The PI3K/mTOR signaling cascade is crucial for
definitive conclusions between HER1/EGFR status and re- cell growth and survival. It is a key component of many
sponse rates could not be assessed due to the small sample cancers. A study documented the rate of activating muta-
size. A large phase III study evaluating the efficacy of gem- tions in PI3K to be approximately 12.5%, exclusively in
citabine and oxaliplatin with the addition of erlotinib was gallbladder cancer. BKM120, an oral PIK3CA inhibitor, was
conducted among patients with newly diagnosed metastatic recently under clinical investigation, but the study was
biliary tract cancer.53 A total of 268 patients were randomly closed due to poor accrual (NCT01501604). The mTOR
selected to receive either chemotherapy alone or in combi- pathway has been targeted in several malignancies but has
nation with erlotinib. The median progression-free survival shown disappointing results in small phase II studies. In an
was 5.8 months in the combination arm compared with Italian phase II study of everolimus, the median progression-
4.2 months in the chemotherapy-alone arm (p = .087). Overall free survival was 3.2 months and overall survival was
survival was the same at 9.5 months in both arms (p = .611). 7.7 months.62 Everolimus was used for patients with first-
Erlotinib with chemotherapy failed to demonstrate superi- line metastatic disease, with a median progression-free
ority in this study. survival of 6.0 months.63 It is unclear if larger studies will
Monoclonal antibodies targeting EGFR used in combination have more promising results.
with chemotherapy have shown some success. Gruenberger
et al performed a phase II study using cetuximab in combi- FGFR. The fibroblast growth factor (FGF) pathway and
nation with gemcitabine and oxaliplatin (GEMOX).54 The fibroblast growth factor receptor (FGFR) genes are in-
median progression-free survival was 8.3 months and overall volved in multiple biologic processes, ranging from cell
survival was 12.7 months. Interestingly, the response rate transformation, angiogenesis, and tissue repair to embry-
was a remarkable 63%, with 30% of patients undergoing onic development. This pathway has been targeted
curative surgery after treatment. more recently in several malignancies including chol-
angiocarcinoma. Genomic profiling has demonstrated
HER2. HER2 (ERBB2) mediates its signaling via the MAPK that FGFR gene alterations are exclusively seen in intra-
and PI3K pathways. Expression of HER2 is seen in approxi- hepatic cholangiocarcinoma, and its natural history is more
mately 10% of gallbladder cancers and 26% of extrahepatic indolent.49 FGFR mutations and fusions have been observed
cholangiocarcinomas.52 The California Consortium group in 13.6% of intrahepatic cholangiocarcinomas and in 45%
performed a phase II study of lapatinib, a dual HER2 and of intrahepatic cholangiocarcinomas. The documented fu-
EGFR inhibitor, for patients with advanced biliary tract sions to date include: FRGR2-TACC3, FGFR2-BICC1, FGFR2-
cancer.55 Among the 17 patients enrolled, no response was AHCYL1, FGFR2-PPHLN1, and FGFR2-MGEA5.49,64,65 The
seen in any of the patients. Similarly disappointing results fusion protein results in morphologic changes at the
have been reported in other phase I studies of lapatinib. cellular level causing abnormal cellular proliferation.
There are a number of clinical trials currently under-
VEGF. The VEGF pathway mediates its tumorigenic potential way using FGFR antibodies (NCT02508467, NCT02150967,
not just by angiogenesis or vascular permeability, but also by NCT01752920) and specific FGFR2 antibodies (NCT02368951,
cell signaling for tumor initiation.50,56 VEGF expression has NCT02265341, NCT02450136).
been correlated with increasing grade, metastatic potential,
and overall prognosis.57 IDH1. Somatic mutations in IDH1 and IDH2 have been well
Bevacizumab, a recombinant humanized VEGF antibody, documented in several different studies and particularly in
has been studied in phase II trials for its efficacy in treatment intrahepatic cholangiocarcinoma. These mutations cause a
of biliary tract cancers. The combination of gemcitabine, single amino acid change at a conserved arginine residue
oxaliplatin, and bevacizumab (GEMOX-B) was evaluated within the isocitrate binding site of IDH1 (R132) or IDH2
among 35 patients with advanced biliary tract cancer.58 (R172, R140), resulting in decreased enzymatic activity for
FDG-PET scans demonstrated a significant decrease in oxidative decarboxylation of isocitrate to a-ketoglutarate.66
the standardized uptake value (SUV) of the tumors after It has been shown that inhibition of IDH gain of func-
two cycles of treatment (p , .0001). The median tion mutations results in reversal of epigenetic methyla-
progression-free survival was 7.0 months. More recently, a tion and cancer cell differentiation.67-69 Mutation rates for
small first-line study evaluating the combination of IDH1/2 have been observed in 18% to 24% of intrahepatic

e200 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


BILIARY TRACT CANCER

cholangiocarcinomas. Further, the rates of IDH1 (15%23%) the absence of ERK staining was associated with no re-
are far higher than IDH2 (3%4%) across all reported sponse. These results clearly point toward the MEK pathway
intrahepatic cholangiocarcinoma sequencing.47-49,51 Inter- as a potentially promising targetable agent. Clinical studies
estingly, 2-hydroxyglutarate, the by-product of enzymatic using other MEK inhibitors are underway.
activity of IDH1 and IDH2, can be detected in the serum and,
hence, can potentially be used as a biomarker.67,69,70 Studies
evaluating IDH1 inhibitors in cholangiocarcinoma are on- CONCLUSION
going (NCT02481154, NCT02073994, NCT02496741) With dismal overall prognosis for most cases of biliary tract
cancer, new novel therapeutic targets are clearly needed.
MEK pathway. Activated RAS starts a phosphorylation Biliary tract cancer is often associated with hypo- and
cascade, which involves RAF kinase, MEK1/2, and ERK1/2, hypermethylation of promoters. Although several targets
and, ultimately, these affect cellular function. Inhibition of (such as selumetinib and sunitinib) have focused on the
the MEK/ERK signaling pathway lends itself as a therapeutic EGFR/HER2/VEGF pathways, the specificity of drugs tar-
target for biliary tract cancer and other solid malignancies. A geting these agents is likely to be low because the majority of
phase II study of selumetinib in advanced biliary tract cancer biliary tract cancer mediators seem to affect epigenetics and
had a median progression-free survival of 3.7 months and transcription. Understanding the gene expression profile
overall survival of 9.8 months, with a response rate of 12%.71 and mutational burden in biliary tract cancer allows us to
Interestingly, 68% of patients had stable disease; at least better discern the pathogenesis and identify promising new
12% had stable disease for longer than 1 year. In addition, developmental therapeutic targets.

References
1. Patel T. Increasing incidence and mortality of primary intrahepatic chol- 13. Al Ansari N, Kim BS, Srirattanapong S, et al. Mass-forming chol-
angiocarcinoma in the United States. Hepatology. 2001;33:1353-1357. angiocarcinoma and adenocarcinoma of unknown primary: can
2. Sripa B, Kaewkes S, Sithithaworn P, et al. Liver fluke induces chol- they be distinguished on liver MRI? Abdom Imaging. 2014;39:
angiocarcinoma. PLoS Med. 2007;4:e201. 1228-1240.
3. Zatonski WA, Lowenfels AB, Boyle P, et al. Epidemiologic aspects of 14. Sithithaworn P, Yongvanit P, Duenngai K, et al. Roles of liver fluke
gallbladder cancer: a case-control study of the SEARCH Program of the infection as risk factor for cholangiocarcinoma. J Hepatobiliary Pancreat
International Agency for Research on Cancer. J Natl Cancer Inst. 1997; Sci. 2014;21:301-308.
89:1132-1138. 15. Carpizo DR, DAngelica M. Management and extent of resection for
4. West J, Wood H, Logan RF, et al. Trends in the incidence of primary liver intrahepatic cholangiocarcinoma. Surg Oncol Clin N Am. 2009;18:
and biliary tract cancers in England and Wales 1971-2001. Br J Cancer. 289-305, viii-ix.
2006;94:1751-1758. 16. Jarnagin WR, Shoup M. Surgical management of cholangiocarcinoma.
5. Taylor-Robinson SD, Toledano MB, Arora S, et al. Increase in mortality Semin Liver Dis. 2004;24:189-199.
rates from intrahepatic cholangiocarcinoma in England and Wales 17. Fong Y, Blumgart LH, Lin E, et al. Outcome of treatment for distal bile
1968-1998. Gut. 2001;48:816-820. duct cancer. Br J Surg. 1996;83:1712-1715.
6. von Hahn T, Ciesek S, Wegener G, et al. Epidemiological trends in in- 18. Jarnagin WR, Ruo L, Little SA, et al. Patterns of initial disease recurrence
cidence and mortality of hepatobiliary cancers in Germany. Scand J after resection of gallbladder carcinoma and hilar cholangiocarcinoma:
Gastroenterol. 2011;46:1092-1098. implications for adjuvant therapeutic strategies. Cancer. 2003;98:
7. Alvaro D, Crocetti E, Ferretti S, et al; AISF Cholangiocarcinoma com- 1689-1700.
mittee. Descriptive epidemiology of cholangiocarcinoma in Italy. Dig 19. Ben-David MA, Griffith KA, Abu-Isa E, et al. External-beam radiotherapy
Liver Dis. 2010;42:490-495. for localized extrahepatic cholangiocarcinoma. Int J Radiat Oncol Biol
8. Witjes CD, Karim-Kos HE, Visser O, et al. Intrahepatic chol- Phys. 2006;66:772-779.
angiocarcinoma in a low endemic area: rising incidence and improved 20. Cheng Q, Luo X, Zhang B, et al. Predictive factors for prognosis of hilar
survival. HPB (Oxford). 2012;14:777-781. cholangiocarcinoma: postresection radiotherapy improves survival. Eur
9. Lee T-Y, Lin J-T, Kuo KN, et al. A nationwide population-based study J Surg Oncol. 2007;33:202-207.
shows increasing incidence of cholangiocarcinoma. Hepatol Int. 2013;7: 21. Kim TH, Han SS, Park SJ, et al. Role of adjuvant chemoradiotherapy for
226-232. resected extrahepatic biliary tract cancer. Int J Radiat Oncol Biol Phys.
10. Utada M, Ohno Y, Tamaki T, et al. Long-term trends in incidence and 2011;81:e853-e859.
mortality of intrahepatic and extrahepatic bile duct cancer in Japan. 22. Todoroki T, Ohara K, Kawamoto T, et al. Benefits of adjuvant radio-
J Epidemiol. 2014;24:193-199. therapy after radical resection of locally advanced main hepatic duct
11. Welzel TM, McGlynn KA, Hsing AW, et al. Impact of classification of hilar carcinoma. Int J Radiat Oncol Biol Phys. 2000;46:581-587.
cholangiocarcinomas (Klatskin tumors) on the incidence of intra- and 23. Schoenthaler R, Phillips TL, Castro J, et al. Carcinoma of the extrahepatic
extrahepatic cholangiocarcinoma in the United States. J Natl Cancer bile ducts. The University of California at San Francisco experience. Ann
Inst. 2006;98:873-875. Surg. 1994;219:267-274.
12. Shaib YH, Davila JA, McGlynn K, et al. Rising incidence of intrahepatic 24. Buskirk SJ, Gunderson LL, Schild SE, et al. Analysis of failure after cu-
cholangiocarcinoma in the United States: a true increase? J Hepatol. rative irradiation of extrahepatic bile duct carcinoma. Ann Surg. 1992;
2004;40:472-477. 215:125-131.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e201


BRIDGEWATER ET AL

25. Cameron JL, Pitt HA, Zinner MJ, et al. Management of proximal chol- 43. Wolkov HB, Graves GM, Won M, et al. Intraoperative radiation therapy
angiocarcinomas by surgical resection and radiotherapy. Am J Surg. of extrahepatic biliary carcinoma: a report of RTOG-8506. Am J Clin
1990;159:91-97. Oncol. 1992;15:323-327.
26. Gonzalez Gonzalez D, Gerard JP, Maners AW, et al. Results of radiation 44. Rea DJ, Heimbach JK, Rosen CB, et al. Liver transplantation with
therapy in carcinoma of the proximal bile duct (Klatskin tumor). Semin neoadjuvant chemoradiation is more effective than resection for hilar
Liver Dis. 1990;10:131-141. cholangiocarcinoma. Ann Surg. 2005;242:451-461.
27. Schoenthaler R, Castro JR, Halberg FE, et al. Definitive postoperative 45. Marino D, Leone F, Cavalloni G, et al. Biliary tract carcinomas: from
irradiation of bile duct carcinoma with charged particles and/or pho- chemotherapy to targeted therapy. Crit Rev Oncol Hematol. 2013;85:
tons. Int J Radiat Oncol Biol Phys. 1993;27:75-82. 136-148.
28. Bonet Beltran M, Allal AS, Gich I, et al. Is adjuvant radiotherapy needed 46. Zhu AX, Hezel AF. Development of molecularly targeted therapies in
after curative resection of extrahepatic biliary tract cancers? A sys- biliary tract cancers: reassessing the challenges and opportunities.
tematic review with a meta-analysis of observational studies. Cancer Hepatology. 2011;53:695-704.
Treat Rev. 2012;38:111-119. 47. Ross JS, Wang K, Catenacci DVT, et al. Comprehensive genomic profiling
29. Vern-Gross TZ, Shivnani AT, Chen K, et al. Survival outcomes in resected of biliary tract cancers to reveal tumor-specific differences and genomic
extrahepatic cholangiocarcinoma: effect of adjuvant radiotherapy in a alterations. J Clin Oncol. 2015;33(suppl; abstr 231).
surveillance, epidemiology, and end results analysis. Int J Radiat Oncol 48. Simbolo M, Fassan M, Ruzzenente A, et al. Multigene mutational
Biol Phys. 2011;81:189-198. profiling of cholangiocarcinomas identifies actionable molecular sub-
30. Ben-Josef E, Guthrie KA, El-Khoueiry AB, et al. SWOG S0809: a phase II groups. Oncotarget. 2014;5:2839-2852.
Intergroup trial of adjuvant capecitabine and gemcitabine followed by 49. Churi CR, Shroff R, Wang Y, et al. Mutation profiling in chol-
radiotherapy and concurrent capecitabine in extrahepatic chol- angiocarcinoma: prognostic and therapeutic implications. PLoS One.
angiocarcinoma and gallbladder carcinoma. J Clin Oncol. 2015;33: 2014;9:e115383.
2617-2622. 50. Merla A, Liu KG, Rajdev L. Targeted therapy in biliary tract cancers. Curr
31. Shinohara ET, Mitra N, Guo M, et al. Radiation therapy is associated with Treat Options Oncol. 2015;16:48.
improved survival in the adjuvant and definitive treatment of intra- 51. Jiao Y, Pawlik TM, Anders RA, et al. Exome sequencing identifies fre-
hepatic cholangiocarcinoma. Int J Radiat Oncol Biol Phys. 2008;72: quent inactivating mutations in BAP1, ARID1A and PBRM1 in intra-
1495-1501. hepatic cholangiocarcinomas. Nat Genet. 2013;45:1470-1473.
32. Ghafoori AP, Nelson JW, Willett CG, et al. Radiotherapy in the treatment 52. Pignochino Y, Sarotto I, Peraldo-Neia C, et al. Targeting EGFR/HER2
of patients with unresectable extrahepatic cholangiocarcinoma. Int J pathways enhances the antiproliferative effect of gemcitabine in biliary
Radiat Oncol Biol Phys. 2011;81:654-659. tract and gallbladder carcinomas. BMC Cancer. 2010;10:631.
33. Tao R, Krishnan S, Bhosale PR, et al. Ablative radiotherapy doses lead 53. Lee J, Park SH, Chang HM, et al. Gemcitabine and oxaliplatin with or
to a substantial prolongation of survival in patients with inoperable without erlotinib in advanced biliary-tract cancer: a multicentre, open-
intrahepatic cholangiocarcinoma: a retrospective dose response label, randomised, phase 3 study. Lancet Oncol. 2012;13:181-188.
analysis. J Clin Oncol. 2016;34:219-226. 54. Gruenberger B, Schueller J, Heubrandtner U, et al. Cetuximab, gem-
34. Jung DH, Kim MS, Cho CK, et al. Outcomes of stereotactic body ra- citabine, and oxaliplatin in patients with unresectable advanced or
diotherapy for unresectable primary or recurrent cholangiocarcinoma. metastatic biliary tract cancer: a phase 2 study. Lancet Oncol. 2010;11:
Radiat Oncol J. 2014;32:163-169. 1142-1148.
35. Mahadevan A, Dagoglu N, Mancias J, et al. Stereotactic body radio- 55. Ramanathan RK, Belani CP, Singh DA, et al. A phase II study of lapatinib
therapy (SBRT) for intrahepatic and hilar cholangiocarcinoma. J Cancer. in patients with advanced biliary tree and hepatocellular cancer. Cancer
2015;6:1099-1104. Chemother Pharmacol. 2009;64:777-783.
36. Barney BM, Olivier KR, Miller RC, et al. Clinical outcomes and toxicity 56. Goel HL, Mercurio AM. VEGF targets the tumour cell. Nat Rev Cancer.
using stereotactic body radiotherapy (SBRT) for advanced chol- 2013;13:871-882.
angiocarcinoma. Radiat Oncol. 2012;7:67. 57. Quan ZW, Wu K, Wang J, et al. Association of p53, p16, and vascular
37. Ibarra RA, Rojas D, Snyder L, et al. Multicenter results of stereotactic endothelial growth factor protein expressions with the prognosis and
body radiotherapy (SBRT) for non-resectable primary liver tumors. Acta metastasis of gallbladder cancer. J Am Coll Surg. 2001;193:380-383.
Oncol. 2012;51:575-583. 58. Zhu AX, Meyerhardt JA, Blaszkowsky LS, et al. Efficacy and safety of
38. Polistina FA, Guglielmi R, Baiocchi C, et al. Chemoradiation treat- gemcitabine, oxaliplatin, and bevacizumab in advanced biliary-tract
ment with gemcitabine plus stereotactic body radiotherapy for cancers and correlation of changes in 18-fluorodeoxyglucose PET
unresectable, non-metastatic, locally advanced hilar cholangiocarcinoma. with clinical outcome: a phase 2 study. Lancet Oncol. 2010;11:
Results of a five year experience. Radiother Oncol. 2011;99:120-123. 48-54.
39. Goodman KA, Wiegner EA, Maturen KE, et al. Dose-escalation study of 59. Bengala C, Bertolini F, Malavasi N, et al. Sorafenib in patients with
single-fraction stereotactic body radiotherapy for liver malignancies. Int advanced biliary tract carcinoma: a phase II trial. Br J Cancer. 2010;102:
J Radiat Oncol Biol Phys. 2010;78:486-493. 68-72.
40. Tse RV, Hawkins M, Lockwood G, et al. Phase I study of individu- 60. Moehler M, Kanzler S, Woerns S, et al. A randomized, double-blind,
alized stereotactic body radiotherapy for hepatocellular carci- multicenter phase II AIO trial with gemcitabine plus sorafenib versus
noma and intrahepatic cholangiocarcinoma. J Clin Oncol. 2008;26: gemcitabine plus placebo in patients with chemotherapy-naive ad-
657-664. vanced or metastatic biliary tract cancer: first safety and efficacy data. J
41. Hong TS, Wo JY, Yeap BY, et al. Multi-institutional phase II study of Clin Oncol. 2011;29 (suppl; abstr 4077).
high-dose hypofractionated proton beam therapy in patients with 61. Yi JH, Thongprasert S, Lee J, et al. A phase II study of sunitinib as a
localized, unresectable hepatocellular carcinoma and intrahepatic second-line treatment in advanced biliary tract carcinoma: a multi-
cholangiocarcinoma. J Clin Oncol. Epub 2015 Dec 14. centre, multinational study. Eur J Cancer. 2012;48:196-201.
42. Jarnagin WR. Cholangiocarcinoma of the extrahepatic bile ducts. Semin 62. Buzzoni R, Pusceddu S, Bajetta E, et al. Activity and safety of RAD001
Surg Oncol. 2000;19:156-176. (everolimus) in patients affected by biliary tract cancer progressing

e202 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


BILIARY TRACT CANCER

after prior chemotherapy: a phase II ITMO study. Ann Oncol. 2014;25: through broad-based tumor genotyping. Oncologist. 2012;17:
1597-1603. 72-79.
63. Yeung YH, Chionh FJM, Price TJ, et al. Phase II study of everolimus 67. Razumilava N, Gores GJ. Cholangiocarcinoma. Lancet. 2014;383:2168-2179.
monotherapy as first-line treatment in advanced biliary tract cancer: 68. Rohle D, Popovici-Muller J, Palaskas N, et al. An inhibitor of mutant IDH1
RADichol. J Clin Oncol. 2014;32:5s (suppl; abstr 4101). delays growth and promotes differentiation of glioma cells. Science.
64. Sia D, Losic B, Moeini A, et al. Massive parallel sequencing uncovers 2013;340:626-630.
actionable FGFR2-PPHLN1 fusion and ARAF mutations in intrahepatic 69. Keum YS, Choi BY. Isocitrate dehydrogenase mutations: new oppor-
cholangiocarcinoma. Nat Commun. 2015;6:6087. tunities for translational research. BMB Rep. 2015;48:266-270.
65. Arai Y, Totoki Y, Hosoda F, et al. Fibroblast growth factor receptor 2 70. Reitman ZJ, Parsons DW, Yan H. IDH1 and IDH2: not your typical on-
tyrosine kinase fusions define a unique molecular subtype of chol- cogenes. Cancer Cell. 2010;17:215-216.
angiocarcinoma. Hepatology. 2014;59:1427-1434. 71. Bekaii-Saab T, Phelps MA, Li X, et al. Multi-institutional phase II study of
66. Borger DR, Tanabe KK, Fan KC, et al. Frequent mutation of isocitrate selumetinib in patients with metastatic biliary cancers. J Clin Oncol.
dehydrogenase (IDH)1 and IDH2 in cholangiocarcinoma identified 2011;29:2357-2363.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e203


GASTROINTESTINAL (NONCOLORECTAL) CANCER

Designing Clinical Trials to


Achieve Breakthrough Results in
Upper Gastrointestinal
Malignancies

CHAIR
Lynn M. Matrisian, PhD, MBA
Pancreatic Cancer Action Network
Manhattan Beach, CA

SPEAKERS
Jordan D. Berlin, MD
Vanderbilt-Ingram Cancer Center
Nashville, TN

Markus Frederic Renschler, MD


Celgene Corporation
Summit, NJ
LESSONS FROM PANCREATIC CANCER CLINICAL TRIALS

The Past, Present, and Future of Pancreatic Cancer Clinical


Trials
Lynn M. Matrisian, PhD, MBA, and Jordan D. Berlin, MD

OVERVIEW

Upper gastrointestinal malignancies comprise half of the deadliest cancers as defined by those with a 5-year survival rate less
than 50%. Using pancreatic adenocarcinoma (PAC) as an example, we retrospectively evaluated the success of phase III
clinical trials, examined the current landscape of clinical trials, and identified emerging areas that foretell the future for this
disease. Pancreatic and liver cancers are on the rise and will be the second and third leading causes of cancer deaths in 2030.
A total of 35 different agents or combinations have been tested in randomized phase III clinical trials for patients with
advanced PAC over the past 25 years, but only 11% have been incorporated into clinical practice. There has been a 37%
increase in the number of PAC trials open in the United States between 2011 to 2012 and 2014 to 2015. Enrollment has also
increased slightly, from 3.85% of the newly diagnosed cases in 2011 to 4.15% in 2014. However, the demand for patients far
exceeds the number of patients available for these trials. On the horizon is the realization that stratification of patients with
PAC using biomarkers that predict a high probability of a response could reallocate patients to faster, smaller trials with a
greater chance of a survival benefit. The current landscape of PAC clinical trials and the launch of the Pancreatic Cancer
Action Networks Know Your Tumor initiative indicate this shift is starting to occur, with particular emphasis on targeted
therapies, immunotherapies, and agents that disrupt the stroma.

H istorically, the big four cancers in the United States have


been considered lung, colorectal, breast, and prostate
because of their high incidence and mortality rates. How-
rise. It is the only major cancer with a 5-year relative survival
in the single digits. Despite pancreatic cancer being the 11th
most commonly diagnosed cancer in men and 9th in women,
ever, trends are changing dramatically, and cancers that deaths from it surpassed breast cancer and moved to the
have not been top of mind are becoming increasing third leading cause of cancer-related death in the United
threats to public health in the United States. Eight major States this year. An estimated 53,070 Americans will be
cancers are considered the deadliest, characterized by diagnosed with pancreatic cancer in 2016 and 41,780 are
5-year survival rates below 50%.1,2 These eight cancers will expected to lose their lives to the disease.3
account for half of the cancer deaths in the United States this The threat of pancreatic cancer is only expected to in-
year, and four of the eight cancers are diseases of the upper crease in the coming years. A 2014 study predicted that
gastrointestinal tract (i.e., pancreas, liver, esophagus, and pancreatic cancer deaths would surpass breast cancer and
stomach).3 This article focuses on PAC, representing 95% of further projects that pancreatic cancer deaths will exceed
pancreatic cancer diagnoses and defines the problem from those caused by colorectal cancer around 2020, positioning
the perspective of a patient with PAC through the lens of an pancreatic cancer as the second leading cause of cancer-
advocacy organization and a medical oncologist working in related deaths in the United States before 2030 (Fig. 1).4
this field for many years. We outline some of the historical Moreover, deaths from liver cancer have surpassed prostate
challenges to successful clinical trial design and execution, cancer this year, placing it among the top-five cancer killers
describe efforts currently underway, and highlight future in the United States. Projections show that liver cancer
directions that may bring urgently needed progress to this deaths will continue to rise quickly, leading to it becoming
deadly disease. These insights provide lessons learned the third leading cause of cancer-related death in the United
relevant to deadly upper gastrointestinal malignancies in States by 2030.4
general. The clinical advances required to slow the alarming tra-
Although overall incidence and death rates for cancer in jectory of pancreatic cancer deaths will be made as a result
the United States are declining, pancreatic cancer is on the of clinical research. National Comprehensive Cancer Network

From the Pancreatic Cancer Action Network, Manhattan Beach, CA; Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Lynn M. Matrisian, PhD, MBA, The Pancreatic Cancer Action Network, 1500 Rosecrans Ave., Suite 200, Manhattan Beach, CA 90266; email: lmatrisian@pancan.org.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e205


MATRISIAN AND BERLIN

FIGURE 1. Projected Number of Cancer Deaths, 2010 to 2030

guidelines indicate that a clinical trial is the preferred course following sections evaluate the past, present, and future of
of treatment at all stages of PAC.5 The Pancreatic Cancer pancreatic cancer clinical trials.
Action Network encourages all patients who call its Patient
Central call center to consider clinical trials when making
treatment decisions. But enrollment rates remain low, and THE PAST
clinical trials do not always match patients needs, leading to Progress in advanced PAC can be measured by treatment
slow progress and continued poor outcomes for patients. The options considered clinically meaningful enough to be
available as standard of care to patients: gemcitabine (ap-
proved by the U.S. Food and Drug Administration [FDA]
in 1996), FOLFIRINOX (positive phase III data published in
KEY POINTS 2011),6 gemcitabine with nab-paclitaxel (FDA approved in
2013) for first-line treatment, and the recent FDA approval
Deaths from pancreatic cancer are rising, making the of irinotecan liposome injection (2015) for patients pre-
disease the third leading cause of cancer deaths in the viously treated with gemcitabine. To achieve this degree of
United States in 2016. success, a total of 35 different agents or combinations were
Randomized phase III trials over the past 25 years have tested in 39 phase III clinical trials in advanced-stage PAC
resulted in a new standard of care for PAC 11% of the between 1997 and 2015 (Fig. 2, unpublished data, LR, PhD
time. 2016). Twenty-seven of these were tested in patients with
The number of clinical trials for PAC open in the United treatment-naive disease, and eight of the 35 agents or
States has increased by 37% over the past 5 years.
combinations were tested in patients with previously
Although there has been a slight increase in the number
of patients who enroll in PAC trials from 3.85% in 2011 to
treated disease. Although the FDA approved the combina-
4.15% in 2014, the demand for patients to enroll in trials tion of gemcitabine and erlotinib in 2005 based on a hazard
far exceed the number of patients available, and it will ratio (HR) of 0.82 for overall survival, the modest improve-
take, on average, more than 6 years to accrue those ment in median survival of 6.2 months compared with
trials open in 2014. 5.9 months with gemcitabine alone7 is generally viewed as
The landscape of current PAC clinical trials is changing, being insufficient to warrant the additional toxicity and
with an increase in the number of neoadjuvant trials, expense of erlotinib. Thus, the overall success rate of PAC
particularly focusing on radiation therapy and an phase III trials is a dismal 11%, despite a prior phase II trial in
increase in the number of trials for previously treated the majority of cases.
patients, such as for targeted therapies. In the purest sense of trial design, the phase II trial is
The Pancreatic Cancer Action Networks Know Your
written with the ultimate goal that (1) if the primary end-
Tumor initiative has offered molecular profiling
opportunities to more than 500 individuals from 38
point is positive, it will lead to a phase III trial, and (2) if
states; 40% of them demonstrate an actionable negative, the drug or regimen will not go further in that
alteration that suggests a therapeutic option that setting. However, in 85% of the cases where we could
would not otherwise be identified for these identify a prior phase II trial, a phase III trial was pursued
patients. irrespective of the phase II not meeting its primary endpoint
(Rahib et al, unpublished data, 2016). The decision to

e206 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


LESSONS FROM PANCREATIC CANCER CLINICAL TRIALS

FIGURE 2. Agents/Combinations in Phase III Clinical Trials for Pancreatic Adenocarcinoma, 1992 to 2015

Abbreviation: FDA, U.S. Food and Drug Administration.


Adapted from Rahib et al (unpublished data, 2016).

proceed was based on encouraging secondary endpoints, Networks Patient Central were ineligible for 90% of the
subset analyses results, or a second phase II trial conducted clinical trials that were open because they had already
elsewhere that showed promise. Factors that are not started treatment.9 The increase in clinical trials that include
mentioned but contribute to corporate go/no-go decisions patients with previously treated disease opens up more
involve economic and logistic considerations by the trial options and encourages a multistep treatment plan that
sponsors that are influenced by whether the trial is the first provides several opportunities for interventions that may
indication for a new drug and by the size of the company.8 If improve survival. In this respect, the study of patients with
the past is prelude to the future, there is considerable more refractory disease in later line is encouraging, but we
concern that the trend of negative phase III trials will acknowledge that due to the severity of this illness risks
continue and thwart the chance for any major advancement missing potentially active agents if had they been studied in
in PAC survival. an earlier line of therapy.
For patients with early-stage resectable disease, 13% of
trials were active for neoadjuvant treatment and 10% for
THE PRESENT adjuvant treatment from 2011 to 2012. From 2014 to
The Pancreatic Cancer Action Network maintains a data- 2015, there was an approximately two fold increase in
base of pancreatic cancer clinical trials in the United States trials for neoadjuvant treatment (24% of active trials)
that is constantly updated to be able to give those who and a slight decrease to 7% of the trials requiring patients
contact the Patient Central call center accurate, up-to-date for adjuvant studies (Fig. 4). The increase in neoadjuvant
information on clinical trials that match the patients stage trials may be attributed to the recognition that even
of disease and that are open within a distance they are patients with resectable PAC have dismal outcomes be-
willing to travel. We analyzed the portfolio of pancreatic cause of the rapid development of metastatic disease
cancer clinical trials in 2011 and 2012 by type, phase, a clinical observation that was supported with preclinical
disease stage, and treatment approaches.9 We extended evidence in 2012.10 In addition, the eligibility criteria for
this study to include the years 2013 to 2015 and report the neoadjuvant trials have shown an expansion to patients
results in this article. We are encouraged to find that the with borderline resectable disease with the hope that
number of PAC-specific clinical trials open in the United neoadjuvant treatment shrinks the tumor and permits a
States has steadily increased between 2011 and 2015 negative margin resection.
(Fig. 3A). This trend is observed for all phases of clinical The landscape of PAC clinical trials was also examined by
trials, with the most dramatic growth observed in phase I treatment type (Fig. 5A). Trials that focused on therapies
trials (Fig. 3B). targeted to specific molecular pathways increased markedly
During 2011 to 2012, the majority of PAC clinical trials between 2011 and 2015, comprising 29% of the PAC
were designed for patients with treatment-naive, metastatic landscape from 2011 to 2012 and 40% from 2014 to 2015.
disease (51% of total PAC trials for these 2 years; Fig. 4). The majority of these targeted trials are for patients with
Since then, there has been a dramatic shift toward a greater refractory disease and those who received previous treat-
number of trials to accommodate patients who have already ments (Fig. 5B) and include targets for PARP, JAK, WEE1,
been treated. In fact, the majority of the trials during 2014 to mTOR, and several others.
2015 focused on patients with refractory and previously In addition, trials focused on radiation therapy increased
treated disease (38%), whereas the percentage of first-line from 13% of total PAC trials during 2011 to 2012 to 18%
metastatic trials during 2014 to 2015 (22%) decreased by during 2014 to 2015 (Fig. 5A). The majority of these trials
more than two fold compared with 2011 to 2012. This shift were in the neoadjuvant setting, which comprised 58% of
from metastatic/treatment-naive trials to trials for patients the total radiation trials from 2014 to 2015 (Fig. 5B). Many of
with previously treated disease is viewed very positively these neoadjuvant trials (. 50%) used intensity-modulated
from a patient perspective. In 2011, we observed that two- radiotherapy or stereotactic body radiotherapy in combi-
thirds of the patients that call the Pancreatic Cancer Action nation with chemotherapy.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e207


MATRISIAN AND BERLIN

FIGURE 3. Pancreatic Adenocarcinoma Trials Open in the United States from 2011 to 2015

(A) Trials open (2011-2015) (B) Trials open by phase.

Trials focused on an immunotherapeutic approach were As a result of regularly updating clinical trial information
relatively steady, with an average of 14 trials per year stored in the organizations comprehensive database, the
over the past 5 years (Fig. 5A). There has been a change, Pancreatic Cancer Action Network has established strong
however, in the stage of disease of treatment. From 2011 to relationships with trial sponsors. These relationships
2012, the majority of trials were for patients with meta- allowed the accumulation of data to estimate the number of
static disease (52%), whereas from 2014 to 2015 the patients with pancreatic cancer in the United States who
majority of immunotherapy trials were for patients with were accrued to clinical trials in 2011,9 and this analysis was
refractory disease and those who received previous repeated for 2014 clinical trials. Considering only those
treatments (62%; Fig. 5B). Approximately 60% of these patients with a PAC diagnosis who enrolled in trials (in
trials involved a vaccine treatment, 20% included T-cell contrast to the previous analysis, which considered all
modified therapy, and 7% included checkpoint blockade pancreatic cancer diagnoses), 1,612 patients with PAC en-
therapy. rolled in clinical trials in 2011 (93% of trials reporting), and

e208 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


LESSONS FROM PANCREATIC CANCER CLINICAL TRIALS

FIGURE 4. Pancreatic Adenocarcinoma Clinical Trials Open in the United States from 2011 to 2015 by Disease
Stage

1,835 patients with PAC enrolled in clinical trials in 2014 received prior chemotherapy. Irinotecan liposome injection
(94% of trials reporting). As a percentage of the estimated was shown to improve overall survival by nearly 2 months
number of new cases of PAC in that year (95% of the es- when administered in combination with fluorouracil and
timated total pancreatic cancer incidence),3 we calculate leucovorin. This approval was a historic achievement, as it
that 3.85% of patients with PAC went on a clinical trial in was the first drug combination approved with an indication
2011, and 4.16% went on a trial in 2014. specific to this patient population.11 As described above, in
Although the slight increase is encouraging, the time it will the past few years, the amount of clinical trials focused on
take to enroll patients with PAC in clinical trials based on this patients with previously treated disease has increased. As
accrual rate is unacceptably slow. For example, the number of oncologists and patients are often eager to begin chemo-
newly diagnosed patients with resectable disease who could therapy treatments as soon as possible upon diagnosis,
be eligible for clinical trials for neoadjuvant or adjuvant providing expanded options for patients with treatment-
treatment approaches in 2014 is estimated to be 4,178 refractory PAC fills a critical gap and has the potential to
(Table 1). However, the 47 trials that were open in these positively affect patient outcomes.
categories required 3,437 patients, indicating that 82% of the Clinical research provides the evidence for treatments that
eligible patients in the United States would need to go on one prolong survival. The randomized phase III trial is the gold
of these clinical trials to complete enrollment during that year. standard for FDA approval of new treatments and accep-
The actual percentage of enrollment for these trials for 2014 tance as standard of care. However, progress in pancreatic
was 10%, indicating that it would take 9.7 years to complete cancer using this paradigm has been agonizingly slow. A
enrollment of the neoadjuvant/adjuvant trials open in 2014. transformative shift in approach is emerging as a result of
On the other hand, it is encouraging that the time necessary to the incorporation of molecular profiling as a basis of iden-
complete enrollment of trials for patients with locally ad- tifying subsets of patients with cancer with a high probability
vanced and previously treated metastatic disease has de- of responding to a specific treatment. For example, the care
creased, from 9.0 years to 3.8 years and 7.1 years to 6.0 years, of patients with breast cancer is routinely stratified by their
respectively (Table 1). Nevertheless, for a disease with a 71% estrogen receptor and HER2 status, and the genotyping of
1-year mortality, it is clear we must identify a new paradigm nonsmall cell lung cancer for ALK translocations as an in-
for clinical advancement to accelerate progress. dicator of specific chemotherapeutic treatments is be-
coming routine.
The ability to identify those patients whose disease has a
THE FUTURE high probability of responding strongly to a specific treat-
There has been a palpable shift away from the nihilistic ment has enormous implications for clinical trial design. For
attitude toward PAC that bodes well for the future for these example, a treatment with a true HR of 0.4 requires only
patients. In October 2015, the FDA approved irinotecan li- 70 patients if all the patients enrolled in the trial have the
posomal injection for patients with metastatic PAC who had drug target.12 However, if the trial occurs in an unselected

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e209


MATRISIAN AND BERLIN

FIGURE 5. Pancreatic Adenocarcinoma Clinical Trials Open in the United States from 2011 to 2015 by Treatment
Type
A
5
60 4

Phase 0 Phase I Phase II


50 21
22 Phase III Phase IV
Number of Active Trials

2
24
40

2 19
2
30 2
1 2 1
3 2
18
18
20 39 2
36 19
10 17
1 19
31 21 16 19
1
1 5 1
24 7 3
2 1 6 3 6 17 0
10 7 1 4
3 4 0 5
15 3 0 2 4 2 1
11 3 2 12 12
9 0
1 3 0 9 9 3 1 0
1 10 4
7 8 2 8 2 6 6 8 7
6 6 6 0
1 5 6 5 1 3 5 4 6
0
1 4 3 4 3 3 4 3
1 2 2 1 2 1 1 1
0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0 0
2011
2012
2013
2014
2015
2011
2012
2013
2014
2015
2011
2012
2013
2014
2015
2011
2012
2013
2014
2015
2011
2012
2013
2014
2015
2011
2012
2013
2014
2015
2011
2012
2013
2014
2015
2011
2012
2013
2014
2015
Immunotherapy Targeted Stroma Metabolism Radiaon Delivery Opmizaon Miscellaneous

B 70

60
Neoadjuvant
Adjuvant
50 Locally Advanced
31 35
Metastac; treatment nave
Number of Active Trials

27
40 7 Metastac; previously treated

5 3
30 0
4
4 7 1
25 2 2
1 5
2 6 5 3 8 4 6
20 19 24 21 2
21 7 6 1
5 6 3 2
4 6
1 0
3 3 1 2 5 1
1 4 4 6
10 9 19 2 5 1 9
8 8 6 8 5 1
0 0 4 3 4 11 4
7 15 16 1
0
1 0 5
2 2 10 0 1 1 5 5 2 12 6
1 1 3 0
1 1 1 8 9 2 0 3 3 3 7 7 6 2 0
2 3 9 0 3 2 6
3 3 3 2 2 0 5 1 0 2 7 7 7 2
2
1 1 2 1 1 1 2
4 4 0
1
0
2 2 2 0
3 0
1
2 2 2
1 0
1
2
0
1 1 1 1 1
1 2 1
1 2
2 2 0 2
0 0
0 0 0 0 0 0 0 0 0 0 1 1 1
0
2011
2012
2013
2014
2015
2011
2012
2013
2014
2015
2011
2012
2013
2014
2015
2011
2012
2013
2014
2015
2011
2012
2013
2014
2015
2011
2012
2013
2014
2015
2011
2012
2013
2014
2015
2011
2012
2013
2014
2015

Immunotherapy Targeted Stroma Metabolism Radiation Delivery Optimization Miscellaneous

(A) Trials open by treatment type and phase (B) Trials open by treatment type and disease stage

patient population, the number of patients needed is with pancreatic cancer, have resulted in relatively small
proportional to the prevalence of the target. Further, the observed HR and improvement in survival. Given that
observed HR decreases with proportion of patients. The pancreatic cancer is heterogeneous, it seems possible that
same treatment above requiring 70 selected patients, if a missing component is identification of patients who are
evaluated in an unselected population containing 20% of most likely to respond to specific treatments. Continuing
patients with the target, requires 1,750 patients, and the with the paradigm of treating all patients with the same
HR observed is only 0.84. A trial requires 29,620 patients if chemotherapeutic agents will continue to reap low rewards
the marker is present in only 5% of the unselected pop- and squander the precious resource of patients willing and
ulation. Past trials, in large numbers of unselected patients eligible to enroll in clinical trials.

e210 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


LESSONS FROM PANCREATIC CANCER CLINICAL TRIALS

TABLE 1. Anticipated Total and Actual Accrual in the United States in 2011 and 2014
Distribution Est. No. No. of Potential Enrollment Capacity Enrolled Years to
Stage at Diagnosis Year (%) Patients* Trials Enrollment (%)** (%) Completion
Localized ([Neo] 2011 9 3,963 29 2,874 73 15 6.9
Adjuvant)
2014 9 4,178 47 3,437 82 10 9.7
Regional (Locally 2011 28 12,328 16 756 6 13 9.0
Advanced)
2014 28 12,998 12 800 6 27 3.8
Distant (Metastatic) 2011 53 23,336 62 6,394 27 18 6.2
2014 53 24,603 38 3,535 14 15 6.6
Metastatic; Previously 2011 27 11,668 12 524 4 14 7.1
Treated
2014 27 12,301 59 4,364 35 17 6.0
The distribution by disease stage was determined using the Surveillance, Epidemiology, and End Results (SEER) 18 2005-2011 Registry, All Races, both Sexes by SEER Summary Stage
2000 for localized, locally advanced, and metastatic. The number of estimated patients diagnosed with pancreatic cancer for each year was multiplied by 95% to account for pancreatic
adenocarcinomas only. The distribution of previously treated metastatic disease was determined by multiplying the percentage of patients with metastatic disease by 50%, as it is
reported that about 50% of patients with pancreatic cancer are eligible for second-line therapy.36-38
*The number of new patient cases of pancreatic cancer was estimated to be 44,030 for 2011 and 46,420 for 2014.
**Enrollment capacity is total potential enrollment for open trials divided by the estimated number of available patients for 2011 and 2014.

Percentage enrolled is the number of patients enrolled divided by the potential enrollment for trials within that subgroup for which accrual numbers were available for 2011 and
2014.

Years to completion is the number of potential enrollment divided by the accrual numbers (for each subgroup) for which accrual numbers were available for 2011 and
2014.

Is molecular stratification feasible in pancreatic cancer? PEGPH20, nab-paclitaxel, and gemcitabine (PAG) or nab-
Recent genomics analyses have revealed high numbers of paclitaxel plus gemcitabine (AG) were presented at the
genetic changes that contribute to the initiation and pro- 2015 American Society of Clinical Oncology Annual Meet-
gression of PAC. Although we know that oncogenic KRAS ing. Among patients with high HA levels treated with PAG,
drives 95% of PAC cases, more recent studies have shown median progression-free survival was 9.2 months, which
greater genetic diversity than previously expected. Efforts was more than double that observed with AG alone
remain underway to devise methods to directly attack KRAS, (4.3 months).20 This ongoing trial represents an example of
which would certainly be game-changing for the treatment customizing treatment to a patients tumor characteristics
of PAC.13 In the meantime, identifying genetic changes and and provides promising preliminary evidence in favor of this
signatures that could predict patient response to targeted or approach.
more traditional therapies could have immediate and major Finally, multiple studies are underway to investigate im-
effect on patient care. munotherapy for the treatment of PAC. Pancreatic adeno-
Results from the Australian Pancreatic Cancer Genome carcinoma is known to be a highly immunosuppressive
Initiative within the International Cancer Genome Consor- disease, rendering immunotherapeutic approaches that
tium defined four subtypes of PAC: stable, locally rear- have shown success in other cancer types ineffective as
ranged, scattered, and unstable. Of particular interest is the single agents in PAC. To combat this immunosuppression
unstable phenotype, which was observed in 14% of the and recruit T cells to the PAC microenvironment, immune
100 patient samples analyzed. Unstable genomes were found checkpoint inhibitors such as antiCTLA-4, antiPD-1, and
to contain more than 200 structural variation events, which antiPD-L1 antibodies are being tested. Preliminary results
often suggests damage to the DNA repair pathways.14 This suggest that these tactics can be successful in combating
group and others have shown that patients with PAC who the immunosuppression but do not lead to cancer cell
have DNA damage repair alterations may be particularly death. Therefore, focus has shifted to combination of check-
sensitive to platinum-containing chemotherapeutics and/or point inhibitors with a therapeutic vaccine strategy or
PARP inhibitors. 15-17 A recent paper further refines the other agents such as chemotherapy or radiation (e.g.,
molecular subtypes of pancreatic cancer based on genomic NCT02303990).21-25
analysis.18 Correlative studies are revealing stratification approaches
An additional targeted therapeutic approach involves that indicate patients most likely to respond to immuno-
assessing patients levels of hyaluronan (HA), which is a therapeutic approaches, including antiCTLA-4 and anti
glycosaminoglycan present in the microenvironment sur- PD-1 therapy. A study published in 2015 showed that the
rounding PAC tumors. Hyaluronan contributes to elevated overall mutational load, neoantigen load, and expression of
interstitial pressure, and its inhibition with an agent called cytolytic markers in the immune microenvironment of pa-
PEGPH20 leads to the expansion of tumor-associated blood tients with metastatic melanoma were significantly associ-
vessels, allowing delivery of other drugs to the tumor.18,19 ated with clinical benefit to ipilimumab.26 Another study,
Interim analyses of a phase II randomized study of patients also published in 2015 that included patients with nonsmall
with previously untreated metastatic PAC treated with cell lung cancer, melanoma, and renal cell cancer, showed

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e211


MATRISIAN AND BERLIN

that nine out of 25 patients with PD-L1positive tumors had outcomes registry to obtain more detailed information on
an objective response to antiPD-1 therapy, while all 17 their experience and the outcome of treatment. Since its
patients with PD-L1negative tumors had no response.27,28 inception in June 2014 to December 2015, Know Your Tumor
Standardized testing of PD-L1 on tumor cells for the pre- has enrolled 496 patients with pancreatic cancer from 38
diction and evaluation of treatment efficacy is needed. In different states.32
addition, mismatch repairdeficient tumors may be sensi- There were 175 molecular profiles from Know Your Tumor
tive to immune checkpoint blockade. Currently, multiple participants evaluated as of December 31, 2015. Of the 175
trials are underway testing the sensitivity of antiPD-1 patients with completed reports, 58% were treated at an
therapy in this subset of patients in several gastrointes- academic institution or high-volume center, while 42% were
tinal cancers, including a phase II trial in patients with treated at a community center (Fig. 6). The distribution of
previously treated advanced colorectal cancer,29 a phase III pathologic genomic alterations as determined by analysis
first-line randomized trial comparing antiPD-1 therapy to with the Foundation One panel of 343 genes is shown in
standard chemotherapy in patients with metastatic co- Fig. 7.32 Forty percent of patients had at least one actionable
lorectal cancer,30 and a phase II trial for patients with alteration as defined based on a high response rate in pa-
previously treated colorectal and other gastrointestinal tients with an identified molecular abnormality in any cancer
cancers. This phase II trial reported promising response to type, or based on a mechanism/pathway-defined implica-
antiPD-1 therapy in noncolorectal gastrointestinal cancers tion of response to treatment.33 For example, 18% of pa-
with mismatch repair deficiency, including ampullary, pan- tients tumors had alterations in genes involved in DNA
creas, biliary small bowel, and gastric cancers.31 damage repair, including BRCA2, PALB2, ATM, and others
As we gather more genomic, proteomic, and immune cell indicated in Fig. 7. These alterations suggest treatment
information about PAC and accumulate evidence from this with a DNA-damaging agent such as platinum and/or a PARP
and other cancer types about molecularly targeted treat- inhibitor. Based on patient follow-up as of December 31,
ment approaches, efforts such as the Pancreatic Cancer 2015, 34 patients received their next line of treatment
Action Networks Know Your Tumor precision medicine consistent with the Perthera report, eight of whom enrolled
initiative will play an important role in providing patients in a clinical trial and three of whom used an off-label
access to personalized treatment approaches so that hy- treatment. In addition, six patients received therapy that
potheses on the predictive value of these markers can be was not indicated by the report. Although the numbers are
tested. Patients with pancreatic cancer across the United small, preliminary analysis suggests an improved outcome
States are able to access Know Your Tumor for multi-omic for patients who followed options based on the Perthera
molecular profiling information on their tumor by contact- report compared with those who received treatment not
ing the Pancreatic Cancer Action Networks Patient Central. listed on the report (median overall survival 45 vs.
A highly trained Patient Central associate determines the 32 weeks).33
eligibility and interest of the patient in enrolling in the Clinical trials for pancreatic cancer are incorporating
program and transfers suitable patients to a partnering biomarkers for patient selection. In January 2016, an analysis
organization, Personalized Cancer Therapy, Inc. (doing of the Pancreatic Cancer Action Networks clinical trial da-
business as [d.b.a] Perthera). Perthera obtains patient tabase revealed 21 of 174 (12%) clinical trials required a
consent, talks to the treating physician, facilitates obtaining molecular indicator for enrollment. It is anticipated that this
suitable biopsy tissue according to standard operating percentage will increase over time as more knowledge is
procedures, and sends the sample to appropriate diagnostic gained and a targeted approach is applied to the deadliest
laboratories. Multi-omic testing is performed, including cancers.
genomic, protein, and phosphoprotein analysis. The results
are returned to Perthera for data analysis and integration,
and an expert medical review is conducted that combines OTHER UPPER GASTROINTESTINAL CANCERS
this information with current literature and knowledge of Similar to pancreatic cancer, other upper gastrointestinal
pancreatic cancer clinical trial results and the patients cancers have also had limited successes. Biliary tract cancer
history. The medical review team includes a medical on- and hepatocellular cancers each have only one standard
cologist with considerable experience and expertise in regimen. After initial success with sorafenib as first-line
treating pancreatic cancer. Treatment options are ranked therapy for hepatocellular carcinoma, multiple other vas-
and include clinical trials with up-to-date information on cular endothelial growth factor receptor (VEGFR) inhibitors
availability from the Pancreatic Cancer Action Network were unsuccessfully tested in phase III trials.34 Although it is
database, solid tumor phase I trials, off-label treatments, not surprising that other VEGFR inhibitors were tested, it is
and standard of care options. The report is sent to the remarkable that the sheer number of trials were conducted
treating physician, the patient, and the Pancreatic Cancer and no other targets were evaluated in a substantive manner.
Action Network, and outcomes data are collected from both Similarly, based on almost no preclinical evidencesimply
the patient and the treating physician. All patients using overexpression of the proteinEGFR inhibitors were tested
Know Your Tumor are encouraged to participate in the in multiple randomized studies in gastroesophageal cancer,
Pancreatic Cancer Action Networks patient-reported all of which were negative.

e212 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


LESSONS FROM PANCREATIC CANCER CLINICAL TRIALS

FIGURE 6. Know Your Tumor Recruitment Across the United States and Community and Academic/High-Volume
Sites

The question for the future of all upper gastrointestinal afterthought used as rationalization for a planned study. As
cancersand all cancers in generalis whether we can start noted above, designing clinical trials in the era of person-
conducting smarter trials. In 2009, the State of the Science alized medicine may allow for smaller trials with greater
meeting results for pancreatic cancer were published.35 The effect. Others have recommended n of 1 studies, testing
meeting emphasized having preclinical data guide the de- different agents or combinations for an individual patient,
velopment of the clinical trials, rather than becoming an but how this can be done without simply being an anecdote

FIGURE 7. Pathogenic Alterations Observed by Next-Generation Sequencing in Know Your Tumor Participants

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e213


MATRISIAN AND BERLIN

has yet to be defined. Smarter trials can be smaller, but it molecular characteristics of the tumor from patients across
unlikely that one patient can define the utility of a drug. the United States and learn from them. This will set the stage
for vast improvements in the current approach and rapid
CONCLUSION advances toward improving the survival of those with the
The lessons we have learned from pancreatic cancer reflect deadliest cancers.
the potential that exists in upper gastrointestinal malig-
nancies in general. Although we celebrate the few successes, ACKNOWLEDGMENT
it is essential that we learn from the failures. Clinical trials Pancreatic Cancer Action Network staff and associates that
must be smarter in their design to conserve the precious contributed to this work include Lynn Matrisian, PhD, MBA
resource of patients willing to enroll in clinical trials. Ap- (concepts, writing, oral presentation); Lola Rahib, PhD (data
plying biomarkers that identify patients with a high prob- analysis, writing, figure preparation); Porsha James, MPH,
ability of response is one way to leverage remarkable and Kyla Holmes (data collection for clinical trial landscape
advances in cancer research with the practical outcome of and accrual); Allison Rosenzweig, PhD (writing); William
reducing the number of patients needed to identify a Hoos, MBA (concepts); and our partners from Personalized
clinically meaningful result. Initiatives such as the Pancre- Cancer Therapy, Inc. (Perthera) and the Know Your Tumor
atic Cancer Action Networks Know Your Tumor provide a initiative. We thank all trial sponsors that provided accrual
mechanism to disseminate the practice of interrogating the information.

References
1. Deadliest Cancers Coalition. http://www.deadliestcancers.org/. Accessed 16. Lohse I, Borgida A, Cao P, et al. BRCA1 and BRCA2 mutations sensitize to
February 1, 2016. chemotherapy in patient-derived pancreatic cancer xenografts. Br J
2. Pancreatic Cancer Action Network. The Recalcitrant Cancer Research Cancer. 2015;113:425-432.
Act: An Important Step Toward Improving Pancreatic Cancer Survival. 17. Kindler HL, Locker GY, Mann H, et al. POLO: a randomized phase III trial
https://www.pancan.org/wp-content/uploads/2015/07/fact-sheet- of olaparib tablets in patients with metastatic pancreatic cancer (mPC)
Recalcitrant-Cancer-Research-Act.pdf. Accessed February 2, 2016. and a germline BRCA1/2mutation (gBRCAm) who have not progressed
3. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin. following first-line chemotherapy. J Clin Oncol. 2015;33:15s (suppl;
2016;66:7-30. abstr TPS4149).
4. Rahib L, Smith BD, Aizenberg R, et al. Projecting cancer incidence and 18. Bailey P, Chang DK, Nones K, et al. Genomic analyses identify molecular
deaths to 2030: the unexpected burden of thyroid, liver, and pancreas subtypes of pancreatic cancer. Nature. 2016;531:47-52.
cancers in the United States. Cancer Res. 2014;74:2913-2921. 19. Jacobetz MA, Chan DS, Neesse A, et al. Hyaluronan impairs vascular
5. National Comprehensive Cancer Network. Pancreatic Adenocarcinoma, function and drug delivery in a mouse model of pancreatic cancer. Gut.
version 2.2015. http://www.nccn.org/. Accessed February 2, 2016. 2013;62:112-120.
6. Conroy T, Desseigne F, Ychou M, et al. FOLFIRINOX versus gemcitabine 20. Hingorani SR, Harris PW, Hendifar AE, et al. High response rate and PFS
for metastatic pancreatic cancer. N Engl J Med. 2011;364:1817-1825. with PEGPH20 added to nab-paclitaxel/gemcitabine in stage IV pre-
7. Moore MJ, Goldstein D, Hamm J, et al. Erlotinib plus gemcitabine viously untreated pancreatic cancer patients with high-HA tumors:
compared with gemcitabine alone in patients with advanced pancreatic Interim results of a randomized phase II study. J Clin Oncol. 2015;33
cancer: a phase III trial of the National Cancer Institute of Canada Clinical (suppl; abstr 4006).
Trials Group. J Clin Oncol. 2007;25:1960-1966. 21. Foley K, Kim V, Jaffee E, Zheng L. Current progress in immunotherapy for
8. Feuerstein A, Ratain MJ. Oncology micro-cap stocks: caveat emptor! pancreatic cancer. Cancer Lett. Epub 2015 Dec 23.
J Natl Cancer Inst. 2011;103:1488-1489. 22. Le DT, Lutz E, Uram JN, et al. Evaluation of ipilimumab in combination
9. Hoos WA, James PM, Rahib L, et al. Pancreatic cancer clinical trials and with allogeneic pancreatic tumor cells transfected with a GM-CSF
accrual in the United States. J Clin Oncol. 2013;31:3432-3438. gene in previously treated pancreatic cancer. J Immunother. 2013;36:
10. Rhim AD, Mirek ET, Aiello NM, et al. EMT and dissemination precede 382-389.
pancreatic tumor formation. Cell. 2012;148:349-361. 23. Le DT, Wang-Gillam A, Picozzi V, et al. Safety and survival with GVAX
11. Wang-Gillam A, Li CP, Bodoky G, et al. Nanoliposomal irinotecan with pancreas prime and Listeria Monocytogenes-expressing mesothelin
fluorouracil and folinic acid in metastatic pancreatic cancer after (CRS-207) boost vaccines for metastatic pancreatic cancer. J Clin Oncol.
previous gemcitabine-based therapy (NAPOLI-1): a global, randomised, 2015;33:1325-1333.
open-label, phase 3 trial. Lancet. 2016;387:545-557. 24. Le DT, Crocenzi TS, Uram JN, et al. Randomized phase II study of the
12. Sleijfer S, Bogaerts J, Siu LL. Designing transformative clinical trials in safety, efficacy, and immune response of GVAX pancreas (with cyclo-
the cancer genome era. J Clin Oncol. 2013;31:1834-1841. phosphamide) and CRS-207 with or without nivolumab in patients with
13. National Cancer Institute. The RAS Initiative. http://www.cancer.gov/ previously treated metastatic pancreatic adenocarcinoma (STELLAR). J
research/key-initiatives/ras. Accessed February 2, 2016. Clin Oncol. 2016;34:4s (suppl; abstr TPS486).
14. Waddell N, Pajic M, Patch A-M, et al. Whole genomes redefine the 25. Twyman-Saint Victor C, Rech AJ, Maity A, et al. Radiation and dual
mutational landscape of pancreatic cancer. Nature. 2015;518:495-501. checkpoint blockade activate non-redundant immune mechanisms in
15. Kaufman B, Shapira-Frommer R, Schmutzler RK, et al. Olaparib mon- cancer. Nature. 2015;520:373-377.
otherapy in patients with advanced cancer and a germline BRCA1/2 26. Van Allen EM, Miao D, Schilling B, et al. Genomic correlates of response to
mutation. J Clin Oncol. 2015;33:244-250. CTLA-4 blockade in metastatic melanoma. Science. 2015;350:207-211.

e214 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


LESSONS FROM PANCREATIC CANCER CLINICAL TRIALS

27. Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune 33. Pishvaian MJ, Brody JR, Matrisian LM, et al. Multi-omic profiling (MoP)
correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366: for patients (pts) with pancreatic cancer (PDA): initial results of the
2443-2454. Know Your Tumor (KYT) initiative. J Clin Oncol. 2016;34:4s (suppl; abstr
28. Zheng P, Zhou Z. Human cancer immunotherapy with PD-1/PD-L1 282).
blockade. Biomark Cancer. 2015;7:15-18. 34. Llovet JM, Villanueva A, Lachenmayer A, et al. Advances in targeted
29. Le DT, Yoshino T, Jager D, et al. KEYNOTE-164: phase II study of therapies for hepatocellular carcinoma in the genomic era. Nat Rev Clin
pembrolizumab (MK-3475) for patients with previously treated, Oncol. 2015;12:436.
microsatellite instability-high advanced colorectal carcinoma. J Clin 35. Philip PA, Mooney M, Jaffe D, et al. Consensus report of the national
Oncol. 2016;34;4s (suppl; abstr TPS787). cancer institute clinical trials planning meeting on pancreas cancer
30. Diaz LAJ, Le DT, Yoshino T, et al. KEYNOTE-177: first-line, open-label, treatment. J Clin Oncol. 2009;27:5660-5669.
randomized, phase III study of pembrolizumab (MK-3475) versus 36. Rahma OE, Duffy A, Liewehr DJ, et al. Second-line treatment in ad-
investigator-choice chemotherapy for mismatch repair deficient or vanced pancreatic cancer: a comprehensive analysis of published
microsatellite instability-high metastatic colorectal carcinoma. J Clin clinical trials. Ann Oncol. 2013;24:1972-1979.
Oncol. 2016;34;4s (suppl; abstr TPS789). 37. Teague A, Lim KH, Wang-Gillam A. Advanced pancreatic adenocarci-
31. Le DT, Uram JN, Wang H, et al. PD-1 blockade in mismatch repair noma: a review of current treatment strategies and developing ther-
deficient non-colorectal gastrointestinal cancers. J Clin Oncol. 2016;34: apies. Ther Adv Med Oncol. 2015;7:68-84.
4s (suppl; abstr 195). 38. Schrag D, Archer L, Wang X, et al. A patterns-of-care study of post-
32. Engebretson A, Brody JR, Rahib L, et al. The Know Your Tumor (KYT) progression treatment (Rx) among patients (pts) with advanced pan-
initiative: a national program of multi-omic molecular profiling (MoP) creas cancer (APC) after gemcitabine therapy on Cancer and Leukemia
for patients (Pts) with pancreatic cancer (PDA). J Clin Oncol. 2016;34;4s Group B (CALGB) study #80303. J Clin Oncol. 2007;25:18s (suppl; abstr
(suppl; abstr 279). 4524).

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e215


GASTROINTESTINAL (NONCOLORECTAL) CANCER

HER2, VEGFR, and Beyond:


Genomic Profiling of Upper
Gastrointestinal Tract Cancers
and the Future of Personalized
Treatment

CHAIR
Jaffer A. Ajani, MD
The University of Texas MD Anderson Cancer Center
Houston, TX

SPEAKERS
Adam J. Bass, MD
Dana-Farber Cancer Institute
Boston, MA

Jeeyun Lee, MD, PhD


Samsung Medical Center
Seoul, South Korea
LEE, BASS, AND AJANI

Gastric Adenocarcinoma: An Update on Genomics, Immune


System Modulations, and Targeted Therapy
Jeeyun Lee, MD, PhD, Adam J. Bass, MD, and Jaffer A. Ajani, MD

OVERVIEW

Gastric adenocarcinoma (GAC) is a global health burden on all societies, and it was the third-leading cause of cancer-related
mortality in 2012, causing 723,000 deaths worldwide. The prognosis of patients with metastatic GAC remains poor, with a
median overall survival of less than 1 year in patients treated with currently available therapies. A limited number of
therapeutic agents is currently available. Recent additions to the armamentarium include trastuzumab and ramucirumab,
which have shown some survival advantage when added to cytotoxic(s). Genomic analyses have defined various genotypes
of GACs. The novel genomic knowledge can lead to discovery of novel targets and novel therapeutic agents. In this update,
we focus on the current genomic data, targeted therapies including immune system modulators, and expand on HER2/neu
testing and the use of agents against this target. Several other facets of GAC and its therapy are not to be included in this
review but have been discussed elsewhere.

D eeper analysis of the genomic and molecular features of


GAC can provide multiple benefits to the study and
development of therapy for GAC that is clearly a hetero-
whole-exome sequencing, microarray profiling of genomic
amplifications and deletions, messenger RNA (mRNA) and
micro-RNA expression analysis, and genome-wide DNA
geneous entity. Gastric adenocarcinoma has been divided methylation analysis with some samples also analyzed with
into two primary histologic variants: the intestinal and whole-genome sequencing or proteomic analysis. By con-
diffuse types. Beyond histology, there is discussion regarding trast, the ACRG GAC study accrued from a single referral
the differences in the GAC in Eastern and Western patients, hospital in South Korea, providing a more homogenously
different features of GAC in the distal versus proximal treated cohort with richer clinical annotation relative to the
stomach, and differences relating to distinct molecular TCGA study. The ACRG samples were subjected to more
etiologies and molecular subtypes of GACs. In this context, limited analysis including mRNA expression, somatic copy
modern genomic and molecular analyses can help delineate number, and focused gene sequencing.
subtypes of GAC and identify salient molecular features of Each of these projects developed a four-group classifi-
these distinct classes, information that is increasingly es- cation structure for GAC, with these classification ap-
sential to guide therapy, especially emerging targeted and proaches having some overlapping but also distinct features.
biologic agents.1-9 The ACRG categories were determined on the basis of gene
Recent years have witnessed a revolution in the capacity to expression profiling with the following features of GAC: (1)
genomically and molecularly describe cancer. As these microsatellite instability, (2) epithelial-to-mesenchymal tran-
technologies have become less costly, there have been sition (EMT) phenotype, (3) a p53 signature (CDKN1A and
several large-scale efforts to molecularly detail this cancer. MDM2 expressing), or (4) no p53 signature. Of these groups,
Here, we will discuss two recent substantial efforts in this the cohort with microsatellite instability had improved sur-
realm, studies emerging from the Asian Cancer Research vival, whereas the EMT group, which was composed mostly of
Group10 (ACRG) and from The Cancer Genome Atlas11 diffuse-type tumors, showed inferior survival. Furthermore,
(TCGA). TCGA, a consortium from the National Institutes of after surgical resection, those with the EMT signatures also
Health, collected primary untreated gastric samples from showed greater rates of recurrence and an association with
institutions across the world. These samples were all sub- subsequent peritoneal metastases. Although this EMT group
jected to a diverse array of molecular analyses including was largely diffuse-type tumors, the tumors in this group

From the Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Kangnamgu, Seoul, South Korea;
Division of Molecular and Cellular Oncology, Dana-Farber Cancer Institute, Boston, MA; Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson
Cancer Center, Houston, TX.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Jaffer A. Ajani, MD, Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston,
TX 77030; email: jajani@mdanderson.org.

2016 by American Society of Clinical Oncology.

104 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


GASTRIC ADENOCARCINOMA

lacked other features that have been seen commonly in Microsatellite instability was found as a distinct group by
diffuse GAC, such as mutation of CDH1, the gene responsible the TCGA as well as ACRG effort. Microsatellite insta-
for hereditary diffuse GAC, and recently identified DGC mu- bilitypositive GACs possess marked hypermutation as a
tations of the RHOA GTPase.12,13 As predicted, the cohort of result of the underlying defect in DNA mismatch repair.
samples with deficient p53 activity possessed greater aneu- These tumors also possess a relatively bland genomic copy
ploidy and accompanying amplifications of oncogenes in- number pattern lacking frequent genomic amplifications of
cluding ERBB2 (HER2). genes such as ERBB2. However, a number of likely path-
The TCGA classification derivation differed from the ACRG ogenic mutations exist within this cohort, including mu-
in that it was based upon an integration of six distinct mo- tations in targetable genes such as ERBB2, ERBB3, and
lecular platforms as opposed to the reliance upon mRNA PIK3CA, as well as oncogenes such as KRAS that lack ef-
expression. Based upon observations from the molecular fective inhibitors. A notable contrast with MSI-positive
data, tumors were divided into four groups: (1) GACs positive colorectal cancer was the lack of activating BRAF mutations
for Epstein-Barr virus (EBV), (2) microsatellite instability, (3) within these GACs. One notable question regarding
chromosomally unstable, and (4) genome stable. The latter microsatellite instability tumors, whose high mutation
genome stable group represented the remnants after the burdens generate large number of neoantigens, will be the
groups with EBV and microsatellite instability were excluded potential efficacy of immune checkpoint inhibitors as have
and the remaining GACs were divided based upon possession shown efficacy in microsatellite instabilitypositive co-
of aneuploidy (chromosomally unstable) or its absence (ge- lorectal cancers.15
nome stable). A first notable difference compared with the Like the EMT group from the ACRG, the TCGAs genome
ACRG was that EBV-positive tumors emerged as a distinct stable group primarily consist of those with diffuse histology.
entity. This difference likely follows the inclusion of DNA The GACs in this so-called genome stable group get their name
methylation analysis, which revealed a strong and highly from their lack of the hypermethylation prevalent in EBV-
distinguishing methylation signature in this cohort. Beyond positive, the hypermutation of microsatellite instability, or the
methylation, however, EBV-positive GACs possess other marked aneuploidy in chromosomally unstable GACs. Al-
clinically relevant findings, including highly recurrent PIK3CA though both the genome stable and EMT tumors were largely
mutations and genomic amplification of the genes encoding diffuse type, their molecular features did differ as evidenced
PD-1 pathway activators PD-L1 and PD-L2, suggesting the by the common finding of both CDH1 and RHOA mutations in
possible roles for PI3K pathway inhibitors, emerging immune the TCGA genome stable set. Despite these differences, results
checkpoint agents, and, perhaps, DNA hypomethylating from these two classification schemes provide insights into
agents. Like microsatellite instability GACs, EBV-positive GACs diffuse GAC. First, GACs with diffuse histology span molecular
have also been described to have a favorable phenotype.14 subtypes including those with microsatellite instability, EBV,
and marked aneuploidy. However, diffuse types are generally
less enriched in classic oncogenes and therapeutic targets such
as KRAS or ERBB2 that are more prevalent in other groups of
KEY POINTS GACs, speaking to the clear remaining need to identify new
therapeutic approaches and vulnerabilities for these highly
Patients with advanced GAC continue to have poor aggressive tumors.
quality of life, short survival, and limited therapeutic The largest group in the TCGA analysis, the chromosomally
choices. Many patients do not have access to clinical unstable tumors, matches most closely the p53-deficient
trials when standard regimens have failed.
tumors from the ACRG schema. As their names suggest,
The empiric approaches that assume one-size-fits-all
have been ineffective methods to improve the outcome
the salient features of these GACs are the high degree of
of patients with GAC. Alternatives and customized structural genomic aberrations, leading to a high frequency of
approaches must be considered. genomic amplifications of bona fide oncogenes such as re-
The Cancer Genome Atlas study of GACs has uncovered ceptor tyrosine kinases ERBB2, EGFR, MET, or FGFR2; cell cycle
four genotypes with unique features. This discovery mediators such as CCND1, CCNE1, and CDK6; and transcription
needs to be fully exploited to find novel targets and factor oncogenes such as GATA4, GATA6, and MYC. These
effective drugs. genomic data speak for the potential for inhibitors for targets
Trastuzumab and ramucirumab (in combination with other than HER2 to ultimately have roles in GACs. Indeed, the
cytotoxic[s]) have produced only minimum/modest emergence of approved inhibitors targeting specific cell cycle
benefit for patients with GAC, emphasizing the need for kinases, such as the CDK4/6-directed agent palbociclib, should
better therapies.
be evaluated in specific sets of patients guided by genomic
Since early results from checkpoint inhibitors show
promising results in some patients with GAC, rationally
data.
designed multiprong approaches that combine immune Comparing the TCGA chromosomally unstable group and
modulation, targeted agent(s), and conventional the ACRG p53-negative group, a notable difference is that
cytotoxics may prove more advantageous than all the TCGA chromosomally unstable class comprises a much
previous approaches. more substantial fraction of the total tumors than does the
p53-negative group. This difference may reflect not only the

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 105


LEE, BASS, AND AJANI

TABLE 1. Major Clinical Trials in Gastric Adenocarcinoma With Targeted Agents

Patient Results
Type of Selection (Primary
Target Trial Study/Line Method Regimen Endpoint) Reference
HER2 ToGa Phase III/first HER2 IHC 5-FU/capecitabine + Positive (OS) Bang et al 20105
cisplatin 6 trastuzumab
HER2 LOGIC Phase III/first HER2 Lapatinib + XELOX Negative (OS) Hecht et al16
amplification
XELOX
HER2 TYTAN Phase III/second HER2 Paclitaxel + lapatinib vs. paclitaxel Negative (OS) Bang et al (2013)17
amplification
EGFR EXPAND Phase III/first All comer Cetuximab/XP vs. placebo/XP Negative (OS) Lordick et al18
EGFR REAL-III Phase III/first All comer Panitumumab/EOC vs. EOC Negative (OS) Waddell et al19
EGFR Nimotuzumab Phase II/second All comer Nimotuzumab/irinotecan vs. Negative Kim et al20
irinotecan
VEGF AVAGAST Phase III/first All comer XP/bevacizumab vs. XP Negative (OS) Van Cutsem et al21
MET RILOMET-1 Phase III/first MET IHC Rilotumumab/ECX vs. ECX Negative (OS) Iveson et al22
MET METGastric Phase III MET IHC Onartuzumab/FOLFOX vs. FOLFOX Negative (OS) Shah et al23
FGFR2 SHINE R-Phase II/second FGFR2 AZD4547/paclitaxel vs. paclitaxel Negative (PFS) Bang et al (2015)24
amplification
mTOR GRANITE Phase III/second All comer Everolimus vs. placebo Negative (OS) Ohtsu et al25
or third
AKT MK2206 Phase II/second All comer MK-2206 Response rate, 1% Ramanathan et al26
ATM Olaparib R-Phase II/second ATM IHC Paclitaxel/olaparib vs. Negative (PFS) Bang et al (2015)27
paclitaxel/placebo
VEGF MEGA R-Phase II/first All comer FOLFOX/aflibercept vs. FOLFOX Negative (6-mo PFS) Enzinger et al28
HER2 GATSBY Phase II/III/ second HER2 IHC TDM1 vs. paclitaxel or docetaxel Negative (OS) Kang et al29
VEGFR-2 RAINBOW Phase III/second All comer Paclitaxel/ramucirumab vs. Positive (OS) Wilke et al30
paclitaxel/placebo
VEGFR-2 REGARD Phase III/third All comer Ramucirumab vs. placebo Positive (OS) Fuchs et al31
Abbreviations: OS, overall survival; XP, capecitabine (Xeloda) and cisplatin; EOC, epirubicin, oxaliplatin, and capecitabine; IHC, immunohistochemistry; ECX, epirubicin, cisplatin, and
capecitabine; PFS, progression-free survival.

different classification schema but also the differences in the to developing new therapies, we will have to take into ac-
populations studied. Chromosomally unstable tumors were count this clear molecular heterogeneity that relates to
more prevalent in the proximal stomach, whereas other different etiologies of genomic instability, different micro-
groups were more prevalent in more distal regions. It is bial etiologies, and GACs possessing distinct driving somatic
well recognized that GAC in Western populations has, in alterations. These differences will impact our approaches
recent years, become increasingly a disease of the proximal both to standard conventional therapy, emerging targeted
stomach, mirroring the rise of esophageal adenocarcinoma. therapies, and immunotherapy.
As this subset may be a more aneuploidy variant of GACs, a
cohort such as the TCGA group with more Western patients TARGETED THERAPIES AND IMMUNE SYSTEM
would be expected to harbor higher degrees of aneuploidy. MODULATION TO TREAT GASTRIC
Indeed, in the ACRG cohort, the percentage of diffuse-type ADENOCARCINOMA
GACs was higher and proximal GACs lower than in the TCGA Table 1 summarizes representative important clinical trial
study. Regardless of this difference, the prevalence of dis- results in GAC.
tinct oncogene amplification events in these tumors does Negative trials with targeted agents have substantially
speak for the potential of these different alterations to serve outnumbered the positive trials (the ToGA, REGARD, and
as biomarkers to guide therapy. RAINBOW trials) in GAC in the past decade. Among other
Overall, the most important lesson of these differences is factors, major factors accounting for this negative outcome
that it is beyond the time to try to devise singular approaches may be (1) many trials did not select the patient population
for GAC. There are substantial molecular and etiologic dif- based on specific target (lacking any biomarker) and did not
ferences across these GACs. Furthermore, even within a account for distinct molecular subtypes and how they may
single molecular subtype, there remains highly relevant impact response to therapy, (2) inaccurate biomarker for
heterogeneity. For example, even among the highly aneu- selection patient selection (i.e., HER2, fluorescence in situ
ploidy GACs, optimal treatment of an ERBB2-amplified and hybridization [FISH] vs. HER2 immunohistochemistry [IHC];
MET-amplified GACs will clearly differ. As we move forward FGFR2 FISH vs. FGFR2 IHC; MET FISH vs. MET IHC). There may

106 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


GASTRIC ADENOCARCINOMA

be many other factors influencing the failure of various drugs plasma DNA and high FGFR2 copy number gain,37 suggesting
when investigated in the clinic. that optimal use of biomarkers may mark those for whom
In general, for the targeted agents, clinical trials in GAC for this target may be effective. Hence, although the SHINE trial
unselected patient population have been largely disap- did not meet its primary endpoint, it is not yet conclusive to
pointing. One clear example is the EXPAND trial, where EGFR preclude FGFR2 inhibitor from clinical development, but
inhibitors were given to all patients despite the prevalence of more optimized biomarker selection criteria are needed for
EGFR gene amplification occurs in only approximately 5% of the future FGFR2 inhibitor trials.
GACs and EGFR protein overexpression in approximately 15% There is emerging evidence that pembrolizumab/nivolumab
to 20% of patients with GAC. Another EGFR-directed study blocks binding of PD-L1 to its receptor, PD-1, resulting in T-cell
in unselected patients, the nimotuzumab/irinotecan second- activation/proliferation. KEYNOTE-012, a phase IB study of
line trial, also failed to demonstrate survival benefit. However, pembrolizumab in patients with metastatic GAC and PD-L1
in subgroup analysis of the EXPAND trial, a subset of patients tumor cell positivity by IHC demonstrated promising results.38
with high EGFR overexpression by IHC showed survival benefit MEDI4736, another antiPD-L1 antibody, also has demon-
from cetuximab compared with placebo.32 In line with this, strated promising activity in GAC in an initial analysis.
in the randomized phase II trial of nimotuzumab/irinotecan
versus irinotecan, a subset of patients with EGFR 2+/3+ by Is It Time for Umbrella Trials in GAC?
IHC demonstrated superior progression-free survival (PFS) Recently, even further complicating the clinical develop-
and overall survival (OS).33 Based on this finding, a phase III ment of targeted agents in GAC, TCGA and ACRG10,11
trial of nimotuzumab/irinotecan versus irinotecan with pa- published comprehensive genomic data, and both groups
tients selected based upon EGFR2/3-positive IHC is currently have reported on molecular subtypes of GAC (detailed
ongoing. earlier). Given the tumor heterogeneity among patients
Recently, another disappointing result came from the with GAC and within each patient, an umbrella trial has
phase III RILOMET-1 trial that compared rilotumumab in been recently introduced.39-41 An umbrella (or basket) trial
combination with epirubicin/cisplatin/capecitabine (ECX) investigates a single tumor type selected according to the
versus ECX alone as first-line chemotherapy administered biomarkers relevant to one or more of the candidate drugs
in patients with MET-positive GAC. There was worse sur- and patients and directed toward different arms of the study
vival in the arm containing the anti-HGFtargeted agent according to the molecular characteristics of the tumor.39
rilotumumab.34 Moreover, there were no subgroups shown The Personalized Antibodies from Gastro-Esophageal Ade-
to benefit from rilotumumab including those patients with nocarcinoma (PANGEA) trial selects patients with GAC
higher percentages of cells with greater than or equal to +1 according to biomarker status, such as MET amplification,
MET expression by IHC.34 It is not yet reported whether FGFR2 amplification, and KRAS amplification, to different
genomic amplification of the gene encoding MET influenced arms in first-line setting. Another trial, the Targeted Agent
survival or response to this agent. However, a phase I trial of Evaluation in Gastric Cancer Basket Korea (VIKTORY), assigns
the MET kinase inhibitor AMG337 with 13 patients with patients with metastatic GAC according to RAS amplification,
MET-amplified GAC demonstrated a response rate of 62%.35 RAS mutation, TP53 mutation, PIK3CA mutation, MEK high
Intriguingly, 40% to 50% of patients with MET-amplified GAC versus low signature, MET amplification, and MET protein
enrolled on AMG337 trial harbored coamplification of HER2 overexpression to corresponding treatment arms in second-
and/or EGFR concurrently, suggestive of patients who might line treatment. Patients without any of the designated
benefit from combination targeted therapy.36 To further add markers receive either mTOR inhibitor or Akt inhibitor with
to the complexity, there was a substantial heterogeneity and paclitaxel.
discordance in MET amplifications between the primary and
metastatic lesions in a single studied patient with GAC. HER2/NEU AS TARGET IN GASTRIC
Hence, molecular profiling of a single tumor biopsied may ADENOCARCINOMA AND AN UPDATE ON HER2/
not be inadequate to guide targeted therapy in GAC. NEU TESTING
Another lesson can be learned from the SHINE trial that A small fraction of GAC in patients overexpresses HER2/neu
compared AZD4547, an FGFR2 inhibitor, administered in protein or have increase copy number of the ERBB2 gene.
patients with FGFR2-amplified GAC in the second-line The HER2/neu receptor is one of the four receptors in the
setting. Despite strong preclinical evidence for AZD4547 EGFR family. The intracellular domain has tyrosine kinase
in FGFR2-amplified GAC preclinical models, the SHINE trial activity. The HER2/neu receptor can be targeted by an
failed to meet the primary endpoint, PFS, when AZD4547 antibody (such as trastuzumab or pertuzumab, which can
was added to paclitaxel.24 The trial screened 960 patients prevent homodimerization or heterodimerization with other
and randomly assigned 71 patients with 9% prevalence for family members) against the extracellular domain or re-
FGFR2 amplification. Although that trial was negative, in an- ceptor tyrosine kinase small-molecule inhibitors (such as
other phase II trial with AZD4547 in FGFR1-, 2-, and 3-amplified lapatinib). There have been several prospective phase III
cancer, three of nine (33%) patients with FGFR2-amplified trials that have investigated inhibition of HER2/neu signaling
GAC experienced a partial response. Notably, all responding in patients with HER2/neu-positive GAC.5,16,42 However,
patients had FGFR2 copy number gain demonstrated in only the ToGA trial showed statistically significant benefit

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 107


LEE, BASS, AND AJANI

(p = .0046).5 Therefore, the use of trastuzumab in combi- In a biopsy specimen of GAC (or gastroesophageal ade-
nation with chemotherapy in patients with HER2/neu-pos- nocarcinoma), if 5% of cells adequately stain for HER2/neu,
itive metastatic GAC is now considered a standard. We this is currently considered adequate to designate a positive
discuss various aspects of targeting HER2/neu pathway and HER2/neu status; however, the presence of positive staining
HER2 testing. in such a small fraction of the sampled area raises obvious
questions of sampling and whether such few cells really
The Receptor represent the entire tumor (including metastatic tumor).
The HER2/neu receptor is unique in that it lacks the ligand However, from surgical specimen, at least 10% of cells must
binding cleft for extracellular growth factors that other stain for HER2/neu designation. This is a bit better but still
ERBB3 family members have. Its dimerization loop is ex- raises the same questions.
tended like others, and the intracellular domain has ATP-
binding cleft.43 The ERBB2 gene, encoding HER2/neu, is Clinical Trials
often amplified in many cancers including a fraction of GACs The ToGA trial screened 3,803 patients with untreated,
(including in some squamous cell carcinoma of the esoph- advanced gastroesophageal adenocarcinoma and randomly
agus). The drugs that can inhibit HER2/neu receptors are assigned 594 patients with HER2/neu positive tumors (as
well described in a recent review.43 However, primary and defined for the purposes of this trial) to receive either
secondary resistance to HER2/neu inhibition, as with other chemotherapy (fluoropyrimidine/cisplatin) and trastuzu-
single targets, is a likely inevitable and universal phenom- mab or chemotherapy. The primary endpoint of improved
enon.44 Common mechanisms of HER2/neu directed ther- overall survival was achieved (13.8 months vs. 11.1 months;
apy resistance have not been extensively studied in GAC, p = .0046). Other endpoints (PFS and increase in response
but, in breast cancer, it has been seen because of PTEN loss, rate) were also met. Trastuzumab was found safe to deliver
truncation of the receptor, activating mutations in PIK3CA, with standard chemotherapy. This trial led to worldwide
or upregulation of other oncogenes (IGF1 or ERBB3).43,44 regulatory approval of trastuzumab and clinical adoption.
However, the benefit from trastuzumab diminished con-
siderably when the results were reanalyzed after a longer
HER2/neu Test Results in Various Settings follow-up by the U.S. Food and Drug Administration (the
In routine use, the presence of strong (3+) IHC staining has hazard ratio increased from 0.73 to 0.80, and the overall
been shown to have strong concordance with the presence survival difference narrowed to a meager 1.4 months). This
of amplification of ERBB2, leading this protein test alone to would suggest considerable heterogeneity among patients
be sufficient to guide the use of trastuzumab.5,45 However, with HER2/neu-positive GACs and need for better agents.
one issue to consider is the considerable heterogeneity in This modest impact also suggests the need to refine our
staining for HER2/neu and the potential adequacy of a single identification of possible gradations of HER2/neu-positive
small biopsy to accurately assess HER2 status (as opposed GACs. Furthermore, these results speak strongly to the need
to a surgical specimen46-48). How many unique specimens to develop additional treatments for patients with HER2/
should be tested also remains unresolved.49-51 Similarly, neu-positive GAC, both for up-front therapies and when the
technical issues regarding the optimal copy numbers cutoffs resistance emerges.
to use to guide use of anti HER2/neu agents are also a subject In contrast to the ToGA trial of the therapeutic antibody
of debate.52 In addition, whether there is concordance in trastuzumab, results have been disappointing for HER2-
HER2/neu results if metastatic cancer or primary cancer is directed kinase inhibitors. Two trials studied the re-
tested, remains generally unresolved.53-55 Regarding prog- versible EGFR/ERBB2 inhibitor lapatinib in the second-line
nostic significance, HER2/neu positivity in a GAC does not setting42 and in the first-line setting,16 both yielding
confer poor prognosis, but there are reports to refute this as negative results. Ado-trastuzumab emtansine (T-DM1) is
well.56-63 an antibody-drug conjugate consisting of antibody
(trastuzumab) and cytotoxic (DM1). The EMILIA trial for
Testing for HER2/neu patients with HER2/neu-positive breast cancer was pos-
Currently, we recommend the guidelines outlined by the itive, resulting in a survival benefit of 5.8 months; how-
National Comprehensive Cancer Network (NCCN)64,65 until ever, a T-DM1 trial for HER2/neu-positive GAC in the
formal guidelines to be published jointly by the American second-line setting was a disappointment.66 Currently,
Society of Clinical Oncology, College of American Patholo- trials are ongoing with higher doses of trastuzumab to
gists, and American Society of Clinical Pathologists become improve exposure and another that adds the second
available in early 2016. Patients with gastroesophageal antibody pertuzumab to trastuzumab in comparison with
adenocarcinoma who have unresectable, advanced cancer trastuzumab (the JACOB trial). Additional mechanistic
should be tested for HER2/neu, starting with IHC. If it is 3+ study into the molecular effects of specific classes of anti-
(positive), 1+ (negative), or 0 (negative), no further testing is HER2/neu agents and the mechanistic etiologies of both
required. However, if it is 2+ (equivocal), then FISH for de novo and acquired resistance are required to
amplification of ERBB2 is recommended. Currently, any establish a foundation for rational development of new
available tumor tissue should be tested. approaches for to target HER2-positive GACs.

108 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


GASTRIC ADENOCARCINOMA

FUTURE DIRECTIONS the development of effective combination therapies to


Gastric adenocarcinoma remains a deadly disease with exploit these targets. Furthermore, we will have to eval-
significant molecular and histologic heterogeneity. Trials of uate the potential for tumor heterogeneity and tumor
empirical chemotherapy, largely with a one-size-fits-all evolution/resistance to be a barrier to these therapies.
approach have led to modest survival benefits and limited Moreover, emerging immunotherapies possess great
options. Newer insights into the underlying etiology of promise. Ultimately, linking immune, cytotoxics, and other
GACs does bring possible promise of new, improved targeted therapies will likely be needed as part of multi-
therapies. However, substantial work needs to be com- pronged approaches to provide maximum benefits to our
pleted to identify optimal targets, optimal biomarkers, and patients.

References
1. Torre LA, Bray F, Siegel RL, et al. Global cancer statistics, 2012. CA Cancer 16. Hecht JR, Bang YJ, Qin SK, et al. Lapatinib in combination with cape-
J Clin. 2015;65:87-108. citabine plus oxaliplatin in human epidermal growth factor receptor
2. Al-Batran SE, Hartmann JT, Probst S, et al; Arbeitsgemeinschaft 2-positive advanced or metastatic gastric, esophageal, or gastro-
Internistische Onkologie. Phase III trial in metastatic gastroesophageal esophageal adenocarcinoma: TRIO-013/LOGiC-A randomized phase III
adenocarcinoma with fluorouracil, leucovorin plus either oxaliplatin or trial. J Clin Oncol. 2016;34:443-451.
cisplatin: a study of the Arbeitsgemeinschaft Internistische Onkologie. 17. Bang Y-J RX, Taroh S, et al. A randomized, open-label, phase III study of
J Clin Oncol. 2008;26:1435-1442. lapatinib in combination with weekly paclitaxel versus weekly paclitaxel
3. Cunningham D, Starling N, Rao S, et al; Upper Gastrointestinal Clinical alone in the second-line treatment of HER2 amplified advanced gastric
Studies Group of the National Cancer Research Institute of the United cancer in Asian population: TyTAN study. J Clin Oncol. 2013;30 (suppl 34;
Kingdom. Capecitabine and oxaliplatin for advanced esophagogastric abstr 11).
cancer. N Engl J Med. 2008;358:36-46. 18. Lordick F, Kang YK, Chung HC, et al; Arbeitsgemeinschaft Internistische
4. Kang YK, Kang WK, Shin DB, et al. Capecitabine/cisplatin versus 5-fluorouracil/ Onkologie and EXPAND Investigators. Capecitabine and cisplatin with or
cisplatin as first-line therapy in patients with advanced gastric cancer: without cetuximab for patients with previously untreated advanced
a randomised phase III noninferiority trial. Ann Oncol. 2009;20:666-673. gastric cancer (EXPAND): a randomised, open-label phase 3 trial. Lancet
5. Bang YJ, Van Cutsem E, Feyereislova A, et al; ToGA Trial Investigators. Oncol. 2013;14:490-499.
Trastuzumab in combination with chemotherapy versus chemotherapy 19. Waddell T, Chau I, Cunningham D, et al. Epirubicin, oxaliplatin, and
alone for treatment of HER2-positive advanced gastric or gastro- capecitabine with or without panitumumab for patients with previously
oesophageal junction cancer (ToGA): a phase 3, open-label, rando- untreated advanced oesophagogastric cancer (REAL3): a randomised,
mised controlled trial. Lancet. 2010;376:687-697. open-label phase 3 trial. Lancet Oncol. 2013;14:481-489.
6. Wilke H, Muro K, Van Cutsem E, et al; RAINBOW Study Group. 20. Kim YH, Sasaki Y, Lee KH, et al. Randomized phase II study of nimo-
Ramucirumab plus paclitaxel versus placebo plus paclitaxel in patients tuzumab, an anti-EGFR antibody, plus irinotecan in patients with 5-
with previously treated advanced gastric or gastro-oesophageal junc- fluorouracil-based regimen-refractory advanced or recurrent gastric
tion adenocarcinoma (RAINBOW): a double-blind, randomised phase 3 cancer in Korea and Japan: preliminary results. J Clin Oncol. 2011;29:4s
trial. Lancet Oncol. 2014;15:1224-1235. (suppl 4; abstr 87).
7. Shah MA. Update on metastatic gastric and esophageal cancers. J Clin 21. Van Cutsem E, de Haas S, Kang YK, et al. Bevacizumab in combination
Oncol. 2015;33:1760-1769. with chemotherapy as first-line therapy in advanced gastric cancer:
8. Cohen DJ, Leichman L. Controversies in the treatment of local and a biomarker evaluation from the AVAGAST randomized phase III trial.
locally advanced gastric and esophageal cancers. J Clin Oncol. 2015;33: J Clin Oncol. 2012;30:2119-2127.
1754-1759. 22. Iveson T, Donehower RC, Davidenko I, et al. Rilotumumab in
9. Tan P, Yeoh KG. Genetics and molecular pathogenesis of gastric ade- combination with epirubicin, cisplatin, and capecitabine as first-line
nocarcinoma. Gastroenterology. 2015;149:1153-1162 e3. treatment for gastric or oesophagogastric junction adenocarci-
10. Cristescu R, Lee J, Nebozhyn M, et al. Molecular analysis of gastric noma: an open-label, dose de-escalation phase 1b study and a
cancer identifies subtypes associated with distinct clinical outcomes. double-blind, randomised phase 2 study. Lancet Oncol. 2014;15:
Nat Med. 2015;21:449-456. 1007-1018.
11. Cancer Genome Atlas Research Network. Comprehensive molecular 23. Shah MA, Bang Y-J, Lordick F, et al. METGastric: A phase III study of
characterization of gastric adenocarcinoma. Nature. 2014;513:202-209. onartuzumab plus mFOLFOX6 in patients with metastatic HER2-
12. Kakiuchi M, Nishizawa T, Ueda H, et al. Recurrent gain-of-function negative (HER22) and MET-positive (MET+) adenocarcinoma of the
mutations of RHOA in diffuse-type gastric carcinoma. Nat Genet. 2014; stomach or gastroesophageal junction (GEC). J Clin Oncol. 2015;33
46:583-587. (suppl; abstr 4012).
13. Wang K, Yuen ST, Xu J, et al. Whole-genome sequencing and com- 24. Bang Y-J, Cutsem EV, Mansoor W, et al. A randomized, open-label phase
prehensive molecular profiling identify new driver mutations in gastric II study of AZD4547 (AZD) versus paclitaxel (P) in previously treated
cancer. Nat Genet. 2014;46:573-582. patients with advanced gastric cancer (AGC) with fibroblast growth
14. van Beek J, zur Hausen A, Klein Kranenbarg E, et al. EBV-positive gastric factor receptor 2 (FGFR2) polysomy or gene amplification (amp): SHINE
adenocarcinomas: a distinct clinicopathologic entity with a low fre- study. J Clin Oncol. 2015;33 (suppl; abstr 4014).
quency of lymph node involvement. J Clin Oncol. 2004;22:664-670. 25. Ohtsu A, Ajani JA, Bai YX, et al. Everolimus for previously treated ad-
15. Le DT, Uram JN, Wang H, et al. PD-1 blockade in tumors with mismatch- vanced gastric cancer: results of the randomized, double-blind, phase III
repair deficiency. N Engl J Med. 2015;372:2509-2520. GRANITE-1 study. J Clin Oncol. 2013;31:3935-3943.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 109


LEE, BASS, AND AJANI

26. Ramanathan RK, McDonough SL, Kennecke HF, et al. Phase 2 study of 40. Catenacci DV. Next-generation clinical trials: Novel strategies to address
MK-2206, an allosteric inhibitor of AKT, as second-line therapy for the challenge of tumor molecular heterogeneity. Mol Oncol. 2015;9:
advanced gastric and gastroesophageal junction cancer: A SWOG co- 967-996.
operative group trial (S1005). Cancer. Epub 2015 Mar 30. 41. Lee J, Kim KM, Kang WK, et al. Innovative personalized medicine in
27. Bang YJ, Im SA, Lee KW, et al. Randomized, double-blind phase II trial gastric cancer: time to move forward. Clin Genet. 2014;86:37-43.
with prospective classification by ATM protein level to evaluate the 42. Satoh T, Xu RH, Chung HC, et al. Lapatinib plus paclitaxel versus pac-
efficacy and tolerability of olaparib plus paclitaxel in patients with litaxel alone in the second-line treatment of HER2-amplified advanced
recurrent or metastatic gastric cancer. J Clin Oncol. 2015;33:3858-3865. gastric cancer in Asian populations: TyTANa randomized, phase III
28. Enzinger PC, McCleary NJ, Zheng H, et al. Multicenter double-blind study. J Clin Oncol. 2014;32:2039-2049.
randomized phase II: FOLFOX + ziv-aflibercept/placebo for patients (pts) 43. Yarden Y, Pines G. The ERBB network: at last, cancer therapy meets
with chemo-naive metastatic esophagogastric adenocarcinoma systems biology. Nat Rev Cancer. 2012;12:553-563.
(MEGA). J Clin Oncol. 2016;34 (suppl 4S; abstr 4). 44. Holohan C, Van Schaeybroeck S, Longley DB, et al. Cancer
29. Kang Y-K, Shah MA, Ohtsu A, et al. A randomized, open-label, multi-
drug resistance: an evolving paradigm. Nat Rev Cancer. 2013;13:
center, adaptive phase 2/3 study of trastuzumab emtansine (T-DM1)
714-726.
versus a taxane (TAX) in patients (pts) with previously treated HER2-
45. Chen M, Li Y, Ming Z, et al. Comparison of HER2 status by fluorescence
positive locally advanced or metastatic gastric/gastroesophageal
in situ hybridisation and immunohistochemistry in gastric cancer.
junction adenocarcinoma (LA/MGC/GEJC). J Clin Oncol. 2016;34
Contemp Oncol (Pozn). 2014;18:95-99.
(suppl 4S; abstr 5).
46. Yoshida H, Yamamoto N, Taniguchi H, et al. Comparison of HER2 status
30. Wilke H, Van Cutsem E, Oh SC, et al. RAINBOW: A global, phase III,
between surgically resected specimens and matched biopsy specimens
randomized, double-blind study of ramucirumab plus paclitaxel versus
of gastric intestinal-type adenocarcinoma. Virchows Arch. 2014;465:
placebo plus paclitaxel in the treatment of metastatic gastroesophageal
junction (GEJ) and gastric adenocarcinoma following disease progres- 145-154.
sion on first-line platinum- and fluoropyrimidine-containing combi- 47. Wang T, Hsieh ET, Henry P, et al. Matched biopsy and resection
nation therapy rainbow IMCL CP12-0922 (I4T-IE-JVBE). J Clin Oncol. specimens of gastric and gastroesophageal adenocarcinoma show high
2014;32 (suppl 3; abstr LBA7). concordance in HER2 status. Hum Pathol. 2014;45:970-975.
31. Fuchs CS, Tomasek J, Yong CJ, et al; REGARD Trial Investigators. 48. van Hagen P, Biermann K, Boers JE, et al. Human epidermal growth
Ramucirumab monotherapy for previously treated advanced gastric or factor receptor 2 overexpression and amplification in endoscopic bi-
gastro-oesophageal junction adenocarcinoma (REGARD): an in- opsies and resection specimens in esophageal and junctional adeno-
ternational, randomised, multicentre, placebo-controlled, phase 3 trial. carcinoma. Dis Esophagus. 2015;28:380-385.
Lancet. 2014;383:31-39. 49. Grillo F, Fassan M, Ceccaroli C, et al. The reliability of endoscopic bi-
32. Lordick F, Kang Y-K, Salman P, et al. Clinical outcome according to tumor opsies in assessing HER2 status in gastric and gastroesophageal junction
HER2 status and EGFR expression in advanced gastric cancer from the cancer: a study comparing biopsies with surgical samples. Transl Oncol.
EXPAND study. J Clin Oncol. 2013;31 (suppl; abstr 4021). 2013;6:10-16.
33. Satoh T, Lee KH, Rha SY, et al. Randomized phase II trial of nimotuzumab 50. Ge X, Wang H, Zeng H, et al. Clinical significance of assessing Her2/neu
plus irinotecan versus irinotecan alone as second-line therapy for expression in gastric cancer with dual tumor tissue paraffin blocks. Hum
patients with advanced gastric cancer. Gastric Cancer. 2015;18: Pathol. 2015;46:850-857.
824-832. 51. Qiu Z, Sun W, Zhou C, et al. HER2 expression variability between primary
34. Cunningham D, Tebbutt NC, Davidenko I, et al. Phase III, randomized, gastric cancers and corresponding lymph node metastases. Hep-
double-blind, multicenter, placebo (P)-controlled trial of rilotumumab atogastroenterology. 2015;62:231-233.
(R) plus epirubicin, cisplatin and capecitabine (ECX) as first-line therapy 52. Kumarasinghe MP, de Boer WB, Khor TS, et al. HER2 status in gastric/
in patients (pts) with advanced MET-positive (pos) gastric or gastro- gastro-oesophageal junctional cancers: should determination of gene
esophageal junction (G/GEJ) cancer: RILOMET-1 study. J Clin Oncol. amplification by SISH use HER2 copy number or HER2: CEP17 ratio?
2015;33 (suppl; abstr 4000).
Pathology. 2014;46:184-187.
35. Kwak EL, LoRusso P, Hamid O, et al. Clinical activity of AMG 337, an oral
53. Kochi M, Fujii M, Masuda S, et al. Differing deregulation of HER2 in
MET kinase inhibitor, in adult patients (pts) with MET-amplified gas-
primary gastric cancer and synchronous related metastatic lymph
troesophageal junction (GEJ), gastric (G), or esophageal (E) cancer. J Clin
nodes. Diagn Pathol. 2013;8:191.
Oncol. 2015;33 (suppl 3; abstr 1).
54. Cho EY, Park K, Do I, et al. Heterogeneity of ERBB2 in gastric carcinomas:
36. Kwak EL, Ahronian LG, Siravegna G, et al. Molecular heterogeneity and
a study of tissue microarray and matched primary and metastatic
receptor coamplification drive resistance to targeted therapy in MET-
carcinomas. Mod Pathol. 2013;26:677-684.
amplified esophagogastric cancer. Cancer Discov. 2015;5:1271-1281.
55. Bozzetti C, Negri FV, Lagrasta CA, et al. Comparison of HER2 status in
37. Smyth EC, Turner NC, Pearson A, et al. Phase II study of AZD4547 in FGFR
amplified tumours: gastroesophageal cancer (GC) cohort pharmaco- primary and paired metastatic sites of gastric carcinoma. Br J Cancer.
dynamic and biomarker results. J Clin Oncol. 2016;34 (suppl 4S; abstr 2011;104:1372-1376.
154). 56. Chen XZ, Zhang WH, Yao WQ, et al. Immunohistochemical HER2 ex-
38. Muro K, Bang Y-J, Shankaran V, et al. Relationship between PD-L1 pression not associated with clinicopathological characteristics of stage I-
expression and clinical outcomes in patients (Pts) with advanced gastric III gastric cancer patients. Hepatogastroenterology. 2014;61:1817-1821.
cancer treated with the anti-PD-1 monoclonal antibody pembrolizumab 57. Geng Y, Chen X, Qiu J, et al. Human epidermal growth factor receptor-2
(Pembro; MK-3475) in KEYNOTE-012. J Clin Oncol. 2015;33 (suppl 3, expression in primary and metastatic gastric cancer. Int J Clin Oncol.
abstr 3). 2014;19:303-311.
39. Biankin AV, Piantadosi S, Hollingsworth SJ. Patient-centric trials for 58. Geppert CI, Rummele P, Sarbia M, et al. Multi-colour FISH in oeso-
therapeutic development in precision oncology. Nature. 2015;526: phageal adenocarcinoma-predictors of prognosis independent of stage
361-370. and grade. Br J Cancer. 2014;110:2985-2995.

110 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


GASTRIC ADENOCARCINOMA

59. Aizawa M, Nagatsuma AK, Kitada K, et al. Evaluation of HER2-based 63. Janjigian YY, Werner D, Pauligk C, et al. Prognosis of metastatic gastric and
biology in 1,006 cases of gastric cancer in a Japanese population. Gastric gastroesophageal junction cancer by HER2 status: a European and USA
Cancer. 2014;17:34-42. International collaborative analysis. Ann Oncol. 2012;23:2656-2662.
60. Gomez-Martin C, Plaza JC, Pazo-Cid R, et al. Level of HER2 gene am- 64. Ajani JA, Bentrem DJ, Besh S, et al; National Comprehensive Cancer
plification predicts response and overall survival in HER2-positive ad- Network. Gastric cancer, version 2.2013: featured updates to the NCCN
vanced gastric cancer treated with trastuzumab. J Clin Oncol. 2013;31: Guidelines. J Natl Compr Canc Netw. 2013;11:531-546.
4445-4452. 65. Ajani JA, DAmico TA, Almhanna K, et al; National Comprehensive
61. Okines AF, Thompson LC, Cunningham D, et al. Effect of HER2 on Cancer Network. Esophageal and esophagogastric junction cancers,
prognosis and benefit from peri-operative chemotherapy in early version 1.2015. J Natl Compr Canc Netw. 2015;13:194-227.
oesophago-gastric adenocarcinoma in the MAGIC trial. Ann Oncol. 66. Kang YK, Shah MA, Ohtsu A, et al. A randomized, open-label, multi-
2013;24:1253-1261. center, adaptive phase 2/3 study of trastuzumab emtasine (T-DM1)
62. Yoon HH, Shi Q, Sukov WR, et al. Adverse prognostic impact of versus a taxane (TAX) in patients with previously treated Her2 positive
intratumor heterogeneous HER2 gene amplification in patients with locally advanced or metastatic gastric/gastroesophageal junction ad-
esophageal adenocarcinoma. J Clin Oncol. 2012;30:3932-3938. enocarcinoma. J Clin Oncol. 2016;34 (suppl 4S; abstr 5).

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 111


GASTROINTESTINAL (NONCOLORECTAL) CANCER

Localized Pancreatic Cancer:


Multidisciplinary Management
With Incorporation of ASCO
Guidelines

CHAIR
E. Gabriela Chiorean, MD
Fred Hutchinson Cancer Research Center
Seattle, WA

SPEAKERS
Diane M. Simeone, MD
University of Michigan
Ann Arbor, MI

Joseph M. Herman, MD, MSc


Johns Hopkins University School of Medicine
Baltimore, MD
MULTIDISCIPLINARY TREATMENT IN PANCREATIC CANCER

Localized Pancreatic Cancer: Multidisciplinary Management


Andrew L. Coveler, MD, Joseph M. Herman, MD, MSc, Diane M. Simeone, MD, and E. Gabriela Chiorean, MD

OVERVIEW

Pancreatic cancer is an aggressive cancer that continues to have single-digit 5-year mortality rates despite advancements in
the field. Surgery remains the only curative treatment; however, most patients present with late-stage disease deemed
unresectable, either due to extensive local vascular involvement or the presence of distant metastasis. Resection guidelines
that include a borderline resectable group, as well as advancements in neoadjuvant chemotherapy and radiation that
improve resectability of locally advanced disease, may improve outcomes for patients with more invasive disease. Multi-
agent chemotherapy regimens fluorouracil, leucovorin, irinotecan, and oxaliplatin (FOLFIRINOX) and nab-paclitaxel with
gemcitabine improved response rates and survival in metastatic pancreatic cancer and are now being used in earlier
stages for patients with localized potentially resectable and unresectable disease, with goals of downstaging tumors to allow
margin-negative resection and reducing systemic recurrence. Chemoradiotherapy, although still controversial for both
resectable and unresectable pancreatic cancer, is being used in the context of contemporary chemotherapy backbone
regimens, and novel radiation techniques such as stereotactic body frame radiation therapy (SBRT) are studied on the premise
of maintaining or improving efficacy and reducing treatment duration. Patient selection for optimal treatment designation is
currently provided by multidisciplinary tumor boards, but biomarker discovery, in blood, tumors, or through novel imaging, is
an area of intense research. Results to date suggest that some patients with unresectable disease at the outset have survival
rates as good as those with initially resectable disease if able to undergo surgical resection. Long-term follow-up and improved
clinical trials options are needed to determine optimal treatment modalities for patients with localized pancreatic cancer.

A pproximately 277,000 new cases of pancreatic cancer


are diagnosed each year in the world,1 among which
approximately 49,000 occur in both the United States2 and
Cancer Network (NCCN) guidelines recommend surgery fol-
lowed by adjuvant chemotherapy with gemcitabine or fluo-
rouracil (5-FU) as the current standard of care.4 Despite
Europe.3 Localized disease accounts for approximately 50% perioperative mortality rates of less than 2% in experienced
of newly diagnosed patients, with the largest fraction, 35%, centers, patients undergoing surgical resection followed by
being locally advanced, unresectable pancreatic cancer adjuvant chemotherapy (gemcitabine or 5-FU) or chemo-
(LAUPC) and 10% to 15% resectable or borderline resectable therapy plus chemoradiotherapy experience survival rates of
pancreatic cancer (BRPC). The goals of care for patients with 24 months and 5-year overall survival (OS) of 20% on average,
localized pancreatic cancer are increasing the likelihood of unchanged in the past few decades, mostly due to high rates
margin-negative resection, preventing metastatic spread, of systemic relapse.58 The EORTC 40013 randomized phase
converting unresectable into potentially resectable disease, II trial compared adjuvant gemcitabine (four cycles) with
and overall improving patients quality of life. In this update gemcitabine (two cycles) plus gemcitabine-based radio-
on multidisciplinary treatment of localized pancreatic can- therapy and showed similar OS rates (24 months), but local
cer, we highlight standard and novel approaches, overview relapse was improved in the chemoradiotherapy arm (15%
ongoing controversies, and emphasize current clinical trials. vs. 31%).9 Because 30% of patients with pancreatic cancer
experience relapse only locally after surgery,5 the potential
INTEGRATING MULTIMODALITY THERAPY IN benefit of adjuvant chemoradiotherapy needs to be addressed
THE TREATMENT OF LOCALIZED AND LOCALLY in a larger randomized trial. With many historical trials highly
ADVANCED PANCREATIC CANCER criticized due to suboptimal chemoradiotherapy treatment
Resectable Pancreatic Cancer delivery (e.g., GITSG, EORTC, and ESPAC-1),6,10,11 the phase III
Localized resectable pancreatic cancer represents less than study RTOG-0848 (NCT01013649) in the United States is
15% of all new diagnoses, and the National Comprehensive currently evaluating the role of adjuvant chemoradiotherapy

From the Division of Hematology-Oncology, University of Washington, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Radiation Oncology and Molecular
Radiation Sciences, Johns Hopkins University, Baltimore, MD; University of Michigan, Ann Arbor, MI.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: E. Gabriela Chiorean, MD, Fred Hutchinson Cancer Research Center, 825 Eastlake Ave. E, G4-833, Seattle, WA 98109-6248; email: gchiorea@uw.edu.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e217


COVELER ET AL

when added after 6 months of gemcitabine compared with assumption of several advantages: prompt use of systemic
gemcitabine alone (6 months). Until results from RTOG-0848 therapy to treat occult micrometastases, allowing a period of
more clearly define the benefit of adjuvant chemoradiotherapy time to observe the tumor biology during chemotherapy
in this patient population, in clinical practice, adjuvant exposure and avoiding unnecessary resection for rapidly
chemoradiotherapy is most commonly reserved for pa- metastasizing tumors, improving the likelihood of margin-
tients with margin-positive (R1 or R2) resection and/or negative resection, and improved chemotherapy delivery
lymph nodepositive disease.12,13 Intensifying systemic compared with postresection, when complications may
chemotherapy after pancreatic cancer resection is being delay or worsen chemotherapy tolerability. In a few in-
addressed in the adjuvant APACT study (NCT01964430), stitutional prospective studies of neoadjuvant chemother-
using nab-paclitaxel with gemcitabine versus gemcitabine apy (gemcitabine/cisplatin and gemcitabine/oxaliplatin)
alone, and the ACCORD 24 study (NCT01526135) with with or without chemoradiotherapy in radiographic re-
modified FOLFIRINOX (mFOLFIRINOX) versus gemcitabine. sectable disease, approximately 70% of patients underwent
None of these latter studies incorporates chemoradiotherapy surgical resection, and OS reached 27 to 34 months for
in the adjuvant regimen. resected patients, possibly due to patient selection.1517
Data in genetically engineered mouse models of pancre- However, most patients in these studies still experienced
atic cancer suggest metastatic spread precedes local tumor relapsed with metastatic disease. It is therefore not clear
formation.14 Combined with evidence that there is a high that neoadjuvant therapy with standard chemotherapy
rate of disease recurrence following resection of stage I/II regimens confers higher OS compared with adjuvant ap-
disease suggests that even in the absence of radiologic proaches, and with no randomized studies to date, the NCCN
evidence of metastasis, more effective systemic chemo- guidelines do not yet endorse neoadjuvant therapy for
therapy should confer an increased likelihood of cure after patients with clearly resectable disease on baseline staging
resection by successfully treating micrometastases as early CT scans, outside of clinical trials. For patients who receive
as possible. On this premise, neoadjuvant approaches have neoadjuvant therapy followed by resection, further adjuvant
been tested in resectable pancreatic cancer with the therapy is recommended. This approach is being adopted by
the recently opened SWOG S1505 randomized phase II
study (NCT02243007), which compares mFOLFIRINOX with
nab-paclitaxel with gemcitabine for 3 months before and
KEY POINTS 3 months after surgery. A recent report suggests that a
subset of patients treated with neoadjuvant therapy who
Neoadjuvant multimodality therapy should be
considered for patients with borderline resectable and
undergo surgical resection and are found to have low lymph
locally advanced unresectable pancreatic cancer. node ratio (number of positive lymph nodes/number total
Induction chemotherapy for at least 2 to 3 months lymph nodes resected , 0.15) may have better survival
selects patients without rapid disease metastasis who outcomes when also treated with adjuvant chemotherapy
may benefit most from the addition of radiotherapy. (OS: 72 vs. 33 months; p = .008).18 In the absence of definitive
Ongoing clinical trials will help define the role of guidelines, clinical criteria for considering neoadjuvant
adjuvant radiotherapy for resectable patients therapy in resectable pancreatic cancer are: large primary
(RTOG0848), as well as for locally advanced tumors, high carbohydrate antigen 19-9 (CA19-9) levels
unresectable patients (RTOG1201). (. 1,000 U/mL), peripancreatic lymph node involvement by
Select patients with initially localized but unresectable cross-sectional imaging or endoscopic ultrasound, or equiv-
disease may undergo potentially curable surgical
ocal radiologic features.19 Randomized phase II/III studies are
resection after neoadjuvant therapy and have outcomes
comparable to patients with initially resectable disease.
ongoing in resectable pancreatic cancer that may help clarify
Radiologic RECIST response does not adequately predict the role of neoadjuvant versus adjuvant therapies: NEOPAC
resectability for patients with borderline resectable or (NCT01314027) compares adjuvant gemcitabine versus
locally advanced unresectable pancreatic cancer. neoadjuvant gemcitabine/oxaliplatin followed by adjuvant
Pathologic complete or near-complete response (often gemcitabine, NEPAFOX (NCT02172976) compares adjuvant
defined as < 5% viable cells) to neoadjuvant therapy may gemcitabine versus neoadjuvant/adjuvant FOLFIRINOX,
predict increased survival. and NEONAX (NCT02047513) compares adjuvant versus
It is currently unknown whether borderline resectable neoadjuvant/adjuvant nab-paclitaxel with gemcitabine.
or locally advanced unresectable patients undergoing Defining a more effective neoadjuvant or adjuvant regi-
induction (neoadjuvant) multi-agent chemotherapy will men would provide great benefit in improving survival
derive additional benefit with the addition of
among resectable patients, in whom the therapy is geared
radiotherapy.
Novel combinations of systemic treatment and
toward targeting micrometastatic disease.
radiotherapy modalities such as SBRT and IMRT may
provide superior resectability and local control.
Blood and tissue biomarkers may assist in determining Borderline Resectable Pancreatic Cancer
optimal neoadjuvant strategies. Patients with borderline-resectable pancreatic cancer (BRPC)
have localized disease, but less likely to be resected with

e218 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


MULTIDISCIPLINARY TREATMENT IN PANCREATIC CANCER

negative margins due to proximity or direct involvement of patients with BRPC and 80% to 90% R0 resections among
venous and/or arterial vascular structures (# 180).20 In this those resected.30-40 Patients undergoing surgical resection
setting, although no particular chemotherapy with or without had median progression-free survival (PFS) rates of 16 to
a chemoradiotherapy regimen can be considered superior, the 23 months and median OS rates of 24 to 30 months, similar to
NCCN guidelines do not recommend up-front surgery in this historical results for patients with up-front resectable disease
category of patients when there is a high likelihood of positive (Table 1); in contrast, for patients whose disease remained
margins and instead recommend neoadjuvant therapy, unresectable after neoadjuvant treatment, PFS and OS rates
preferably at a high-volume center. Consensus opinions for were 9 to 10 and 12 to 15 months, respectively.30,32 Never-
BRPC have recommended neoadjuvant multimodality treat- theless, many of the retrospective studies included both pa-
ment mostly based on historical retrospective studies, which tients with localized resectable and BRPC/LAUPC, making it
showed increased probability of response (10%30%), re- difficult to identify distinct results for individual subsets of
sectability (30%60%), and R0 resection (80%90%) with this patients.
approach. 21-26 It is well described that R0 resections The prospective Alliance A021101 study (NCT01821612)
are associated with improved OS rates (1823 months) used neoadjuvant mFOLFIRINOX (four cycles) followed by
when compared with R1 (1415 months) or R2 resections capecitabine-based chemoradiotherapy for patients with
(1011 months).27-29 Recent retrospective studies with con- BRPC.41 Resectable patients underwent surgery followed by
temporary therapies FOLFIRINOX or nab-paclitaxel with two cycles of adjuvant gemcitabine. Fifteen of the 22 treated
gemcitabine with or without chemoradiotherapy, often in- patients (68%) underwent surgical resection, among which
cluding both patients BRPC or LAUPC, note encouraging results 93% were R0 resections. The pathologic complete response
with both regimens: resectability rates of 60% to 80% for (pCR) and near pCR (, 5% residual tumor cells) were 9% and

TABLE 1. Multimodality Clinical Trials for BRPC and LAUPC With Emphasis on FOLFIRINOX and Nab-Paclitaxel/
Gemcitabine
Study No. of BRPC/LAUPC Treatment Resected (%) OS (Overall) Months OS (Resected) Months
26
Katz et al 129/0 Gem-platinum + CRT 66 22 33
Hosein et al30 14/4 FOLFIRINOX 6 CRT 75 (BRPC) Not reported Not reported
43 (LAUPC)
Kim et al25 39/6 Gem-oxaliplatin 6 CRT 63 18.4 (BRPC) 25.4
9.4 (LAUPC)
Faris et al51 0/22 FOLFIRINOX + CRT 23 3 year, 7% Not reached
Boone et al 31
12/13 FOLFIRINOX 6 SBRT 64 (BRPC) Not reported Not reported
20 (LAUPC)
Christians36 18/0 FOLFIRINOX 6 CRT 67 Not reported 22 months, 58%
Rose et al24 64/0 Gem-docetaxel 6 CRT 48 23.6 22 months, 81%
Paniccia et al 37
18/0 FOLFIRINOX 6 CRT 94 25 Not reported
Katz et al41 23/0 FOLFIRINOX + CRT 68 18 months, 50% Not reported
Blazer et al38 18/25 FOLFIRINOX 61 (BRPC) 21.2 Not reached
44 (LAUPC)
Khushman et al50 11/40 FOLFIRINOX 6 CRT 22 35.4 3-year 67%
Ferrone et al39 14/33 FOLFIRINOX 6 CRT 85 34 Not reached
Marthey et al 52
0/77 FOLFIRINOX 6 CRT 36 22 months, 77% 24.9 months, not reached
Nitsche et al53 0/14 FOLFIRINOX 29 1 year, 64% 31 months, not reached
Nanda et al40 15/29 FOLFIRINOX + CRT 41 1 year, 66% 18.6 months, not reached
Sadot et al55 0/101 FOLFIRINOX 6 CRT 30 26, if no PD on chemo Not reached
Idrees et al 32
58/28 FOLFIRINOX 6 CRT 84 27.4 Not reached
nab-P/Gem 6 CRT
Portales et al33 16/19 FOLFIRINOX 6 CRT 63 (BRPC) 24 53
37 (LAUPC)
Kim et al34 26/0 FOLFIRINOX 77 PFS 22.5 months Not reported
Peterson et al35 14/6 nab-P/Gem 25 Not reported Not reported
Dean et al 54
0/42 nab-P/Gem 6 CRT 19 Not reported 27.5
Abbreviations: BRPC, borderline resectable pancreatic cancer; LAUPC, locally advanced unresectable pancreatic cancer; OS, overall survival; CRT, chemoradiotherapy; SBRT,
stereotactic body radiotherapy; PD, progressive disease; chemo, chemotherapy; PFS, progression-free survival; nab-P, nab-paclitaxel; Gem, gemcitabine.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e219


COVELER ET AL

47%, respectively, and the RECIST response after pre- Similar to the efforts of intensifying multimodality therapy
operative therapy was 28% (two CR/four PR). Overall sur- in localized and BRPC, both FOLFIRINOX and nab-paclitaxel
vival at 18 months was 50%. Based on the encouraging with gemcitabine, with the addition of chemoradiotherapy
resectability results, the prospective randomized phase II for select patients, have been extensively adopted for pa-
Alliance A021501 study in BRPC using mFOLFIRINOX with or tients with LAUPC, to increase the chance of downstaging
without hypofractionated radiotherapy, either SBRT or and the potential for curative resection. Several institutional
hypofractionated image-guided radiotherapy (HIGRT), is to and multi-institutional studies with FOLFIRINOX or nab-
begin shortly. Some reports indicate that near-complete paclitaxel with gemcitabine reported radiologic response
pathologic complete response (near-pCR, with , 5% re- rates of 20% to 40%, resectability rates of 20% to 44%, with
sidual viable cells) or pCR to neoadjuvant therapy is asso- R0 resection rates of 40% to 80% among resected patients
ciated with long-term survival for resected patients (OS: (Table 1).30-33,35,38-40,50-55 Notably, patients whose disease
53 months or longer)32,42,43; however, analysis of larger becomes resectable after induction/neoadjuvant chemo-
cohorts of patients will be needed to verify this observation. therapy with or without chemoradiotherapy have an en-
Although it has been assumed that multi-agent chemo- couraging median OS of approximately 30 months and
therapy will confer high response rates, radiologic RECIST 2-year OS of 60% to 70%.38,39,49,50,54,55 A Massachusetts
response after 2 to 3 months of neoadjuvant chemotherapy General Hospital retrospective report compared patients
for localized disease has not consistently shown meaningful with initially resectable pancreatic cancer who underwent
downsizing.26,41 At this time, lack of downstaging after neo- surgery (87 patients) with patients who had initially unre-
adjuvant chemotherapy with or without chemoradiotherapy sectable disease with either BRPC or LAUPC and who were
for patients with BRPC should not deter exploration and treated with neoadjuvant FOLFIRINOX with or without
attempt at resection in the absence of local or metastatic chemoradiotherapy followed by surgery with or without
progression or other clinical deterioration factors in ap- intraoperative radiation therapy if deemed resectable
propriate surgical candidates. (40 patients), and the authors noted that R0 resections were
86% versus 92%, and OS rates were longer in the initially
unresectable group (2-year OS: 5% vs. 70%; p = .008).39
Locally Advanced Unresectable Pancreatic Cancer As is the case with retrospective data, most of the in-
It has been observed in an autopsy series that up to one-third stitutional studies were heterogenous and included both
of patients with pancreatic cancer die due to complications patients with BRPC and LAUPC. Prior to the use of novel
from local progression and not metastatic disease, suggesting multi-agent chemotherapy regimens, a meta-analysis of
that more aggressive local therapy may be appropriate in a 111 studies of 4,394 patients with resectable or BRPC/LAUPC
subset of patients.44 Locally advanced unresectable pancre- treated with neoadjuvant chemotherapy (with or without
atic cancer has traditionally been treated with both che- chemoradiotherapy) noted overall responses of 34%, re-
motherapy and chemoradiotherapy, in various sequencing section rates of 74% in initially resectable disease, and 33%
approaches, with OS rates of 9 to 13 months and with some in BRPC/LAUPC, among which R0 resection rates were ap-
studies (ECOG 4201: gemcitabine-based chemoradiotherapy proximately 80%.22 Interestingly, OS after resection was
followed by gemcitabine versus gemcitabine alone),45 but not similar in both the initially resectable and unresectable
others (FFCD-SFRO: 5-FU/cisplatin-based chemoradiotherapy groups (23.3 months [range 1254 months] versus
followed by gemcitabine vs. gemcitabine alone),46 showing 20.5 months [range 962 months]). Nevertheless, patients
OS benefit with the addition of up-front chemoradiotherapy with disease deemed unresectable after neoadjuvant
(ECOG 4201: OS 11.1 vs. 9.2 months, p = .017 in favor of the therapy had poor outcomes, with OS of 8.4 months (range
chemoradiotherapy arm, and FFCD-SFRO: OS of 13 vs. 614 months) among those initially resectable and
8.6 months, p = .03 in favor of the chemotherapy arm). In a 10.2 months (range 621 months) for those with initial
meta-analysis of GERCOR studies, Huguet et al47 noted that BRPC/LAUPC. A more recent meta-analysis of FOLFIRINOX-
induction chemotherapy for 3 to 4 months may select patients based neoadjuvant therapy (with or without chemoradiotherapy)
without rapid metastatic progression who may be the most in BRPC and LAUPC among 253 patients across 13 studies noted
likely to benefit from additional chemoradiotherapy compared an overall resection rate of 43% (of which 85% were R0 re-
with continuing chemotherapy alone. Based on these data, the sections) in all patients and 26% among those with LAUPC, but
randomized phase III LAP-07 trial used 4 months of induction long-term outcomes are not available.56
chemotherapy with gemcitabine with or without erlotinib Compared with historical chemotherapy regimens that
and, if no progression, randomly assigned patients to con- rendered approximately 20% to 30% of the patients with
tinuing chemotherapy (for an additional 2 months) versus locally advanced and borderline pancreatic cancers resect-
capecitabine-based chemoradiotherapy. The OS rates for able, but with high recurrence rates and OS rates less than
patients able to undergo the randomization after induction 24 months,22 preliminary evidence from new retrospective
chemotherapy were similar: 16.7 versus 15.3 months (p = .83) and small prospective studies using the highly active regi-
in the chemotherapy arm compared with the chemotherapy mens FOLFIRINOX and nab-paclitaxel with gemcitabine
plus chemoradiotherapy arm.48 Nevertheless, local progression seem to confer 10% to 20% higher resection rates and more
rates were improved with CRT, 34% vs. 65% (p , .0001).49 durable PFS and OS outcomes.

e220 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


MULTIDISCIPLINARY TREATMENT IN PANCREATIC CANCER

In the context of the novel multi-agent chemotherapy benefit of incorporating an immunotherapy to standard
backbones, the role of adding chemoradiotherapy has not therapies to reduce pancreatic cancer recurrence.60 The GVAX
yet been defined. DPC4/SMAD4 has been identified as a pancreas vaccine, composed of granulocyte macrophage
putative biomarker predictive of local progression when colony-stimulating factorsecreting pancreatic cancer cell
intact or metastatic dissemination when mutated.44 The lines, is currently being studied in combination with SBRT and
RTOG1201 phase II randomized study (NCT01921751) will FOLFIRINOX for patients with resected pancreatic cancer
prospectively test whether the DPC4 gene status identifies (NCT01595321). Given potential synergism, neoadjuvant and
patients with LAUPC more likely to benefit from chemo- adjuvant GVAX will be studied with and without immune
radiotherapy. In this study, 346 patients will be randomly checkpoint inhibitors and chemoradiotherapy for patients
assigned to nab-paclitaxel with gemcitabine alone versus with resectable disease (NCT02451982), and in LAUPC, GVAX
nab-paclitaxel with gemcitabine plus standard-dose 50 Gy with an immune checkpoint inhibitor will be added to SBRT
of capecitabine-based radiotherapy (three-dimensional (NCT02648282). Therapies targeting the tumor stroma, in-
conformal or intensity modulated radiotherapy [IMRT]) cluding the immune-suppressive compartment, are being
versus nab-paclitaxel with gemcitabine plus high-dose actively investigated for resectable and unresectable localized
60 Gy of capecitabine-based IMRT. In Europe, CONKO-007 pancreatic cancer. For example, tumor-associated macro-
(NCT01827553) will assess induction FOLFIRINOX or gem- phages (TAM)targeted therapies such as the CD40 agonist
citabine followed by gemcitabine-based radiotherapy ver- RO7009789 alone and with nab-paclitaxel with gemcitabine
sus continuing chemotherapy, and the SCALOP-2 study are being studied as neoadjuvant/adjuvant therapy in re-
(NCT02024009) will use nab-paclitaxel with gemcitabine sectable pancreatic cancer (NCT02588443). PF-04136309, a
(three cycles) followed by capecitabine-based radiotherapy novel chemokine (C-C motif) receptor 2 inhibitor capable of
with or without the addition of nelfinavir, an HIV protease depleting inflammatory immunosuppressive TAM in combi-
inhibitor with radiosensitizing activity, versus nab-paclitaxel nation with FOLFIRINOX, showed encouraging preliminary
with gemcitabine alone (six cycles). LAPACT (NCT02301143) is results, with 52% radiologic response rates among patients
an international randomized phase II study that will evaluate with BRPC/LAUPC,61 and further studies are anticipated.
induction nab-paclitaxel with gemcitabine for six cycles Stroma-depleting pegylated recombinant human hyaluroni-
and, if no progression, allow investigator choice of surgical dase, which targets tumor hyaluronan to potentially improve
resection, chemoradiotherapy, or continued nab-paclitaxel chemotherapy delivery, is being tested in a neoadjuvant trial
with gemcitabine. with nab-paclitaxel with gemcitabine for patients with BRPC
Choosing the best radiosensitizing agent has been the subject (NCT02487277). Several other agents, including PARP in-
of many investigations. Gemcitabine has been found to be as hibitors, are being studied with the goal of improved radio-
effective but better tolerated than 5-FU.57,58 More recently, the sensitization by preventing DNA repair (NCT01908478) or
randomized phase II SCALOP study using induction gemcitabine/ among patients with BRCA mutations who may be more
capecitabine followed by capecitabine-based or gemcitabine- therapeutically responsive to this class of inhibitors.62-65 Two
based radiotherapy found capecitabine to be more effective recent studies of large numbers of pancreatic cancers dem-
than gemcitabine in prolonging OS (15.2 vs. 13.4 months; onstrating novel molecular subtypes, including an immune
p = .012) and improving tolerability.59 subtype, may help us direct more personalized, effective
At this time, the NCCN guidelines recommend multi-agent regimens to patients most likely to benefit.66,67
chemotherapy (at least 23 months and up to 68 months)
with the possibility of adding chemoradiotherapy for select LOCALIZED PANCREATIC CANCER: HOW DOES
patients with LAPC who do not progress systemically after a THE RADIATION ONCOLOGIST CONTRIBUTE?
course of induction chemotherapy. The use of up-front What Is SBRT?
chemoradiotherapy should be reserved for patients pre- Radiation therapy has historically been delivered to pa-
senting with poorly controlled pain or local invasion with tients with pancreatic cancer to improve local control,
bleeding (NCCN guidelines). delay disease progression, and ameliorate locally obstructive
symptoms such as pain. SBRT is a novel technique that allows
precise delivery of radiation to the tumor while minimizing
Novel Therapies in Pancreatic Cancer dose to surrounding normal structures. Recent advances in
Several studies have incorporated immunotherapeutics in image guidance, respiratory motion management, and ra-
early-stage pancreatic cancer to prevent metastasis and im- diation technology over the past few decades allow for
prove survival. IMPRESS (NCT01072981) was a phase III ran- millimeter accuracy that is necessary for this emerging
domized study among 722 patients with resected pancreatic modality. Due to the high doses of radiation delivered in
cancer treated with gemcitabine and fluoropyrimidine-based such a short amount of time, SBRT requires close supervi-
radiotherapy with and without algenpantucel-L, a hyperacute sion, and advanced measures have been acquired to mini-
vaccine of two allogenic pancreatic cancer cell lines transduced mize treatment-related toxicity. Image guidance throughout
with the a-galactosyl transferase gene. Although results are the process of SBRT planning and delivery is achieved with
eagerly awaited (median OS: at least 28.5 in both arms gold fiducial markers (or fiducials) that are placed in or
combined), this study will be the first to show the potential near the pancreatic tumor under endoscopic guidance.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e221


COVELER ET AL

A four-dimensional CT simulation scan can be used to de- SBRT (2530 Gy) at Moffitt Cancer Center was published
termine which patients require management of respiratory in 2013.75 Most patients received induction gemcitabine/
motion to safely achieve an ablative dose to the tumor. docetaxel/capecitabine (66%) or gemcitabine alone (25%),
Personalization of treatment plans with dose modification whereas only 5% received induction FOLFIRINOX. Among the
based on the location of the tumor to critical structures such patients with BRPC, 56% underwent surgery, with 97%
as the stomach or duodenum may be used to reduce ra- achieving a margin-negative (R0) resection and 9% pCR.
diotherapy exposure to those tissues. Radiotherapy quality Median OS among the patients who underwent margin-
assurance is also used to maintain consistent setup each day negative resection was significantly higher than for unre-
such that SBRT can be safely and accurately delivered. sected patients (19.3 vs. 12.3 months; p = .03). Furthermore,
this approach was well tolerated with no acute grade 3 or
What Are the Potential Advantages of SBRT worse toxicity reported and only 5.3% late grade 3 or worse
Compared With Standard Chemoradiotherapy? toxicity. More recently, this single-institution series was
SBRT has become a standard treatment modality in several updated to include a total of 159 patients (110 with BRPC
malignancies such as lung, liver, brain, spine, and prostate and 49 with LAPC).76 Among the patients with BRPC, 51%
cancers, but it has not yet been established as such in the underwent surgery with 97% achieving a R0 resection and
management of pancreatic cancer. Nevertheless, SBRT has 7% pCR. Median OS in the patients with resected BRPC was
been increasingly adopted in institutions worldwide for 34 months. Acute grade 3 toxicity or worse was 2%, and late
patients with pancreatic cancer due to several advantages: grade 3 toxicity or worse was 5%. The Johns Hopkins Uni-
(1) SBRT can be delivered as a hypofractionated regimen versity School of Medicine reported on their study among
over 3 to 5 days (with dose ranging from 2540 Gy) in 88 patients (14 with BRPC and 74 with LAPC) who received
comparison with 25 to 30 days with conventional chemo- induction chemotherapy followed by SBRT (2533 Gy). The
radiotherapy (dose range, 5054 Gy), (2) SBRT allows for majority of patients (76%) received gemcitabine-based
optimal local control while limiting the delay of alternative chemotherapy as opposed to 5-FU-based (specifically,
therapies such as systemic full-dose chemotherapy or sur- FOLFIRINOX) chemotherapy. Of the 19 patients (22%) who
gical resection, (3) SBRT results in minimal acute side effects underwent surgery following SBRT, 84% had a R0 resection,
and has been shown to improve pain while preserving and 16% of patients had pCR.77 Median OS of resected
quality of life,68 and (4) the radiobiology of SBRT along with patients was 20.2 versus 12.3 months in unresected patients
the ability to escalate the dose to at least 50 Gy at the tumor (p = .07). Of note, only 11% of resected patients had BRPC.
vessel interface may increase the likelihood of a margin-
negative resection. Therefore, patients with pancreatic
cancer, especially those with localized disease, have an Role of SBRT for Locally Advanced, Unresectable
increased likelihood of achieving aggressive trimodality Pancreatic Cancer
therapy with a hope for prolonged survival. Because SBRT is Results of SBRT among patients with LAUPC are encour-
well tolerated and only involves a week of treatment, pa- aging. Early phase I/II studies using single-fraction SBRT
tients are able to focus their energy on achieving personal (25 Gy in one fraction) demonstrated excellent freedom
treatment goals, whether this includes extending survival by from local progression (FFLP) at 1 year (. 90%) and minimal
resuming chemotherapy, undergoing surgical resection, or acute toxicity, but high rates of late grade 2 to 4 gastroin-
maximizing quality of life by returning to daily activities. testinal toxicity were reported.70,71,78-80 A single-arm phase
II multi-institutional study evaluated whether gemcitabine
with fractionated SBRT (in five fractions of 6.6 Gy, for a total
SBRT in Localized Pancreatic Cancer 33.0 Gy) could achieve reduced late grade 2 to 4 gastroin-
Early studies reporting high rates of late gastrointestinal testinal toxicity compared with a historical cohort of patients
toxicity sparked controversy over pancreatic SBRT69-71; treated with gemcitabine and a single 25 Gy-fraction of
however, the abovementioned methods to prioritize safety SBRT.68 Forty-nine patients with LAUPC received up to three
have emerged, and SBRT is now gaining traction as a fa- doses of gemcitabine (1,000 mg/m2) followed by a 1-week
vorable and highly effective local technique. Recent studies break and SBRT (33.0 Gy in five fractions). Following SBRT,
report acceptable rates of late toxicity in concordance with patients continued gemcitabine until progression or toxicity.
minimal acute toxicity.69,72-74 Thus far, there have been Rates of acute and late toxicities (primary endpoint) grade
more than 800 published cases of pancreatic cancer treated 2 or worse gastritis, fistula, enteritis, or ulcers were 2% and
with SBRT, and the toxicity, survival, and patterns of failure 11%, respectively, and QLQ-C30 global quality-of-life scores
data are promising. remained stable. Patients reported a notable improvement
in pancreatic pain (p , .001) 4 weeks after SBRT on the QLQ-
Role of Neoadjuvant SBRT in Borderline Resectable PAN26 questionnaire. Median OS was 13.9 months (95% CI,
Pancreatic Cancer 10.216.7), and FFLP at 1 year was 78%. Four patients (8%)
A retrospective study included 73 patients with localized with LAUPC at diagnosis underwent R0 resections and had
pancreatic cancer (57 boderline resectable and 16 LAUPC) who node-negative status; a fifth patient who was deemed re-
received induction chemotherapy followed by five-fraction sectable declined surgery.

e222 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


MULTIDISCIPLINARY TREATMENT IN PANCREATIC CANCER

With the consideration that patients originally deemed downstaging or tumor response observed on imaging.26,89
unresectable may become resectable, a shift toward neo- The University of Texas MD Anderson Cancer Center re-
adjuvant therapy in LAUPC has recently emerged. Of the ported on 122 patients with BRPC who were restaged after
49 patients with LAUPC who were treated with induction neoadjuvant therapy, with 85 patients (66%) undergoing
chemotherapy and SBRT at Moffitt Cancer Center, five (10%) surgery despite only one patient (1%) having been down-
were resected with a 100% R0 resection rate.76 Patients with staged to have resectable disease.26 Patients received
LAUPC survived a median of 13.2 months. Johns Hopkins gemcitabine-based induction chemotherapy followed by
reported 15 of 74 (20%) patients with LAUPC who went to conventional chemoradiotherapy. Similarly, Johns Hopkins
surgery with an 80% R0 resection rate and 13% pCR.77 revealed that 29 of 50 patients (58%) with BRPC (25 patients)
Median OS was 18.4 months. and LAPC (four patients) underwent successful surgical re-
section despite no considerable change in vessel involve-
How Can We Improve the Impact of SBRT in Localized ment after neoadjuvant therapy.89 Interestingly, three (15%)
Pancreatic Cancer? patients were noted to have progressive disease on imaging
In an initial pilot cooperative group study in BRPC (Alliance but were successfully resected. The difficulty in evaluating
A021101), FOLFIRINOX-based multimodality therapy was tumor response after neoadjuvant therapy is presumably
shown to be well tolerated, and resectability and preliminary due to the amount of treatment-related fibrosis that
OS were encouraging.41 The follow-up study (Alliance develops following the delivery of therapy, particularly
A021501) will evaluate survival among patients with BRPC chemoradiotherapy.
randomly assigned to neoadjuvant mFOLFIRINOX with or Given the morbidity and mortality associated with a pan-
without hypofractionated radiotherapy, either SBRT or createctomy, careful patient selection is warranted; however,
HIGRT. as shown above, it is important to consider patients who
As we await results from cooperative group studies, it is appear to otherwise be unresectable after therapy. If the
essential to pursue further understanding of the tumor patient is willing, surgery can be attempted, and is recom-
biology and immune response associated with pancreatic mended, in the following circumstances following a course of
cancer. It has been shown that radiotherapy not only induces neoadjuvant therapy of at least 3 months, ideally: (1) absence
cell death through DNA damage, but also promotes anti- of distant progression, (2) stable or improved CA 19-9, and
tumor reactivity in certain solid tumors.81-83 SBRT specifi- (3) good performance status (ECOG # 1). Insight into
cally, with its highly ablative doses over a short course, has predictive and prognostic biomarkers and imaging mo-
been shown to be associated with radiation-induced dalities such as PET may allow us to further guide man-
immunomodulation.84-86 Preclinical data suggest that radia- agement decisions and practice personalized care.
tion increases antigen presentation by altering the immune-
phenotype of tumor infiltrating inflammatory cells, increases CONCLUSION
T-cell infiltration, and induces proinflammatory cytokines that Although the results of ongoing clinical trials are likely to
promote tumor cell death.87,88 reshape clinical practice, improvement in systemic and
chemoradiotherapy modalities and the development and
How Can We Optimally Measure Response to routine use of more predictive blood, tumor, and imaging
Therapy, and Which Patients Should Be Taken to biomarkers for the selection of patients most likely to
Surgery? benefit are areas of high need and intense research. New
The difficulty of selecting patients with localized pancreatic understanding of the genetic complexity and intertumoral
cancer who are candidates for surgery following neo- heterogeneity among patients and the development of
adjuvant therapy lies in the inadequacy of radiographic biomarker-driven clinical trials using novel combinations of
imaging to depict treatment response and vessel involve- promising therapeutic agents are likely to begin to change
ment. Two series have published on patients with BRPC who the landscape to improved outcomes in patients with this
have been successfully resected despite the lack of tumor dreadful disease.

References
1. Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer incidence and mor- 4. Tempero MA, Malafa MP, Behrman SW, et al. Pancreatic adenocarci-
tality worldwide: sources, methods and major patterns in GLOBOCAN noma, version 2.2014: featured updates to the NCCN guidelines. J Natl
2012. Int J Cancer. 2015;136:E359-E386. Compr Canc Netw. 2014;12:1083-1093.
2. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin. 5. Oettle H, Neuhaus P, Hochhaus A, et al. Adjuvant chemotherapy with
2015;65:5-29. gemcitabine and long-term outcomes among patients with resected pan-
3. Bosetti C, Bertuccio P, Malvezzi M, et al. Cancer mortality in Europe, creatic cancer: the CONKO-001 randomized trial. JAMA. 2013;310:1473-1481.
2005-2009, and an overview of trends since 1980. Ann Oncol. 2013;24: 6. Neoptolemos JP, Stocken DD, Friess H, et al; European Study Group
2657-2671. for Pancreatic Cancer. A randomized trial of chemoradiotherapy and

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e223


COVELER ET AL

chemotherapy after resection of pancreatic cancer. N Engl J Med. 2004; 24. Rose JB, Rocha FG, Alseidi A, et al. Extended neoadjuvant chemotherapy
350:1200-1210. for borderline resectable pancreatic cancer demonstrates promising
7. Regine WF, Winter KA, Abrams R, et al. Fluorouracil-based chemo- postoperative outcomes and survival. Ann Surg Oncol. 2014;21:
radiation with either gemcitabine or fluorouracil chemotherapy after 1530-1537.
resection of pancreatic adenocarcinoma: 5-year analysis of the U.S. 25. Kim EJ, Ben-Josef E, Herman JM, et al. A multi-institutional phase 2 study
Intergroup/RTOG 9704 phase III trial. Ann Surg Oncol. 2011;18: of neoadjuvant gemcitabine and oxaliplatin with radiation therapy in
1319-1326. patients with pancreatic cancer. Cancer. 2013;119:2692-2700.
8. Liao WC, Chien KL, Lin YL, et al. Adjuvant treatments for resected 26. Katz MH, Fleming JB, Bhosale P, et al. Response of borderline pancreatic
pancreatic adenocarcinoma: a systematic review and network meta- cancer to neoadjuvant therapy is not reflected by radiographic criteria.
analysis. Lancet Oncol. 2013;14:1095-1103. Cancer. 2012;118:5749-5756.
9. Van Laethem JL, Hammel P, Mornex F, et al. Adjuvant gemcitabine alone 27. Fatima J, Schnelldorfer T, Barton J, et al. Pancreatoduodenectomy for
versus gemcitabine-based chemoradiotherapy after curative resection ductal adenocarcinoma: implications of positive margin on survival.
for pancreatic cancer: a randomized EORTC-40013-22012/FFCD-9203/ Arch Surg. 2010;145:167-172.
GERCOR phase II study. J Clin Oncol. 2010;28:4450-4456. 28. Winter JM, Cameron JL, Campbell KA, et al. 1423 pancreatico-
10. Gastrointestinal Tumor Study Group. Further evidence of effective duodenectomies for pancreatic cancer: A single-institution experience.
adjuvant combined radiation and chemotherapy following curative J Gastrointest Surg. 2006;10:1199-1210, discussion 1210-1211.
resection of pancreatic cancer. Cancer. 1987;59:2006-2010. 29. Konstantinidis IT, Warshaw AL, Allen JN, et al. Pancreatic ductal ade-
11. Klinkenbijl JH, Jeekel J, Sahmoud T, et al. Adjuvant radiotherapy and nocarcinoma: is there a survival difference for R1 resections versus
5-fluorouracil after curative resection of cancer of the pancreas and locally advanced unresectable tumors? What is a true R0 resection?
periampullary region: phase III trial of the EORTC gastrointestinal tract Ann Surg. 2013;257:731-736.
cancer cooperative group. Ann Surg. 1999;230:776-782, discussion 782-784. 30. Hosein PJ, Macintyre J, Kawamura C, et al. A retrospective study of
12. Herman JM, Swartz MJ, Hsu CC, et al. Analysis of fluorouracil-based neoadjuvant FOLFIRINOX in unresectable or borderline-resectable lo-
adjuvant chemotherapy and radiation after pancreaticoduodenectomy cally advanced pancreatic adenocarcinoma. BMC Cancer. 2012;12:199.
for ductal adenocarcinoma of the pancreas: results of a large, pro- 31. Boone BA, Steve J, Krasinskas AM, et al. Outcomes with FOLFIRINOX for
spectively collected database at the Johns Hopkins Hospital. J Clin Oncol. borderline resectable and locally unresectable pancreatic cancer. J Surg
2008;26:3503-3510. Oncol. 2013;108:236-241.
13. You DD, Lee HG, Heo JS, et al. Prognostic factors and adjuvant che- 32. Idrees K, Parikh AA, McLendon Postlewait L, et al. Treatment of bor-
moradiation therapy after pancreaticoduodenectomy for pancreatic derline resectable and locally advanced pancreatic cancer in the era of
adenocarcinoma. J Gastrointest Surg. 2009;13:1699-1706. FOLFIRINOX and gemcitabine plus nab-paclitaxel: a multi-institutional
14. Rhim AD, Mirek ET, Aiello NM, et al. EMT and dissemination precede study. J Clin Oncol. 2016;34 (suppl 4S; abstr 451).
pancreatic tumor formation. Cell. 2012;148:349-361. 33. Portales F, Gagniard B, Thezenas S, et al. Feasibility and impact on
15. Evans DB, Varadhachary GR, Crane CH, et al. Preoperative gemcitabine- resectability of FOLFIRINOX in locally-advanced and borderline pan-
based chemoradiation for patients with resectable adenocarcinoma of creatic cancer. J Clin Oncol. 2016;34 (suppl 4S; abstr 318).
the pancreatic head. J Clin Oncol. 2008;26:3496-3502. 34. Kim SS, Nakakura EK, Wang ZJ, et al. Is neoadjuvant chemoradiation
16. Varadhachary GR, Wolff RA, Crane CH, et al. Preoperative gemcitabine important in borderline resectable pancreatic cancer (BRPC)? Clinical
and cisplatin followed by gemcitabine-based chemoradiation for re- and surgical outcomes associated with preoperative FOLFIRINOX alone
sectable adenocarcinoma of the pancreatic head. J Clin Oncol. 2008;26: in BRPC. J Clin Oncol. 2016;34 (suppl 4S; abstr 351).
3487-3495. 35. Peterson S, Loaiza-Bonilla A, Ben-Josef E, et al. Neoadjuvant nab-
17. OReilly EM, Perelshteyn A, Jarnagin WR, et al. A single-arm, non- paclitaxel and gemcitabine (AG) in borderline resectable (BR) or
randomized phase II trial of neoadjuvant gemcitabine and oxaliplatin in unresectable (UR) locally advanced pancreatic adenocarcinoma
patients with resectable pancreas adenocarcinoma. Ann Surg. 2014; (LAPC) in patients ineligible for FOLFIRINOX. J Clin Oncol. 2016;34
260:142-148. (suppl 4S; abstr 328).
18. Roland CL, Katz MH, Tzeng CW, et al. The addition of postoperative 36. Christians KK, Tsai S, Mahmoud A, et al. Neoadjuvant FOLFIRINOX for
chemotherapy is associated with improved survival in patients with borderline resectable pancreas cancer: a new treatment paradigm?
pancreatic cancer treated with preoperative therapy. Ann Surg Oncol. Oncologist. 2014;19:266-274.
2015;22:1221-1228 (suppl 3). 37. Paniccia A, Edil BH, Schulick RD, et al. Neoadjuvant FOLFIRINOX ap-
19. Heestand GM, Murphy JD, Lowy AM. Approach to patients with pan- plication in borderline resectable pancreatic adenocarcinoma: a ret-
creatic cancer without detectable metastases. J Clin Oncol. 2015;33: rospective cohort study. Medicine (Baltimore). 2014;93:e198.
1770-1778. 38. Blazer M, Wu C, Goldberg RM, et al. Neoadjuvant modified FOLFIRINOX
20. Katz MH, Marsh R, Herman JM, et al. Borderline resectable pancreatic for locally advanced unresectable and borderline resectable adeno-
cancer: need for standardization and methods for optimal clinical trial carcinoma of the pancreas. Ann Surg Oncol. 2015;22:1153-1159.
design. Ann Surg Oncol. 2013;20:2787-2795. 39. Ferrone CR, Marchegiani G, Hong TS, et al. Radiological and surgical
21. Abrams RA, Lowy AM, OReilly EM, et al. Combined modality treatment implications of neoadjuvant treatment with FOLFIRINOX for locally
of resectable and borderline resectable pancreas cancer: expert con- advanced and borderline resectable pancreatic cancer. Ann Surg. 2015;
sensus statement. Ann Surg Oncol. 2009;16:1751-1756. 261:12-17.
22. Gillen S, Schuster T, Meyer Zum Buschenfelde C, et al. Preoperative/ 40. Nanda RH, El-Rayes B, Maithel SK, et al. Neoadjuvant modified
neoadjuvant therapy in pancreatic cancer: a systematic review and FOLFIRINOX and chemoradiation therapy for locally advanced pancreatic
meta-analysis of response and resection percentages. PLoS Med. 2010; cancer improves resectability. J Surg Oncol. 2015;111:1028-1034.
7:e1000267. 41. Katz MHG, Shi Q, Ahmad SA, et al. Preoperative modified FOLFIRINOX
23. Katz MH, Pisters PW, Evans DB, et al. Borderline resectable pancreatic followed by chemoradiation for borderline resectable pancreatic
cancer: the importance of this emerging stage of disease. J Am Coll Surg. cancer: initial results from Alliance trial A021101. J Clin Oncol. 2015;33
2008;206:833-846, discussion 846-848. (suppl; abstr 4008).

e224 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


MULTIDISCIPLINARY TREATMENT IN PANCREATIC CANCER

42. Chatterjee D, Katz MH, Rashid A, et al. Histologic grading of the extent of chemoradiation in locally advanced pancreatic cancer? Int J Radiat
residual carcinoma following neoadjuvant chemoradiation in pancre- Oncol Biol Phys. 2002;52:1293-1302.
atic ductal adenocarcinoma: a predictor for patient outcome. Cancer. 58. Huang J, Robertson JM, Margolis J, et al. Long-term results of full-dose
2012;118:3182-3190. gemcitabine with radiation therapy compared to 5-fluorouracil with
43. Zhao Q, Rashid A, Gong Y, et al. Pathologic complete response to radiation therapy for locally advanced pancreas cancer. Radiother
neoadjuvant therapy in patients with pancreatic ductal adenocarci- Oncol. 2011;99:114-119.
noma is associated with a better prognosis. Ann Diagn Pathol. 2012;16: 59. Mukherjee S, Hurt CN, Bridgewater J, et al. Gemcitabine-based or
29-37. capecitabine-based chemoradiotherapy for locally advanced pancreatic
44. Iacobuzio-Donahue CA, Fu B, Yachida S, et al. DPC4 gene status of the cancer (SCALOP): a multicentre, randomised, phase 2 trial. Lancet
primary carcinoma correlates with patterns of failure in patients with Oncol. 2013;14:317-326.
pancreatic cancer. J Clin Oncol. 2009;27:1806-1813. 60. Coveler AL, Rossi GR, Vahanian NN, et al. Algenpantucel-L immu-
45. Loehrer PJ Sr, Feng Y, Cardenes H, et al. Gemcitabine alone versus notherapy in pancreatic adenocarcinoma. Immunotherapy. 2016;8:
gemcitabine plus radiotherapy in patients with locally advanced pan- 117-125.
creatic cancer: an Eastern Cooperative Oncology Group trial. J Clin 61. Wang-Gillam A, Nyweing TM, Sanford DE, et al. Phase IB study of
Oncol. 2011;29:4105-4112. FOLFIRINOX plus PF-04136309 in patients with borderline resectable
46. Chauffert B, Mornex F, Bonnetain F, et al. Phase III trial comparing and locally advanced pancreatic adenocarcinoma. J Clin Oncol. 2015;33
intensive induction chemoradiotherapy (60 Gy, infusional 5-FU and (suppl 3; abstr 338).
intermittent cisplatin) followed by maintenance gemcitabine with 62. Salo-Mullen EE, OReilly EM, Kelsen DP, et al. Identification of germline
gemcitabine alone for locally advanced unresectable pancreatic cancer. genetic mutations in patients with pancreatic cancer. Cancer. 2015;121:
Definitive results of the 2000-01 FFCD/SFRO study. Ann Oncol. 2008;19: 4382-4388.
1592-1599. 63. Lowery MA, Kelsen DP, Smith SC, et al. Phase II trial of veliparib in
47. Huguet F, Andre T, Hammel P, et al. Impact of chemoradiotherapy after patients with previously treated BRCA or PALB2 mutated pancreas
disease control with chemotherapy in locally advanced pancreatic adenocarcinoma. J Clin Oncol. 2015;33 (suppl 3; abstr 358).
adenocarcinoma in GERCOR phase II and III studies. J Clin Oncol. 2007; 64. Pishvaian MJ, Wang H, Zhuang T, et al. A phase I/II study of ABT-888 in
25:326-331. combination with 5-fluorouracil and oxaliplatin in patients with met-
48. Hammel P, Huguet F, van Laethem JL, et al. Comparison of chemo- astatic pancreatic cancer. J Clin Oncol. 2013;31 (suppl 4; abstr 147).
radiotherapy (CRT) and chemotherapy (CT) in patients with locally 65. OReilly EM, Lowery MA, Segal MF, et al. Phase Ib trial of cisplatin,
advanced pancreatic cancer (LAPC) controlled after 4 months of gemcitabine, and veliparib in patients with known or potential BRCA or
gemcitabine with or without erlotinib: final results of the international PALB2-mutated pancreas adenocarcinoma. J Clin Oncol. 2014;32 (suppl
phase III LAP 07 study. J Clin Oncol. 2013;33:15s (suppl; abstr LBA4003). 5S; abstr 4023).
49. Huguet F, Hammel P, Vernerey D, et al. Impact of chemoradiation on 66. Waddell N, Pajic M, Patch AM, et al; Australian Pancreatic Cancer
local control and time without treatment in patients with locally ad- Genome Initiative. Whole genomes redefine the mutational landscape
vanced pancreatic cancer included in the international phase III LAP 07 of pancreatic cancer. Nature. 2015;518:495-501.
study. J Clin Oncol. 2014;32 (suppl 5S; abstr 4001). 67. Bailey P, Chang DK, Nones K, et al; Australian Pancreatic Cancer Genome
50. Khushman M, Dempsey N, Maldonado JC, et al. Full dose neoadjuvant Initiative. Genomic analyses identify molecular subtypes of pancreatic
FOLFIRINOX is associated with prolonged survival in patients with lo- cancer. Nature. 2016;531:47-52.
cally advanced pancreatic adenocarcinoma. Pancreatology. 2015;15: 68. Herman JM, Chang DT, Goodman KA, et al. Phase 2 multi-institutional
667-673. trial evaluating gemcitabine and stereotactic body radiotherapy for
51. Faris JE, Blaszkowsky LS, McDermott S, et al. FOLFIRINOX in locally patients with locally advanced unresectable pancreatic adenocarci-
advanced pancreatic cancer: the Massachusetts General Hospital noma. Cancer. 2015;121:1128-1137.
Cancer Center experience. Oncologist. 2013;18:543-548. 69. Hoyer M, Roed H, Sengelov L, et al. Phase-II study on stereotactic
52. Marthey L, Sa-Cunha A, Blanc JF, et al. FOLFIRINOX for locally advanced radiotherapy of locally advanced pancreatic carcinoma. Radiother
pancreatic adenocarcinoma: results of an AGEO multicenter pro- Oncol. 2005;76:48-53.
spective observational cohort. Ann Surg Oncol. 2015;22:295-301. 70. Schellenberg D, Goodman KA, Lee F, et al. Gemcitabine chemotherapy
53. Nitsche U, Wenzel P, Siveke JT, et al. Resectability after first-line and single-fraction stereotactic body radiotherapy for locally advanced
FOLFIRINOX in initially unresectable locally advanced pancreatic can- pancreatic cancer. Int J Radiat Oncol Biol Phys. 2008;72:678-686.
cer: a single-center experience. Ann Surg Oncol. 2015;22:1212-1220 71. Schellenberg D, Kim J, Christman-Skieller C, et al. Single-fraction ste-
(suppl 3). reotactic body radiation therapy and sequential gemcitabine for the
54. Dean A, McGrath A, Youd J, Spry N. Nab-paclitaxel plus gemcitabine treatment of locally advanced pancreatic cancer. Int J Radiat Oncol Biol
followed by radiotherapy with concurrent 5-FU in locally advanced Phys. 2011;81:181-188.
unresectable pancreatic cancer. J Clin Oncol. 2016;34 (suppl 4S; abstr 72. Tozzi A, Comito T, Alongi F, et al. SBRT in unresectable advanced
430). pancreatic cancer: preliminary results of a mono-institutional experi-
55. Sadot E, Doussot A, OReilly EM, et al. FOLFIRINOX induction therapy for ence. Radiat Oncol. 2013;8:148.
stage 3 pancreatic adenocarcinoma. Ann Surg Oncol. 2015;22: 73. Mahadevan A, Jain S, Goldstein M, et al. Stereotactic body radiotherapy
3512-3521. and gemcitabine for locally advanced pancreatic cancer. Int J Radiat
56. Petrelli F, Coinu A, Borgonovo K, et al; Gruppo Italiano per lo Studio dei Oncol Biol Phys. 2010;78:735-742.
Carcinomi dellApparato Digerente (GISCAD). FOLFIRINOX-based neo- 74. Timmerman RD, Kavanagh BD, Cho LC, et al. Stereotactic body radiation
adjuvant therapy in borderline resectable or unresectable pancreatic therapy in multiple organ sites. J Clin Oncol. 2007;25:947-952.
cancer: a meta-analytical review of published studies. Pancreas. 2015; 75. Chuong MD, Springett GM, Freilich JM, et al. Stereotactic body radiation
44:515-521. therapy for locally advanced and borderline resectable pancreatic
57. Crane CH, Abbruzzese JL, Evans DB, et al. Is the therapeutic index better cancer is effective and well tolerated. Int J Radiat Oncol Biol Phys. 2013;
with gemcitabine-based chemoradiation than with 5-fluorouracil-based 86:516-522.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e225


COVELER ET AL

76. Mellon EA, Hoffe SE, Springett GM, et al. Long-term outcomes of 83. Gupta A, Probst HC, Vuong V, et al. Radiotherapy promotes tumor-
induction chemotherapy and neoadjuvant stereotactic body radio- specific effector CD8+ T cells via dendritic cell activation. J Immunol.
therapy for borderline resectable and locally advanced pancreatic 2012;189:558-566.
adenocarcinoma. Acta Oncol. 2015;54:979-985. 84. Zeng J, Harris TJ, Lim M, Drake CG, Tran PT. Immune modulation and
77. Moningi S, Dholakia AS, Raman SP, et al. The role of stereotactic body stereotactic radiation: Improving local and abscopal responses. Biomed
radiation therapy for pancreatic cancer: A single-institution experience. Res Int. 2013;2013:658126.
Ann Surg Oncol. 2015;22:2352-2358. 85. Neefjes J, Jongsma ML, Paul P, et al. Towards a systems understanding
78. Koong AC, Le QT, Ho A, et al. Phase I study of stereotactic radiosurgery in of MHC class I and MHC class II antigen presentation. Nat Rev Immunol.
patients with locally advanced pancreatic cancer. Int J Radiat Oncol Biol 2011;11:823-836.
Phys. 2004;58:1017-1021. 86. Finkelstein SE, Timmerman R, McBride WH, et al. The confluence of
79. Koong AC, Christofferson E, Le QT, et al. Phase II study to assess the stereotactic ablative radiotherapy and tumor immunology. Clin Dev
efficacy of conventionally fractionated radiotherapy followed by a Immunol. 2011;2011:439752.
stereotactic radiosurgery boost in patients with locally advanced 87. Sharabi AB, Nirschl CJ, Kochel CM, et al. Stereotactic radiation therapy
pancreatic cancer. Int J Radiat Oncol Biol Phys. 2005;63:320-323. augments antigen-specific PD-1-mediated antitumor immune re-
80. Chang DT, Schellenberg D, Shen J, et al. Stereotactic radiotherapy for sponses via cross-presentation of tumor antigen. Cancer Immunol
unresectable adenocarcinoma of the pancreas. Cancer. 2009;115:665-672. Res. 2015;3:345-355.
81. Reits EA, Hodge JW, Herberts CA, et al. Radiation modulates the peptide 88. Matsumura S, Demaria S. Up-regulation of the pro-inflammatory
repertoire, enhances MHC class I expression, and induces successful chemokine CXCL16 is a common response of tumor cells to ionizing
antitumor immunotherapy. J Exp Med. 2006;203:1259-1271. radiation. Radiat Res. 2010;173:418-425.
82. Gameiro SR, Jammeh ML, Wattenberg MM, et al. Radiation-induced 89. Dholakia AS, Hacker-Prietz A, Wild AT, et al. Resection of borderline
immunogenic modulation of tumor enhances antigen processing and resectable pancreatic cancer after neoadjuvant chemoradiation does
calreticulin exposure, resulting in enhanced T-cell killing. Oncotarget. not depend on improved radiographic appearance of tumor-vessel
2014;5:403-416. relationships. J Radiat Oncol. 2013;2:413-425.

e226 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


GENITOURINARY (NONPROSTATE) CANCER

Elderly Patients With Bladder


Cancer: Perspectives From a
Surgeon, Medical, and Radiation
Oncologist

CHAIR
Supriya Gupta Mohile, MD, MS
University of Rochester Medical Center
Rochester, NY

SPEAKERS
Eila C. Skinner, MD
Stanford University School of Medicine
Stanford, CA

Nicholas D. James, BSc, MBBS, PhD


University of Warwick and Queen Elizabeth Hospital
Coventry, United Kingdom
EILA C. SKINNER

Treatment of Muscle-Invasive Bladder Cancer in Older


Patients
Eila C. Skinner, MD

OVERVIEW

Treatment of muscle-invasive bladder cancer in older patients is challenging. Definitive therapy of localized disease requires
either surgery or radiation therapy, ideally combined with systemic chemotherapy. However, current population data
suggest that less than half of patients older than age 70 are offered such treatments. We will review tools available to assess
the fitness of older patients for surgery, alternatives, and tips for perioperative patient treatment.

B ladder cancer is a disease of older patients, with in-


creasing incidence with age, and rarely seen before age
40. The majority of patients present with noninvasive tu-
mixed reports regarding the advantage of PET/CT over
standard CT with contrast, PET may improve detection of
occult metastases, which might be particularly useful in
mors that have a high recurrence rate but rarely develop older patients in whom the risk of surgery may not be
metastases. These are managed with transurethral resection warranted if cure is unlikely.3
and intravesical therapy, both of which are generally well- Patient evaluation in older patients requires some as-
tolerated in older patients. However, 20% to 30% of patients sessment of fitness for surgery and a relative estimate of
will present with an invasive, high-grade tumor that has a complications related to the operation. There has been
high potential for metastasis. In general, muscle-invasive considerable progress in this regard over the past few years
tumors that are not treated will cause increasingly severe with the development of tools for assessment of older
local symptoms and, in most cases, death from metastatic patients. Ideally, the patient may be evaluated by a trained
disease within 2 years. In recent years, urologists and ra- geriatrician to perform a comprehensive geriatric assess-
diation oncologists have been called on to treat an increasing ment.4 However, such specialists may not be readily avail-
number of patients older than age 75 with this type of able and the full assessment is not practical for the average
aggressive disease. Definitive therapy requires either radical urologic oncology clinic.
cystectomy or radiation therapy, with or without systemic A number of health domains have been shown to con-
chemotherapy. However, current reports indicate that only tribute to overall frailty in older patients. In addition to
65% of patients age 71 to 80 and 35% of patients age 81 to 90 chronological age, these include cognitive function, de-
receive these treatments.1 A multidisciplinary approach is pendence in activities of daily living, nutritional status,
required to identify older patients who should be offered physical fitness, medical comorbidities, social support,
such treatments. mood, and history of falls. Several authors have combined
factors that were predictive of morbidity and mortality from
PATIENT EVALUATION surgery into simplified tests that could be applied in a
Initial evaluation of the patient with high-risk bladder cancer urology clinic with minimal extra training.5-14 Some of these
requires careful evaluation of both the tumor and the pa- assessments focus on medical comorbidities, functional
tient. Tumor evaluation is identical regardless of age, and status, cognition, and laboratory tests such as serum al-
it includes review of prior treatments, complete tumor bumin. These assessments also have been used in retro-
staging, review of pathology slides, and possible repeat spective studies and applied to large databases such as the
resection. Repeat resection with examination under anes- National Surgery Quality Improvement Program.10,11,14
thesia has been shown to improve accuracy of local staging.2 Others use actual tests of physical strength and mobility
A full radiographic staging workup of a muscle-invasive (such as the get up and go test and grip-strength measure),
cancer should include a CT chest/abdomen/pelvis with cognitive testing, and patient-reported signs such as weight
contrast and bone scan, or PET/CT scan. Although there are loss. These measures require a trained clinician to directly

From Stanford University School of Medicine, Stanford, CA.

Disclosures of potential conflicts of interest provided by the author are available with the online article at asco.org/edbook.

Corresponding author: Eila C. Skinner, MD, Stanford University School of Medicine, 300 Pasteur Dr., Suite S-287, Stanford, CA 94305; email: skinnere@stanford.edu.

2016 by American Society of Clinical Oncology.

e228 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


TREATMENT OF MUSCLE-INVASIVE BLADDER CANCER IN OLDER PATIENTS

evaluate the patient prospectively.9,13 Regardless of the 10% of patients with reasonable life expectancy are can-
particular measures used, most of these assessments have didates for either of these approaches.
correlated with surgical outcomes and have improved
predictive power compared with the surgeons intuition
Observation
after seeing the patient seated on the examination table.
The primary advantage of observation is avoiding the po-
Table 1 shows an example of one such tool that is relatively
tential risks and side effects associated with treatment, with
straightforward to administer.9
the implicit hope/expectation that the patient will die of
Patients who score poorly on these tests have been shown
another illness before the bladder cancer causes morbidity.
to have higher complications and higher mortality from
Local symptoms may include bleeding, frequency and urgency,
major abdominal surgery and, even in some cases, in less-
incontinence, bladder pain, outlet obstruction, and upper-
invasive procedures such as transurethral resection of the
tract obstruction. Symptom management may include repeat
prostate or sling.11 The results from these evaluations must
TURBT or fulguration for bleeding, anticholinergics, catheter
be interpreted in light of the severity of patient symptoms
drainage, and stents or percutaneous nephrostomy tube
and the likelihood that a nonoperative approach will be
placement. Unfortunately, once local symptoms become
successful. In addition, the degree to which these factors are
problematic they often are refractory to these treatments
modifiable in a reasonably short time is unclear. However,
and patients may require multiple trips to the emergency
the knowledge about risks of surgery can help patients and
department, hospital admissions, transfusions, and painful
families decide on treatment options.
procedures. It is not uncommon for the 88-year-old patient
who is not thought to be a surgical candidate to end up
requiring a palliative cystectomy a year later when he or she
CHOICE OF TREATMENT has metastatic disease. Thus, a patient who is believed to
Three options should be discussed with every older patient
have a 2-year life expectancy and is likely to survive the
who presents with invasive localized bladder cancer: (1)
surgery is often best treated with definitive therapy with
observation with symptom management alone; (2) radiation
cystectomy or radiation.
therapy, with or without concomitant chemotherapy; and
(3) radical cystectomy, with or without chemotherapy
Partial cystectomy is appealing in this age group because it Radiation Therapy
has so much less morbidity, but it is only effective in rare The advantage of radiation therapy is the avoidance of the
cases of an isolated tumor near the dome of the bladder and morbidity of surgery and the requirement for a urostomy.
is often associated with local recurrence. Similarly, tran- Radiation therapy is most effective when combined with
surethral resection of the bladder tumor (TURBT) alone has sensitizing chemotherapy.16 The two most common regi-
been associated with at least 50% local recurrence and mens are either weekly cisplatin or 5-fluorouracil and mi-
progression, even in highly selected patients.15 Only 5% to tomycin C for patients who are ineligible for cisplatin.16 The
two regimens have not been compared directly, but they
appear to have similar outcomes. There are also no ran-
domized studies directly comparing cystectomy and che-
moradiation, and retrospective comparisons are fraught
KEY POINTS with selection bias. Nevertheless, overall survival rates ap-
pear similar to cystectomy series. Age alone does not appear
Less than 50% of patients older than age 70 are currently
to affect the success of chemoradiation.17
receiving definitive therapy for localized muscle-
invasive bladder cancer.
Initial response to induction chemoradiation is approxi-
Mortality from radical cystectomy is approximately 10% mately 70% in most patients.16-19 Local recurrence is observed
in older patients by 90 days. in 30% to 40% with approximately 10% to 30% of patients
A number of combined measures of frailty have been requiring a subsequent cystectomy for persistent or recurrent
shown to be predictive of morbidity and mortality of invasive disease. Noninvasive cancers also are common
major surgeries in older patients, including radical during follow-up, emphasizing the need for continued cys-
cystectomy. toscopic surveillance. Endoscopic evaluation of the bladder
Combination treatment with chemotherapy plus after radiation therapy can be difficult because of mucosal
radiation therapy has been shown to be effective in ulceration and hyperemia that may require biopsy to dif-
older patients, with similar overall survival to most ferentiate from recurrent cancer, especially carcinoma in situ.
cystectomy series. However, toxicity may be significant,
Short-term bladder and bowel side effects are common
and 10% to 20% of patients will require delayed
cystectomy because of invasive local recurrence or
during radiation treatments, with grade 3 to 4 genito-
complications from radiation. urinary toxicity ranging from 2% to 20%, and gastroin-
A multidisciplinary approach with surgeons, medical testinal toxicity approximately 10%. Approximately 10%
oncologists, and radiation oncologists can optimize to 20% of patients also will have grade 3 to 4 hematologic
outcomes in this group of patients. complications from the chemotherapy. Long-term grade 1
to 2 genitourinary side effects are observed in 10% to 25%

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e229


EILA C. SKINNER

TABLE 1. The Edmonton Frail Scale9


Domain Item 0 Points 1 Point 2 Points
Cognition Imagine that this pre-drawn circle is a clock. No errors Minor spacing errors Other errors
I would like you to place the numbers in
correct position then place the hands to
indicate a time of 10 after 11.
General Health Status In the past year how many times have you 0 12 .2
been admitted to a hospital?
In general how would you describe your Excellent Fair Poor
health?
Very good
Good
Functional Independence With how many of the following activities 01 24 58
do you require help (meal preparation,
transportation, telephone, house-
keeping, laundry, managing money,
taking medication)?
Social Support When you need help, can you count on Always Sometimes Never
someone who is willing and able to
meet your needs?
Medications Do you use five or more different No Yes
prescription medications on a regular
basis?
At times do you forget to take your No Yes
prescription medications?
Nutrition Have you recently lost weight such that No Yes
your clothing has become looser?
Mood Do you often feel sad or depressed? No Yes
Continence Do you have a problem with losing control No Yes
of urine when you dont want to?
Functional Performance I would like you to sit in this chair with 010 seconds 1120 seconds . 20 seconds,
your back and arms resting. When I say unable or
Go please get up and walk at a safe requires
and comfortable pace to the mark on assistance
the floor (approximately 3 m) and then
return to the chair and sit down.
Total Score (of 17)

of patients, with grade 3 to 4 symptoms in 3% to 8%. schedule also may be less intense. Salvage cystectomy after
Approximately 2% to 3% require cystectomy for severe chemoradiation is feasible but has increased complications
local symptoms.20 compared with nonirradiated patients, and neobladder re-
Radiation therapy is most effective in patients with lower- construction is only possible in selected patients.21
volume disease. Clinical cT2 (versus cT3), visibly complete
transurethral resection, and absence of hydronephrosis and
carcinoma in situ have been shown to be predictors of Radical Cystectomy
better initial response.17,20 A number of radiation series in The advantages of cystectomy include management of local
the literature have excluded patients with these high-risk symptoms and the potential for cure. The latter depends on
characteristics. the pathologic stage of the tumor. For tumors that are organ-
Shipley and colleagues popularized endoscopic re-evaluation confined on final pathology, the overall cure rate is above 70%
of patients undergoing chemoradiation after approximately at 5 years. On the other hand, tumors that are already
4 weeks of treatment (40 Gy).17 At that point, salvage cys- metastatic to pelvic lymph nodes are only cured about 30%
tectomy has minimally increased morbidity from the radiation of the time with surgery, even with the addition of adjuvant
effects. This may be a reasonable approach in an older patient chemotherapy.22 Neoadjuvant chemotherapy has been
who is still a good surgical candidate. This approach has not been shown to result in a 5% absolute improvement in overall
directly compared with a more standard practice of completing survival in patients undergoing cystectomy, and it is partic-
all therapy and offering cystectomy for persistent or recurrent ularly helpful in patients with hydronephrosis, lymphovascular
disease. If a patient truly is not a surgical candidate, then there is invasion, a palpable mass under anesthesia, or invasion of the
no rationale for an intermediate assessment and the follow-up prostate or uterus (cT3 or T4 disease). However, extravesical

e230 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


TREATMENT OF MUSCLE-INVASIVE BLADDER CANCER IN OLDER PATIENTS

extension is encountered on final pathology in 50% of patients Because the average age of patients undergoing cystectomy
even in the absence of any preoperative risk factors.23 today is approximately age 70, most centers performing a
Fitness of the patient to receive a cisplatin-based chemo- large number of cystectomies have experience with treat-
therapy regimen should be assessed by a medical oncologist ment of older patients. Careful fluid management is critical in
because those regimens have the highest effectiveness. this age group, especially in patients with cardiac disease.
However, up to 60% of patients older than age 70 may have an Postoperative delirium is also more common in older patients
estimated glomerular filtration rate of less than 60, which may and avoidance of narcotics may be helpful.34 A low threshold
make them ineligible for cisplatin therapy.24 Other challenges for diagnosis of infections is necessary because fever and
of delivery of effective systemic therapy include cardiac dis- leukocytosis may be suppressed in this age group. Early
ease, hearing loss, neuropathy, and cognitive dysfunction.25 ambulation and aggressive physical therapy is helpful in
Expert geriatric assessment and treatment by oncologists ex- avoiding progressive weakness from inactivity, and often the
perienced with treating older patients are both helpful. Pa- physical therapy should be continued at home.
tients of all ages tolerate chemotherapy better before surgery Perioperative management strategies recently have fo-
than afterward. In the patient who is not able to receive a cused on a multimodality approach called the ERAS (En-
standard cisplatin combination regimen, the decision between hanced Recovery After Surgery) system. This was first
surgery alone versus neoadjuvant chemotherapy with an al- described in the colorectal literature and it has been adapted
ternate regimen, such as split-dose cisplatin, gemcitabine plus to patients undergoing cystectomy.35 The ERAS system in-
carboplatin, or a clinical trial, must be made on an individual cludes aspects of preoperative, intraoperative, and post-
basis with consultation among the patient, oncologist, and operative management. The addition of an antagonist to
surgeon. the m morphine receptor in the gut (alvimopam) has been a
The morbidity of cystectomy and diversion is substantial, critical element of this system, and it has been shown in a
with approximately 60% of patients suffering some type of randomized cystectomy trial to decrease hospital stay by at
complication and about 15% to 20% suffering high-grade least 1 day and markedly decrease ileus and the need for
complications.26-28 Overall mortality rate is about 1% to 2%, nasogastric suction.36 Daneshmand and colleagues reported
but, in patients older than age 75, it is approximately 10% by on 110 patients treated with a modified ERAS protocol and
90 days following surgery.7,27-30 The most common major observed a median hospital stay of 4 days without an in-
complications include cardiovascular events, sepsis, and bowel crease in readmission.37 We have observed a similar im-
complications. The use of minimally invasive techniques gen- provement in average postoperative hospital stay at
erally has not improved outcomes except for decreased trans- Stanford from a median of 8 to 5 days with adoption of these
fusion requirements.31,32 Full recovery can take several months, techniques.
especially for older patients. A major concern is the potential loss Discharge management is also critical to the patients full
of capacity for independent living, both as a result of the basic recovery from such major surgery. Patients of all ages who
surgical recovery and difficulty in managing the urinary diversion. have undergone cystectomy require assistance with basic
activities of daily living for at least 2 to 3 weeks after surgery.
Perioperative management. Once a patient and his or her
Many older patients live with an older or frail spouse who is
family have decided to pursue surgery, optimizing physio-
unable to provide such support, or live alone without family
logic function before surgery is critical to minimizing risk.
or other resources for such assistance. They may need to be
These might include the following:
discharged to a convalescent facility. Discharge planning
1. Nutritional assessment with improvement of malnu-
should begin before surgery with a frank discussion of the
trition if possible with dietary supplementation help required and resources available to the patient.
2. Cardiovascular and/or pulmonary evaluation to rule
out reversible ischemia or chronic obstructive pul-
monary disease OUTCOMES
3. Optimization of other medical conditions such as di- Major complications and death following radical cystectomy
abetes and renal dysfunction (e.g., with drainage of an are more common in patients older than age 70 than in
obstructed kidney) younger patients. A number of recent reports identify 90-day
4. Smoking cessation at least 8 weeks before surgery mortality after cystectomy to be in the range of 10% to as high
(ideally). There is no clear benefit and there may be an as 32% for patients over age 85.7,27,28 Nevertheless, most older
increased risk of quitting for less than 8 weeks.33 patients recover after cystectomy, and the primary risk of
Specific conditions that significantly increase risk of cystectomy death in the first 2 to 3 years after surgery is metastatic disease
in older patients include cirrhosis, malnutrition, morbid obesity, rather than competing causes of mortality. Continent diversion
and chronic immunosuppression. None of these conditions is has been performed successfully in selected older patients
easily reversed in the short period of time required for cancer who appear to have outcomes similar to younger patients.
treatment. However, patients and their families must be aware Regaining continence may take longer in older patients who
of the increased risk and, together with their physicians, should undergo neobladder reconstruction. Although overall survival,
take this into consideration when making decisions about of course, is worse in older patients, there is controversy over
treatment. whether cancer-specific survival is worse in this age group.38,39

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e231


EILA C. SKINNER

CONCLUSION helping patients and families decide about treatment op-


Older patients with muscle-invasive bladder cancer tions and the advantage or disadvantage of aggressive
represent a particularly challenging group of patients to treatment. Careful geriatric assessment and a multidisci-
treat. Patient evaluation and risk assessment are critical in plinary approach can optimize patient outcomes.

References
1. Gray PJ, Fedewa SA, Shipley WU, et al. Use of potentially curative combined-modality therapy: a pooled analysis of Radiation Therapy
therapies for muscle-invasive bladder cancer in the United States: Oncology Group protocols 8802, 8903, 9506, 9706, 9906, and 0233.
results from the National Cancer Data Base. Eur Urol. 2013;63:823-829. J Clin Oncol. 2014;32:3801-3809.
2. Herr HW. Role of repeat resection in non-muscle-invasive bladder 19. Rodel C, Grabenbauer GG, Kuhn R, et al. Combined-modality treatment
cancer. J Natl Compr Canc Netw. 2015;13:1041-1046. and selective organ preservation in invasive bladder cancer: long-term
3. Rais-Bahrami S, Pietryga JA, Nix JW. Contemporary role of advanced results. J Clin Oncol. 2002;20:3061-3071.
imaging for bladder cancer staging. Urol Oncol. 2016;34:124-133. 20. Ploussard G, Daneshmand S, Efstathiou JA, et al. Critical analysis of
4. Hurria A, Cirrincione CT, Muss HB, et al. Implementing a geriatric as- bladder sparing with trimodal therapy in muscle-invasive bladder
sessment in cooperative group clinical cancer trials: CALGB 360401. cancer: a systematic review. Eur Urol. 2014;66:120-137.
J Clin Oncol. 2011;29:1290-1296. 21. Eisenberg MS, Dorin RP, Bartsch G, et al. Early complications of
5. Kim SW, Han HS, Jung HW, et al. Multidimensional frailty score for the cystectomy after high dose pelvic radiation. J Urol. 2010;184:
prediction of postoperative mortality risk. JAMA Surg. 2014;149: 2264-2269.
633-640. 22. Stein JP, Lieskovsky G, Cote R, et al. Radical cystectomy in the treatment
6. Revenig LM, Canter DJ, Kim S, et al. Report of a simplified frailty score of invasive bladder cancer: long-term results in 1,054 patients. J Clin
predictive of short-term postoperative morbidity and mortality. J Am Oncol. 2001;19:666-675.
Coll Surg. 2015;220:904-911.e1. 23. Culp SH, Dickstein RJ, Grossman HB, et al. Refining patient selection for
7. Morgan TM, Keegan KA, Barocas DA, et al. Predicting the probability of neoadjuvant chemotherapy before radical cystectomy. J Urol. 2014;
90-day survival of elderly patients with bladder cancer treated with 191:40-47.
radical cystectomy. J Urol. 2011;186:829-834. 24. Dash A, Galsky MD, Vickers AJ, et al. Impact of renal impairment on
8. Robinson TN, Wu DS, Pointer L, et al. Simple frailty score predicts eligibility for adjuvant cisplatin-based chemotherapy in patients
complications across surgical specialties. Am J Surg. 2013;206:544-50. with urothelial carcinoma of the bladder. Cancer. 2006;107:
9. Rolfson DB, Majumdar SR, Tsuyuki RT, et al. Validity and reliability of the 506-513.
Edmonton Frail Scale. Age Ageing. 2006;35:526-529. 25. Galsky MD. How I treat bladder cancer in elderly patients. J Geriatr
10. Lascano D, Pak JS, Kates M, et al. Validation of a frailty index in patients Oncol. 2015;6:1-7.
undergoing curative surgery for urologic malignancy and comparison 26. Shabsigh A, Korets R, Vora KC, et al. Defining early morbidity of radical
with other risk stratification tools. Urol Oncol. 2015;33:426.e1-426. cystectomy for patients with bladder cancer using a standardized
e.12. reporting methodology. Eur Urol. 2009;55:164-174.
11. Suskind AM, Walter LC, Jin C, et al. Impact of frailty on complications in 27. Comploj E, West J, Mian M, et al. Comparison of complications from
patients undergoing common urological procedures: a study from the radical cystectomy between old-old versus oldest-old patients. Urol Int.
American College of Surgeons National Surgical Quality Improvement 2015;94:25-30.
database. BJU Int. Epub. 2016 Jan 17. 28. Berger I, Martini T, Wehrberger C, et al. Perioperative complications and
12. Grubmueller B, Seitz C, Shariat SF. The treatment of muscle-invasive 90-day mortality of radical cystectomy in the elderly (75+): a retro-
bladder cancer in geriatric patients. Curr Opin Urol. 2016;26:160-164. spective, multicentre study. Urol Int. 2014;93:296-302.
13. Fried LP, Tangen CM, Walston J, et al; Cardiovascular Health Study 29. Schiffmann J, Gandaglia G, Larcher A, et al. Contemporary 90-day
Collaborative Research Group. Frailty in older adults: evidence for a mortality rates after radical cystectomy in the elderly. Eur J Surg Oncol.
phenotype. J Gerontol A Biol Sci Med Sci. 2001;56:M146-M157. 2014;40:1738-1745.
14. Amrock LG, Neuman MD, Lin HM, et al. Can routine preoperative data 30. Zakaria AS, Santos F, Tanguay S, et al. Radical cystectomy in patients
predict adverse outcomes in the elderly? Development and validation over 80 years old in Quebec: a population-based study of outcomes.
of a simple risk model incorporating a chart-derived frailty score. J Am J Surg Oncol. 2015;111:917-922.
Coll Surg. 2014;219:684-694. 31. Yuh BE, Nazmy M, Ruel NH, et al. Standardized analysis of frequency and
15. Cha EK, Donahue TF, Bochner BH. Radical transurethral resection alone, severity of complications after robot-assisted radical cystectomy. Eur
robotic or partial cystectomy, or extended lymphadenectomy: can we Urol. 2012;62:806-813.
select patients with muscle invasion for less or more surgery? Urol Clin 32. Bochner BH, Dalbagni G, Sjoberg DD, et al. Comparing open radical
North Am. 2015;42:189-199, viii. cystectomy and robot-assisted laparoscopic radical cystectomy: a ran-
16. James ND, Hussain SA, Hall E, et al; BC2001 Investigators. Radiotherapy domized clinical trial. Eur Urol. 2015;67:1042-1050.
with or without chemotherapy in muscle-invasive bladder cancer. 33. Warner MA, Offord KP, Warner ME, et al. Role of preoperative cessation
N Engl J Med. 2012;366:1477-1488. of smoking and other factors in postoperative pulmonary complica-
17. Efstathiou JA, Spiegel DY, Shipley WU, et al. Long-term outcomes of tions: a blinded prospective study of coronary artery bypass patients.
selective bladder preservation by combined-modality therapy for in- Mayo Clin Proc. 1989;64:609-616.
vasive bladder cancer: the MGH experience. Eur Urol. 2012;61:705-711. 34. Large MC, Reichard C, Williams JT, et al. Incidence, risk factors, and
18. Mak RH, Hunt D, Shipley WU, et al. Long-term outcomes in patients with complications of postoperative delirium in elderly patients undergoing
muscle-invasive bladder cancer after selective bladder-preserving radical cystectomy. Urology. 2013;81:123-129.

e232 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


TREATMENT OF MUSCLE-INVASIVE BLADDER CANCER IN OLDER PATIENTS

35. Cerantola Y, Valerio M, Persson B, et al. Guidelines for perioperative 37. Daneshmand S, Ahmadi H, Schuckman AK, et al. Enhanced recovery pro-
care after radical cystectomy for bladder cancer: Enhanced Recovery tocol after radical cystectomy for bladder cancer. J Urol. 2014;192:50-56.
After Surgery (ERAS()) society recommendations. Clin Nutr. 2013;32: 38. Horovitz D, Turker P, Bostrom PJ, et al. Does patient age affect survival
879-887. after radical cystectomy? BJU Int. 2012;110(11 Pt B):E486-E493.
36. Lee CT, Chang SS, Kamat AM, et al. Alvimopan accelerates gastroin- 39. Leveridge MJ, Siemens DR, Mackillop WJ, et al. Radical cystectomy and
testinal recovery after radical cystectomy: a multicenter randomized adjuvant chemotherapy for bladder cancer in the elderly: a population-
placebo-controlled trial. Eur Urol. 2014;66:265-272. based study. Urology. 2015;85:791-798.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e233


GENITOURINARY (NONPROSTATE) CANCER

Renal Cell Carcinoma: Systemic


Treatment, Evolving TKIs, and
Immuno-Oncology

CHAIR
Thomas E. Hutson, DO, PharmD, FACP
Texas Oncology
Dallas, TX

SPEAKERS
Brian I. Rini, MD, FACP
Cleveland Clinic Taussig Cancer Institute
Cleveland, OH

Robert A. Figlin, MD, FACP


Cedars-Sinai Medical Center
Los Angeles, CA
THE EVOLUTION OF SYSTEMIC THERAPY IN mRCC

The Evolution of Systemic Therapy in Metastatic Renal Cell


Carcinoma
Thomas E. Hutson, DO, PharmD, FACP, Gregory R. Thoreson, MD, Robert A. Figlin, MD, FACP, and
Brian I. Rini, MD, FACP

OVERVIEW

The treatment landscape for renal cell carcinoma (RCC) is a dynamic process that has seen considerable change in recent
years. We have seen a rebirth of original breakthroughs with immune checkpoint inhibitors showing promise in patients with
treatment-refractory disease. The optimal sequencing of treatments and incorporation of novel therapeutics are actively
being investigated and have yet to be determined. The clinical challenges of this evolving treatment paradigm can be
attributed to cost considerations, toxicity, and defining endpoints in the management of advanced RCC. As novel thera-
peutics emerge, finding the optimal treatment regimen for patients will have an increasing focus on patient-centered
outcomes and improvement in quality of life in addition to improving survival.

M anagement of RCC continues to evolve, with many of


the recent advancements occurring in the treatment
of patients with metastatic disease. Approximately one in
trials over the past decade have explored combinations,
largely of VEGF inhibitors plus VEGF inhibitors or VEGF in-
hibitors plus mTOR inhibitors.10-14 Collectively, these trials
three patients present with either regionally advanced or failed because of either excessive toxicity preventing ad-
metastatic disease, excluding candidacy for extirpative ministration of full doses of each agent/toxicity in general, or
surgery with curative intent and ultimately requiring sys- lack of a proven efficacy advantage over monotherapy.
temic therapy as their only option.1 The earliest forms of On this landscape, recent data regarding the combination
systemic therapy emerged in the 1980s and were based on of a VEGF receptor inhibitor (lenvatinib) and an mTOR in-
adoptive immunotherapy. Observations at the National Institutes hibitor (everolimus) is notable.15 A phase II study randomly
of Health showed objective responses with the administration of assigned 153 patients with mRCC who had received one
lymphokine-activated killer cells and recombinant interleukin-2 prior VEGF-targeted therapy to lenvatinib plus everolimus
(IL-2) across four histologic cell types, including RCC.2 By the early compared with lenvatinib monotherapy compared with ever-
1990s, IL-2, interferon, or a combination of the two were widely olimus monotherapy with a primary endpoint of progression-
adopted, and in the early 2000s, this became the standard of free survival (PFS). Progression-free survival as initially reported
care.3,4 To date, the only durable complete responses observed in per investigator assessment, was 14.6 months for the combi-
metastatic RCC (mRCC) are with this modality of treatment.5 The nation arm, 7.4 months for the lenvatinib monotherapy arm,
latter part of the decade ushered in the era of targeted therapies, and 5.5 months for the everolimus arm. Both lenvatinib-
with pivotal clinical trials showing improved survival when containing arms were significantly longer in terms of PFS
compared with interferon.6-8 This line of therapy is predicated than the everolimus arm (lenvatinib plus everolimus vs.
on inhibiting VEGF or mTOR. Recently, the treatment paradigm everolimus, hazard ratio [HR] 0.40; p = .0005; lenvatinib
has shifted back toward immunotherapy, with an immune vs. everolimus, HR 0.61; p = .048) but not significantly
checkpoint inhibitor showing superiority over an mTOR in- different from each other (lenvatinib plus everolimus vs.
hibitor in a select patient population that demonstrated dis- lenvatinib, HR 0.66; p = .12). Objective response rates also
ease progression while on antiangiogenic therapy.9 favored the combination arm (43% vs. 27% vs. 6%). There was
This influx of new anticancer therapies in mRCC has considerable toxicity in the combination arm necessitating
provided new opportunities to explore the therapeutic that 71% of patients reduce their lenvatinib dose and 24%
potential of combination therapies. To date, combination discontinue for toxicity. Importantly, updated data using in-
therapy in mRCC has not provided clinical benefit. Several dependent radiologic review of CT scans has been published.16

From the Texas A&M Health Science Center, Bryan TX; Baylor-Sammons Cancer Center, Dallas, TX; US Oncology, Texas Oncology, Charles A. Sammons Cancer Center, Baylor University
Medical Center, Dallas, TX; Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA; Cleveland Clinic Taussig Cancer Institute, Cleveland, OH.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Thomas E. Hutson, DO, PharmD, FACP, Texas Oncology, 3410 Worth St., Suite 400, Dallas, TX 75254; email: thomas.hutson@usoncology.com.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 113


HUTSON ET AL

These data demonstrate a reduction in the combination arm This combination, however, was not further pursued
PFS (to 12.8 months) and the response rate (to 35%). Thus, the clinically because of development of the nivolumab plus ipi-
benefit of this specific combination over monotherapy remains limumab combination. This latter combination was evaluated
to be established in larger trials. The biology of why this as part of the same clinical trial and also demonstrated re-
combination could have benefit where other similar combi- markable clinical activity (Table 1). Toxicity was higher in the
nations have failed is unknown and also requires further study. arm with the higher ipilimumab dosing and was also
Cabozantinib, an oral small molecule inhibitor of tyrosine unacceptably high in a small cohort (six patients) who
kinases including MET, VEGF, and AXL, has shown recent received a higher dose of both drugs. This combination is
promise in patients with mRCC that has failed first-line being further pursued in a front-line phase III trial com-
therapy with VEGFR-targeting tyrosine kinase inhibitors. pared with sunitinib. Accrual to this trial has been com-
In a phase III clinical trial with 658 patients, cabozantinib pleted and results are pending.
demonstrated improved PFS (7.4 vs. 3.8 months), overall Additional PD-1 or PD-L1 inhibitors have been initially
survival (HR 0.67), and objective response rates (21% vs. 5%) combined with anti-VEGF therapy. Pembrolizumab, a PD-1
when compared with everolimus.17 Cabozantinib, which is inhibitor, was combined with pazopanib. Several patients
currently approved by the U.S. Food and Drug Administration experienced LFT abnormalities necessitating interruption of
(FDA) for the treatment of patients with progressive meta- therapy, and other patients have been treated with a lower
static medullary thyroid cancer, has previously been granted initial dose of pazopanib. The safety of this particular combi-
Breakthrough Therapy and Fast Track designations and is nation awaits further experience. Pembrolizumab has also
likely to be approved for treatment of refractory disease.18 been combined with axitinib. A small safety cohort of 11 pa-
As noted, checkpoint inhibition has undergone rapid tients demonstrated tolerability and early signs of efficacy. An
clinical development in mRCC with the FDA approval of nivo- expansion cohort has been accrued to further clarify safety and
lumab for refractory disease. Several combination regimens of efficacy.
immunotherapy or immunotherapy plus VEGF therapy have An alternative approach to checkpoint inhibition is an
been initially explored (Table 1). There is clinical rationale for antibody that blocks the ligand for the PD-1 receptor, PD-L1.
such combinations given the activity of each approach as Atezolizumab is a monoclonal antibody that has been
monotherapy, but also a biologic rationale, as VEGF may lead to studied in many solid tumors including RCC. An initial phase I
immune suppression and thus VEGF blockade may allow for study of atezolizumab demonstrated the safety of mono-
enhanced effect of immunotherapy.19,20 Nivolumab was therapy in RCC and other solid tumors, and a small expansion
combined in a multicohort phase II trial with sunitinib and cohort in RCC tested the combination of atezolizumab and
separately with pazopanib. The pazopanib cohort was not bevacizumab. This combination was well tolerated, with
expanded because of liver function test (LFT) abnormalities early signs of efficacy in the form of tumor burden reduction.
in the initial cohort of patients. The sunitinib cohort ex- A randomized phase II trial was then undertaken in front-line
panded and demonstrated antitumor activity as evidenced mRCC randomly assigning patients to this combination,
by a robust objective response rate. atezolizumab monotherapy, or standard sunitinib with a
primary endpoint of PFS. This trial has completed accrual
with results pending. Importantly, this trial contains an
arm with antiPD-L1 monotherapy. Clinical and correlate
KEY POINTS results from this arm may help identify if there is a subset
of patients with mRCC who can safely and effectively receive
The treatment of advanced renal cell carcinoma is a
single-agent checkpoint inhibition. A randomized phase III
dynamic process that has undergone considerable
change in the past 40 years with a recent rebirth of
trial has also begun with this combination in front-line mRCC,
immuno-oncology drugs. randomly assigning patients to the combination or sunitinib
Optimal sequence and combinations of novel therapies with a PFS/overall survival coprimary endpoint.
are currently being investigated and pose considerable Challenges for managing RCC are not only considerations
challenges to providers. of clinical algorithm development but also opportunities to
Toxicity profiles of targeted therapies and immuno- demonstrate value-based cancer care, especially in a disease
oncology agents are unique, and effective therapy with multiple treatment approvals.21-23 The Institute of Medicine
management must balance treatment dosing, duration, has delineated six elements of value in cancer care: safety,
and adverse event management. effectiveness, patient centeredness, timeliness, efficiency, and
A value-based framework for cancer care requires the equity. The American Society of Clinical Oncology has selected
incorporation of effectiveness, safety, and efficiency
three of these for its value-based framework for cancer care:
while maintaining incentives for the continued research
and development of novel oncologic therapeutics.
clinical benefit (effectiveness), toxicity (safety), and cost (effi-
Defining endpoints for patients with metastatic renal ciency). The Memorial Sloan Kettering Cancer Center has added
cell carcinoma continue to evolve, with an increasing to the dialogue by including research and development costs and
emphasis on patient-centered outcomes in addition to population health burden, and the National Comprehensive
survival data. Cancer Network has included elements of affordability. Health
economics research has started to delineate how patients with

114 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


THE EVOLUTION OF SYSTEMIC THERAPY IN mRCC

TABLE 1. Select Studies of Combination Therapy in Renal Cell Carcinoma


No. Patient
Trial Patients Population Study Design ORR Median PFS Comments
Immunotherapy Plus Anti-VEGF Nivolumab Plus TKI
NCT01472081, 33 Front-line and Nivolumab 2 mg/kg every 52% 49 weeks; 78%
Phase I/II34 refractory mRCC 3 weeks + sunitinib 50 mg 4/2 progression-free
at 24 weeks
20 Nivolumab 2 mg/kg every 45% 31 weeks; 55% Pazopanib cohort not
3 weeks + pazopanib 800 mg/day progression-free expanded because of
at 24 weeks hepatic toxicity
Pembrolizumab Plus TKI
NCT02014636, 20 Front-line mRCC Pembrolizumab plus 40% NR AST and/or ALT elevations
Phase I/II35 pazopanib 600 mg every day . 5x ULN observed in
(10 patients) or 800 mg every 13 patients
day (10 patients)
NCT02133742, 11 Front-line mRCC Pembrolizumab 2 mg/kg 55% NR 41 additional patients
Phase IB36 every 3 weeks plus enrolled for further
axitinib 5 mg twice daily safety/efficacy
investigation
Atezolizumab Plus Bevacizumab
NCT01633970, 10 Front-line mRCC 1,200 mg IV every 3 weeks atezolizumab + 40% SD $ 24 weeks 9/10 patients remain
Phase IB37 bevacizumab 15 mg/kg every in 4 patients on treatment
3 weeks
NCT01984242 300 Front-line mRCC 1,200 mg IV every 3 weeks NR NR Accrual complete
Randomized atezolizumab +/2 bevacizumab
Phase II 15 mg/kg every 3 weeks vs. sunitinib
NCT02420821, 550 Front-line mRCC 1,200 mg IV every 3 weeks atezolizumab + NR NR Accrual ongoing
Phase III bevacizumab 15 mg/kg every
3 weeks vs. sunitinib
NCT02420821, 550 Front-line mRCC 1,200 mg IV every 3 weeks atezolizumab + NR NR Accrual ongoing
Phase III bevacizumab 15 mg/kg every
3 weeks vs. sunitinib
Immunotherapy Plus Immunotherapy
Nivolumab Plus Ipilimumab
NCT01472081, 47 Front-line and N3 + I1* 38% 33.3 weeks
Phase I/II38 refractory mRCC
47 N1 + I3* 40% 47.1 weeks Increased toxicity in
the I3
NCT02231749, 1,070 Front-line mRCC N3 + I1 NR NR Accrual complete
Phase III
*Arm N3 + I1; nivolumab 3 mg/kg + ipilimumab 1 mg/kg; Arm N1 + I3; nivolumab 1 mg/kg + ipilimumab 3 mg/kg.
Abbreviations: AST, aspartate transaminase; ALT, alanine aminotransferase; NR, no response; PFS, progression-free survival; ORR, overall response rate; ULN, upper limit of normal;
TKI, tyrosine kinase inhibitor; mRCC, metastatic renal cell carcinoma.

cancer value hope and the implications for cost-effectiveness (i.e., everolimus, temsirolimus), and more recently immuno-
assessments of high-cost cancer therapies.24 A patients will- oncology drugs (i.e., nivolumab). Effective therapy manage-
ingness to pay for a hopeful therapy may be influenced by their ment must be a balance between dosing, treatment duration,
economic realities.25 In addition, consideration of quality- and adverse event management.
adjusted cost of care has been used to measure the growth The common adverse events associated with VEGF in-
and new innovation costs associated with novel therapies.26,27 hibitors include fatigue, hand/foot syndrome, hypertension,
Unfortunately, none of these growing health economic ap- hepatotoxicity, diarrhea, stomatitis, myelosuppression, and
proaches to cancer care have been applied uniformly to the proteinuria. For the mTOR inhibitors, fatigue, diarrhea, sto-
growing portfolio of cancer treatments for RCC. This remains a matitis, myelosuppression, pneumonitis, and infections are
unique challenge in a disease with nine new drugs approved by the most common adverse events. Management of these
the FDA over the past decade. chronic toxicities is associated with improved dosing and
The toxicity profiles of targeted therapies and immuno- treatment duration and is best supported by aggressive ad-
oncology agents are quite different, and their management verse event management.
continues to evolve. Toxicities can generally be broken down Recent evidence has suggested that, for example, with
into three categories: side effects produced by the VEGF sunitinib, adjusting the dosing schedule to a 2:1 schedule
inhibitors/tyrosine kinase inhibitors (i.e., sunitinib, pazopanib, from the original 4:2 schedule allows for full dosing
axitinib, sorafenib, and bevacizumab), mTOR inhibitors maintenance of concentrations of drug with a possible

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 115


HUTSON ET AL

amelioration of toxicity. Black box warnings for pazopanib and optimizing toxicity management will mean keeping the
and sunitinib for potential liver toxicity must guide treat- patient on drug while maintaining quality of life. The early
ment management, as well as pneumonitis, a class effect recognition and intervention for the successful management
from mTOR inhibition characterized by noninfectious, of immune-related adverse events is critical with this new
nonmalignant infiltrates associated with cough, dyspnea, class of drugs.
and radiographic findings of ground glass opacities and The traditional endpoints for approval of targeted ther-
focal consolidation. Several published summaries will guide apies in RCC have been PFS. For patients treated with tar-
recommended adverse event management in mTOR-treated geted agents, it is clear that improvements in PFS have been
RCC, especially for noninfectious pneumonitis, stomatitis, and associated with improvements in overall survival. It is in-
infections. The side effect profile from targeted agents is teresting to note that with the newer immuno-oncology
usually quite predictable, and the optimal management will agents, overall survival has been the primary endpoint. With
increase drug exposure and improve overall outcome. Toxicity the approval of nivolumab, there is evidence that overall
is often treatable and can be managed with schedule changes survival is benefited, while there is no apparent difference in
and treatment interruptions before dose reduction is PFS, and as such, we may be seeing an evolution in endpoint
considered. evaluations for patients with RCC.28-30 It is clear from a
With the recently approved immuno-oncology agent, we are patient perspective that overall survival benefits are critical.
now presented with a different spectrum of toxicitiesusually This presents a challenge to the practicing physician because
of low frequencycalled immune-related adverse events, we are typically used to using imaging as a surrogate in-
which have timing that is less predictable than with adverse termediate endpoint for the evaluation of benefit for pa-
events with targeted agents, can appear without warning, and tients. With circumstances such as pseudo-progression or
often require immune suppression with corticosteroids where early growth followed by regression, we now must re-
holding the drug is typically insufficient. consider the use of imaging in deciding when and if to take a
Several websites now have guides to immune-mediated patient off of systemic therapies.21 Further evolution in
adverse reaction management, and physicians should fa- endpoint evaluation in RCC will evolve, especially if bio-
miliarize themselves with these side effects. Interestingly, markers become available to help identify populations of
the time to onset of selected treatment-related adverse patients most likely to benefit from systemic therapy.
events for immuno-oncology agents such as nivolumab can The treatment landscape for RCC is a dynamic process that
sometimes be weeks to months, and, as such, careful and has seen considerable change in recent years. We have
continual monitoring of the patient is required. Although seen a rebirth of original breakthroughs with immune
most side effects are low grade, grade 3 and 4 toxicities checkpoint inhibitors showing promise in treatment re-
can occur, especially immune-mediated pulmonary, he- fractory disease, which may play a crucial role in combi-
patic, gastrointestinal, and renal adverse reactions. Immune- nation therapies. The clinical challenges of this evolving
mediated endocrinopathies can be life threatening if not treatment paradigm can be attributed to cost consider-
approached appropriately. Recent evidence suggests that ations, toxicity, and defining endpoints in the management
immune suppression may not reduce the efficacy of immuno- of advanced RCC. As novel therapeutics emerge, identifying
oncology approaches and, as such, might allow for the con- the optimal sequence and combinations of therapies will be
tinued administration of these agents. These new therapies based on individual patient characteristics with an em-
have the potential to improve and prolong patients lives, phasis on improving quality of life and survival.

References
1. Howlader NA, Krapcho M, Garshell J. SEER Cancer Statistics Review, in patients with metastatic renal cell carcinoma. J Clin Oncol. 2005;23:
1975--2012. Bethesda: National Cancer Institute; 2014. 133-141.
2. Rosenberg SA, Lotze MT, Muul LM, et al. Observations on the systemic 6. Motzer RJ, Hutson TE, Tomczak P, et al. Sunitinib versus interferon alfa
administration of autologous lymphokine-activated killer cells and in metastatic renal-cell carcinoma. N Engl J Med. 2007;356:115-124.
recombinant interleukin-2 to patients with metastatic cancer. N Engl J 7. Escudier B, Eisen T, Stadler WM, et al; TARGET Study Group. Sorafenib in
Med. 1985;313:1485-1492. advanced clear-cell renal-cell carcinoma. N Engl J Med. 2007;356:125-134.
3. Negrier S, Escudier B, Lasset C, et al. Recombinant human interleukin-2, 8. Hudes G, Carducci M, Tomczak P, et al; Global ARCC Trial. Temsirolimus,
recombinant human interferon alfa-2a, or both in metastatic renal-cell interferon alfa, or both for advanced renal-cell carcinoma. N Engl J Med.
carcinoma. Groupe Franais dImmunotherapie. N Engl J Med. 1998; 2007;356:2271-2281.
338:1272-1278. 9. Motzer RJ, Escudier B, McDermott DF, et al; CheckMate 025 In-
4. Yang JC, Sherry RM, Steinberg SM, et al. Randomized study of high-dose vestigators. Nivolumab versus everolimus in advanced renal-cell car-
and low-dose interleukin-2 in patients with metastatic renal cancer. cinoma. N Engl J Med. 2015;373:1803-1813.
J Clin Oncol. 2003;21:3127-3132. 10. Rini BI, Garcia JA, Cooney MM, et al. Toxicity of sunitinib plus bev-
5. McDermott DF, Regan MM, Clark JI, et al. Randomized phase III trial of acizumab in renal cell carcinoma. J Clin Oncol. 2010;28:e284-e285;
high-dose interleukin-2 versus subcutaneous interleukin-2 and interferon author reply e286-e287.

116 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


THE EVOLUTION OF SYSTEMIC THERAPY IN mRCC

11. Rini BI, Bellmunt J, Clancy J, et al. Randomized phase III trial of tem- 25. Lakdawalla DN, Romley JA, Sanchez Y, et al. How cancer patients value
sirolimus and bevacizumab versus interferon alfa and bevacizumab in hope and the implications for cost-effectiveness assessments of high-
metastatic renal cell carcinoma: INTORACT trial. J Clin Oncol. 2014;32: cost cancer therapies. Health Aff (Millwood). 2012;31:676-682.
752-759. 26. Lakdawalla D, Shafrin J, Lucarelli C, et al. Quality-adjusted cost of care:
12. Ravaud A, Barrios CH, Alekseev B, et al. RECORD-2: phase II randomized a meaningful way to measure growth in innovation cost versus the value
study of everolimus and bevacizumab versus interferon a-2a and of health gains. Health Aff (Millwood). 2015;34:555-561.
bevacizumab as first-line therapy in patients with metastatic renal cell 27. Stevens W, Philipson TJ, Khan ZM, et al. Cancer mortality reductions
carcinoma. Ann Oncol. 2015;26:1378-1384. were greatest among countries where cancer care spending rose the
13. Negrier S, Gravis G, Perol D, et al. Temsirolimus and bevacizumab, or most, 1995-2007. Health Aff (Millwood). 2015;34:562-570.
sunitinib, or interferon alfa and bevacizumab for patients with ad- 28. Hoos A, Wolchok JD, Humphrey RW, et al. CCR 20th Anniversary
vanced renal cell carcinoma (TORAVA): a randomised phase 2 trial. Commentary: immune-related response criteriacapturing clinical ac-
Lancet Oncol. 2011;12:673-680. tivity in immuno-oncology. Clin Cancer Res. 2015;21:4989-4991.
14. Flaherty KT, Manola JB, Pins M, et al. BEST: a randomized phase II study 29. Wolchok JD, Hoos A, ODay S, et al. Guidelines for the evaluation of
of vascular endothelial growth factor, RAF kinase, and mammalian immune therapy activity in solid tumors: immune-related response
target of rapamycin combination targeted therapy with bevacizumab, criteria. Clin Cancer Res. 2009;15:7412-7420.
sorafenib, and temsirolimus in advanced renal cell carcinomaa trial of 30. Nishino M, Giobbie-Hurder A, Gargano M, et al. Developing a common
the ECOG-ACRIN Cancer Research Group (E2804). J Clin Oncol. 2015;33: language for tumor response to immunotherapy: immune-related re-
2384-2391. sponse criteria using unidimensional measurements. Clin Cancer Res.
15. Motzer RJ, Hutson TE, Glen H, et al. Lenvatinib, everolimus, and the 2013;19:3936-3943.
combination in patients with metastatic renal cell carcinoma: a rand- 31. Sonpavde G, Choueiri TK, Escudier B, et al. Sequencing of agents for
omised, phase 2, open-label, multicentre trial. Lancet Oncol. 2015;16: metastatic renal cell carcinoma: can we customize therapy? Eur Urol.
1473-1482. 2012;61:307-316.
16. Motzer RJ, Hutson TE, Ren M, et al. Independent assessment of len- 32. Motzer RJ, Hutson TE, Cella D, et al. Pazopanib versus sunitinib in
vatinib plus everolimus in patients with metastatic renal cell carcinoma. metastatic renal-cell carcinoma. N Engl J Med. 2013;369:722-731.
Lancet Oncol. 2016;17:e4-e5. 33. Escudier B, Szczylik C, Porta C, et al. Treatment selection in metastatic
17. Choueiri TK, Escudier B, Powles T, et al; METEOR Investigators. renal cell carcinoma: expert consensus. Nat Rev Clin Oncol. 2012;9:
Cabozantinib versus everolimus in advanced renal-cell carcinoma. 327-337.
N Engl J Med. 2015;373:1814-1823. 34. Amin A, Plimack ER, Infante JR, et al. Nivolumab (anti--PD--1; BMS--
18. Viola D, Cappagli V, Elisei R. Cabozantinib (XL184) for the treatment of 936558, ONO--4538) in combination with sunitinib or pazopanib in
locally advanced or metastatic progressive medullary thyroid cancer. patients (pts) with metastatic renal cell carcinoma (mRCC). J Clin Oncol.
Future Oncol. 2013;9:1083-1092. 2014;32:5s (suppl; abstr 5010).
19. Shrimali RK, Yu Z, Theoret MR, et al. Antiangiogenic agents can 35. McDermott DF, Infante JR, Chowdhury S, et al. A phase I/II study to
increase lymphocyte infiltration into tumor and enhance the effec- assess the safety and efficacy of pazopanib (paz) and pembrolizumab
tiveness of adoptive immunotherapy of cancer. Cancer Res. 2010;70: (pembro) in patients (pts) with advanced renal cell carcinoma (aRCC).
6171-6180. Eur J Cancer. 2015;51:S519-S520.
20. Kusmartsev S, Su Z, Heiser A, et al. Reversal of myeloid cell-mediated 36. Atkins MB, Gupta S, Choueiri TK, et al. Phase lb dose--finding study of
immunosuppression in patients with metastatic renal cell carcinoma. axitinib plus pembrolizumab in treatment--naive patients with ad-
Clin Cancer Res. 2008;14:8270-8278. vanced renal cell carcinoma. J Immunother Cancer. 2015;3:P353.
21. Zon RT, Frame JN, Neuss MN, et al. American Society of Clinical On- 37. McDermott, D, Sznol M, Sosman JA, et al. 809O -- Immune correlates
cology Policy Statement on Clinical Pathways in Oncology. J Oncol Pract. and long term follow up of a phase Ia study of MPDL3280A, an engi-
2016;12:261-266. neered PD--L1 antibody, in patients with metastatic renal cell carcinoma
22. Saltz LB. The value of considering cost, and the cost of not considering (mRCC). Ann Oncol. 2014;25:iv280-iv304.
value. J Clin Oncol. 2016;34:659-660. 38. Hammers H, Plimack ER, Infante JR, et al. Expanded cohort results from
23. Young RC. Value-based cancer care. N Engl J Med. 2015;373:2593-2595. CheckMate 016: A phase I study of nivolumab in combination with
24. Neumann PJ, Cohen JT. Measuring the value of prescription drugs. ipilimumab in metastatic renal cell carcinoma (mRCC). J Clin Oncol.
N Engl J Med. 2015;373:2595-2597. 2015;33 (suppl; abstr 4516).

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 117


GENITOURINARY (PROSTATE) CANCER

Contemporary Active Surveillance


for Prostate Cancer: Do We Need
Better Imaging and Molecular
Testing?

CHAIR
Edward M. Schaeffer, MD, PhD
Northwestern University
Chicago, IL

SPEAKERS
Jonathan I. Epstein, MD
Johns Hopkins University School of Medicine
Baltimore, MD

Peter L. Choyke, MD
National Cancer Institute at the National Institutes of Health
Bethesda, MD

Stacy Loeb, MD
New York University Langone Medical Center
New York, NY
ACTIVE SURVEILLANCE FOR PROSTATE CANCER

Active Surveillance of Prostate Cancer: Use, Outcomes,


Imaging, and Diagnostic Tools
Jeffrey J. Tosoian, MD, MPH, Stacy Loeb, MD, MSc, Jonathan I. Epstein, MD, Baris Turkbey, MD,
Peter L. Choyke, MD, and Edward M. Schaeffer, MD, PhD

OVERVIEW

Active surveillance (AS) has emerged as a standard management option for men with very low-risk and low-risk prostate
cancer, and contemporary data indicate that use of AS is increasing in the United States and abroad. In the favorable-risk
population, reports from multiple prospective cohorts indicate a less than 1% likelihood of metastatic disease and prostate
cancerspecific mortality over intermediate-term follow-up (median 56 years). Higher-risk men participating in AS appear
to be at increased risk of adverse outcomes, but these populations have not been adequately studied to this point. Although
monitoring on AS largely relies on serial prostate biopsy, a procedure associated with considerable morbidity, there is a need
for improved diagnostic tools for patient selection and monitoring. Revisions from the 2014 International Society of Urologic
Pathology consensus conference have yielded a more intuitive reporting system and detailed reporting of low-intermediate
grade tumors, which should facilitate the practice of AS. Meanwhile, emerging modalities such as multiparametric magnetic
resonance imaging and tissue-based molecular testing have shown prognostic value in some populations. At this time,
however, these instruments have not been sufficiently studied to consider their routine, standardized use in the AS setting.
Future studies should seek to identify those platforms most informative in the AS population and propose a strategy by
which promising diagnostic tools can be safely and efficiently incorporated into clinical practice.

A ctive surveillance of prostate cancer with curative intent


was described in the mid-1990s, and early AS experi-
ences were reported in 2002.1,2 Under the AS approach, men
As such, there is a substantial need for more accurate methods
of patient selection and monitoring. An ideal diagnostic tool
would impart valuable diagnostic and prognostic information
with favorable-risk cancers are monitored, and curative with limited associated morbidity at a reasonable cost. Although
intervention is pursued upon evidence of higher-risk the ideal platform does not currently exist, advances in tech-
disease. Over the last 2 decades, AS has emerged as a nology and improved understanding of the molecular basis of
standard management option for men with very low-risk prostate cancer have initiated progress toward that goal.13-16 For
and low-risk prostate cancer.3,4 Observations from two example, the use and utility of multiparametric MRI (mpMRI)
large, prospective AS cohorts reach nearly 20 years of of the prostate has increased substantially in recent years.17
follow-up and indicate very low likelihood of metastatic Furthermore, clinically validated molecular tests have estab-
disease or prostate cancerspecific mortality in appro- lished a prognostic role in some clinical contexts.18 These
priately selected men.5,6 Despite its utility in reducing platforms, along with a recently updated system of pathologic
overtreatment, AS is not without morbidity. 7 The con- grading, present a unique opportunity to improve the practice of
temporary practice of AS remains largely based on fre- AS.19,20 This article aims to review the contemporary practice of
quent clinical examination, serum prostate-specific antigen AS, including trends in use, outcomes, pathologic grading, MRI,
(PSA) testing, and prostate biopsy,8 a procedure associ- and tissue-based molecular testing.
ated with patient discomfort and serious complications
including infection.9,10 Furthermore, these methods lack TRENDS IN USE OF ACTIVE SURVEILLANCE
sensitivity for detection of higher-risk disease, as evi- Although the AS approach was previously underutilized,21,22
denced by the substantial proportion of men meeting AS recent data from multiple countries have confirmed in-
criteria who demonstrate high-risk features at radical creasing use, corresponding with its inclusion in multiple
prostatectomy.11,12 national guidelines and the availability of more data on

From the Brady Urological Institute, Departments of Urology and Pathology, Johns Hopkins University School of Medicine, Baltimore, MD; Department of Urology and Population Health,
New York University, New York, NY; Molecular Imaging Program, National Cancer Institute, Bethesda, MD; Department of Urology, Northwestern University, Chicago, IL.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Edward M. Schaeffer, MD, PhD, Northwestern University, 303 East Chicago Ave., Tarry Building 16-713, Chicago, IL 60611; email: e-schaeffer@
northwestern.edu.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e235


TOSOIAN ET AL

long-term outcomes.4-6 In the United States, as of 2006, low- at 5 years and beyond.8 In Toronto, Klotz et al5 initiated a
risk prostate cancer was managed conservatively for only 10% program of AS aimed at low-risk and select intermediate-risk
of men.23 Since that time, however, there has been a major patients. Their monitoring protocol includes PSA measure-
expansion in use. By 2011, the New Hampshire State Cancer ments every 3 months for 2 years and then every 6 months,
Registry reported that this disease was managed expectantly with a confirmatory biopsy during the first year and then
for 42% of low-risk patients.24 In a large registry from every 3 to 4 years until age 80. Intervention was initially
Michigan, 49% of low-risk prostate cancers diagnosed in 2012 recommended for histologic upgrading on rebiopsy and/or
to 2013 were managed on AS.25 Finally, new data from the PSA doubling time less than 3 years, but PSA changes now
CaPSURE clinical practice registry reported an increase in trigger additional work-up rather than immediate in-
conservative management for up to 40% of low-risk cases tervention. The most recent update of this cohort was
from 2010 to 2013.26 Similarly, studies from Canada have published in 2015, with a median follow-up of 6.4 years
reported a reduction in the proportion of low-risk patients (ranging up to 19.8 years). Of the 993 men to enroll in the
undergoing radical prostatectomy.27 program since 1995, 28 (2.8%) developed metastases and 15
Corroborative findings have been observed in several (1.5%) died of prostate cancer. Meanwhile, treatment rates
European studies. In the National Prostate Cancer Register at 10 and 15 years were 36% and 45%, respectively.
of Sweden from 2007 to 2011, AS was selected by 59% of More recently, extended follow-up was reported from the
very low-risk patients and 41% of low-risk patients.28 Johns Hopkins AS program, which was also initiated in 1995.6
Meanwhile, data from Germany demonstrated a decline in This program aims to enroll very low-risk patients (with a PSA
the proportion of men with Gleason score 6 disease at density , 0.15, clinical stage T1c, and Gleason score # 6 in a
radical prostatectomy from 2000 to 2014.29 Despite these maximum of two cores with # 50% cancer involvement in
favorable trends suggesting a reduction in the overtreat- any core). The protocol includes PSA and digital rectal ex-
ment of low-risk prostate cancer, there continues to be amination (DRE) every 6 months and a yearly prostate bi-
substantial variability in management patterns between and opsy. At the most recent analysis, the cohort was composed
within various clinical practice settings.30,31 Furthermore, in of 71% very low-risk men and 29% low-risk men. Triggers for
some parts of the world, the use of AS continues to remain treatment included an increase in grade (Gleason score . 6)
more limited.32 Nevertheless, the indications for AS con- or volume of cancer on biopsy. The 10- and 15-year cu-
tinue to expand in national guidelines, 33 and it can be mulative incidence of treatment was 50% and 57%, re-
expected that this management paradigm will become spectively, with a median treatment-free survival of 8.5
increasingly offered for favorable-risk disease in the future. years. Meanwhile, 15-year metastasis-free and cancer-
specific survival rates were 99.4% and 99.9%, respectively.
OUTCOMES OF ACTIVE SURVEILLANCE Table 1 presents published data from these two large
The modern approach to AS was first described in 1995, and prospective cohorts.
numerous programs have now reported follow-up outcomes Other U.S. centers have similarly reported intermediate-
term outcomes from AS. At the University of California, San
Francisco, prostate cancer has been managed among 810
men using an AS protocol based on quarterly PSA testing,
KEY POINTS repeat biopsy within 12 months, and follow-up biopsy every
1 to 2 years depending on medical risk.34 Of these men, 69%
Active surveillance represents a standard management met strict eligibility criteria (PSA # 10 ng/ml, clinical stage
option for men with very low-risk and low-risk prostate
T1/T2, Gleason score # 6, # 33% positive biopsy cores,
cancer.
With follow-up extending to 20 years (median 56
and # 50% involvement of any core with cancer). The au-
years), multiple AS cohorts report metastasis and thors reported a 5-year treatment-free survival of 60%, and
prostate cancer mortality in less than 1% of favorable- there were no prostate cancer deaths during a median
risk men; these outcomes appear to be more frequent in follow-up of 60 months.
men who do not meet low-risk criteria, but higher-risk At Royal Marsden in the United Kingdom, the eligibility
populations have not been adequately studied. criteria for AS include age 5080, clinical stage T1/T2 disease,
Revisions to pathologic grading from the 2014 PSA less than 15 ng/ml, Gleason score of 6 or less (as well as
International Society of Urologic Pathology consensus Gleason 3 + 4 in men older than age 65), and 50% positive
conference should facilitate a clearer understanding of biopsy cores or less.35 PSA and DRE are performed every
pathologic data and more accurate assessment of risk in 3 months in year 1, every 4 months in year 2, and every
men bordering AS criteria.
6 months thereafter. A biopsy is performed at 18 to
Multiparametric MRI and tissue-based molecular
testing may improve extant monitoring protocols but
24 months from diagnosis and then every 2 years. Triggers
first require additional study and validation. for treatment are a PSA velocity greater than 1 ng/ml per
As the practice of AS continues to evolve, it is important year, a Gleason score above 3 + 4, and/or more than 50%
that modifications of protocols are based on evidence of positive cores on repeat biopsy. Among 471 men in the
patient benefit. program, 323 (68.6%) remained on AS at a median follow-up
of 5.7 years, and there were two prostate cancer deaths.

e236 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


ACTIVE SURVEILLANCE FOR PROSTATE CANCER

TABLE 1. Basic Characteristics of Two Large Prospective Active Surveillance Cohorts


Patient Selection Monitoring Outcomes
Frequency of Median Follow-up, Metastatic
Cohort No. of Patients GS 7, % Biopsy, Years Years 10-Year Treated, % 10-Year PCSM, % Disease, %*
Toronto5 993 13 34 6.4 36 1.9 2.8
JHH6 1,298 0 1 5.0 50 0.1 0.4
Abbreviations: JHH, Johns Hopkins Hospital; GS, Gleason score; PCSM, prostate cancerspecific mortality.
*Proportion of patients during total follow-up; time-adjusted values not available.

Finally, 6-year follow-up data were reported for 439 men and then to 2005, reported Gleason scores 24 decreased
from the Goteborg randomized trial of prostate cancer from 2.7% to 0% and reported Gleason score 5 decreased
screening who elected AS.36 The cohort composition was from 12.2% to 0.3%.
51% very low risk, 36.7% low risk, 21% intermediate risk, and Another major divergence from the original Gleason
1.4% high risk. The monitoring protocol included PSA testing system is in the assignment of grade to cribriform glands.
every 3 to 6 months and rebiopsy at varying intervals, Within Gleasons original illustrations of his cribriform
depending on clinical characteristics. Nearly one-half of the pattern 3, he depicts large cribriform glands.45 Cases graded
cohort remained free from treatment at 10 years, and a prior to 2005 as Gleason pattern 3 included large cribriform
single patient died of prostate cancer at 12.7 years from glands that today would uniformly be called Gleason pattern
diagnosis. The most common reason for discontinuing AS 4.46,47 Historically, a diagnosis of Gleason score 6 cancer was
was an increase in grade or cancer involvement on repeat not as predictive of good behavior, with a higher rate of
biopsy, followed by increases in PSA. progression and some men even dying of prostate can-
In summary, these results demonstrate the durability and cer.47,48 Currently, it is recommended that all cribriform
safety of AS with careful patient selection and follow-up. At 5 glands be considered as Gleason pattern 4. Ill-defined glands
years, the majority of men enrolling in these programs re- with poorly formed glandular lumina were not discussed or
main free from treatment, allowing preservation of quality depicted by Gleason as either Gleason pattern 3 or 4. It was
of life. Furthermore, the development of metastatic disease the consensus of the 2005 conference that poorly formed
or prostate cancer death is rare within the first 10 to 15 years glands should not be considered Gleason pattern 3. Con-
of AS. Available data suggest that inclusion of higher-risk sequently, only individual well-formed glands are currently
men and less frequent monitoring biopsy may be associated graded as Gleason pattern 3.
with increased risk of adverse outcomes. An understanding The consequence of the above changes is that contem-
of these principles is crucial to appropriate patient education poraneously graded Gleason score 3 + 3 = 6 cancers are
and counseling. incapable of metastatic behavior.47 Although occasional
publications (usually from large prostate cancer databases)
UPDATED INTERNATIONAL SOCIETY OF infrequently report otherwise, these reports are subjected
UROLOGIC PATHOLOGY GRADING AND to incomplete submission of the prostate for histologic
IMPLICATIONS ON ACTIVE SURVEILLANCE examination, errors in data records, use of the older grading
In 1966, Donald Gleason37 developed the classification of system, and lack of recorded tertiary patterns, among other
prostatic carcinomas using the architectural pattern rather limitations.49 Gleason pattern 3 in the setting of surrounding
than cytology for assigning the grade. The underlying Gleason pattern 4 may have metastatic potential as recently
principles of the Gleason grading system and its contribu- shown in a case report supported by molecular evidence of
tions to prostate cancer clinical management retain rele- the clonal origin of the lethal cancer from a focus of Gleason
vance and influence over half a century from the time of pattern 3 in the middle of higher-grade cancer.50
its development. However, a number of new clinical and There has been a call to change the grading system for
pathologic discoveries, changes in prostate cancer screening prostate cancer, replacing Gleason score 3 + 3 = 6 disease
and detection, and development of new methodologies (and lower grades) with the term indolent lesion of epi-
justified revisions of the original grading system at the 2005 thelial origin to mitigate anxiety and curtail overtreatment
and 2014 International Society of Urologic Pathology (ISUP) of potentially indolent cancers.51 There exist numerous
consensus conferences.38,39 clinical and morphologic reasons why Gleason pattern 3
As it relates to AS, there are several major changes to the should be classified as cancer.52 Rather than changing
Gleason grading system. The reporting of Gleason scores 25 Gleason score 6 to a noncancerous diagnosis, there is a
has virtually disappeared from current clinical practice. In need to change the confusing and inconsistent manner in
Gleasons original data, Gleason scores 25 were seen in which the pathologic grade is reported. One of the major
27.9% of patients.40 Pathologists rendered a diagnosis of deficiencies of the current Gleason grading system is that the
Gleason score 24 in 22% to 24% of patients in the early lowest grade assigned in the contemporary setting is 6,
1990s compared with just 1.6% to 2.4% a decade later.41-43 despite a scale which ranges from 2 to 10; such practice
Helpap and Egevad44 demonstrated that from 1996 to 2000 implies that a Gleason score 6 is in the middle of the grading

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e237


TOSOIAN ET AL

scale in terms of aggressiveness. Similarly, patients with TABLE 2. Histologic Definition of the Updated Grading
Gleason score 3 + 4 = 7 may be overly concerned because a System
score of 7 is closer to the maximum score of 10 than the
lowest score of 2. Grade Gleason Score
Group Equivalent Characteristic Features
To address these deficiencies with the original Gleason
grading system, along with other problems not relevant to 1 3+3=6 Only individual discrete well-formed glands
the practice of AS, a new grading system has been proposed. 2 3+4=7 Predominantly well-formed glands with
a lesser component of poorly formed/
This system is founded in the original Gleason system, yet it fused/cribriform glands
is based on extensive subsequent research that has im-
3 4+3=7 Predominantly poorly formed/fused/cribri-
proved the original system in its definition and application. If form glands with a lesser component of
one were developing a new prostate cancer grading system well-formed glands*
de novo, the goal would be a simple system with the fewest 4 8 Only poorly formed/fused/cribriform
number of distinct grades that can adequately represent glands, predominantly well-formed
glands and a lesser component lacking
established differences in prognosis. glands, or predominantly lacking glands
The updated grade groups were originally developed in and a lesser component of well-formed
2013 by Dr. Jonathan Epstein based on data from 7,869 glands
patients who underwent radical prostatectomy at the Johns 5 910 Lack of gland formation (or necrosis) with or
Hopkins Hospital.19 This contemporary grading system was without poorly formed/fused/cribriform
glands*
subsequently validated in a cohort of 20,845 patients from
*For patients with greater than 95% poorly formed/fused/cribriform glands or lack of
five academic institutions.20 Based on these data, the rates of glands on a core or at RP, the component of less than 5% well-formed glands is not
5-year biochemical recurrence-free survival for grade groups factored into the grade.

Poorly formed/fused/cribriform glands can be a more minor component.
15 based on radical prostatectomy grade were 96%, 88%,
63%, 48%, and 26%, respectively. The grade groups were also
predictive of outcomes based on biopsy grade when biopsy AS is to determine the appropriateness of patient candidates.55
was followed by radical prostatectomy or radiation therapy. For instance, patients with apparently small, low-grade tumors
These grade groups were recently validated in a population- on random biopsy may in fact harbor larger, clinically significant
based setting using data from the National Prostate Cancer cancers that would render them inappropriate for AS. If
Registry of Sweden.53 With the new grading system, patients concerning lesions are detected on mpMRI, they can be ac-
can be assured that they have a grade group 1 of 5, which is curately sampled using various image-guided methods such as
the lowest grade, or a grade group 2 of 5, which is still a transrectal ultrasonography (TRUS)/MRI fusion.
relatively low grade. The new grades would, for the fore- Multiparametric MRI has a high negative predictive value
seeable future, be used in conjunction with the Gleason for intermediate- and high-risk prostate cancers. Thus, a
system. For example, Gleason score 3 + 3 = 6 is equivalent to negative MRI scan is good evidence that a clinically signif-
grade group 1. These definitions are listed in Table 2. icant cancer is not present. Furthermore, when a lesion is
Another recommendation of the 2014 ISUP Consensus seen, mpMRI can provide insight regarding tumor behav-
conference was for pathologists to record the percentage of ior.56,57 For instance, mpMRI has been used to predict
Gleason pattern 4 present in Gleason score 7 cancers. This Gleason scores mainly through apparent diffusion coeffi-
recommendation is directly related to the practice of AS. cient (ADC) values derived from diffusion-weighted MRI.
Traditionally, low-volume Gleason score 3 + 3 = 6 cancers Although there are strong inverse correlations between ADC
have been widely considered for a surveillance approach. and Gleason grade, mpMRI cannot be considered a re-
Depending on patient age, comorbidity, and other clinico- placement for biopsy at this time.58,59 Nevertheless, ADC
pathologic characteristics, however, there may be a pro- values can provide useful information in stratifying patient
portion of Gleason score 3 + 4 = 7 cancers that could be risk prior to biopsy. For instance, a lesion with very low ADC
reasonably considered for AS if pattern 4 disease is minimal. values that is read as a low-grade tumor on biopsy should be
Currently, this information is not transparent in pathology considered for rebiopsy based on the high risk of such a
reports, in which the percentage pattern 4 in Gleason score lesion harboring a higher-grade tumor.
3 + 4 = 7 cancers can range from 1% to close to 50%. Based on Despite its utility in lesion detection and targeting, mpMRI
these and other improvements, the new grading system has does not routinely appear in decision-making algorithms and
been accepted by the World Health Organization for the clinical nomograms for AS. To date, there are no long-term
2016 edition of Pathology and Genetics of Tumours of the prospective studies regarding the use of mpMRI in AS. Using a
Urinary System and Male Genital Organs.39,54 220 patient cohort, Shukla-Dave et al60 developed a nomo-
gram based on clinicopathological data and mpMRI findings;
MULTIPARAMETRIC MRI IN ACTIVE this nomogram demonstrated a high area under the receiver
SURVEILLANCE operating characteristic curve (AUC) of 0.854 for selecting
Multiparametric MRI offers the ability to detect, measure, and patients appropriate for AS. A follow-up study of 181 patients
monitor prostate cancers in vivo, as well as direct biopsies to a revealed an AUC of only 0.738 for low-risk disease (defined
specific target or lesion.17 Currently, the main role of mpMRI in as # pT2, Gleason grade , 4, and tumor volume # 0.5 cc)

e238 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


ACTIVE SURVEILLANCE FOR PROSTATE CANCER

for those who underwent radical prostatectomy.61 The 17-gene panel, including 12 genes from four pathways as-
National Cancer Institute nomogram, which uses only sociated with carcinogenesis and five reference genes. Gene
mpMRI criteria, generated an AUC of 0.71 for predicting expression levels are incorporated into an algorithm yielding
candidates for AS in a cohort of 85 patients, 60 of whom had the GPS, which is measured from 0 to 100.66 The GPS is then
very low-risk disease (# cT1c, PSA density , 0.15, biopsy considered in the setting of traditional clinicopathologic risk
Gleason score # 6, # 2 positive biopsy cores, and # 50% categories to determine a predicted likelihood of favorable
cancer involvement in any biopsy core).62 This nomogram was pathology. In an initial validation study, GPS from 395 men
developed with biopsy pathology as an endpoint, which is a with low- to low intermediaterisk disease were incor-
method that has not been validated. Thus, although mpMRI porated with preoperative models including the Cancer of
appears to provide some incremental value in selecting the Prostate Risk Assessment (CAPRA) score and National
candidates for AS, it is far from perfect and remains limited by Comprehensive Cancer Network (NCCN) risk categories.67 In
an inability to consistently detect clinically significant lesions, both models, a 20-unit increase in GPS was associated with
with reported false-negative rates of up to 16%.63 an approximate twofold increased odds of adverse pa-
The utility of mpMRI for monitoring men on AS has not thology at prostatectomy (model with CAPRA: odds ratio
been well established. Walton-Diaz et al64 reported a series [OR], 2.1; 95% CI, 1.43.2; model with NCCN risk classifi-
of 58 patients on AS with a median follow-up of 16.1 months cation: OR,1.9; 95% CI, 1.32.8), defined as primary Gleason
who underwent serial mpMRI with TRUS/MRI fusion-guided pattern 4, any Gleason pattern 5, or nonorgan-confined
biopsy. Upgrading to Gleason score 3 + 4 = 7 was docu- disease. In a subsequent study of 402 men with low to
mented for 17 men (29%), and the positive and negative intermediate-risk disease,68 a 20-point increase in GPS
predictive values of mpMRI for Gleason score progression similarly conveyed a significant increase in risk of bio-
were 53% (95% CI, 28%77%) and 80% (95% CI, 65%91%), chemical recurrence after treatment (hazard ratio [HR] 2.93;
respectively. One recent retrospective study by Felker et al65 95% CI, 2.034.15; p , .001) over a median follow-up of 5.2
included 49 consecutive men with Gleason score 6 prostate years; GPS was also an independent predictor of biochemical
cancer who underwent mpMRI and targeted prostate biopsy recurrence (BCR) in multivariable models with baseline
at baseline and again more than 6 months later. Over a mean clinical factors. Although the test is yet to be assessed in an
follow-up of 28.3 months (range, 1143 months), Gleason AS cohort, one clinical utility study demonstrated a 10%
score progression occurred for 19 patients (39%). The ad- increased use of AS when GPSs were incorporated into
dition of serial mpMRI improved the detection of Gleason clinical decision making.69 However, the long-term effect of
score progression during follow-up. these changes in management are unknown.
Preliminary data indicate that mpMRI and targeted biopsy
provide incremental value in the selection of candidates for Prolaris: Cell Cycle Progression
AS. Performing mpMRI prior to entering AS can reduce the The Prolaris platform (Myriad Genetics, Salt Lake City, UT) is
number of inappropriate AS candidates by 29% (Figs. 1 and based on tumor cell proliferation as measured by quan-
2).62 Once a patient has initiated AS, mpMRI can be useful in titative reverse-transcription polymerase chain reaction
demonstrating lesion stability (Fig. 3) but currently lacks the of a 46-gene panel, consisting of 31 cell cycle genes and
sensitivity to reliably detect higher-risk lesions during follow- 15 housekeeping genes.70 The output is reported as a cell
up. As such, continued serial biopsies are recommended cycle progression (CCP) score categorized clinically as less
until mature data indicate that a stable mpMRI may in fact than 0, 01, 12, 23, and greater than 3. CCP scores were
preclude subsequent biopsy. Ultimately, additional studies retrospectively assessed for 349 men conservatively
with longer follow-up are needed to establish the role of managed within the Transatlantic Prostate Group.71
mpMRI in the context of monitoring. This practice will Ten-year rates of prostate cancer death associated with
continue to evolve as larger studies are published. CCP score groups less than 0, 01, 12, 23, and greater
than 3 were 19.3%, 19.8%, 21.1%, 48.2%, and 74.9%, re-
spectively. In multivariable analysis, the CCP score (HR for
MOLECULAR TESTING IN ACTIVE SURVEILLANCE one-unit change: 1.65; 95% CI, 1.312.09), Gleason score
Several molecular profiling tests have been presented for use in
greater than 7 (HR 1.90; 95% CI, 1.183.07), and PSA (HR
the AS setting. Unfortunately, these tools have yet to be
1.37; 95% CI, 1.051.79) significantly predicted prostate
prospectively assessed in AS populations or with repeat lon-
cancer death. Perhaps most pertinent to the AS setting,
gitudinal measures. Based on existing data, however, some of
however, the CCP score was not independently predic-
which are derived from conservatively managed cohorts, it is
tive of death for men with Gleason score 6 cancers (HR
both reasonable and biologically plausible to consider some
1.30; 95% CI, 0.612.77). Two subsequent analyses
molecular tests in the AS setting.18 Here, we briefly review the
have been performed among treated populations. In one
molecular tests proposed for use in decisions about AS (Table 3).
multi-institutional study of men treated with radical
prostatectomy,72 a single-unit increase in CCP score on
Oncotype DX: Genomic Prostate Score biopsy was associated with increased risk of BCR (HR 1.47;
The Oncotype DX (Genomic Health, Redwood City, CA) 95% CI, 1.231.76) and metastasis (HR 4.19; 95% CI,
genomic prostate score (GPS) measures expression of a 2.088.45) on multivariable analysis. In men treated with

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e239


TOSOIAN ET AL

FIGURE 1. Use of MRI Prior to Enrollment in Active Surveillance

A 71-year-old man with a serum PSA of 5.47 ng/ml was found to have Gleason score 3 + 3 cancer (5%) on random biopsy. Due to rising PSA levels, he underwent MRI scan
prior to enrollment in active surveillance. (A) An axial T2-weighted MRI scan shows a PI-RADS 5 lesion in the left mid-anterior transition zone (arrow). (BD) The lesion is
positive on the apparent diffusion coefficient map (B), diffusion-weighted MRI (b = 2,000 mm/s2) (C), and dynamic contrast-enhanced MRI (D; arrows). A TRUS/MRI fusion-guided
biopsy revealed a high volume Gleason 3 + 4 score (100% core involvement with cancer with 30% Gleason pattern 4 of the entire cancer). The patient became an active treatment
candidate after multiparametric MRI and a TRUS/MRI fusion-guided biopsy approach.
Abbreviations: PI-RADS, Prostate Imaging Reporting and Data System; PSA, prostate-specific antigen; TRUS, transrectal ultrasonography.

radiation therapy, a one-unit increase in the CCP score was second using the lowest.76,77 Both TMAs were tested using a
similarly associated with a twofold increased risk of BCR 12-protein panel that demonstrated similar ability to dis-
(HR 2.11; 95% CI, 1.054.25).73 Based on surveys of or- criminate tumor aggressiveness (Gleason score $ 7, pT3b,
dering physicians, use of the CCP score led to a change in N1, or M1) based on the AUC (using the highest Gleason
management selection in one-third to two-thirds of cases; pattern: AUC, 0.70; 95% CI, 0.620.77; using the lowest
however, as with the clinical utility studies of Oncotype DX, Gleason pattern: AUC, 0.72; 95% CI, 0.640.79).77 This initial
the long-term effect of these changes in management panel was reduced to an eight-protein assay and validated
based on Prolaris results is unknown.74,75 for predicting unfavorable pathology (Gleason score $ 4 + 3
or nonorgan-confined disease) in a cohort of matched
ProMark biopsy and prostatectomy specimens.78 Remarkably, using a
The ProMark test (Metamark Genetics Inc, Augusta, GA) is threshold score of less than 0.33 the test was 90% sensitive
based on a quantitative proteomics profile uniquely for predicting favorable pathology, and above a score of 0.8
designed to yield prognostic value regardless of sampling the test was 95% specific for unfavorable pathology. On
error. To do this, two tissue microarrays (TMAs) were de- multivariable analysis, a score increase of 0.25 was associ-
rived from a series of prostatectomy specimens, one using ated with 3.1-fold increased odds of unfavorable disease
the highest Gleason pattern observed in the specimen and a (OR, 3.1; 95% CI, 2.24.4).

FIGURE 2. Use of Various Modalities to Determine Treatment Approach

A comparison of pathology-defined candidates (top row) for active treatment (yellow) and active surveillance (orange) versus definitions based on multiparametric MRI, the
Epstein criteria, the DAmico criteria, and CAPRA score. Each patient is represented by a column of the array. Note that multiparametric MRI results most closely mirrored pathology
results with fewest false-positive decisions for active treatment in good active surveillance candidates.
Abbreviations: CAPRA, Cancer of the Prostate Risk Assessment; MP, multiparametric.
Reprinted from Turkbey et al55 with permission from the publisher.

e240 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


ACTIVE SURVEILLANCE FOR PROSTATE CANCER

FIGURE 3. Use of MRI in Monitoring a Prostate Lesion During Active Surveillance

A 59-year-old man presenting with a serum PSA of 4.68 ng/ml. (AC) An axial T2-weighted MRI scan (A), apparent diffusion coefficient map (B), and b1500 diffusion-weighted MRI
(C) show a lesion in the left mid-peripheral zone (arrows). This lesion was biopsied and found to harbor Gleason 3 + 3 prostate cancer (10%). The patient elected active surveillance.
(DF) A 1-year follow-up MRI scan (serum PSA = 5.10 ng/ml) shows that the lesion is stable in size and MRI signal features. The follow-up biopsy also revealed Gleason 3 + 3 prostate
cancer (15%). MRI may become a tool for monitoring patients on active surveillance.
Abbreviation: PSA, prostate-specific antigen.

Phosphatase and Tensin Homolog Testing prostate cancer death (HR 8.13; 95% CI, 2.823.2). Limiting
The phosphatase and tensin homolog (PTEN) gene is located its utility, however, PTEN loss was only present in 3% of this
on chromosome 10q and functions as a tumor suppressor in population. Other contemporary studies have described
the phosphoinositide 3-kinase/AKT pathway.79 PTEN in- PTEN loss in 11% to 12% of favorable-risk study pop-
activation is an early event in carcinogenesis observed in up ulations.84,85 Findings from these studies have consistently
to 40% of prostate cancers. Specifically, PTEN loss appears to supported an association between PTEN loss on biopsy and
be associated with increasing likelihood of advanced grade adverse clinicopathologic outcomes.84,85 Considering a
and stage, as well as early recurrence after treatment.80-82 As strong correlation with poor outcomes but its less than 15%
such, PTEN loss detected by either fluorescence in situ prevalence in potential AS candidates, there may be a role
hybridization or immunohistochemical staining has been for PTEN testing whereby PTEN loss is considered a high-risk
proposed for prognostic use. One report from the Trans- feature that may preclude management on AS.
atlantic Prostate Group investigated PTEN in men with
conservatively managed prostate cancer with favorable Current Status of Molecular Testing
disease characteristics.83 In the population of men with a The true promise of molecular tests lies in their ability to
Gleason score below 7, PTEN was highly predictive of both improve upon and expand beyond biopsy. In addition

TABLE 3. Molecular Tests Proposed for Use in Active Surveillance

Test Manufacturer Platform Biologic Process Endpoints Assessed


Oncotype DX GPS Genomic Health (Redwood City, CA) qRT-PCR RNA quantification (gene expression) Likelihood of favorable
pathology at RP*
Prolaris CCP score Myriad Genetics (Salt Lake City, UT) qRT-PCR RNA quantification (gene expression) 10-year risk of PCSM
ProMark Metamark Genetics Inc. (Augusta, GA) QMPI Protein quantification Likelihood of unfavorable
pathology
Abbreviations: GPS, genomic prostate score; CCP, cell cycle progression; qRT-PCR, quantitative reverse-transcription polymerase chain reaction; QMPI, quantitative multiplex
proteomics imaging; RP, radical prostatectomy; PCSM, prostate cancerspecific mortality.
*Freedom from: primary Gleason pattern 4, any Gleason pattern 5, and nonorgan-confined disease.

Gleason score of 4 + 3 or greater or nonorgan-confined disease ($ pT3, N1, or M1).

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e241


TOSOIAN ET AL

to practical considerations, the use of biopsy is limited by criteria may in fact harbor indolent cancers that will not
sampling error and a dependence on subjective grading.86 threaten their quantity or quality of life. Thus, there is
Conversely, Long et al87 have demonstrated consistent gene substantial need for more accurate diagnostic and prog-
expression across multiple biopsy cores of a single patient, nostic tools to identify both those patients who are ap-
supporting previous work revealing limited variation in gene propriate candidates for AS and those who are not.
expression levels of highly expressed genes across multiple We have herein outlined the updated grading system for
biopsy cores, including both malignant and benign stromal prostate cancer as determined according to the ISUP con-
tissue.88 As others have noted, such consistent gene ex- sensus conference, most recently in 2014. As in the past,
pression, despite tissue and tumor heterogeneity, supports prognostic grade groups were determined based on long-
the notion that one can be confident in the overall prostate term oncological outcomes. Perhaps most clinically impor-
biology based on analysis of a limited sample.89 tant is the conversion from a complex Gleason score scale,
Certainly there are many reasons for optimism in exploring traditionally measured from 2 to 10, to a straightforward 15
molecular tests in the AS setting. Contrary to pathologic scale. This modification should allow that the information
grading, molecular tests provide an objective result that conveyed by pathologic grade is more clearly understood by
remains consistent across institutions. Beyond the platforms both patients and physicians. In the realm of AS, the most
discussed above, additional tests continue to emerge and substantial change in pathologic grading is the recom-
may prove useful. For example, the Decipher genomic mendation for uniform recording of the percent pattern 4 in
classifier (GenomeDx Biosciences, Vancouver, BC, Canada), Gleason score 7 cancers. Although the safety of AS in higher-
which has been previously validated for predicting metas- risk populations is not well established, the prognostic risk
tasis after surgery based on prostatectomy tissue,90-92 has associated with Gleason score 3 + 4 = 7 cancer varies sub-
more recently demonstrated prognostic value in biopsy stantially between samples with 1% Gleason pattern 4 and
specimens.93 At the same time, the majority of current 50% Gleason pattern 4.97,98 Measuring such differences will
evidence has been gleaned from retrospective analyses of enable better understanding of these risks, and, when
cohorts that were not established with a surveillance ap- clinically appropriate, consideration of careful expansion of
proach in mind. Furthermore, these tests remain expensive AS to populations with minimal Gleason pattern 4 disease.
and their incremental value in the context of other di- We are at a critical point in the evolution of AS. Emerging
agnostics such as serum markers and MRI is unknown. For technologies such as mpMRI and molecular testing un-
example, the serum Prostate Health Index has demonstrated doubtedly add to the bevy of tools available for patient
predictive ability using baseline and longitudinal samples in selection and monitoring. These tools, however, currently
the AS setting and currently represents a less-invasive, more lack a template for use. Now more than ever, prostate cancer
affordable option.94,95 Acknowledging the entirety of the is diagnosed using TRUS/MRI fusion-guided biopsy, yet there
data, we lack a definitive answer to the most fundamental exists no standard by which cancers diagnosed on targeted
questions about molecular testing: does it work, and is it biopsy can be incorporated into conventional risk classifi-
cost-effective? Additional evidence for these important cation schemes such as that of the NCCN. Similarly, more
questions is necessary and will ideally stem from pro- complex issues remain unaddressed, such as when and how
spectively designed studies. to incorporate mpMRI and genomic tests into the AS par-
adigm. A body of retrospective data exists to begin shaping
ACTIVE SURVEILLANCE: PRESENT AND FUTURE these approaches, but more evidence is needed. With
An abundance of data indicate that the use of AS is rising greater experience, the paradigm for incorporating these
rapidly and that it is a safe option to minimize overtreatment tests into current management approaches should continue
in men with favorable-risk prostate cancer.8 Nonetheless, to take shape.
based on current methods of patient selection and moni- A future in which AS protocols are largely based on mpMRI
toring, more than one in five men eligible for AS will have and molecular testing is easy to envision, but many questions
evidence of more aggressive disease on prostatectomy.96 lie between here and there. How soundly these questions are
Conversely, some men who fail to meet conventional AS answered will dictate ifand whenwe arrive.

References

1. Choo R, Klotz L, Danjoux C, et al. Feasibility study: watchful waiting 3. Heidenreich A, Bastian PJ, Bellmunt J, et al; European Association of
for localized low to intermediate grade prostate carcinoma with Urology. EAU guidelines on prostate cancer. part 1: screening, di-
selective delayed intervention based on prostate specific anti- agnosis, and local treatment with curative intent-update 2013. Eur Urol.
gen, histological and/or clinical progression. J Urol. 2002;167: 2014;65:124-137.
1664-1669. 4. Mohler JL, Armstrong AJ, Bahnson RR, et al; National Comprehensive
2. Carter HB, Walsh PC, Landis P, et al. Expectant management of non- Cancer Network. NCCN Clinical Practice Guidelines in Oncology (NCCN
palpable prostate cancer with curative intent: preliminary results. Guidelines): Prostate Cancer, version 1. Fort Washington, PA: National
J Urol. 2002;167:1231-1234. Comprehensive Cancer Network Inc; 2015.

e242 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


ACTIVE SURVEILLANCE FOR PROSTATE CANCER

5. Klotz L, Vesprini D, Sethukavalan P, et al. Long-term follow-up of a large 25. Womble PR, Montie JE, Ye Z, et al; Michigan Urological Surgery Im-
active surveillance cohort of patients with prostate cancer. J Clin Oncol. provement Collaborative. Contemporary use of initial active surveil-
2015;33:272-277. lance among men in Michigan with low-risk prostate cancer. Eur Urol.
6. Tosoian JJ, Mamawala M, Epstein JI, et al. Intermediate and longer-term 2015;67:44-50.
outcomes from a prospective active-surveillance program for favorable- 26. Cooperberg MR, Carroll PR. Trends in management for patients with
risk prostate cancer. J Clin Oncol. Epub 2015 Aug 31. localized prostate cancer, 1990-2013. JAMA. 2015;314:80-82.
7. Bergman J, Litwin MS. Quality of life in men undergoing active sur- 27. Louis AS, Kalnin R, Maganti M, et al. Oncologic outcomes following
veillance for localized prostate cancer. J Natl Cancer Inst Monogr. 2012; radical prostatectomy in the active surveillance era. Can Urol Assoc J.
2012:242-249. 2013;7:E475-E480.
8. Tosoian JJ, Carter HB, Lepor A, et al. Active surveillance for prostate 28. Loeb S, Berglund A, Stattin P. Population based study of use and de-
cancer: current evidence and contemporary state of practice. Nat Rev terminants of active surveillance and watchful waiting for low and
Urol. 2016. In press. intermediate risk prostate cancer. J Urol. 2013;190:1742-1749.
9. Loeb S, Vellekoop A, Ahmed HU, et al. Systematic review of compli- 29. Huland H, Graefen M. Changing trends in surgical management of
cations of prostate biopsy. Eur Urol. 2013;64:876-892. prostate cancer: the end of overtreatment? Eur Urol. 2015;68:175-178.
10. Bokhorst LP, Lepisto I, Kakehi Y, et al. Complications after prostate 30. Weerakoon M, Papa N, Lawrentschuk N, et al. The current use of active
biopsies in men on active surveillance and its effect on receiving further surveillance in an Australian cohort of men: a pattern of care analysis
biopsies in the Prostate Cancer Research International: Active Sur- from the Victorian Prostate Cancer Registry. BJU Int. 2015;115:50-56.
veillance (PRIAS) study. BJU Int. Epub 2016 Jan 14. 31. Filson CP, Schroeck FR, Ye Z, et al. Variation in use of active surveillance
11. Filippou P, Welty CJ, Cowan JE, et al. Immediate versus delayed radical among men undergoing expectant treatment for early stage prostate
prostatectomy: updated outcomes following active surveillance of cancer. J Urol. 2014;192:75-81.
prostate cancer. Eur Urol. 2015;68:458-463. 32. Mitsuzuka K, Koga H, Sugimoto M, et al. Current use of active sur-
12. Tosoian JJ, Sundi D, Trock BJ, et al. Pathologic outcomes in favorable-risk veillance for localized prostate cancer: a nationwide survey in Japan. Int
prostate cancer: comparative analysis of men electing active surveil- J Urol. 2015;22:754-759.
lance and immediate surgery. Eur Urol. Epub 2015 Oct 5. 33. Mohler JL, Armstrong AJ, Bahnson RR, et al. Prostate cancer, version
13. Stephenson SK, Chang EK, Marks LS. Screening and detection advances 1.2016. J Natl Compr Canc Netw. 2016;14:19-30.
in magnetic resonance image-guided prostate biopsy. Urol Clin North 34. Welty CJ, Cowan JE, Nguyen H, et al. Extended followup and risk factors
Am. 2014;41:315-326. for disease reclassification in a large active surveillance cohort for
14. Vargas HA, Hotker AM, Goldman DA, et al. Updated prostate imaging localized prostate cancer. J Urol. 2015;193:807-811.
reporting and data system (PIRADS v2) recommendations for the de- 35. Selvadurai ED, Singhera M, Thomas K, et al. Medium-term outcomes of
tection of clinically significant prostate cancer using multiparametric active surveillance for localised prostate cancer. Eur Urol. 2013;64:
MRI: critical evaluation using whole-mount pathology as standard of 981-987.
reference. Eur Radiol. Epub 2015 Sep 22. 36. Godtman RA, Holmberg E, Khatami A, et al. Outcome following active
15. Barbieri CE, Bangma CH, Bjartell A, et al. The mutational landscape of surveillance of men with screen-detected prostate cancer. Results from
prostate cancer. Eur Urol. 2013;64:567-576. the Goteborg randomised population-based prostate cancer screening
16. van den Bergh RCN, Ahmed HU, Bangma CH, et al. Novel tools to trial. Eur Urol. 2013;63:101-107.
improve patient selection and monitoring on active surveillance for 37. Gleason DF, Mellinger GT. Prediction of prognosis for prostatic ade-
low-risk prostate cancer: a systematic review. Eur Urol. 2014;65: nocarcinoma by combined histological grading and clinical staging.
1023-1031. J Urol. 1974;111:58-64.
17. Turkbey B, Brown AM, Sankineni S, et al. Multiparametric prostate 38. Epstein JI, Allsbrook WC Jr, Amin MB, et al; ISUP Grading Committee.
magnetic resonance imaging in the evaluation of prostate cancer. CA The 2005 International Society of Urological Pathology (ISUP) Con-
Cancer J Clin. Epub 2015 Nov 23. sensus Conference on Gleason Grading of Prostatic Carcinoma. Am J
18. Ross AE, DAmico AV, Freedland SJ. Which, when and why? Rational use Surg Pathol. 2005;29:1228-1242.
of tissue-based molecular testing in localized prostate cancer. Prostate 39. Epstein JI, Egevad L, Amin MB, et al; Grading Committee. The 2014
Cancer Prostatic Dis. 2016;19:1-6. International Society of Urological Pathology (ISUP) Consensus Con-
19. Pierorazio PM, Walsh PC, Partin AW, et al. Prognostic Gleason grade ference on Gleason Grading of Prostatic Carcinoma: definition of
grouping: data based on the modified Gleason scoring system. BJU Int. grading patterns and proposal for a new grading system. Am J Surg
2013;111:753-760. Pathol. 2016;40:244-252.
20. Epstein JI, Zelefsky MJ, Sjoberg DD, et al. A contemporary prostate 40. Gleason DF, Tannenbaum M (eds). Histological Grading and Staging
cancer grading system: a validated alternative to the Gleason score. Eur of Prostatic Carcinoma: Urologic Pathology. Philadelphia, PA: Lea &
Urol. 2016;69:428-435. Feibiger; 1977.
21. Cooperberg MR, Broering JM, Carroll PR. Time trends and local variation 41. Ghani KR, Grigor K, Tulloch DN, et al. Trends in reporting Gleason score
in primary treatment of localized prostate cancer. J Clin Oncol. 2010;28: 1991 to 2001: changes in the pathologists practice. Eur Urol. 2005;47:
1117-1123. 196-201.
22. Carter HB. Active surveillance for prostate cancer: an underutilized 42. Fine SW, Epstein JI. A contemporary study correlating prostate needle
opportunity for reducing harm. J Natl Cancer Inst Monogr. 2012;2012: biopsy and radical prostatectomy Gleason score. J Urol. 2008;179:
175-183. 1335-1338, discussion 1338-1339.
23. Cooperberg MR, Broering JM, Kantoff PW, et al. Contemporary trends in 43. Steinberg DM, Sauvageot J, Piantadosi S, et al. Correlation of prostate
low risk prostate cancer: risk assessment and treatment. J Urol. 2007; needle biopsy and radical prostatectomy Gleason grade in academic
178:S14-S19. and community settings. Am J Surg Pathol. 1997;21:566-576.
24. Ingimarsson JP, Celaya MO, Laviolette M, et al. Trends in initial man- 44. Helpap B, Egevad L. Correlation of modified Gleason grading of prostate
agement of prostate cancer in New Hampshire. Cancer Causes Control. carcinoma with age, serum prostate specific antigen and tumor extent
2015;26:923-929. in needle biopsy specimens. Anal Quant Cytol Histol. 2008;30:133-138.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e243


TOSOIAN ET AL

45. Gleason DF. Histologic grading of prostate cancer: a perspective. Hum 66. Knezevic D, Goddard AD, Natraj N, et al. Analytical validation of the
Pathol. 1992;23:273-279. Oncotype DX prostate cancer assay - a clinical RT-PCR assay optimized
46. McNeal JE, Yemoto CE. Spread of adenocarcinoma within prostatic for prostate needle biopsies. BMC Genomics. 2013;14:690.
ducts and acini. Morphologic and clinical correlations. Am J Surg Pathol. 67. Klein EA, Cooperberg MR, Magi-Galluzzi C, et al. A 17-gene assay to
1996;20:802-814. predict prostate cancer aggressiveness in the context of Gleason grade
47. Ross HM, Kryvenko ON, Cowan JE, et al. Do adenocarcinomas of the heterogeneity, tumor multifocality, and biopsy undersampling. Eur
prostate with Gleason score (GS) #6 have the potential to metastasize Urol. 2014;66:550-560.
to lymph nodes? Am J Surg Pathol. 2012;36:1346-1352. 68. Cullen J, Rosner IL, Brand TC, et al. A biopsy-based 17-gene genomic
48. Nakabayashi M, Hayes J, Taplin M-E, et al. Clinical predictors of survival prostate score predicts recurrence after radical prostatectomy and
in men with castration-resistant prostate cancer: evidence that Gleason adverse surgical pathology in a racially diverse population of men with
score 6 cancer can evolve to lethal disease. Cancer. 2013;119: clinically low- and intermediate-risk prostate cancer. Eur Urol. 2015;68:
2990-2998. 123-131.
49. Kryvenko ON, Epstein JI. Re: nationwide prevalence of lymph node 69. Badani KK, Kemeter MJ, Febbo PG, et al. The impact of a biopsy based
metastases in Gleason score 3 + 3 = 6 prostate cancer. Pathology. 17-gene genomic prostate score on treatment recommendations in
2015;47:394. men with newly diagnosed clinically prostate cancer who are candidates
50. Haffner MC, De Marzo AM, Yegnasubramanian S, et al. Diagnostic for active surveillance. Urol Pract. 2015;2:181-189.
challenges of clonal heterogeneity in prostate cancer. J Clin Oncol. 2015; 70. Cuzick J, Swanson GP, Fisher G, et al; Transatlantic Prostate Group.
33:e38-e40. Prognostic value of an RNA expression signature derived from cell cycle
51. Esserman LJ, Thompson IM, Reid B, et al. Addressing overdiagnosis and proliferation genes in patients with prostate cancer: a retrospective
overtreatment in cancer: a prescription for change. Lancet Oncol. 2014; study. Lancet Oncol. 2011;12:245-255.
15:e234-e242. 71. Cuzick J, Berney DM, Fisher G, et al; Transatlantic Prostate Group.
52. Carter HB, Partin AW, Walsh PC, et al. Gleason score 6 adenocarcinoma: Prognostic value of a cell cycle progression signature for prostate cancer
should it be labeled as cancer? J Clin Oncol. 2012;30:4294-4296. death in a conservatively managed needle biopsy cohort. Br J Cancer.
53. Loeb S, Folkvaljon Y, Robinson D, et al. Evaluation of the 2015 Gleason 2012;106:1095-1099.
grade groups in a nationwide population-based cohort. Eur Urol. Epub 72. Bishoff JT, Freedland SJ, Gerber L, et al. Prognostic utility of the cell cycle
2015 Dec 17. progression score generated from biopsy in men treated with pros-
54. Eble JN, Sauter G, Epstein JI, et al (eds). Pathology and Genetics of tatectomy. J Urol. 2014;192:409-414.
Tumours of the Urinary System and Male Genital Organs. Lyon, France: 73. Freedland SJ, Gerber L, Reid J, et al. Prognostic utility of cell cycle
International Agency for Research on Cancer; 2004. progression score in men with prostate cancer after primary external
55. Turkbey B, Mani H, Aras O, et al. Prostate cancer: can multiparametric beam radiation therapy. Int J Radiat Oncol Biol Phys. 2013;86:848-853.
MR imaging help identify patients who are candidates for active sur- 74. Crawford ED, Scholz MC, Kar AJ, et al. Cell cycle progression score and
veillance? Radiology. 2013;268:144-152. treatment decisions in prostate cancer: results from an ongoing reg-
56. Siddiqui MM, Rais-Bahrami S, Turkbey B, et al. Comparison of istry. Curr Med Res Opin. 2014;30:1025-1031.
MR/ultrasound fusion-guided biopsy with ultrasound-guided biopsy for 75. Shore N, Concepcion R, Saltzstein D, et al. Clinical utility of a biopsy-
the diagnosis of prostate cancer. JAMA. 2015;313:390-397. based cell cycle gene expression assay in localized prostate cancer. Curr
57. Panebianco V, Barchetti F, Sciarra A, et al. Multiparametric magnetic Med Res Opin. 2014;30:547-553.
resonance imaging vs. standard care in men being evaluated for 76. Shipitsin M, Small C, Giladi E, et al. Automated quantitative multiplex
prostate cancer: a randomized study. Urol Oncol. 2015;33:17.e1-17.e7. immunofluorescence in situ imaging identifies phospho-S6 and
58. Turkbey B, Shah VP, Pang Y, et al. Is apparent diffusion coefficient phospho-PRAS40 as predictive protein biomarkers for prostate cancer
associated with clinical risk scores for prostate cancers that are visible lethality. Proteome Sci. 2014;12:40.
on 3-T MR images? Radiology. 2011;258:488-495. 77. Shipitsin M, Small C, Choudhury S, et al. Identification of proteomic
59. Vargas HA, Akin O, Franiel T, et al. Diffusion-weighted endorectal MR biomarkers predicting prostate cancer aggressiveness and lethality
imaging at 3 T for prostate cancer: tumor detection and assessment of despite biopsy-sampling error. Br J Cancer. 2014;111:1201-1212.
aggressiveness. Radiology. 2011;259:775-784. 78. Blume-Jensen P, Berman DM, Rimm DL, et al. Development and clinical
60. Shukla-Dave A, Hricak H, Kattan MW, et al. The utility of magnetic validation of an in situ biopsy-based multimarker assay for risk strat-
resonance imaging and spectroscopy for predicting insignificant ification in prostate cancer. Clin Cancer Res. 2015;21:2591-2600.
prostate cancer: an initial analysis. BJU Int. 2007;99:786-793. 79. Leslie NR, Downes CP. PTEN function: how normal cells control it and
61. Shukla-Dave A, Hricak H, Akin O, et al. Preoperative nomograms tumour cells lose it. Biochem J. 2004;382:1-11.
incorporating magnetic resonance imaging and spectroscopy for 80. Lotan TL, Gurel B, Sutcliffe S, et al. PTEN protein loss by immuno-
prediction of insignificant prostate cancer. BJU Int. 2012;109: staining: analytic validation and prognostic indicator for a high risk
1315-1322. surgical cohort of prostate cancer patients. Clin Cancer Res. 2011;17:
62. Stamatakis L, Siddiqui MM, Nix JW, et al. Accuracy of multiparametric 6563-6573.
magnetic resonance imaging in confirming eligibility for active sur- 81. Krohn A, Diedler T, Burkhardt L, et al. Genomic deletion of PTEN is
veillance for men with prostate cancer. Cancer. 2013;119:3359-3366. associated with tumor progression and early PSA recurrence in ERG
63. Serrao EM, Barrett T, Wadhwa K, et al. Investigating the ability of fusion-positive and fusion-negative prostate cancer. Am J Pathol. 2012;
multiparametric MRI to exclude significant prostate cancer prior to 181:401-412.
transperineal biopsy. Can Urol Assoc J. 2015;9:E853-E858. 82. Liu W, Xie CC, Thomas CY, et al. Genetic markers associated with early
64. Walton Diaz A, Shakir NA, George AK, et al. Use of serial multiparametric cancer-specific mortality following prostatectomy. Cancer. 2013;119:
magnetic resonance imaging in the management of patients with prostate 2405-2412.
cancer on active surveillance. Urol Oncol. 2015;33:202.e1-202.e7. 83. Cuzick J, Yang ZH, Fisher G, et al; Transatlantic Prostate Group. Prog-
65. Felker ER, Wu J, Natarajan S, et al. Serial MRI in active surveillance of nostic value of PTEN loss in men with conservatively managed localised
prostate cancer: incremental value. J Urol. Epub 2015 Dec 7. prostate cancer. Br J Cancer. 2013;108:2582-2589.

e244 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


ACTIVE SURVEILLANCE FOR PROSTATE CANCER

84. Lotan TL, Carvalho FL, Peskoe SB, et al. PTEN loss is associated with 91. Karnes RJ, Bergstralh EJ, Davicioni E, et al. Validation of a genomic
upgrading of prostate cancer from biopsy to radical prostatectomy. classifier that predicts metastasis following radical prostatectomy in an
Mod Pathol. 2015;28:128-137. at risk patient population. J Urol. 2013;190:2047-2053.
85. Mithal P, Allott E, Gerber L, et al. PTEN loss in biopsy tissue predicts poor 92. Den RB, Feng FY, Showalter TN, et al. Genomic prostate cancer classifier
clinical outcomes in prostate cancer. Int J Urol. 2014;21:1209-1214. predicts biochemical failure and metastases in patients after postoperative
86. Amin MB, Lin DW, Gore JL, et al. The critical role of the pathologist in radiation therapy. Int J Radiat Oncol Biol Phys. 2014;89:1038-1046.
determining eligibility for active surveillance as a management option in 93. Klein EA, Haddad Z, Yousefi K, et al. Decipher genomic classifier measured on
patients with prostate cancer: consensus statement with recommen- prostate biopsy predicts metastasis risk. Urology. Epub 2016 Jan 22.
dations supported by the College of American Pathologists, In- 94. Tosoian JJ, Loeb S, Feng Z, et al. Association of [-2]proPSA with biopsy
ternational Society of Urological Pathology, Association of Directors of reclassification during active surveillance for prostate cancer. J Urol.
Anatomic and Surgical Pathology, the New Zealand Society of Pa- 2012;188:1131-1136.
thologists, and the Prostate Cancer Foundation. Arch Pathol Lab Med. 95. Hirama H, Sugimoto M, Ito K, et al. The impact of baseline [-2]proPSA-
2014;138:1387-1405. related indices on the prediction of pathological reclassification at
87. Long Q, Xu J, Osunkoya AO, et al. Global transcriptome analysis of 1 year during active surveillance for low-risk prostate cancer: the
formalin-fixed prostate cancer specimens identifies biomarkers of Japanese multicenter study cohort. J Cancer Res Clin Oncol. 2014;140:
disease recurrence. Cancer Res. 2014;74:3228-3237. 257-263.
88. Peng Z, Andersson K, Lindholm J, et al. Operator dependent choice of 96. Tosoian JJ, JohnBull E, Trock BJ, et al. Pathological outcomes in men with
prostate cancer biopsy has limited impact on a gene signature analysis low risk and very low risk prostate cancer: implications on the practice
for the highly expressed genes IGFBP3 and F3 in prostate cancer epi- of active surveillance. J Urol. 2013;190:1218-1223.
thelial cells. PLoS One. 2014;9:e109610. 97. Cooperberg MR, Cowan JE, Hilton JF, et al. Outcomes of active sur-
89. Reichard CA, Stephenson AJ, Klein EA. Applying precision medicine to veillance for men with intermediate-risk prostate cancer. J Clin Oncol.
the active surveillance of prostate cancer. Cancer. 2015;121:3403-3411. 2011;29:228-234.
90. Erho N, Crisan A, Vergara IA, et al. Discovery and validation of a prostate 98. Han M, Partin AW, Zahurak M, et al. Biochemical (prostate specific
cancer genomic classifier that predicts early metastasis following radical antigen) recurrence probability following radical prostatectomy for
prostatectomy. PLoS One. 2013;8:e66855. clinically localized prostate cancer. J Urol. 2003;169:517-523.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e245


GENITOURINARY (PROSTATE) CANCER

Oligometastatic Disease in
Prostate Cancer: Treating the
Patient or the Scan?

CHAIR
Neha Vapiwala, MD
University of Pennsylvania
Philadelphia, PA

SPEAKERS
Christopher J. Sweeney, MBBS
Dana-Farber Cancer Institute/Harvard Cancer Center
Boston, MA

R. Jeffrey Karnes, MD
Mayo Clinic
Rochester, MN
APPROACH TO OLIGOMETASTATIC PROSTATE CANCER

Approach to Oligometastatic Prostate Cancer


Brandon Bernard, MD, Boris Gershman, MD, R. Jeffrey Karnes, MD, Christopher J. Sweeney, MBBS, and
Neha Vapiwala, MD

OVERVIEW

Oligometastatic prostate cancer has increasingly been recognized as a unique clinical state with therapeutic implications. It
has been proposed that patients with oligometastases may have a more indolent course and that outcome may be further
improved with metastasis-directed local ablative therapy. In addition, there are differing schools of thoughts regarding
whether oligometastases represent isolated lesionswhere targeted therapy may render a patient disease freeor
whether they coexist with micrometastases, where targeted therapy in addition to systemic therapy is required for maximal
clinical impact. As such, the approach to the patient with oligometastatic prostate cancer requires multidisciplinary
consideration, with surgery, radiotherapy, and systemic therapy potentially of benefit either singularly or in combination.
Indeed, mounting evidence suggests durable disease-free intervals and, in some cases, possibly cure, may be achieved with
such a multimodal strategy. However, selecting patients that may benefit most from treatment of oligometastases is an
ongoing challenge. Moreover, with the advent of new, highly sensitive imaging technologies, the spectrum based on CT of
the abdomen and pelvis and technetium bone scan of localized to oligometastatic to widespread disease has become
increasingly blurred. As such, new MRI- and PET-based modalities require validation. As some clinical guidelines advise
against routine prostate-specific antigen screening, the possibility of more men presenting with locally advanced or de novo
oligometastatic prostate cancer exists; thus, knowing how best to treat these patients may become more relevant at a
population level. Ultimately, the arrival of prospective clinical data and better understanding of biology will hopefully further
inform how best to treat men with this disease.

O ligometastatic prostate cancer is increasingly being


considered as a unique clinical situation and challenge.
Historically, distinctions with respect to lesser or greater
disease is most likely also associated with micrometastatic
disease; therefore, concurrent systemic therapy with lo-
calized therapy should be considered the optimal treatment.
extent of metastatic disease burden were not typically In the latter case, disease at this stage is considered po-
made, as patients were all treated with androgen depriva- tentially curable as it is with adjuvant systemic therapy (in
tion therapy (ADT). Men with oligometastatic prostate the form of ADT) to radiotherapy (RT) for high-risk localized
cancer were thus deemed incurable and treated systemically disease or microscopic lymph nodepositive disease fol-
with ADT alongside those with widespread metastases. lowing radical prostatectomy.5-8
However, recent advances in imaging are identifying men With this uncertainty, there is provider and patient bias
with potentially isolated metastases, and mounting evi- toward certain treatmentsoptions include ADT alone,
dence suggests that durable control is attainable with focused ablative therapy of radiographic disease alone, or
treatment modalities targeting oligometastases, either with both. Moreover, the role of docetaxel with ADT is also now a
or without the use of systemic therapy.1-3 Such data support matter for consideration. Specifically, some providers and
the hypothesis that oligometastatic prostate cancer may patients will prefer less therapy with possible risk of
represent a unique biologic state with its own natural history undertreatment and poorer cancer control, whereas others
requiring a distinct therapeutic approach.4 That said, there will prefer more therapy with risk of overtreatment and
are currently no clear-cut data or validated tools to guide consequent adverse events. Furthermore, with greater use
optimal therapy for an individual patient. One school of of highly sensitive imaging technologies (e.g., prostate-
thought is to consider isolated lesions on imaging as rep- specific membrane antigen [PSMA], choline, or sodium
resenting the only sites of disease, where local therapy is fluoride [NaF] PET and total body MRI), patients are likely
sufficient. The other viewpoint is that oligometastatic being identified earlier in the natural history of the disease

From the Dana-Farber Cancer Institute, Boston, MA; Harvard Medical School, Boston, MA; Mayo Clinic, Rochester, MN; University of Pennsylvania, Philadelphia, PA.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Neha Vapiwala, MD, Department of Radiation Oncology, Perelman Center for Advanced Medicine, 3400 Civic Center Blvd., TRC 2 West, Philadelphia, PA 19104;
email: vapiwala@uphs.upenn.edu.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 119


BERNARD ET AL

and would have been considered micrometastatic on scan and CT of the abdomen and pelvis remains the gold
standard imaging (CT abdomen and pelvis and technetium standard to define oligometastatic prostate cancer.
bone scan).1 It is clear in light of this latter observation that To help set the stage, the following five cases detail the
incorporating these technologies into the staging and spectrum of patients presenting with oligometastatic dis-
monitoring of men with prostate cancer requires rigorous ease and different management strategies available:
testing to define how best to integrate them into standard 1. A 63-year-old man diagnosed with a Gleason 9 adeno-
clinical care. carcinoma of the prostate, PSA 11, and an isolated
In 2012, the U.S. Preventive Services Task Force updated biopsy-positive lesion at T11. He was started on ADT
their guidelines on prostate cancer screening to recommend with a PSA decline to 2.5 and received a radical pros-
against prostate-specific antigen (PSA) screening in all men9; tatectomy 2 months thereafter with negative margins.
since then, there has been a documented decrease in PSA Stereotactic radiosurgery was subsequently delivered to
screening and in incidence of early-stage prostate cancer.10
T11, and ADT was continued, with a PSA nadir of 0.02
It is yet unknown whether this change has increased the
12 months after radical prostatectomy. At that time, a CT
number of men presenting with more advanced disease and
urogram for nephrolithiasis showed multiple small
consequent higher risk of relapse following local therapy,
however it has been estimated that up to an extra 1,241 men sclerotic bone lesions within the vertebral column
may die of prostate cancer in the United States per year from lack consistent with metastases. PSA has risen to 0.3 with a
of screening.11 Moreover, it is unknown whether there is an testosterone level of less than 7.0 ng/dL (lower limit of
absolute increase in the number of men presenting with de novo normal: 240 ng/dL) 16 months after ADT initiation.
metastatic prostate cancer. As a proportion of such men will have 2. A 66-year-old man presented with localized left hip pain
oligometastatic disease at presentation, having a standardized and a PSA of 1,244. CT showed an abnormal prostate
approach to its assessment and management is necessary. concerning for tumor, as well as an expansile lytic lesion
To design a systematic approach to oligometastatic in the left pubic bone. Prostate biopsy confirmed Gleason
prostate cancer, first and foremost a universally accepted 8 adenocarcinoma, and he was treated with concurrent
definition is required. To that end, variability exists regarding ADT plus RT to the prostate and isolated bone metas-
what constitutes oligometastases, with cutoffs of both tasis. Six months after starting ADT, his PSA had fallen to
three or fewer and five or fewer lesions used in the liter- 0.01. With his PSA still undetectable after 24 months of
ature, and others in actual practice.3,12 Importantly, an therapy, ADT was stopped and he entered surveillance.
accepted definition should require that targeted treatment
As of last follow-up, PSA was 0.04 with a testosterone
of all metastatic lesions with surgery or RT is technically
level of 168 3 years after ADT cessation.
feasible. In addition, standardization of imaging is para-
3. A 60-year-old man with a PSA of 6.6 underwent a
mount; given the uncertainty of the role of newer tech-
nologies in routine care, conventional imaging with bone prostate biopsy identifying Gleason 9 adenocarci-
noma. Subsequent radical prostatectomy found locally
advanced disease (pT3bN1M0) with evidence of
extracapsular extension, seminal vesicle invasion, and a
single positive lymph node. Postoperative PSA was 2.6,
KEY POINTS and the patient received salvage treatment with
7 months of ADT and concurrent intensity-modulated
Oligometastatic prostate cancer is a unique clinical state
with an inherently more indolent tumor biology
radiation therapy (5,040 cGy to the whole pelvis; 7,020
susceptible to metastasis-directed focal ablative cGy to prostatic bed). His PSA became undetectable but
therapy. 12 months later rose to 2.5, prompting reinitiation of
Oligometastatic prostate cancer may represent isolated ADT and the eventual emergence of castration re-
sites of disease or coexist with micrometastases. sistance. Imaging demonstrated a single metastasis in
Modern, highly sensitive imaging technology is the left scapula. He was treated with seven cycles of
potentially identifying patients with oligometastatic
docetaxel; however, PSA continued to rise to 6.5. At that
prostate cancer who would be deemed to have localized
disease or no evidence of radiographic disease by time, left scapular pain and progression of the lesion on
standard imaging of CT of the abdomen and pelvis and bone scan led to treatment with 250 cGy of palliative RT
bone scan. in 20 fractions (5,000-cGy total dose). His PSA has sub-
A multimodal approach to patients with oligometastatic sequently dropped to 0.75 with resolution of symptoms.
disease is needed, with evidence for surgery, 4. A 50-year-old man underwent robotic-assisted radical
radiotherapy, and systemic therapy, alone or in
prostatectomy with pathology demonstrating pT3a NX
combination, improving patient outcomes.
Further prospective data are needed to best select prostate adenocarcinoma. His received radiation ther-
patients with oligometastatic prostate cancer most apy to the prostatic fossa for a persistently elevated
likely to benefit from a given therapeutic approach. PSA postoperatively. Following radiation, PSA remained
elevated. He was placed on ADT but developed

120 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


APPROACH TO OLIGOMETASTATIC PROSTATE CANCER

castration resistance with a rising PSA. When the This review will outline the role of surgery, RT, and sys-
PSA rose to 3, 11C-choline PET/CT demonstrated temic therapy in the management of oligometastatic pros-
node-only disease in the pelvis and retroperitoneum tate cancer. Table 1 shows data from studies of focal therapy
(Fig 1). He elected to undergo combined salvage with surgery or RT alone, systemic therapy with ADT alone,
extended pelvic lymph node dissection and retro- or systemic therapy with ADT plus chemotherapy in men
peritoneal lymph node dissection. Pathology dem- with oligometastatic prostate cancer. Current evidence and
onstrated 9 of 62 lymph nodes involved with prostate existing controversies will be discussed.
adenocarcinoma. His PSA was less than 0.2 at 6 months
after surgery.
5. A 49-year-old man presented with an elevated PSA to
THE ROLE OF SURGERY IN OLIGOMETASTATIC
PROSTATE CANCER
49. Prostate biopsy demonstrated Gleason 9 prostate
The role of surgery in the management of oligometastatic
adenocarcinoma. CT scan revealed pelvic lymphade-
prostate cancer has received increased attention in re-
nopathy (Fig 2). Bone scan was without evidence of cent years. However, the concept of surgery for advanced
osseous metastatic disease. He elected to proceed prostate cancer is not new. For instance, observational
with radical retropubic prostatectomy and extended studies from the 1990s reported improved oncologic out-
bilateral pelvic lymphadenectomy as part of a multi- comes for men with pathologic lymph nodepositive (pN+)
modal management plan. Surgical pathology dem- prostate cancer treated with radical prostatectomy and ADT
onstrated Gleason 9 pT3b N1 (7 of 27 lymph nodes compared with ADT alone, and prior radical prostatectomy
positive) prostate adenocarcinoma. He is receiving 2 has been associated with improved overall survival (OS)
years of adjuvant ADT and underwent a course of for men with metastatic disease.6-8,13 Given that aggres-
adjuvant pelvic radiation therapy at 6 months after sive prostate cancer can persist even after 1 year of sys-
surgery. temic therapy, it is not surprising that surgery may improve

FIGURE 1. Case 4

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 121


BERNARD ET AL

FIGURE 1. Case 4 (Contd)

A 50-year-old man that received salvage extended pelvic and retroperitoneal lymph node dissection for choline PET-positive disease post-RP and adjuvant RT with subsequent
PSA decline.

oncologic outcomes.14 Potential mechanisms include re- and 50% to 75%, respectively, for men treated with radical
ducing tumor burden (i.e., resetting the clock), preventing prostatectomy plus ADT, compared with 50% to 65% and 25%
continued metastatic spread from the primary, and relieving to 30%, respectively, for ADT alone.17,18 Moreover, random-
tumor immune suppression.15 ized data reinforce these figures. Among men treated with
radical prostatectomy and immediate ADT in ECOG 3886, OS
was 64% at a median of 11.9 years versus 45% in those
Surgery for Locally Advanced (Including Pelvic Lymph managed with radical prostatectomy alone.19 Notably among
Node) Disease those managed with ADT alone in EORTC 30846, 10-year OS
For men that have not had prior local therapy, radical was approximately 30%, although approximately 40% of the
prostatectomy has been associated with improved oncologic deaths were due to nonprostate cancer.20
outcomes for men with pN+ disease in a number of ob- Risk stratification is essential for men with pN+ disease,
servational studies. For example, in a matched comparison as some will have durable oncologic control with surgical
of 79 men treated with radical prostatectomy and adju- resection alone, whereas others will require multimodal
vant orchiectomy versus 79 men treated with orchiectomy therapy. One multi-institutional analysis reported that men
alone, 10-year OS was 66% versus 28% in favor of radical with two or fewer positive nodes had significantly better
prostatectomy.8 Similarly, an analysis of the Munich Cancer 15-year CSS than those with more than two nodes (84% vs. 62%;
Registry reported 10-year OS of 64% for completed versus p , .001) when treated with radical prostatectomy and
28% for aborted radical prostatectomy in men with pN+ adjuvant ADT.21 Another study similarly identified more
disease.16 Although there is inherent potential for selection than two positive nodes as an adverse prognostic feature,
bias toward radical prostatectomy for patients with more along with Gleason score greater than 7.22 Most recently,
favorable disease, there is remarkable consistency in reported Moschini et al23 developed a risk score for cancer-specific
oncologic outcomes among observational studies with 10-year mortality (CSM) among pN+ men after radical prostatectomy
cancer-specific survival (CSS) and OS ranging from 70% to 85% with 88% receiving adjuvant ADT. Points are added for three

122 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


APPROACH TO OLIGOMETASTATIC PROSTATE CANCER

FIGURE 2. Case 5 metastases.27 In a multi-institutional report of 106 men who


underwent radical prostatectomy/extended pelvic lymph
node dissection (ePLND) for M1a/M1b disease, CSS was
89% at a median of 22.8 months, and perioperative mor-
bidity was overall similar to that reported for radical
prostatectomy/ePLND for M0 disease.28 Another study
compared 23 patients with low-volume M1b disease (three
or fewer skeletal lesions) who underwent radical
prostatectomy/ePLND with 38 controls treated with ADT
alone.29 Cytoreductive radical prostatectomy was associated
with longer time to castration resistance (40 vs. 29 months),
improved clinical progression-free survival (PFS) (39 vs.
28 months), and improved CSS (96% vs. 84%). Population-
based data likewise support an oncologic benefit to local
therapy with surgery or radiation.30 A randomized trial eval-
uating best systemic therapy with or without local therapy in
the form of surgery or radiation for men with metastatic
prostate cancer should provide important prospective data
(NCT01751438).
Risk stratification to guide patient selection for surgery in
the setting of M1 disease is even more critical than for N+M0
disease. To this end, Fossati et al31 conducted a population-
based analysis of men with metastatic prostate cancer and
reported that baseline CSM risk significantly (p , .0001)
modified the oncologic benefit of local therapy (either
surgery or radiation), with no benefit to local therapy
beyond a specific threshold. Further studies are necessary to
determine which patient populations may derive greatest
A 49-year-old man that received radical prostatectomy and up-front bilateral benefit from cytoreductive radical prostatectomy in the
extended pelvic lymphadenectomy for clinically lymph nodepositive disease
followed by adjuvant RT and long course ADT.
presence of M1 disease.
In the setting of clinical nodal recurrence following de-
finitive local therapy, salvage lymph node dissection (sLND)
or more positive lymph nodes, Gleason 710 disease, positive has gained increased utilization, driven in part by improved
surgical margins, and absence of adjuvant RT to categorize imaging modalities such as 11C-choline PET/CT. Principles of
patients into risk groups with 10-year CSM ranging from 19% surgery for oligometastatic nodal disease include availability
to 46%. of accurate preoperative imaging, complete resection of all
There is increasing evidence that pN+ prostate cancer radiographic disease, and acceptable morbidity. Although
does not uniformly signify systemic disease. In one study of randomized data are lacking, observational studies are
clinical recurrence patterns in 1,011 pN+ men after radical encouraging. One U.S. series reported that, of 52 men who
prostatectomy, 57% of clinical recurrences were identified underwent sLND for nodal recurrence after radical prosta-
at a single location, and of these, 51% were local or node-only at tectomy, 73% achieved undetectable PSA after surgery.32
the time of relapse.24 In light of such data, management of clinical In a follow-up study of 117 patients, the largest single series
lymphadenopathy (cN+), which has traditionally been distin- to date, 5-year biochemical recurrence (BCR)free, radio-
guished from cN0pN+ disease, deserves reconsideration. For logic recurrence-free, and CSS were 39%, 51%, and 97%,
example, the National Comprehensive Cancer Network (NCCN) respectively, after sLND with adjuvant ADT being given to
recommends ADT with or without radiation therapy for men 62.7% of patients.33 In a European series of 59 men who
presenting with cN1M0 disease.25 However, in a recent study of underwent sLND, 59% achieved undetectable PSA, and
302 men with pN+ disease, of whom 17% were cN+, the authors among these, the 8-year BCR-free survival was 23%.34 For the
noted no difference in CSS or OS between cN+ and cN0 pa- overall cohort, 8-year clinical recurrence-free and CSS were
tients.26 Such data suggest that appropriately selected men with 38% and 81%, respectively. A systematic review recently
clinical lymphadenopathy may be treated with surgical resection synthesized the available data on sLND with or without ad-
as an alternative to pelvic radiation as for cN0pN+ disease. juvant ADT from 12 observational studies, 5-year BCR-free
survival of 9% to 22%, and 5-year OS of approximately 75%.35
Favorable prognostic factors included complete PSA response
Surgery in Presence of Distant Metastatic Disease to surgery alone, fewer positive nodes, absence of retroperi-
In the absence of prior local treatment, there is considerably toneal involvement, and lower preoperative PSA. What is not
less data on the role of surgery in the setting of distant known is the optimal utilization and duration of adjuvant

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 123


BERNARD ET AL

TABLE 1. Summary of Key Studies Evaluating Surgery, Radiotherapy, and Systemic Therapy in Oligometastatic
Prostate Cancer in the Post-PSA Era

Study No. of Patients Study Type Imaging Outcome


Surgery Alone
pN+ Disease
Engel et al16 938 Retrospective cohort N/A (pN+) 10-year OS of 64% with RP vs. 28% without RP
ECOG 388619 98 RCT N/A (pN+) 11.9-year OS of 64% with RP + immediate
ADT vs. 45% with RP + deferred ADT
EORTC 3084620 234 RCT N/A (pN+) 10-year CSS of 48% for immediate ADT vs.
44% for delayed ADT (none with RP)
sLND
Zattoni et al33 117 Case series 11
C-Choline PET/CT 5-year 69% BCR-free, 49% radiographic
recurrence-free, and 97% CSS
Suardi et al34 59 Case series 11
C-Choline PET/CT 8-year 23% BCR-free, 38% clinical
recurrence-free, and 81% CSS
M1 Disease
Sooriakumaran et al28 106 Case series Bone scan, CT 89% CSS at median of 22.8 months with
cytoreductive RP for M1 disease
Heidenreich et al29 61 Retrospective cohort Bone scan, CT CSS of 96% with RP + ADT vs. 84% with
ADT alone, at median of 34.5 months
RT Alone
Jereczek-Fossa et al59 19 Case series PET/CT 30-month PFS, 64%
Schick et al60 50 Case series PET/CT; bone scan 3-year OS, 92%
Decaestecker et al61 50 Case series PET/CT Median PFS, 19 months
Picchio et al62 83 Case series PET/CT PSA PR/CR, 83%
ADT Alone
Millikan et al47 84 Phase III RCT Bone scan Median OS, 94 months
S934663 798 Phase III RCT Bone scan; CXR Median OS, 83 months
GETUG 1553 102 Phase III RCT Bone scan; CT Median OS, 83 months
CHAARTED41 143 Phase III RCT Bone scan; CT 4-year OS, 60%
CHT
Millikan et al47 76 Phase III RCT Bone scan Median OS, 93.6 months
GETUG 1553 100 Phase III RCT Bone scan; CT 4-year OS, 70%
CHAARTED41 134 Phase III RCT Bone scan; CT 4-year OS, 70%
Abbreviations: N/A, not applicable; RCT, randomized controlled trial; RP, radical prostatectomy; BCR, biochemical recurrence; ADT, androgen deprivation therapy; PR, partial
response; CR, complete response; CXR, chest x-ray.

systemic therapy with ADT, and whether concurrent docetaxel involvement versus 53% without retroperitoneal disease.37
may possibly further improve these outcomes. The phase II randomized trial STOMP will evaluate the
role of metastasis-directed therapy with either surgery or
Studies on Resection of Distant Metastases After SBRT in oligometastatic recurrence following local therapy
Local Therapy (NCT01558427) compared with active surveillance with ADT
Data reporting outcomes of surgery in this setting have been to be deferred in both arms until more than three meta-
limited to retroperitoneal lymph nodes as part of sLND, static lesions develop.38 This should provide important data
whereas stereotactic body radiation therapy (SBRT) has regarding the potential oncologic benefit of both meta-
generally been used for bone or visceral oligometastatic stastectomy and sLND as monotherapy.
disease.35,36 Although retroperitoneal disease portends a Overall, emerging data suggest that surgery may represent
worse prognosis, data suggest that a subset of patients an important therapeutic modality in the management of
may achieve durable clinical recurrence-free survival with oligometastatic prostate cancer. Ultimately, optimal patient
sLND with or without adjuvant ADT. One study of sLND selection is critical. Radical prostatectomy/ePLND is asso-
reported 8-year clinical recurrence-free survival of 22% ciated with durable oncologic control in the setting of N+M0
for retroperitoneal nodal involvement versus 69% without disease, with emerging data for its use as local therapy in M1
retroperitoneal disease.34 Another study noted 5-year disease. sLND is likewise associated with favorable oncologic
clinical recurrence-free survival of 11% with retroperitoneal outcomes for oligometastatic nodal recurrence after local

124 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


APPROACH TO OLIGOMETASTATIC PROSTATE CANCER

therapy. Prospective studies will further inform these issues p = .01). Also, a nonsignificant trend was observed for improved
and will also inform whether it can be used alone or in median distant PFS with adjuvant ADT (25 vs. 18 months;
combination with systemic therapy. p = .09). As in the review by Ost et al, most of the patients were
staged using PET/CT36; however, as this technique is not
currently considered standard of care, extrapolating and ap-
THE ROLE OF RADIATION THERAPY IN plying these data to clinical practice is difficult.
OLIGOMETASTATIC PROSTATE CANCER An important consideration when applying RT in any disease
Radiation therapy is a well-established curative therapy for setting is the accurate deposition of dose to the target while
intact localized and postoperative locally recurrent prostate avoiding dose-limiting toxicity to surrounding tissue. There are
cancer, and an effective palliative therapy for symptomatic several factors in the evolution of modern RT that are helping to
metastatic disease. Conversely, its role in oligometastatic mitigate this concern. Enhanced imaging modalities and image
disease in an effort to improve survival is unknown. Cur- registration have led to improved accuracy in RT target defi-
rently, the NCCN guidelines recommend considering RT for nition, while advances in both external patient and internal
metastases if symptomatic or residing in a weight-bearing organ immobilization and tracking permit more precise radi-
bone; otherwise, the mainstay for M1 disease remains ation delivery. Furthermore, major technical evolutions in
systemic with either medical or surgical castration, or hardware and software capabilities have enabled safe de-
chemohormonal therapy (CHT).39 However, ADT is associ- livery of larger doses of radiation per treatment fraction
ated with a number of side effects and potential compli- (i.e., hypofractionation); such dosing regimens are not only a
cations, so being able to delay ADT start and/or allow for desirable from a patient convenience standpoint but can carry
longer treatment breaks if given intermittently without greater radiobiological bang for your buck, delivering a po-
compromising survival is a worthy pursuit. Moreover, a tentially higher biologically effective dose over a shorter time
shorter course of ADT with concurrent RT to maximize period.2 Consequently, various hypofractionated radiation
control may be another strategy to limit potential toxicity therapy dosing schedules have been proposed for treatment in
associated with ADT. Plausibly, early detection of oligo- the oligometastatic setting, including one fraction of 20 Gy,
metastases with sensitive, accurate imaging may allow for three fractions of 10 Gy, five to six fractions of 5 or 6 Gy, and 10
more proactive therapy and eradication of locally measur- fractions of 5 Gyall deemed to be of equivalent efficacy.2
able disease, delayed time to ADT, and, perhaps, cure if there In summary, a growing body of retrospective literature
is no associated micrometastatic disease and radiation is suggests RT for oligometastatic prostate cancer is feasible,
able to eradicate the isolated metastatic lesion(s). In the effective, and without significant toxicity. Although un-
setting of postprostatectomy biochemical failure, the me- proven, its role as a solitary therapy or in conjunction with
dian time from PSA recurrence to metastases may be long systemic and/or surgical strategies is evolving, as is the goal
(8 years in one study)40; regular monitoring and prompt and ultimate intent (i.e., aggressive palliation vs. durable
utilization of metastases-directed focal therapy upon their control vs. eradication and cure) of its use. Whether the
development may further improve outcomes and avoid the primary function of radiation in the oligometastatic setting
need for systemic therapy. to primary (if intact) and/or metastasis is to delay time to
Recently, Ost et al36 conducted a review of studies ADT, to consolidate potentially curative multimodal ap-
employing metastases-directed therapy for relapsed pros- proaches, or both, is to be determined. Numerous clinical
tate cancer. Of note, oligometastases were identified using trials are currently in development that will provide pro-
PET/CT in 98% of patients, and 78% of treated metastases spective data to better answer this question (NCT01859221;
were nodal in origin. Although great heterogeneity among NCT01777802; NCT02192788; NCT01558427).
patient populations and variable use of ADT and prophylactic
nodal irradiation made comparison among studies chal-
lenging, the authors found that around one-half of men were THE ROLE OF SYSTEMIC THERAPY IN
progression-free 13 years after metastases-directed ther- OLIGOMETASTATIC PROSTATE CANCER
apy; however, over 60% had adjuvant ADT. Caution should The cornerstone of therapy for metastatic prostate cancer
be taken when evaluating PFS between these various is ADT, either continuous or intermittent. Additionally,
studies, however, as different definitions were used among chemotherapy has been shown to improve OS, both in
them. In addition, although promising, the quality of the the hormone-sensitive and castration-resistant state.41-45 Al-
evidence generated to date is insufficient to recommend this though not curative, goals of systemic therapy include disease
approach as routine standard care. control, symptom reduction, reduction of morbidity, and life
The impact of RT, specifically, on PFS when used to treat prolongation. However, its role in oligometastatic prostate
recurrent, treatment-naive oligometastatic prostate cancer cancer, specifically, is less clear. There is speculation that
was recently the focus of a multi-institutional analysis.3 The control of micrometastatic disease, in conjunction with tar-
study included 163 patients with three or fewer metastases geted treatment to oligometastases, may improve outcome
each. The authors assessed for both local PFS and distant PFS, and, in some, may possibly lead to cure. Chemotherapy may aid
and identified a RT dose of less than or equal to 100 Gy as being by theoretically eliminating less androgen-sensitive tumor cells
associated with a lower local PFS at 3 years (79% vs. 99%; and augment what can be achieved with ADT.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 125


BERNARD ET AL

Prognostically, it has been shown that patients with high- assess the value of adding docetaxel, zoledronic acid, or both
volume disease (visceral metastases and multiple bone drugs combined to standard of care in men with high-risk,
metastases [e.g., four or more]) have worse outcomes.46,47 locally advanced, metastatic or relapsed prostate cancer.42
Furthermore, it was shown that those with five or fewer Standard of care consisted of ADT for all subgroups, with RT
bone metastases fared better than those with more than five optional but later required for those with localized disease.
at time of relapse following RT for localized disease, with This study used a novel, flexible trial design to pragmatically
distinct natural histories apparent for these patient sub- ascertain the impact of therapy along the spectrum of ad-
sets.48 Thus, patients with low-volume metastatic prostate vanced prostate cancer, with 2,265 participants (76%)
cancer at time of relapse appear to innately have more having node-positive, node-unknown, or distant metastases.
indolent biology and more favorable prognoses compared The addition of docetaxel to standard of care resulted in a
with those with more widespread disease. Attempting to 10-month improvement in OS (HR 0.78; 95% CI, 0.660.93;
improve their survival further with metastases-directed p = .006). Importantly, the effect of docetaxel plus standard
therapy, with or without systemic therapy, is an area of of care (ADT for M1 and ADT plus radiation for most of those
great interest. with M0) was consistent across all subgroups, suggesting
Notably, it has been shown that delayed ADT for meta- that locally advanced disease (with high risk of micro-
static prostate cancer does not negatively impact outcome metastases) and node-positive patients also derived a
of patients with oligometastatic prostate cancer, with one benefit from early CHT. However, the OS benefit with CHT
study reporting a median time from metastases to prostate was strongest and significant (p = .005) for those with
cancerspecific death of 82 months.49 Moreover, in men metastatic disease (note that there was no breakdown by
with PSA-only relapse following radical prostatectomy and volume of disease), with nonmetastatic patients deriving a
deferred ADT, it has been demonstrated that metastasis- failure-free survival (FFS) benefit, although not enough
free survival may serve as a surrogate for OS.50 This study events have occurred for an OS benefit at this time. Con-
also identified number of metastases (three or fewer vs. four sistent with the benefit of CHT in localized high-risk setting is
or more, hazard ratio [HR] 0.50; 95% CI, 0.290.85; p = .012) the survival benefit seen with the phase III RTOG 0521 trial,
as an independent prognostic variable for OS, further im- demonstrating a 32% reduction in risk of death at 4 years
plying potential biologic differences between patients with with adjuvant CHT following concurrent ADT plus RT. In
oligometastases and those with diffuse disease.50 It is addition, men that received docetaxel derived an improved
postulated that an improvement in outcome for those 5-year FFS (HR 0.76; 95% CI, 0.571.00; p = .05). Likewise, the
with oligometastatic disease treated with focal ablative phase III GETUG 12 trial showed CHT plus RT conferred a
therapy of their radiographic evident disease may be aug- reduction in risk of relapse or death of 29% at 8 years over
mented with systemic therapy in an attempt to eradicate ADT plus RT alone51; longer follow-up is required to see
micrometastases. whether this benefit extends to OS as well.
Evidence for improved survival with early use of CHT The third trial, GETUG 15, was the first to explore CHT in
consisting of docetaxel chemotherapy plus ADT comes from metastatic hormone-sensitive prostate cancer.52 Although
three randomized, controlled trials. The first to demonstrate the early results did not suggest a benefit from CHT, longer
the benefit, E3805 (CHAARTED), documented a 13.6-month follow-up and harmonizing volume of disease definitions
OS advantage with ADT plus six cycles of docetaxel versus with those of CHAARTED revealed more similar results to
ADT alone (HR for death 0.61; 95% CI, 0.470.80; p , .001).41 those seen in CHAARTED, especially for those with high-
This benefit was even more pronounced in those with high- volume disease.53 Specifically, it was found that CHT resulted
volume disease (defined as visceral metastases, four or more in a 13.5-month improvement in median OS at 7 years
bone metastases with at least one beyond the vertebral (HR 0.88; 95% CI, 0.681.14; p = .3), with a post hoc analysis
column and pelvis, or both), with an OS gain of 17.0 months identifying a 22% reduction in risk of death in men with high-
with CHT (HR 0.60; 95% CI, 0.450.81; p , .001). Further- volume disease, as per the CHAARTED definition.53 Fur-
more, CHT conferred an 8.5-month improvement in me- thermore, radiographic PFS was greater in those that re-
dian time to symptomatic, PSA, or radiographic progression ceived CHT in the total population (HR 0.69;95% CI,
(HR 0.61; 95% CI, 0.510.72; p , .001). When patients with 0.550.87; p = .002) and in those with high-volume disease
protocol-defined low-volume disease were assessed as a (HR 0.61; 95% CI, 0.440.83; p = .002).
separate subgroup, there was also a 39% decrease risk of The utility of CHT in the subgroup of low-volume meta-
deathbut there were far fewer events, and at time of the static hormone-sensitive prostate cancer is less well defined
first report, this was not statistically significant. This in- at early follow-up of the CHAARTED trial. However, when
dicates that these patients have a longer natural history with one considers the totality of the data of adjuvant therapy in
ADT alone with 4-year OS of approximately 60%. Moreover, high-risk localized disease and benefit in the overall pop-
some of these patients with a shorter life expectancy be- ulation of metastatic disease (STAMPEDE and CHAARTED),
cause of age and/or comorbidities may die of a competing it is quite possible that, for patients with oligometastatic
cause. prostate cancer who are at risk for dying of their disease,
These data were supported by those from the STAMPEDE there is a benefit from CHT. At the other end of the
trial, which used a multiarm, multistage platform design to spectrum, studies for patients with low-volume disease

126 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


APPROACH TO OLIGOMETASTATIC PROSTATE CANCER

aimed at avoiding cytotoxic chemotherapy and its associ- DISCUSSION


ated adverse events are currently under investigation, with A number of unanswered questions remain relevant to the
a different hormone-based approach using the competitive discussion of oligometastatic prostate cancer. It clearly has a
androgen receptor antagonist, ARN-509, in combination longer natural history and presumably has a more indolent
with ADT versus ADT alone in chemotherapy-naive pa- biology, most of the time. That said, it is unknown at this time
tients with low-volume mHSPC (NCT02489318). Of note, whether there will be a higher incidence of more advanced and
docetaxel can be prescribed per investigator choice in this de novo metastatic disease with decreased PSA screening in
study. the United States. If this is the case, there will be more patients
Overall, collective data from GETUG 15, CHAARTED, and with metastatic disease and an even greater need at the public
STAMPEDE support the hypothesis that an aggressive ap- health level to improve therapy for metastatic hormone-
proach with systemic therapy early in the metastatic setting sensitive prostate cancer. Related to diagnosis is what, if
improves outcome. Indeed, a recent meta-analyses of data any, impact the new imaging modalities will have on the course
from these studies confirmed the improved survival of CHT of disease. For example, for the patients identified with oli-
over ADT (HR, 0.77; 95% CI, 0.680.87; p , .0001).54 It is gometastases by such highly sensitive tests, is RT to the
therefore logical to apply the principle of systemic therapy metastatic lesion alone, with less systemic effects, sufficient or
with localized/focal ablative therapy in the setting of PSA is the patient best treated with ADT plus RT? Moreover, there
relapse and high-risk localized disease. is a paucity of data regarding whether the total number and
The large phase III RTOG 9601 trial tested whether location of oligometastases (by standard CT and technetium
2 years of anti-androgen therapy plus salvage RT was bone scan imaging) is prognostically important. Identifying the
superior to RT alone in men with PSA relapse follow- patients most likely to benefit from treatment is needed.
ing radical prostatectomy.55 Combined therapy resulted As detailed above, treatment of oligometastases with focal
in a 9% reduction in absolute incidence of metastases (14% ablative therapies appears promising but requires further vali-
vs. 23%; p , .001) and 5% reduction in incidence of prostate dation in prospective studies before it can be recommended
cancerspecific death (2.3% vs. 7.5%; p , .001) at 12 years. as standard of care.36 Data supporting treatment of lymph
Another retrospective study found that patients that re- nodeonly recurrence is also limited, although a recent review
ceived salvage RT plus concurrent ADT had a decreased risk suggested it may confer a therapeutic benefit.35 When designing
of relapse when treated at a PSA relapse of less than or equal such prospective studies, there is differing opinion whether delay
to 0.5 ng/mL following prostatectomy, although the effect in requiring ADT, OS, or both should be the primary endpoint.
appeared limited to those with negative surgical margins.56 Ultimately, the decision regarding when, and how, to treat
This is further evidence that focal ablative therapy with men with oligometastatic prostate cancer requires an as-
systemic therapy improves outcome in the relapsed setting sessment of the competing risks of burden of therapy versus
and supports the role of combined modality therapy in low- those of progressive disease and subsequent therapies. Such an
volume recurrent disease. evaluation requires incorporating factors such as symptoms,
In attempting to select those with oligometastatic disease comorbidities, and life expectancy. Specifically, competing risks
for further metastases-directed therapy, both deep PSA of death are very relevant for patients with oligometastatic
nadir and absence of PSA progression on ADT may predict for hormone-sensitive prostate cancer. Moreover, actively en-
improved survival.57,58 Such patients may be the ones who gaging patients in the therapeutic discussion, defining goals of
benefit the most from targeted treatments to select lesions. care, and enabling shared decision making are all required.
In total, there is growing evidence that more aggressive Further prospective, randomized data are necessary to better
therapy early in the course of disease leads to improved characterize the nature of oligometastatic prostate cancer and
outcome, and therefore utilizing systemic therapy as part define the role and value of therapies in this state. These trials
of a multimodal approach with focal ablative therapy to should take into account the gaps in knowledge described
radiographic evident disease for patients with oligometa- herein, and help to address the most pressing quandaries
static prostate cancer should be considered. facing clinicians and patients today.

References
1. Reyes DK, Pienta KJ. The biology and treatment of oligometastatic 4. Weichselbaum RR, Hellman S. Oligometastases revisited. Nat Rev Clin
cancer. Oncotarget. 2015;6:8491-8524. Oncol. 2011;8:378-382.
2. Conde Moreno AJ, Ferrer Albiach C, Muelas Soria R, et al. Oligome- 5. Bolla M, Van Tienhoven G, Warde P, et al. External irradiation with or
tastases in prostate cancer: restaging stage IV cancers and new ra- without long-term androgen suppression for prostate cancer with high
diotherapy options. Radiat Oncol. 2014;9:258. metastatic risk: 10-year results of an EORTC randomised study. Lancet
3. Ost P, Jereczek-Fossa BA, As NV, et al. Progression-free survival Oncol. 2010;11:1066-1073.
following stereotactic body radiotherapy for oligometastatic prostate 6. Cheng CW, Bergstralh EJ, Zincke H. Stage D1 prostate cancer. A non-
cancer treatment-naive recurrence: a multi-institutional analysis. Eur randomized comparison of conservative treatment options versus
Urol. 2016;69:9-12. radical prostatectomy. Cancer. 1993;71(3, Suppl):996-1004.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 127


BERNARD ET AL

7. Frohmuller HG, Theiss M, Manseck A, et al. Survival and quality of life of 25. Mohler JL, Kantoff PW, Armstrong AJ, et al; National Comprehensive
patients with stage D1 (T1-3 pN1-2 M0) prostate cancer. Radical Cancer Network. Prostate cancer, version 2.2014. J Natl Compr Canc
prostatectomy plus androgen deprivation versus androgen deprivation Netw. 2014;12:686-718.
alone. Eur Urol. 1995;27:202-206. 26. Moschini M, Briganti A, Murphy CR, et al. Outcomes for patients with
8. Ghavamian R, Bergstralh EJ, Blute ML, et al. Radical retropubic pros- clinical lymphadenopathy treated with radical prostatectomy. Eur Urol.
tatectomy plus orchiectomy versus orchiectomy alone for pTxN+ 2016;69:193-196.
prostate cancer: a matched comparison. J Urol. 1999;161:1223-1227, 27. Bayne CE, Williams SB, Cooperberg MR, et al. Treatment of the primary
discussion 1227-1228. tumor in metastatic prostate cancer: current concepts and future
9. U.S. Preventive Services Task Force. Final Update Summary: Prostate Cancer: perspectives. Eur Urol. Epub 2015 May 20.
Screening. U.S. Preventive Services Task Force. May 2012. http://www. 28. Sooriakumaran P, Karnes J, Stief C, et al. A multi-institutional analysis of
uspreventiveservicestaskforce.org/Page/Document/UpdateSummaryFinal/ perioperative outcomes in 106 men who underwent radical prosta-
prostate-cancer-screening. Accessed January 15, 2016. tectomy for distant metastatic prostate cancer at presentation. Eur
10. Jemal A, Fedewa SA, Ma J, et al. Prostate cancer incidence and PSA Urol. Epub 2015 May 30.
testing patterns in relation to USPSTF screening recommendations. 29. Heidenreich A, Pfister D, Porres D. Cytoreductive radical prostatectomy
JAMA. 2015;314:2054-2061. in patients with prostate cancer and low volume skeletal metastases:
11. Penson DF. The pendulum of prostate cancer screening. JAMA. 2015; results of a feasibility and case-control study. J Urol. 2015;193:832-838.
314:2031-2033. 30. Culp SH, Schellhammer PF, Williams MB. Might men diagnosed with
12. Niibe Y, Chang JY. Novel insights of oligometastases and oligo- metastatic prostate cancer benefit from definitive treatment of the
recurrence and review of the literature. Pulm Med. 2012;2012:261096. primary tumor? A SEER-based study. Eur Urol. 2014;65:1058-1066.
13. Thompson IM, Tangen C, Basler J, et al. Impact of previous local 31. Fossati N, Trinh QD, Sammon J, et al. Identifying optimal candidates for
treatment for prostate cancer on subsequent metastatic disease. J Urol. local treatment of the primary tumor among patients diagnosed with
2002;168:1008-1012. metastatic prostate cancer: a SEER-based study. Eur Urol. 2015;67:3-6.
14. Tzelepi V, Efstathiou E, Wen S, et al. Persistent, biologically meaningful 32. Karnes RJ, Murphy CR, Bergstralh EJ, et al. Salvage lymph node dis-
prostate cancer after 1 year of androgen ablation and docetaxel section for prostate cancer nodal recurrence detected by 11C-choline
treatment. J Clin Oncol. 2011;29:2574-2581. positron emission tomography/computerized tomography. J Urol.
15. Swanson G, Thompson I, Basler J, et al. Metastatic prostate cancer- 2015;193:111-116.
does treatment of the primary tumor matter? J Urol. 2006;176: 33. Zattoni F, Nehra A, Murphy CR, et al. Mid-term outcomes following
1292-1298. salvage lymph node dissection for prostate cancer nodal recurrence
16. Engel J, Bastian PJ, Baur H, et al. Survival benefit of radical prosta- status post-radical prostatectomy. Eur Urol Focus. Epub 2016 Feb 10.
tectomy in lymph node-positive patients with prostate cancer. Eur Urol. 34. Suardi N, Gandaglia G, Gallina A, et al. Long-term outcomes of salvage
2010;57:754-761. lymph node dissection for clinically recurrent prostate cancer: results
17. Briganti A, Blute ML, Eastham JH, et al. Pelvic lymph node dissection in of a single-institution series with a minimum follow-up of 5 years. Eur
prostate cancer. Eur Urol. 2009;55:1251-1265. Urol. 2015;67:299-309.
18. Gakis G, Boorjian SA, Briganti A, et al. The role of radical prostatectomy 35. Ploussard G, Almeras C, Briganti A, et al. Management of node only
and lymph node dissection in lymph node-positive prostate cancer: recurrence after primary local treatment for prostate cancer: a sys-
a systematic review of the literature. Eur Urol. 2014;66:191-199. tematic review of the literature. J Urol. 2015;194:983-988.
19. Messing EM, Manola J, Yao J, et al; Eastern Cooperative Oncology Group 36. Ost P, Bossi A, Decaestecker K, et al. Metastasis-directed therapy of
study EST 3886. Immediate versus deferred androgen deprivation regional and distant recurrences after curative treatment of prostate
treatment in patients with node-positive prostate cancer after radical cancer: a systematic review of the literature. Eur Urol. 2015;67:852-863.
prostatectomy and pelvic lymphadenectomy. Lancet Oncol. 2006;7: 37. Rigatti P, Suardi N, Briganti A, et al. Pelvic/retroperitoneal salvage lymph
472-479. node dissection for patients treated with radical prostatectomy with
20. Schroder FH, Kurth KH, Fossa SD, et al. Early versus delayed endocrine biochemical recurrence and nodal recurrence detected by [11C]choline
treatment of T2-T3 pN1-3 M0 prostate cancer without local treatment positron emission tomography/computed tomography. Eur Urol. 2011;
of the primary tumour: final results of European Organisation for the 60:935-943.
Research and Treatment of Cancer Protocol 30846 after 13 years of 38. Decaestecker K, De Meerleer G, Ameye F, et al. Surveillance or
follow-up (a randomised controlled trial). Eur Urol. 2009;55:14-22. metastasis-directed therapy for oligometastatic prostate cancer re-
21. Briganti A, Karnes JR, Da Pozzo LF, et al. Two positive nodes represent a currence (STOMP): study protocol for a randomized phase II trial. BMC
significant cut-off value for cancer specific survival in patients with node Cancer. 2014;14:671.
positive prostate cancer. A new proposal based on a two-institution 39. National Comprehensive Cancer Network. Prostate Cancer (Version
experience on 703 consecutive N+ patients treated with radical 1.2016). http://www.nccn.org/professionals/physician_gls/pdf/prostate.
prostatectomy, extended pelvic lymph node dissection and adjuvant pdf. Accessed January 15, 2016.
therapy. Eur Urol. 2009;55:261-270. 40. Pound CR, Partin AW, Eisenberger MA, et al. Natural history of pro-
22. Touijer KA, Mazzola CR, Sjoberg DD, et al. Long-term outcomes of gression after PSA elevation following radical prostatectomy. JAMA.
patients with lymph node metastasis treated with radical prostatec- 1999;281:1591-1597.
tomy without adjuvant androgen-deprivation therapy. Eur Urol. 2014; 41. Sweeney CJ, Chen YH, Carducci M, et al. Chemohormonal therapy in
65:20-25. metastatic hormone-sensitive prostate cancer. N Engl J Med. 2015;373:
23. Moschini M, Sharma V, Zattoni F, et al. Risk stratification of pN+ prostate 737-746.
cancer after radical prostatectomy from a large institutional series with 42. James ND, Sydes MR, Clarke NW, et al; STAMPEDE investigators. Ad-
long-term follow-up. J Urol. Epub 2015 Dec 23. dition of docetaxel, zoledronic acid, or both to first-line long-term
24. Moschini M, Sharma V, Zattoni F, et al. Natural history of clinical re- hormone therapy in prostate cancer (STAMPEDE): survival results from
currence patterns of lymph node-positive prostate cancer after radical an adaptive, multiarm, multistage, platform randomised controlled
prostatectomy. Eur Urol. 2016;69:135-142. trial. Lancet. Epub 2015 Dec 21.

128 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


APPROACH TO OLIGOMETASTATIC PROSTATE CANCER

43. Tannock IF, de Wit R, Berry WR, et al; TAX 327 Investigators. Docetaxel 54. Vale CL, Burdett S, Rydzewska LH, et al; STOpCaP Steering Group. Addition
plus prednisone or mitoxantrone plus prednisone for advanced pros- of docetaxel or bisphosphonates to standard of care in men with localised
tate cancer. N Engl J Med. 2004;351:1502-1512. or metastatic, hormone-sensitive prostate cancer: a systematic review
44. Petrylak DP, Tangen CM, Hussain MH, et al. Docetaxel and estramustine and meta-analyses of aggregate data. Lancet Oncol. 2016;17:243-256.
compared with mitoxantrone and prednisone for advanced refractory 55. Shipley WU, Seiferheld W, Lukka H, et al. Report of NRG Oncology/RTOG
prostate cancer. N Engl J Med. 2004;351:1513-1520. 9601, a phase 3 trial in prostate cancer: anti-androgen therapy (AAT)
45. de Bono JS, Oudard S, Ozguroglu M, et al; TROPIC Investigators. with bicalutamide during and after radiation therapy (RT) in patients
Prednisone plus cabazitaxel or mitoxantrone for metastatic castration- following radical prostatectomy (RP) with pT2-3pN0 disease and an
resistant prostate cancer progressing after docetaxel treatment: elevated PSA. Int J Radiat Oncol Biol Phys. 2016;94:3.
a randomised open-label trial. Lancet. 2010;376:1147-1154. 56. Parekh A, Chen MH, Graham P, et al. Role of androgen deprivation
46. Eisenberger MA, Blumenstein BA, Crawford ED, et al. Bilateral orchi- therapy in early salvage radiation among patients with prostate-
ectomy with or without flutamide for metastatic prostate cancer. N Engl specific antigen level of 0.5 or less. Clin Genitourin Cancer. 2015;
J Med. 1998;339:1036-1042. 13:e1-e6.
47. Millikan RE, Wen S, Pagliaro LC, et al. Phase III trial of androgen ablation 57. Hussain M, Tangen CM, Higano C, et al; Southwest Oncology Group Trial
with or without three cycles of systemic chemotherapy for advanced 9346 (INT-0162). Absolute prostate-specific antigen value after an-
prostate cancer. J Clin Oncol. 2008;26:5936-5942. drogen deprivation is a strong independent predictor of survival in new
48. Singh D, Yi WS, Brasacchio RA, et al. Is there a favorable subset of metastatic prostate cancer: data from Southwest Oncology Group Trial
patients with prostate cancer who develop oligometastases? Int J 9346 (INT-0162). J Clin Oncol. 2006;24:3984-3990.
Radiat Oncol Biol Phys. 2004;58:3-10. 58. Hussain M, Goldman B, Tangen C, et al. Prostate-specific antigen
49. Makarov DV, Humphreys EB, Mangold LA, et al. The natural history of progression predicts overall survival in patients with metastatic prostate
men treated with deferred androgen deprivation therapy in whom cancer: data from Southwest Oncology Group Trials 9346 (Intergroup
metastatic prostate cancer developed following radical prostatectomy. Study 0162) and 9916. J Clin Oncol. 2009;27:2450-2456.
J Urol. 2008;179:156-161, discussion 161-162. 59. Jereczek-Fossa BA, Beltramo G, Fariselli L, et al. Robotic image-guided
50. Schweizer MT, Zhou XC, Wang H, et al. Metastasis-free survival is as- stereotactic radiotherapy, for isolated recurrent primary, lymph node
sociated with overall survival in men with PSA-recurrent prostate or metastatic prostate cancer. Int J Radiat Oncol Biol Phys. 2012;82:
cancer treated with deferred androgen deprivation therapy. Ann Oncol. 889-897.
2013;24:2881-2886. 60. Schick U, Jorcano S, Nouet P, et al. Androgen deprivation and high-dose
51. Fizazi K, Faivre L, Lesaunier F, et al. Androgen deprivation therapy plus radiotherapy for oligometastatic prostate cancer patients with less
docetaxel and estramustine versus androgen deprivation therapy alone than five regional and/or distant metastases. Acta Oncol. 2013;52:
for high-risk localised prostate cancer (GETUG 12): a phase 3 rando- 1622-1628.
mised controlled trial. Lancet Oncol. 2015;16:787-794. 61. Decaestecker K, De Meerleer G, Lambert B, et al. Repeated stereotactic
52. Gravis G, Fizazi K, Joly F, et al. Androgen-deprivation therapy alone or with body radiotherapy for oligometastatic prostate cancer recurrence.
docetaxel in non-castrate metastatic prostate cancer (GETUG-AFU 15): Radiat Oncol. 2014;9:135.
a randomised, open-label, phase 3 trial. Lancet Oncol. 2013;14:149-158. 62. Picchio M, Berardi G, Fodor A, et al. 11C-Choline PET/CT as a guide to
53. Gravis G, Boher JM, Joly F, et al; GETUG. Androgen deprivation therapy radiation treatment planning of lymph-node relapses in prostate cancer
(ADT) plus docetaxel versus ADT alone in metastatic non castrate patients. Eur J Nucl Med Mol Imaging. 2014;41:1270-1279.
prostate cancer: impact of metastatic burden and long-term survival 63. Hussain M, Tangen CM, Berry DL, et al. Intermittent versus continuous
analysis of the randomized phase 3 GETUG-AFU15 trial. Eur Urol. Epub androgen deprivation in prostate cancer. N Engl J Med. 2013;368:
2015 Nov 21. 1314-1325.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 129


GENITOURINARY (PROSTATE) CANCER

Precision Medicine in Advanced


Prostate Cancer: Understanding
Genomics, Androgen Receptor
Splice Variants, and Imaging
Biomarkers

CHAIR
Himisha Beltran, MD
Weill Cornell Medical College
New York, NY

SPEAKERS
Emmanuel S. Antonarakis, MD
The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
Baltimore, MD

Gerhardt Attard, MD, PhD


The Institute of Cancer Research and The Royal Marsden Hospital
London, United Kingdom

Michael J. Morris, MD
Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College
New York, NY
TISSUE, LIQUID, AND IMAGING BIOMARKERS IN CRPC

Emerging Molecular Biomarkers in Advanced Prostate Cancer:


Translation to the Clinic
Himisha Beltran, MD, Emmanuel S. Antonarakis, MD, Michael J. Morris, MD, and
Gerhardt Attard, MD, PhD

OVERVIEW

Recent clinical and preclinical studies focused on understanding the molecular landscape of castration-resistant prostate
cancer (CRPC) have provided insights into mechanisms of treatment resistance, disease heterogeneity, and potential
therapeutic targets. This work has served as a framework for several ongoing clinical studies focused on bringing novel
observations into the clinic in the form of tissue, liquid, and imaging biomarkers. Resistance in CRPC typically is driven
through reactivation of androgen receptor (AR) signaling, which can occur through AR-activating point mutations, am-
plification, splice variants (such as AR-V7), or other bypass mechanisms. Detection of AR aberrations in the circulation
negatively impacts response to subsequent AR-directed therapies such as abiraterone and enzalutamide. Other potentially
clinically relevant alterations in CRPC include defects in DNA damage repair (at either the somatic or germline level) in up to
20% of patients (with implications for PARP1 inhibitor therapy), PI3K/PTEN/Akt pathway activation, WNT signaling pathway
alterations, cell cycle gene alterations, and less common but potentially targetable alterations involving RAF and FGFR2.
Imaging biomarkers that include those focused on incorporating overexpressed androgen-regulated genes/proteins, such as
prostate-specific membrane antigen (PSMA) and dihydrotestosterone (DHT) in combination with CT, can noninvasively
identify patterns of AR-driven distribution of CRPC tumor cells, monitor early metastatic lesions, and potentially capture
heterogeneity of response to AR-directed therapies and other therapeutics. This article focuses on the current state of
clinical biomarker development and future directions for how they might be implemented into the clinic in the near term to
improve risk stratification and treatment selection for patients.

the largest size.2 Further, molecular changes can occur in the


S everal drugs are either approved or in clinical develop-
ment for men with metastatic CRPC with varied mech-
anisms of action. Although many drugs have demonstrated
tumor with disease progression and treatment resistance,
and there is recent evidence of polyclonal metastasis-to-
antitumor activity, and even an overall survival benefit in an metastasis seeding.3 Therefore, assessment of the archival
unselected population, the identification of biomarkers to primary tumor in making clinical decisions regarding sys-
help select the best next-line systemic therapy for an in- temic therapies for patients with CRPC may not be adequate
dividual patient is an unmet clinical need. Molecular bio- or fully representative of the tumors current state.
markers have the potential to help clinicians with risk Metastatic biopsies in advanced prostate cancer have not
stratification, inform patient selection for therapy, and assist been considered standard practice. To date, they have been
patient and family counseling (Fig. 1). performed mainly in the context of clinical trials. Metastatic
Primary prostate cancer is commonly multifocal at the biopsies can be challenging to perform because of the high
time of diagnosis. It has a long natural history with often frequency of sclerotic bone-only metastases in CRPC, which
5 years or more before distant metastases are detected and often require special biopsy procedures and tissue handling
castration resistance ensues. In the monoclonal origin model for molecular profiling. Further, single-site metastatic bi-
of clonal evolution supported by several studies,1,2 one opsies do not necessarily represent the entire metastatic
tumor clone/nodule present in a multifocal primary is re- tumor burden of the patient because of intrapatient het-
sponsible for metastasis. However, identification of that erogeneity. Therefore, alternative approaches, such as
dominant lesion at diagnosis is challenging because it may blood-based assays (e.g., circulating tumor cell, circulating
not always be the lesion with the highest Gleason grade or of tumor DNA) and molecular imaging, may be more feasible

From Weill Cornell Medicine, New York, NY, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Memorial Sloan Kettering Cancer Center, Weill Cornell
Medicine, New York, NY; The Institute of Cancer Research, London, United Kingdom, The Royal Marsden Hospital, London, United Kingdom.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding Author: Himisha Beltran, MD, Weill Cornell Medicine, 413 East 69th St., New York, NY 10021; email: hip9004@med.cornell.edu.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 131


BELTRAN ET AL

with the given advantage of performing serial assessments pathways.13 What remains unclear is why some, but not all,
during the course of a patients disease to track tumor tumors in patients develop AR alterations and when they
dynamics and treatment response. occur during the course of AR-directed therapies. As de-
scribed below, liquid biopsies have shown promise as a
means to study AR dynamics during the course of treatment.
THE GENOMIC LANDSCAPE OF CASTRATION- Clinical and preclinical studies investigating therapeutic
RESISTANT PROSTATE CANCER: approaches to better target the AR (and AR splice variants) in
CHARACTERIZATION OF METASTATIC the face of AR-driven resistance are underway; these include
TISSUE BIOPSIES combined CYP17A1 inhibition and AR antagonism, targeting
Several recent studies have independently reported mo- the AR N-terminal or DNA binding domain, or AR degrada-
lecular data of tumors obtained from large cohorts of tion. A subset of CRPC also may lose AR independence be-
patients with clinically localized prostate cancer4-6 and cause of loss of AR expression and/or signaling. Identifying
metastatic CRPC.7-9 Certain genomic alterations, such as ERG patients who will no longer benefit from AR-directed ther-
gene rearrangements and SPOP mutations, are present at apies is also an area of biomarker investigation. One pro-
similar frequency in both localized and metastatic disease, posed mechanism of AR independence includes the
suggesting that these lesions predate metastatic spread. development of epithelial plasticity associated with patho-
Other alterations, such as AR point mutations or amplifi- logic and molecular features of neuroendocrine cancer.14,15
cation, are present primarily in CRPC (50%60%), which There are several other genomic aberrations reported or
suggests they are acquired following androgen depriva- enriched in CRPC, some of which also occur at a lesser
tion.10,11 The acquisition of activating alterations involving frequency in primary disease, many predicted to be ac-
AR (or AR cofactors) seems logical, as multiple studies have tionable (such as PI3K/Akt/PTEN, RAF, FGFR2, WNT pathway,
confirmed that most castration-resistant prostate tumors cell cycle, and DNA repair alterations; recently described in
remain dependent on AR signaling.12 Mechanisms associ- Robinson et al8). This knowledge has served as a framework
ated with reactivation of AR signaling in CRPC include ac- for preclinical testing of novel agents and cotargeting ap-
quisition of AR genomic alterations, AR mRNA splice variant proaches and the design of biomarker-driven clinical studies.
expression (such as AR-V7), cofactor activation, AR bypass Present at similar frequency in approximately 40% to 50% of
(such as glucocorticoid receptor expression), or AR crosstalk primary prostate cancer and CRPC, the TMPRSS2-ERG gene
fusion is prostate cancerspecific and androgen-regulated,
making it an attractive drug target. TMPRSS2-ERG does not
KEY POINTS encode for a chimeric protein; instead, it results in over-
expression of a truncated ERG protein in the setting of active
The study of biopsies from metastatic sites from patients androgen signaling, driving a transcriptional program linked
with advanced prostate cancer has revealed potentially to DNA damage and invasion.16 It is challenging to develop
actionable or prognostic genomic alterations. Common drugs that target transcription factors, but short peptido-
genomic alterations observed in CRPC are: ERG gene mimetics that bind to ERG, bromodomain inhibitors, and
fusion (40%50%), AR gene point mutation or PARP inhibitors have been explored preclinically, and ola-
amplification (50%60%), TP53 mutation or deletion parib currently is being investigated in combination with
(40%50%), PTEN deletion (40%50%), RB1 deletion abiraterone in an ERG-fusion biomarkerstratified phase II
(20%), and alterations in DNA repair genes (20%). clinical trial (NCT01972217). Alteration of genes leading to
Circulating tumor DNA analyses in patients with CRPC
aberrant activation of the PI3K/Akt pathway, most com-
have revealed intrapatient molecular heterogeneity and
the potential to track dynamic changes during the
monly loss of PTEN, is implicated in 40% to 50% of CRPC cases
course of therapeutic response and resistance in and is associated with signaling crosstalk with AR through
patients with advanced prostate cancer. reciprocal negative feedback.17,18 Based on this, there are
Detection of the AR-V7 splice variant in circulating several ongoing phase II trials investigating cotargeting of
tumor cells or AR genomic aberrations in circulating both pathways (NCT01251861, NCT02525068, NCT02215096,
tumor DNA has been associated with decreased clinical NCT02407054).
response to abiraterone or enzalutamide. Prospective Somatic aberrations involving DNA defect repair genes
trials underway are evaluating the prognostic and (such as deletions/mutations involving BRCA1/2, ataxia-
predictive utility of AR-V7 and exploring novel agents telangiectasia mutated [ATM], CHEK2, Fanconi [FANC]
with potential activity against AR-Vexpressing CRPC. genes) are present in up to 20% of CRPC tumors. Somatic loss
Bi-allelic loss of DNA repair genes (e.g., BRCA1/2, ATM)
of the BRCA2 gene, for instance, occurs in approximately
has been associated with clinical response to the PARP
inhibitor olaparib.
3% of localized prostate cancer cases (TCGA)6 and 13% of
Advances in molecular imaging and assessment of bone CRPC cases (SU2C-PCF).8 Notably germline mutations, un-
lesions have shown promise for early detection of commonly present in patients diagnosed with localized
metastasis and detection of biologic heterogeneity disease, are present at higher frequency in patients with
between sites. CRPC (in keeping with their association with more aggressive
disease at diagnosis).8,19,20 Tumors with defects in DNA

132 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


TISSUE, LIQUID, AND IMAGING BIOMARKERS IN CRPC

FIGURE 1. Schematic of Various Types of Molecular Biomarkers

This schematic illustrates various types of molecular biomarkers currently being tested in trials for patients with advanced prostate cancer, with a goal of ultimately
improving clinical decision making. Image credit: L.S. Beltran.

repair would be predicted to have disrupted homologous either radiographic, prostate-specific antigen [PSA], or cir-
recombination; therefore, they would be more sensitive to culating tumor cell [CTC] conversion) was 33% with 12
inhibition of PARP1 or DNA cross-linking agents (such as patients receiving olaparib for more than 6 months and four
platinum chemotherapy) through a mechanism of synthetic patients receiving the drug for more than 12 months. The
lethality. The PARP inhibitor olaparib has shown antitumor majority of responders on the study were found to have
activity in germline carriers of BRCA1/2 gene alterations either germline or somatic alterations in DNA repair genes;
across cancer types, including prostate,21 and it is approved 14 of 16 DNA-repair altered cases (88%) had a response to
for clinical use in patients with BRCA-mutated ovarian olaparib, which was associated with prolonged overall
cancer. survival (median, 13.8 vs. 7.5 months in those without DNA
To investigate whether there are additional patients with repair alterations). Based on these promising results, ola-
CRPC who may also benefit from PARP inhibition, such as parib recently received a U.S. Food and Drug Administration
those with somatic alterations in DNA repair, Mateo et al (FDA) breakthrough designation for patients with BRCA1/2-
developed an adaptive design phase II trial of olaparib where or ATM-mutated metastatic CRPC who have received a prior
pretreatment tissue and blood biomarkers were in- taxane-based chemotherapy and at least either enzaluta-
vestigated prospectively.19 In the 49 evaluable patients mide or abiraterone. A phase III study is planned. Reports
enrolled in this study, overall response rate (defined by linking BRCA2 and FANCA alterations and platinum

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 133


BELTRAN ET AL

sensitivity in patients with CRPC also have been reported prednisone in docetaxel-treated metastatic CRPC, which
recently.22,23 showed that a biomarker panel containing CTC count and
A question now is whether metastatic biopsies will soon be lactate dehydrogenase level was a surrogate for survival at
warranted to look for DNA repair defects and, if so, when the individual-patient level.28 Validation in additional pos-
should this be performed? Can these be detected reliably itive phase III trials is now required.
using circulating tumor DNA or other noninvasive means? Isolating and characterizing rare CTCs in the blood com-
Where will drugs such as PARP inhibitor or platinum che- partment composed of millions of white blood cells in-
motherapy ultimately be used within the growing clinical troduces technologic and logistical challenges. Several more
armamentarium of approved drugs? A higher frequency of recent platforms are showing promise in obtaining impor-
germline alterations involving DNA repair genes in CRPC has tant multiplex phenotypic data,29,30 and planned pro-
not only therapeutic considerations but also familial im- spective studies will investigate associations with clinical
plications. Who and when to screen for germline alterations behavior and outcome. Additionally, as described below, the
and how genetic counseling and testing will be performed AdnaTest platform allows the capture of CTCs with pres-
and reimbursed are areas of active discussion. ervation of mRNA quality, which can then be used to study
Defects in DNA mismatch repair genes such as MLH1 and expression of candidate biomarkers such as AR-V7. Overall,
MSH2 and microsatellite instability also occur at the somatic the number of CTCs is associated with number (and site) of
level in up to 5% of prostate cancers8,24 and in the germline metastases.31,32 Using currently available platforms, the
DNA of Lynch syndrome families.25 This results in a hyper- majority of patients with low-volume metastases have fewer
mutated phenotype with a mutation rate typically greater than 510 CTCs in 10 mL of blood. Newer approaches may
than 10-fold higher than other CRPC tumors. Given obser- improve on this detection rate or allow processing of larger
vations in other tumor types linking mutation rates with volumes of blood. An alternative approach to CRPC mo-
response to immune checkpoint inhibitors,26 identifying this lecular characterization is to study tumor-specific aberra-
small subset of cases may have clinical implications in the tions in cell-free DNA from plasma (i.e., the upper
context of immunotherapies. The design and recruitment for compartment from blood centrifuged to precipitate the
biomarker-enriched trials involving low-frequency subsets, cellular component).
such as those patients with mismatch repair defects, are
challenging and rely on multi-institutional collaboration. Monitoring Plasma DNA to Modify Patient Treatment
Although informative, most of the genomic studies per- Using next-generation sequencing approaches applied to
formed to date in CRPC have represented a snapshot in time. plasma DNA, tumor aberrations are detected in more than
How early events (such as SPOP, PTEN, or ERG) might co- 95% of patients with progressing metastatic CRPC.33,34 These
operate with potentially later events to drive tumor growth, are admixed with normal DNA at variable quantities, and
and how these affect response to subsequent therapies, the their relative abundance can provide information on tumor
tumor microenvironment, and the epigenetic landscape are behavior. Studies in prostate cancer and several other tumor
areas of active investigation. Translation of genomics into the types have reported that changes in circulating tumor DNA
clinic also will require a better understanding of tumor levels are associated with response to treatment and im-
evolution and intrapatient heterogeneity. Longitudinal provements in outcome.35-37 By monitoring sequential
studies, including the two SU2C-PCF Dream Teams, and plasma samples from patients with CRPC treated with
multisample cases (such as rapid autopsies) have started to abiraterone, emergence or selection of an AR mutation
provide new insights that are having an impact on the clinical resulting in either a T878A or an L702H amino acid change
interpretation of single-site biopsies in the context of routine was observed in 15% to 20% of patients, often several months
clinical care and insights into mechanisms of tumor evolution. before manifest clinical or radiologic progression.34,36 Pre-
clinical studies confirm that these two mutations are acti-
DEVELOPMENT OF PROGNOSTIC AND vated by progesterone or prednisolone respectively, both
PREDICTIVE CIRCULATING BIOMARKERS present at increased levels in patients treated with abir-
Because serial metastatic biopsies are not always feasible or aterone and supporting their causative association with
safe for most men with CRPC, noninvasive assays including resistance.36,38 This was followed by a study of pre- and post-
CTCs and/or plasma DNA may be an effective means to treatment plasma samples from 100 patients treated with
assess biomarker status and capture heterogeneity. Pro- abiraterone, the majority of whom were postdocetaxel and/or
spective studies using the CellSearch system confirmed the had previously received potent AR-targeting agents. A mini-
association between CTC count ($ in 7.5 mL) in patients with mum circulating tumor content (7.5%, present in 80 of 97
advanced prostate cancer and worse clinical outcome.27 An pretreatment samples), defined as the abundance or allelic
association with improved outcome for patients who had a frequency of tumor aberrations measured using a custom
CTC conversion (from $ 5 to , 5) compared with those who panel including six informative prostate cancer lesions, was
did not suggested that this approach could be used as an mandated to allow evaluation of AR copy number gain. Data
early indicator of response and as a surrogate endpoint in reported include a strong association between pretreatment
clinical trials. This led to the prospective evaluation of CTC AR copy number gain and/or T878A or L702H AR mutations
count in the phase III COU-301 trial of abiraterone and and PSA decline (4.9 and 7.8 times less likely to have a $ 50

134 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


TISSUE, LIQUID, AND IMAGING BIOMARKERS IN CRPC

or $ 90% decline) and overall survival (HR 7.33; 95% CI, was lowest in men who had not received either agent. Ad-
3.5115.34, p = 1.3 3 1029) and progression-free survival ditionally, when assessing serial CTC samples over time, the
(PFS; HR 3.73; 95% CI, 2.176.41, p = 5.6 3 1027).34 Similarly, authors reported that all men with baseline detection of
an association between AR copy number or an exon 8 AR AR-V7 remained AR-V7positive during the course of
mutation and resistance to enzalutamide in 39 patients with therapy with abiraterone and enzalutamide, whereas 14%
CRPC has been reported.39 Increased AR copy number is of men with negative AR-V7 status at baseline converted
associated with higher AR mRNA transcripts (in tumor tis- to AR-V7positive during treatment; these patients had
sue),40 which drive resistance to androgen suppression intermediate clinical outcomes.52
in vitro. Ongoing studies, as described below, will shed light on Another question is whether the presence of AR-V7 is
the association between AR copy number and AR splice relevant in the setting of taxane chemotherapy, especially
variants and their association with other aberrations that because two preclinical studies have produced conflicting
drive treatment resistance in CRPC. Overall, these data sup- results.48,53 Accordingly, Antonarakis et al54 performed a
port the potential clinical applicability of circulating tumor second prospective study using their CTC-based AR-V7 assay
DNA studies in CRPC. Prospective evaluation of plasma AR in 30 patients beginning treatment with docetaxel and seven
gene aberrations in randomized trials with novel and standard patients beginning treatment with cabazitaxel. The preva-
AR targeting agents, taxanes, and radiopharmaceuticals are lence of AR-V7 in these patients, most of whom had pre-
now planned. Moreover, plasma DNA studies appear to viously received abiraterone and/or enzalutamide, was 46%.
provide additional or complementary molecular data to tumor PSA responses were observed in both AR-V7positive and
biopsies,36,41 introducing the possibility of patient selection AR-V7negative men (41% vs. 65%; p = .19). Similarly, PFS
for single-agent and combination therapeutic strategies tar- was not statistically different in patients who were AR-
geting additional targets, including DNA repair defects and V7positive and negative (p = .11). As a hypothesis-
PI3K/Akt. generating exercise, Antonarakis et al incorporated data
from their prior study of 62 patients treated with abir-
AR-V7 as a Clinical Biomarker aterone and enzalutamide and showed that clinical out-
Androgen receptor splice variants are abnormally truncated comes appeared to be better with taxanes compared with
isoforms of the AR that are capable of activating tran- enzalutamide or abiraterone in men who were AR-V7
scription of androgen-regulated genes without the re- positive, although outcomes did not appear to differ by
quirement of ligand.42-45 Although at least 22 AR splice treatment type in men who were AR-V7negative. More
variants have been reported,8 AR-V7 is the most frequently specifically, in patients who were AR-V7 positive, PSA re-
observed and the most abundant AR-V detected at the sponses were higher in men treated with taxane compared
mRNA and protein level in clinical specimens from patients with enzalutamide or men treated with abiraterone (41% vs.
with CRPC. A number of retrospective studies have sug- 0%, p , .001), and PFS was longer in men treated with
gested that the presence of AR-V7 is associated with more taxane (HR 0.21, p = .003). Intriguingly, 58% of patients with
rapid disease progression and shorter survival in men with baseline AR-V7positive status converted to AR-V7
metastatic CRPC.46-49 The first prospective study to evaluate negative status during treatment with docetaxel or cab-
the prognostic impact of AR-V7 was conducted by Efstathiou azitaxel. Whether such transitions in AR-V7 status may
et al.50 In that study, the detection of AR-V7 by immuno- resensitize such patients to further AR-directed therapies is
histochemistry in bone marrow biopsy specimens of patients unknown.55
with CRPC was associated with resistance to enzalutamide; Taken together, the clinical data to date suggest that the
AR-V7 protein was detected in 57% of men who developed presence of AR-V7 may be a marker of resistance to AR-
disease progression within 4 months of starting enzaluta- directed therapy but not taxane chemotherapy; therefore, it
mide and was not detected in any patient who responded to may represent a treatment selection biomarker in CRPC.
enzalutamide for longer than 6 months (p = .02). These These findings now warrant further prospective validation
findings were further corroborated in a separate study by and clinical qualification before CTC-based AR-V7 testing
the same investigators evaluating the combination of should be used in clinical practice to inform treatment
abiraterone and enzalutamide.51 decisions. A number of AR-V7 biomarker validation studies
More recently, Antonarakis et al52 reported a prospective currently being conducted are summarized in a recent re-
study assessing the prognostic role of AR-V7 detected at the view.56 The Sanofi-sponsored PRIMCAB trial (NCT02379390)
mRNA level by reverse-transcriptase (RT)-PCR from CTCs of is investigating cabazitaxel chemotherapy versus AR-
patients receiving abiraterone or enzalutamide. They found directed therapy in men with metastatic CRPC who have
that the presence of AR-V7 in CTCs was associated with developed disease progression within 6 months of starting
lower PSA response rate, shorter PFS, and shorter overall abiraterone or enzalutamide. As a secondary endpoint, that
survival compared with those patients without detectable trial will prospectively evaluate baseline AR-V7 status from
circulating AR-V7.52 Notably, the prevalence of AR-V7 was CTCs as a putative predictive biomarker in this setting, in
higher in men treated with enzalutamide who previously had which the prevalence of AR-V7 is expected to be about 33%.
received abiraterone and in men treated with abiraterone The Johns Hopkins AR-V7 assay will serve as the central
who had previously received enzalutamide; AR-V7 prevalence laboratory for AR-V7 testing. Exploratory analyses will also

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 135


BELTRAN ET AL

evaluate transitions in AR-V7 status at the time of pro- In a drug library screen aimed at identifying FDA-approved
gression. In a Prostate Cancer Foundation (PCF)-sponsored drugs capable of targeting AR-V7, the antihelminthic drug
prospective biomarker trial (NCT02269982) coordinated by niclosamide emerged as an unexpected success.61 Addi-
the Duke Cancer Institute, three different CTC-based AR-V7 tional mechanistic studies suggested that this agent func-
assays will be evaluated in 120 men with taxane-naive tions by degrading AR-V7 through a proteasome-dependent
metastatic CRPC and high-risk clinical features. In this pathway and that AR-V7 degradation occurred more rapidly
noninterventional trial, patients will receive standard-of- than AR-FL degradation. Niclosamide demonstrated sub-
care abiraterone or enzalutamide and then may also re- stantial antitumor activity in a number of AR-V7expressing
ceive standard-of-care taxane at progression. AR-V7 testing CRPC cell lines and xenograft models resistant to enzalu-
will be performed prior to AR-directed therapy, at pro- tamide, and the combination of niclosamide and enzaluta-
gression on AR-directed therapy, and also at progression on mide produced maximal tumor inhibition. Based on these
chemotherapy. Each patient will undergo AR-V7 testing with preclinical data, a phase I clinical trial was launched in 2015
three assays at each time point: the Johns Hopkins mRNA- (NCT02532114) for men with abiraterone-pretreated CRPC
based assay, the Weill-Cornell mRNA-based assay (which who test positive for AR-V7 using an AR-V7 assay performed
also evaluates other AR variants), and the EPIC Sciences at the University of Washington. In this trial, patients will
protein-based assay. receive enzalutamide plus escalating doses of oral niclosa-
Although no approved agents currently directly target mide. Exploratory analyses will evaluate changes in AR-V7
AR-V7, a number of compounds are now in clinical devel- status during the course of niclosamide treatment and at the
opment; these are summarized in a recent review.56 Gale- time of progression.
terone (Tokai Pharmaceuticals) is an oral drug with three
proposed mechanisms of action, including inhibition of IMAGING METASTATIC DISEASE: CURRENT AND
CYP17 lyase, antagonism of the AR ligand-binding domain, FUTURE PERSPECTIVES
and degradation of both full length AR (AR-FL) and AR splice The tissue and blood-based technologies described above
variants through a proteasome-dependent pathway.56,57 may accelerate drug development and personalize clinical
In a post hoc analysis of the phase II ARMOR2 trial, among decision making by yielding biomarkers for drug sensitivity
seven patients with AR C-terminal loss (as determined using and response. Advances in imaging technology can hasten
an immunohistochemistry-based CTC assay), six men the pace of such progress. Imaging is capable of not only
achieved more than a 50% PSA reduction with galeterone assessing disease extent and distribution, but it can also
treatment. Based on these preliminary data, a registration identify biologic features of a patients totality of lesions
phase III trial (ARMOR3-SV) was launched in 2015 rather than those of a single biopsy or blood draw. Yet, of all
(NCT02438007). Eligible patients are those with AR-V7 of the biomarkers used for prostate cancer staging
positive metastatic CRPC without prior treatment with novel prognostication, prediction, response assessment, and bi-
AR-directed therapies or taxane chemotherapies. AR-V7 ologic characterizationimaging has been one of the most
testing will be conducted using a CLIA-certified assay de- challenging to develop.
veloped by Qiagen. Patients who are CTC-positive and AR- A substantial basis of these difficulties is that prostate
V7positive (148 total patients) will be randomly selected to cancer is a bone-tropic solid tumor, which is difficult to
receive either galeterone or enzalutamide. Notably, assess, much less measure. The Tc-99 MDP tracer is in-
ARMOR3-SV is the first registration trial in prostate cancer to corporated into reactive bone rather than the tumor.
use a biomarker-selection trial design. Consequently, bone scintigraphy is more sensitive to pro-
EPI-506 (ESSA Pharma), an oral prodrug of EPI-002, is the gression than response. Importantly, in 20% to 50% of pa-
first drug reported as capable of targeting the AR N-terminal tients, the bone scan will paradoxically worsen shortly after
domain.58 Specifically, EPI-506 binds covalently to the Tau5 the patient is placed on effective therapy, which can thereby
region of the AF1 domain of the N-terminus, a region that is mislead the inattentive clinician to interpret a worsening
responsible for 99% of the transcriptional activity of AR. scan as disease progression rather than response.62-64 De-
Because the N-terminus is present in both the AR-FL and in spite these limitations, bone scintigraphy is ubiquitously
all of the AR splice variants (including AR-V7), treatment with available, inexpensive, and the existing standard for imaging
EPI-506 would be expected to inhibit all forms of AR sig- bone. Data collection by the Prostate Cancer Working Group
naling. Preclinical studies with EPI-002 have shown that this 2s definition for disease progression was standardized,65
compound has activity in several AR-V7expressing cell lines incorporated into case report forms, and then the definition
and xenograft models.59 A phase I trial (including a sub- and data collection methods were prospectively validated
sequent phase II expansion) using EPI-506 opened in 2016 for reproducibility and tested for an association with overall
(NCT02606123). Eligible patients are those with metastatic survival in three prospective clinical trials involving abir-
CRPC who have previously received either abiraterone or aterone (NCT00887198), enzalutamide (NCT01212991),
enzalutamide; one prior taxane is also permitted but not and orteronel (NCT01193244). These trials demonstrated
required.60 Exploratory analyses of AR-V7 and plasma AR excellent concordance between central and investigator
gene aberrations also will be conducted, but this information reviewers. 66,67 In the first trial assessed, a significant
will not be used for patient selection or stratification. association between radiographic PFS and overall survival

136 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


TISSUE, LIQUID, AND IMAGING BIOMARKERS IN CRPC

was found (Spearmans coefficient, 0.72), and the endpoint upregulated in prostate cancer (especially CRPC), and
was used for regulatory approval of abiraterone for patients present regardless of disease site.83 In 1996, the FDA ap-
with chemotherapy-naive CRPC.65,68,69 Newer methods of proved Indium (111In) capromab pendetide,84 based on an
using bone scintigraphy involve quantitative descriptors of antibody that targets the internal domain of PSMA. Newer
disease burden, such as the bone scan index, which is the tracers, such as the Zr-89 labeled anti-PSMA antibody J591,
fraction of skeletal mass represented by tracer uptake70 and target the external domain of the molecule. A detailed
can be used to describe both response and progression. The lesional analysis of 89Zr-J591 in men with CRPC, verified by
bone scan index has been shown to be prognostic of overall pathology, revealed 89Zr-J591 detected 491 osseous sites
survival in a preliminary retrospective analysis,71 and au- compared with 339 by MDP in 50 patients.85 The overall
tomated systems are undergoing analytic validation.72-74 accuracy of 89Zr-J591 by pathology was 95.2% for osseous
Bone-directed PET/CT imaging using 18F sodium fluoride lesions; however, the performance in soft tissue was less
(NaF) may more accurately localize disease and produce a accurate, at an accuracy of 60%. A major drawback of intact
quantitative output amenable to biomarker development antibodies is a long half-life, resulting in delays between
and modeling. However, it is still subject to flare phenomena injection and imaging, which can be overcome by the use of
and does not directly demonstrate disease. Early studies minibodies86 or small molecules that inhibit the enzymatic
suggested that the modality was superior to both fluo- site of PSMA. An illustrative example of a Zr-89 radiolabeled
rodeoxyglucose (FDG) PET and 99mTc-MDP single photon minibody PET scan (IAB2M, ImaginAb, Inglewood, CA) with
emission computed tomography (SPECT) in detection of detection of bone lesions in a patient with CRPC is shown in
bone metastases.75-77 These findings were supported by Fig. 2. Although molecular imaging has the capacity to
subsequent meta-analyses, such as one analyzing 11 studies demonstrate a greater extent of disease earlier in the dis-
involving 613 patients that concluded, on a per-patient basis, ease course, what to do with that information clinically is
NaF PET had a sensitivity and specificity of 96% (93%98%) unknown.
and 91% (88%94%) versus 88% (84%96%) and 80% The most commonly used anti-PSMA small molecule
(75%84%) for bone scintigraphy.78 The National Oncology ligand is Glu-NH-CO-NH-Lys-(Ahx)-[68Ga(HBED-CC)](68Ga-
PET Registry (NOPR) is examining the use of NaF PET for DKFZ-PSMA-11), which has been extensively studied in
clinical decision making. Data on 1,940 patients with pros- Europe. In a recent retrospective analysis, 319 patients with
tate cancer demonstrated that NaF PET findings prompted a progressive disease across a variety of clinical states were
change in treatment in 41.8% of patients. However, NOPR scanned.87 On a lesional level, sensitivity and specificity were
did not seek to, nor did it define, a standard definition of 76.6% and 100%, respectively. A patient-based analysis
either response or progression, nor assess whether the revealed a sensitivity of 88.1%. Especially relevant for the
change in treatment was justified or associated with patient patient who experienced biochemical relapse was early
benefit. A preliminary multicenter study (NCT01516866) evidence that disease could be detected in patients with very
recently has been completed that delineates the re- low PSA values. Lesions were detected in approximately 47%
producibility of NaF across centers and examines post- (8/17) of patients with a PSA of less than or equal to 0.2, 50%
treatment changes in patients treated with AR-directed (5/10) of cases with PSA were 0.21 to less than or equal to
therapy and taxane chemotherapy. As it stands now, it is 0.5, 58% (14/24) of cases with a PSA between 0.51 and 1,
not clear that NaF PET/CT confers clinically useful in- 72% (28/31) in the range of 0.5 to less than 1, and greater
formation as a response biomarker, and per the Prostate than 90% with higher PSA values (where the number of
Cancer Working Group 2 (and the soon-to-be published patients was much greater). In another large retrospective
Prostate Cancer Working Group 3), should be considered analysis of 248 patients, detection rates were 96.8%, (PSA
investigational as an endpoint in clinical trials.79 value 2 ng/mL or higher), 93.0% (PSA value 1-2 ng/mL),
Assessing the actual disease burden in a patient, as op- 72.7% (PSA value 0.5-1 ng/mL), and 57.9% (PSA value 0.2-0.5
posed to statistical risk assessments of either distant or ng/mL).88 These analyses suggest that PSMA-based imaging
localized disease as determined by mathematical might detect disease at PSA ranges in which decisions about
modeling,80-82 is key to personalized decision making and salvage radiotherapy are indeed highly relevant, and would
clinical trial design and risk stratification. Accurate staging is appear to be more informative than standard imaging
particularly important for men with high-risk localized dis- modalities.89
11
ease and for those whose disease has biochemically relapsed C choline is chemically identical to physiologic choline
following local therapy. In these cases, decision making and is taken up by cancer cells requiring phospholipids to
frequently occurs without the benefit of imaging findings create cell membranes. The detection of prostate cancer by
11
that declare the patients true distribution of disease. New C choline was described in a recent pooled analysis of 12
molecular imaging techniques appear to be superior to studies involving 1,055 patients with biochemical relapse
standard cross-sectional imaging and bone scintigraphy, who underwent either 11C choline or 18F fluorocholine. The
although it is unclear how to best use this information for pooled sensitivity, specificity, and diagnostic odds ratio
clinical care. was 85% (95% CI, 79%89%), 88% (95% CI, 73%95%), and
Prostate-specific membrane antigen is a transmembrane 41.4 (95% CI, 19.7%86.8%), respectively.90 In another
carboxypeptidase that is expressed in prostatic tissue, meta-analysis involving 19 studies and 1,555 patients,91 a

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 137


BELTRAN ET AL

FIGURE 2. Imaging of Bone Lesions in a Patient With Castration-Resistant Prostate Cancer

Imaging of bone lesions of a patient with metastatic CRPC with bone scintigraphy (99mTc-MDP), FDG MIP, and a novel PSMA-directed PET scan using a Zr-89 radiolabeled
minibody (IAB2M, ImaginAb, Inglewood, CA). Although significantly more lesions are identified by IAB2M, whether the informative biomarker of novel imaging modalities, be it some
property of the SUV, lesion number, lesion location, change over time, or some other property, is as yet undefined.
Abbreviations: CRPC, castration-resistant prostate cancer; FDG MIP, fluorodeoxyglucose maximum intensity production; PSMA, prostate-specific membrane antigen; SUV,
standard uptake value.

pooled sensitivity of 85.6% (95% CI, 82.9%88.1%) and and soft-tissue disease. The image and the therapy could
specificity of 92.6% (95% CI, 90.1%94.6%) for all sites of also conceivably target the same molecule, such as PSMA.
disease (prostatic fossa, lymph nodes, and bone) was re- An early example of the power of such technology is 16b-
ported. However, 11C choline was relatively insensitive for 18
F-Fluoro-5a-Dihydrotestosterone (FDHT), a fluorinated ligand
patients with PSA values less than 2 ng/mL, with a 5% de- of the AR.96 FDHT is able to detect the presence of over-
tection rate for PSA levels of less than 1 ng/mL, 15% for PSA expressed AR in CRPC, and can therefore be used to establish
levels of 1 to 2 ng/mL, and 28% for PSA levels of greater than drug targeting, as it was in the early clinical development of
2 ng/mL.92 As PSA increases, and particularly as PSA doubling enzalutamide.97 The tracer also was used to establish the
time decreases, 11C-choline PET is more likely to be posi- phase II dose of the AR antagonist ARN509.98 Although one
tive.93,94 In the context of these European data and a small tracer can establish the targeting of the drug under study,
number of U.S. studies, the FDA approved 11C choline for another could be used to detect post-treatment signaling, as
patients with biochemical relapse after primary therapy has been shown preclinically. For example, PSMA imaging
when conventional imaging does not indicate a definitive can be used to detect changes in AR signaling as a result of
sign of cancer. However, the modality has only limited treatment with enzalutamide.99 Ultimately, molecular im-
availability in the United States, and the indication makes a aging would not only be used as an early drug development
particular point that the PET is not a replacement for tissue tool but as biomarkers of a patients susceptibility or re-
sampling and testing.95 sistance to a given therapy, a means to capture differences
Numerous other tracers are being used or developed for between metastatic lesions, and/or as an indicator of early
disease detection. However, there are limited prospective response.
studies to date that have an underlying hypothesis, adequate
power, and a preplanned strategy for confirming that im- CONCLUSION
aging findings are true or false positives or negatives. Fur- Recent studies focused on integration of preclinical obser-
ther, the actionable findings of these imaging modalities, and vations with clinical information, including specimens and
the clinical pathways that such findings should generate, imaging, and patient response data have started to reveal
remain undefined. Detection of early distant disease pro- the intricacies underlying inter- and intrapatient hetero-
vokes improvised surgical plans for local primary disease, geneity in CRPC, possibly driving differences in clinical re-
lymph node dissections in the salvage setting, irradiation of sponse to systemic agents. Tissue, liquid, and imaging-based
individual metastatic foci for the oligometastatic setting, or biomarkers in prostate cancer hold great potential as bio-
the institution of systemic therapy, in the absence of any data markers for response assessments, staging and early de-
that such maneuvers are beneficial, much less not harmful. tection of disease, and biologic characterization and drug
Trials to define these treatment algorithms must be per- development (Fig. 1). However, translation of such obser-
formed and currently are being planned and opened now. vations into the clinic to eventually impact care requires
As the age of molecular profiling in prostate cancer dawns, prospective studies demonstrating clinical value of bio-
imaging could allow for predictive biomarkers and treatment markers for prognostication or prediction of response. It is
selection by global assessments of a patients bony, nodal, crucial that development be performed with no less rigor

138 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


TISSUE, LIQUID, AND IMAGING BIOMARKERS IN CRPC

than a drug would be developed. For this to happen, more performance characteristics of the modality are defined,
prospective analytic and clinical validation studies must be followed by clinical qualification with hypothesis-driven
performed that are controlled for clinical states and have retrospectively or prospectively collected evidence of an
prespecified endpoints, predefined interventions, and ad- association between the biomarker and a clinically mean-
equate statistical power. The development of clinically ingful event. Although the technologies are ever more
qualified biomarkers is compounded by the regulatory available on a routine basis, the appropriate trials to define
framework for biomarker development, 100,101 which how to use these tools to promote how patients feel,
requires a rigorous analytic validation process in which the function, or survive must be performed.

References
1. Liu W, Laitinen S, Khan S, et al. Copy number analysis indicates 19. Mateo J, Carreira S, Sandhu S, et al. DNA-repair defects and olaparib in
monoclonal origin of lethal metastatic prostate cancer. Nat Med. metastatic prostate cancer. N Engl J Med. 2015;373:1697-1708.
2009;15:559-565. 20. Castro E, Goh C, Olmos D, et al. Germline BRCA mutations are associated
2. Haffner MC, Mosbruger T, Esopi DM, et al. Tracking the clonal origin of with higher risk of nodal involvement, distant metastasis, and poor
lethal prostate cancer. J Clin Invest. 2013;123:4918-4922. survival outcomes in prostate cancer. J Clin Oncol. 2013;31:1748-1757.
3. Gundem G, Van Loo P, Kremeyer B, et al; ICGC Prostate UK Group. The 21. Fong PC, Boss DS, Yap TA, et al. Inhibition of poly(ADP-ribose) po-
evolutionary history of lethal metastatic prostate cancer. Nature. lymerase in tumors from BRCA mutation carriers. N Engl J Med. 2009;
2015;520:353-357. 361:123-134.
4. Baca SC, Prandi D, Lawrence MS, et al. Punctuated evolution of 22. Beltran H, Eng K, Mosquera JM, et al. Whole-exome sequencing of
prostate cancer genomes. Cell. 2013;153:666-677. metastatic cancer and biomarkers of treatment response. JAMA
5. Barbieri CE, Baca SC, Lawrence MS, et al. Exome sequencing identifies Oncol. 2015;1:466-474.
recurrent SPOP, FOXA1 and MED12 mutations in prostate cancer. Nat 23. Cheng HH, Pritchard CC, Boyd T, et al. Biallelic inactivation of BRCA2 in
Genet. 2012;44:685-689. platinum-sensitive metastatic castration-resistant prostate cancer.
6. Cancer Genome Atlas Research Network. The molecular taxonomy of Eur Urol. 2015;S0302-2838(15)01177-X.
primary prostate cancer. Cell. 2015;163:1011-1025. 24. Pritchard CC, Morrissey C, Kumar A, et al. Complex MSH2 and MSH6
7. Grasso CS, Wu YM, Robinson DR, et al. The mutational landscape of mutations in hypermutated microsatellite unstable advanced pros-
lethal castration-resistant prostate cancer. Nature. 2012;487:239-243. tate cancer. Nat Commun. 2014;5:4988.
8. Robinson D, Van Allen EM, Wu YM, et al. Integrative clinical genomics 25. Beltran H. DNA mismatch repair in prostate cancer. J Clin Oncol. 2013;
of advanced prostate cancer. Cell. 2015;161:1215-1228. 31:1782-1784.
9. Beltran H, Yelensky R, Frampton GM, et al. Targeted next-generation 26. Le DT, Uram JN, Wang H, et al. PD-1 blockade in tumors with
sequencing of advanced prostate cancer identifies potential thera- mismatch-repair deficiency. N Engl J Med. 2015;372:2509-2520.
peutic targets and disease heterogeneity. Eur Urol. 2013;63:920-926. 27. de Bono JS, Scher HI, Montgomery RB, et al. Circulating tumor cells
10. Taplin ME, Bubley GJ, Shuster TD, et al. Mutation of the androgen- predict survival benefit from treatment in metastatic castration-
receptor gene in metastatic androgen-independent prostate cancer. resistant prostate cancer. Clin Cancer Res. 2008;14:6302-6309.
N Engl J Med. 1995;332:1393-1398. 28. Scher HI, Heller G, Molina A, et al. Circulating tumor cell biomarker
11. Visakorpi T, Hyytinen E, Koivisto P, et al. In vivo amplification of the panel as an individual-level surrogate for survival in metastatic
androgen receptor gene and progression of human prostate cancer. castration-resistant prostate cancer. J Clin Oncol. 2015;33:1348-1355.
Nat Genet. 1995;9:401-406. 29. Beltran H, Jendrisak A, Landers M, et al. The initial detection and
12. Chen CD, Welsbie DS, Tran C, et al. Molecular determinants of re- partial characterization of circulating tumor cells in neuroendocrine
sistance to antiandrogen therapy. Nat Med. 2004;10:33-39. prostate cancer. Clin Cancer Res. Epub 2015 Dec 15.
13. Watson PA, Arora VK, Sawyers CL. Emerging mechanisms of resistance 30. Stott SL, Lee RJ, Nagrath S, et al. Isolation and characterization of
to androgen receptor inhibitors in prostate cancer. Nat Rev Cancer. circulating tumor cells from patients with localized and metastatic
2015;15:701-711. prostate cancer. Sci Transl Med. 2010;2:25ra23.
14. Beltran H, Rickman DS, Park K, et al. Molecular characterization of 31. Danila DC, Heller G, Gignac GA, et al. Circulating tumor cell number
neuroendocrine prostate cancer and identification of new drug tar- and prognosis in progressive castration-resistant prostate cancer. Clin
gets. Cancer Discov. 2011;1:487-495. Cancer Res. 2007;13:7053-7058.
15. Beltran H, Prandi D, Mosquera JM, et al. Divergent clonal evolution of 32. Olmos, D, Arkenau HT, Ang JE, et al. Circulating tumour cell (CTC)
castration-resistant neuroendocrine prostate cancer. Nat Med. 2016. counts as intermediate end points in castration-resistant prostate
In press. cancer (CRPC): a single-centre experience. Ann Oncol. 2009;20:27-33.
16. Yu J, Yu J, Mani RS, et al. An integrated network of androgen receptor, 33. Heitzer E, Ulz P, Belic J, et al. Tumor-associated copy number changes
polycomb, and TMPRSS2-ERG gene fusions in prostate cancer pro- in the circulation of patients with prostate cancer identified through
gression. Cancer Cell. 2010;17:443-454. whole-genome sequencing. Genome Med. 2013;5:30.
17. Carver BS, Chapinski C, Wongvipat J, et al. Reciprocal feedback reg- 34. Romanel A, Tandefelt DG, Conteduca V, et al. Plasma AR and
ulation of PI3K and androgen receptor signaling in PTEN-deficient abiraterone-resistant prostate cancer. Sci Transl Med. 2015;7:
prostate cancer. Cancer Cell. 2011;19:575-586. 312re10.
18. Mulholland DJ, Tran LM, Li Y, et al. Cell autonomous role of PTEN in 35. Bettegowda C, Sausen M, Leary RJ, et al. Detection of circulating
regulating castration-resistant prostate cancer growth. Cancer Cell. tumor DNA in early- and late-stage human malignancies. Sci Transl
2011;19:792-804. Med. 2014;6:224ra24.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 139


BELTRAN ET AL

36. Carreira S, Romanel A, Goodall J, et al. Tumor clone dynamics in lethal 56. Maughan BL, Antonarakis ES. Clinical relevance of androgen receptor
prostate cancer. Sci Transl Med. 2014;6:254ra125. splice variants in castration-resistant prostate cancer. Curr Treat
37. Dawson SJ, Tsui DW, Murtaza M, et al. Analysis of circulating tumor Options Oncol. 2015;16:57.
DNA to monitor metastatic breast cancer. N Engl J Med. 2013;368: 57. Kwegyir-Afful AK, Ramalingam S, Purushottamachar P, et al. Gale-
1199-1209. terone and VNPT55 induce proteasomal degradation of AR/AR-V7,
38. Chen EJ, Sowalsky AG, Gao S, et al. Abiraterone treatment in induce significant apoptosis via cytochrome C release and suppress
castration-resistant prostate cancer selects for progesterone re- growth of castration resistant prostate cancer xenografts in vivo.
sponsive mutant androgen receptors. Clin Cancer Res. 2015;21: Oncotarget. 2015;6:27440-27460.
1273-1280. 58. Andersen RJ, Mawji NR, Wang J, et al. Regression of castrate-recurrent
39. Azad AA, Volik SV, Wyatt AW, et al. Androgen receptor gene aber- prostate cancer by a small-molecule inhibitor of the amino-terminus
rations in circulating cell-free DNA: biomarkers of therapeutic re- domain of the androgen receptor. Cancer Cell. 2010;17:535-546.
sistance in castration-resistant prostate cancer. Clin Cancer Res. 2015; 59. Myung JK, Banuelos CA, Fernandez JG, et al. An androgen receptor N-
21:2315-2324. terminal domain antagonist for treating prostate cancer. J Clin Invest.
40. Linja MJ, Savinainen KJ, Saramaki OR, et al. Amplification and over- 2013;123:2948-2960.
expression of androgen receptor gene in hormone-refractory prostate 60. Montgomery R, Antonarakis ES, Hussain M, et al. A phase 1/2 open-
cancer. Cancer Res. 2001;61:3550-3555. label study of safety and antitumor activity of EPI-506, a novel AR N-
41. Murtaza M, Dawson SJ, Pogrebniak K, et al. Multifocal clonal evolution terminal domain inhibitor, in men with metastatic castration-resistant
characterized using circulating tumour DNA in a case of metastatic prostate cancer (mCPRC) with progression after enzalutamide or
breast cancer. Nat Commun. 2015;6:8760. abiraterone. J Clin Oncol. 2015;33 (suppl; abstr TPS5072).
42. Dehm SM, Schmidt LJ, Heemers HV, et al. Splicing of a novel androgen 61. Liu C, Lou W, Zhu Y, et al. Niclosamide inhibits androgen receptor
receptor exon generates a constitutively active androgen receptor variants expression and overcomes enzalutamide resistance in
that mediates prostate cancer therapy resistance. Cancer Res. 2008; castration-resistant prostate cancer. Clin Cancer Res. 2014;20:
68:5469-5477. 3198-3210.
43. Hu DG, Hickey TE, Irvine C, et al. Identification of androgen receptor 62. Johns WD, Garnick MB, Kaplan WD. Leuprolide therapy for prostate
splice variant transcripts in breast cancer cell lines and human tissues. cancer. An association with scintigraphic flare on bone scan. Clin
Horm Cancer. 2014;5:61-71. Nucl Med. 1990;15:485-487.
44. Lu J, Lonergan PE, Nacusi LP, et al. The cistrome and gene signature of 63. Pollen JJ, Witztum KF, Ashburn WL. The flare phenomenon on ra-
androgen receptor splice variants in castration resistant prostate dionuclide bone scan in metastatic prostate cancer. AJR Am J
cancer cells. J Urol. 2015;193:690-698. Roentgenol. 1984;142:773-776.
45. Ware KE, Garcia-Blanco MA, Armstrong AJ, et al. Biologic and clinical 64. Ryan CJ, Shah S, Efstathiou E, et al. Phase II study of abiraterone
significance of androgen receptor variants in castration resistant acetate in chemotherapy-naive metastatic castration-resistant
prostate cancer. Endocr Relat Cancer. 2014;21:T87-T103. prostate cancer displaying bone flare discordant with serologic re-
46. Guo Z, Yang X, Sun F, et al. A novel androgen receptor splice variant is sponse. Clin Cancer Res. 2011;17:4854-4861.
up-regulated during prostate cancer progression and promotes an- 65. Morris MJ, Farrelly JS, Fox JJ, et al. The Prostate Cancer Clinical Trials
drogen depletion-resistant growth. Cancer Res. 2009;69:2305-2313. Consortium (PCCTC) bone scan data capture tool for clinical trials
47. Hornberg E, Ylitalo EB, Crnalic S, et al. Expression of androgen receptor using Prostate Cancer Working Group 2 (PCWG2) criteria: effect on
splice variants in prostate cancer bone metastases is associated with data accuracy and workload. J Clin Oncol. 2011;29 (suppl 7; abstr 121).
castration-resistance and short survival. PLoS One. 2011;6:e19059. 66. Ryan CJ, Smith MR, de Bono JS, et al; COU-AA-302 Investigators.
48. Zhang X, Morrissey C, Sun S, et al. Androgen receptor variants occur Abiraterone in metastatic prostate cancer without previous chemo-
frequently in castration resistant prostate cancer metastases. PLoS therapy. N Engl J Med. 2013;368:138-148.
One. 2011;6:e27970. 67. Beer TM, Armstrong AJ, Rathkopf DE, et al; PREVAIL Investigators.
49. Qu Y, Dai B, Ye D, et al. Constitutively active AR-V7 plays an essential Enzalutamide in metastatic prostate cancer before chemotherapy.
role in the development and progression of castration-resistant N Engl J Med. 2014;371:424-433.
prostate cancer. Sci Rep. 2015;5:7654. 68. Morris MJ, Molina A, Small EJ, et al. Radiographic progression-free
50. Efstathiou E, Titus M, Wen S, et al. Molecular characterization of survival as a response biomarker in metastatic castration-resistant
enzalutamide-treated bone metastatic castration-resistant prostate prostate cancer: COU-AA-302 results. J Clin Oncol. 2015;33:
cancer. Eur Urol. 2015;67:53-60. 1356-1363.
51. Efstathiou E, Wen S, San Miguel A, et al. Enzalutamide in combination 69. Kluetz PG, Ning YM, Maher VE, et al. Abiraterone acetate in combi-
with abiraterone acetate in bone-metastatic castration-resistant nation with prednisone for the treatment of patients with metastatic
prostate cancer. J Clin Oncol. 2014;32:5s (suppl; abstr 5000). castration-resistant prostate cancer: U.S. Food and Drug Adminis-
52. Antonarakis ES, Lu C, Wang H, et al. AR-V7 and resistance to enza- tration drug approval summary. Clin Cancer Res. 2013;19:6650-6656.
lutamide and abiraterone in prostate cancer. N Engl J Med. 2014;371: 70. Imbriaco M, Larson SM, Yeung HW, et al. A new parameter for
1028-1038. measuring metastatic bone involvement by prostate cancer: the Bone
53. Thadani-Mulero M, Portella L, Sun S, et al. Androgen receptor splice Scan Index. Clin Cancer Res. 1998;4:1765-1772.
variants determine taxane sensitivity in prostate cancer. Cancer Res. 71. Morris MJ, et al. Post-treatment serial bone scan index (BSI) as an
2014;74:2270-2282. outcome measure predicting for survival. Genitourinary Cancers
54. Antonarakis ES, Lu C, Luber B, et al. Androgen receptor splice Variant 7 Symposium Proceedings (2008).
and efficacy of taxane chemotherapy in patients with metastatic 72. Ulmert D, Kaboteh R, Fox JJ, et al. A novel automated platform for
castration-resistant prostate cancer. JAMA Oncol. 2015;1:582-591. quantifying the extent of skeletal tumour involvement in prostate
55. Nakazawa M, Lu C, Chen Y, et al. Serial blood-based analysis of AR-V7 cancer patients using the Bone Scan Index. Eur Urol. 2012;62:78-84.
in men with advanced prostate cancer. Ann Oncol. 2015;26: 73. Anand A, Morris MJ, Kaboteh R, et al. Analytic validation of the au-
1859-1865. tomated Bone Scan Index as an imaging biomarker to standardize

140 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


TISSUE, LIQUID, AND IMAGING BIOMARKERS IN CRPC

quantitative changes in bone scans of patients with metastatic 88. Eiber, M, Maurer T, Souvatzoglou M, et al. Evaluation of hybrid (6)(8)
prostate cancer. J Nucl Med. 2016;57:41-45. Ga-PSMA ligand PET/CT in 248 patients with biochemical recurrence
74. Armstrong AJ, Kaboteh R, Carducci MA, et al. Assessment of the bone after radical prostatectomy. J Nucl Med. 2015;56:668-674.
scan index in a randomized placebo-controlled trial of tasquinimod in 89. Gotto GT, Yu C, Bernstein M, et al. Development of a nomogram model
men with metastatic castration-resistant prostate cancer (mCRPC). predicting current bone scan positivity in patients treated with
Urol Oncol. 2014;32:1308-1316. androgen-deprivation therapy for prostate cancer. Front Oncol. 2014;
75. Grant, FD, Fahey FH, Packard AB, et al. Skeletal PET with 18F-fluoride: 4:296.
applying new technology to an old tracer. J Nucl Med. 2008;49:68-78. 90. Umbehr MH, Muntener M, Hany T, et al. The role of 11C-choline and
18
76. Schirrmeister, H, Glatting G, Hetzel J, et al. Prospective evaluation of F-fluorocholine positron emission tomography (PET) and PET/CT in
the clinical value of planar bone scans, SPECT, and (18)F-labeled NaF prostate cancer: a systematic review and meta-analysis. Eur Urol.
2013;64:106-117.
PET in newly diagnosed lung cancer. J Nucl Med. 2001;42:1800-1804.
91. Evangelista L, Zattoni F, Guttilla A, et al. Choline PET or PET/CT and
77. Hetzel M, Arslandemir C, Konig HH, et al. F-18 NaF PET for detection of
biochemical relapse of prostate cancer: a systematic review and meta-
bone metastases in lung cancer: accuracy, cost-effectiveness, and
analysis. Clin Nucl Med. 2013;38:305-314.
impact on patient management. J Bone Miner Res. 2003;18:
92. Giovacchini G, Picchio M, Briganti A, et al. [11C]choline positron
2206-2214.
emission tomography/computerized tomography to restage prostate
78. Shen CT, Qiu ZL, Han TT, et al. Performance of 18F-fluoride PET or PET/CT
cancer cases with biochemical failure after radical prostatectomy and
for the detection of bone metastases: a meta-analysis. Clin Nucl Med.
no disease evidence on conventional imaging. J Urol. 2010;184:
2015;40:103-110. 938-943.
79. Scher HI, Halabi S, Tannock I, et al. Trial design and objectives for 93. Giovacchini G, Picchio M, Scattoni V, et al. PSA doubling time for
castration-resistant prostate cancer: updated recommendations from prediction of [(11)C]choline PET/CT findings in prostate cancer pa-
the Prostate Cancer Clinical Trials Working Group 3. J Clin Oncol. 2016; tients with biochemical failure after radical prostatectomy. Eur J Nucl
34:1402-1418. Med Mol Imaging. 2010;37:1106-1116.
80. Stephenson AJ, Scardino PT, Kattan MW, et al. Predicting the outcome 94. Krause BJ, Souvatzoglou M, Tuncel M, et al. The detection rate of [11C]
of salvage radiation therapy for recurrent prostate cancer after radical choline-PET/CT depends on the serum PSA-value in patients with
prostatectomy. J Clin Oncol. 2007;25:2035-2041. biochemical recurrence of prostate cancer. Eur J Nucl Med Mol Im-
81. Stephenson AJ, Scardino PT, Eastham JA, et al. Postoperative no- aging. 2008;35:18-23.
mogram predicting the 10-year probability of prostate cancer re- 95. U.S. Food and Drug Administration. FDA Approves Production of
currence after radical prostatectomy. J Clin Oncol. 2005;23: Imaging Agent That Helps Detect Prostate Cancer. http://www.fda.
7005-7012. gov/NewsEvents/Newsroom/PressAnnouncements/ucm319201.htm.
82. Kattan MW, Zelefsky MJ, Kupelian PA, et al. Pretreatment nomogram Accessed April 27, 2016.
for predicting the outcome of three-dimensional conformal radio- 96. Scher HI, Sawyers CL. Biology of progressive, castration-resistant
therapy in prostate cancer. J Clin Oncol. 2000;18:3352-3359. prostate cancer: directed therapies targeting the androgen-
83. Foss CA, Mease RC, Cho SY, et al. GCPII imaging and cancer. Curr Med receptor signaling axis. J Clin Oncol. 2005;23:8253-8261.
Chem. 2012;19:1346-1359. 97. Scher HI, Beer TM, Higano CS, et al; Prostate Cancer Foundation/
84. Schuster DM, Nieh PT, Jani AB, et al. Anti-3-[(18)F]FACBC positron emission Department of Defense Prostate Cancer Clinical Trials Consortium.
Antitumour activity of MDV3100 in castration-resistant prostate
tomography-computerized tomography and (111)In-capromab pendetide
cancer: a phase 1-2 study. Lancet. 2010;375:1437-1446.
single photon emission computerized tomography-computerized to-
98. Rathkopf DE, Morris MJ, Fox JJ, et al. Phase I study of ARN-509, a novel
mography for recurrent prostate carcinoma: results of a prospective clinical
antiandrogen, in the treatment of castration-resistant prostate can-
trial. J Urol. 2014;191:1446-1453.
cer. J Clin Oncol. 2013;31:3525-3530.
85. Pandit-Taskar N, ODonoghue JA, Durack JC, et al. A phase I/II study for
99. Evans MJ, Smith-Jones PM, Wongvipat J, et al. Noninvasive mea-
analytic validation of 89Zr-J591 ImmunoPET as a molecular imaging
surement of androgen receptor signaling with a positron-emitting
agent for metastatic prostate cancer. Clin Cancer Res. 2015;21: radiopharmaceutical that targets prostate-specific membrane anti-
5277-5285. gen. Proc Natl Acad Sci USA. 2011;108:9578-9582.
86. Pandit-Taskar N, ODonoghue J, Martin D, et al. First in human 89Zr-Df- 100. U.S. Department of Health and Human Services. FDA Guidance:
IAB2M anti-prostate specific membrane antigen (PSMA) minibody in Clinical Trial Imaging Endpoints Process Standards, Draft Guidance.
patients with metastatic prostate cancer. J Nucl Med. 2015;56:400. 2011.
87. Afshar-Oromieh A, Zechmann CM, Malcher A, et al. Comparison of PET 101. U.S. Food and Drug Administration. Guidance for Industry and FDA
imaging with a (68)Ga-labelled PSMA ligand and (18)F-choline-based Staff: Qualification Process for Drug Development Tools. January 2014.
PET/CT for the diagnosis of recurrent prostate cancer. Eur J Nucl Med www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatory
Mol Imaging. 2014;41:11-20. Information/Guidances/UCM230597.pdf. Accessed April 17, 2012.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 141


GYNECOLOGIC CANCER

Paradigm Shift in the


Management Strategy for
Epithelial Ovarian Cancer

CHAIR
Keiichi Fujiwara, MD, PhD
Saitama Medical University International Medical Center
Hidaka, Japan

SPEAKERS
Amit M. Oza, MD
Princess Margaret Cancer Centre
Toronto, ON, Canada

Jessica N. McAlpine, MD
University of British Columbia
Vancouver, BC, Canada
PARADIGM SHIFT IN OVARIAN CANCER STRATEGY

Paradigm Shift in the Management Strategy for Epithelial


Ovarian Cancer
Keiichi Fujiwara, MD, PhD, Jessica N. McAlpine, MD, Stephanie Lheureux, MD, PhD, Noriomi Matsumura,
MD, PhD, and Amit M. Oza, BSc, MD, MBBS, FRCPC

OVERVIEW

The hypothesis on the pathogenesis of epithelial ovarian cancer continues to evolve. Although epithelial ovarian cancer had
been assumed to arise from the coelomic epithelium of the ovarian surface, it is now becoming clearer that the majority of
serous carcinomas arise from epithelium of the distal fallopian tube, whereas clear cell and endometrioid cancers arise from
endometriosis. Molecular and genomic characteristics of epithelial ovarian cancer have been extensively investigated. Our
understanding of pathogenesis of the various histologic types of ovarian cancer have begun to inform changes to the
strategies for management of epithelial ovarian cancer, which represent a paradigm shift not only for treatment but also for
prevention, which previously had not been considered achievable. In this article, we will discuss novel attempts at the
prevention of high-grade serous ovarian cancer and treatment strategies for two distinct entities in epithelial ovarian cancer:
low-grade serous and clear cell ovarian carcinomas, which are relatively rare and resistant to conventional chemotherapy.

W e have seen numerous advances in the field of ovarian


cancer in the past decade, yet overall survival sta-
tistics for this disease are essentially unchanged. Despite the
Alternative opportunities for prevention were inspired by
the appreciation of the role of the fallopian tube in ovarian
cancer (Fig. 1). The distal fallopian tube is the site of origin of
excellent design and execution of multiple screening trials, the majority of the most common histotype of epithelial
the relative impact in terms of number of cases of early ovarian cancerhigh-grade serousin women with he-
ovarian cancer detected has been small and, unfortunately, reditary predisposition (e.g., BRCA1/2 mutation) and in
tempered by the number of unnecessary surgeries performed sporadic occurrences in the general population.11,12 In ad-
as a result of abnormal screening results.1-4 New methods of dition, the fallopian tube serves as a conduit for the passage
disease control are desperately needed, and this has prompted a of endometrial inflammatory cytokines, infections, and/or
push to explore methods of primary prevention. irritants to the ovary and peritoneal cavity.13 Ectopic en-
dometrium may undergo malignant transformation and is
the purported site of origin of the next two most common
PROPHYLACTIC SALGINGECTOMY: SHOULD ALL histotypes of epithelial ovarian cancer: clear cell and
TUBES BE REMOVED? endometrioid cancers.14 Long-substantiated risk factors for
In BRCA1/2-mutation carriers, risk-reducing bilateral salpingo- ovarian cancer make sense in the context of the critical role
oophorectomy has been shown to be highly protective for that the fallopian tube plays in ovarian cancer. Inflammatory
ovarian cancer, with a reduced risk of at least 80% after sur- conditions, such as pelvic inflammatory disease, incessant
gery.5,6 Most high-risk women have reportedly experienced a ovulation, or irritants such as talc that have ascended from
high quality of physical and mental well-being after risk-reducing the lower genital tract via the fallopian tube to the tubo-
bilateral salpingo-oophorectomy,7 and all-cause mortality is ovarian junction, increase mutagenesis and increase the risk
significantly reduced by this procedure.5,8 However, risk-reducing of ovarian cancer. Interventions that decrease these parameters
bilateral salpingo-oophorectomy is not recommended for the (e.g., tubal ligation, ovarian quiescence during pregnancy, or
general/low-risk population, because removal of the ovaries is breast feeding) are associated with a decreased risk of ovarian
reportedly associated with increased mortality, coronary heart cancer.13 Large studies with international cohorts of women
disease, stroke, osteoporosis, and colorectal cancer.9,10 who have undergone tubal ligation have shown a decrease in

From the Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Saitama, Japan; Department of Gynecology and Obstetrics, University of British
Columbia, Vancouver, Canada; Division of Medical Oncology and Hematology, Bras Family Drug Development Program, Princess Margaret Cancer Centre, Toronto, Canada; Department
of Gynecology and Obstetrics, Kyoto University, Kyoto, Japan.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Keiichi Fujiwara, MD, PhD, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka 350-1241, Japan;
email: fujiwara@saitama-med.ac.jp.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e247


FUJIWARA ET AL

risk of developing ovarian carcinoma by 29% overall, with Columbia, which included a review of the rationale for
histotype-specific variation in protective effects. The greatest salpingectomy. This initiative has had a profound effect on
risk reduction was observed in endometrioid (52%) and clear clinical practice in British Columbia: the proportion of
cell (48%) histologies.15 Risk reduction was only 20% in high- hysterectomies that had associated salpingectomies (ex-
grade serous carcinoma; however, given that this histotype cluding hysterectomies in which ovaries were removed, the
encompasses 70% of all epithelial ovarian cancers and accounts rate of which remains unchanged) increased from 8% in
for 90% of deaths, this impact is still substantial. A recent meta- 2008 to 63% in 2011 and to 75% in 2013, and the proportion
analysis of published series with tubal ligation data reported of sterilizations by bilateral salpingectomy increased from
similar findings,16 and, importantly, the protective effect (60% 0.5% in 2008 to 33% in 2011 and to 48% in 2013.18 The
risk reduction) of tubal ligation has also been observed in high- Society of Gynecologic Oncologists of Canada, U.S. Society
risk populations (BRCA1/2-mutation carriers).17 Although not for Gynecologic Oncology, American College of Obstetrics
the focus of this article, oral contraception pills have arguably and Gynecology, and Royal Australian and New Zealand
had the greatest success in ovarian cancer risk reduction globally College of Obstetricians and Gynecologists have all since
and across all histotypes; however, the mechanism for the published statements that recommend consideration of
protective effect of the oral contraception pills is not fully salpingectomy for all women at the time of gynecologic
understood. Past and current hypotheses again suggest an surgery,19-22 and physician surveys show willingness to un-
important role of the fallopian tube, and the influence of the dertake practice change.23-26 It is essential to clarify that this
progesterone component of oral contraception pills on tubal campaign is aimed at women in the general (low-risk) population
epithelial cell proliferation (differentiated and/or stem cell) is who have a lifetime risk of developing ovarian cancer of ap-
under active investigation. proximately 1.7%; importantly, risk-reducing bilateral salpingo-
oophorectomy at age 40 or after completion of childbearing27 is
The Intervention: Is There an Opportunity to Remove still recommended and represents the standard of care in high-
Fallopian Tubes? risk populations, such as in BRCA1/2-mutation carriers. A two-
Beginning in 2008, and then as a formal initiative in 2010, we, step procedure of risk-reducing salpingectomy followed by
as members of the British Columbia Ovarian Cancer Re- oophorectomy in high-risk women, although feasible, carries
search Team (OVCARE) that is based in Vancouver, have increased surgical risks, and there is insufficient data to support
suggested to gynecologic surgeons that they should consider optimal timing or protective effect (if any) for breast and ovarian
the following: perform bilateral salpingectomy in all women cancers; thus, the two-step procedure is reserved for women
at the time of hysterectomy (even when the ovaries are who are unwilling to undergo risk-reducing bilateral salpingo-
being preserved) and perform bilateral salpingectomy in oophorectomy.28,29
place of tubal ligation for sterilization. In September 2010, We have termed this intervention in the low-risk pop-
we circulated a DVD to all gynecologic surgeons in British ulation opportunistic salpingectomy, which suggests that
surgeons should take the opportunity to remove the fal-
lopian tubes if there is access to these anatomic structures
during other scheduled gynecologic procedures but should
KEY POINTS not perform a procedure solely for the purpose of tubal
removal for ovarian cancer prevention. We have published
Appreciation of the changing landscape of epithelial data from a time period 2 years prior and 2 years after the
ovarian cancer enables the development of novel formal introduction of this campaign, including data from
strategies in treatment and prevention.
procedures performed on 43,931 women. We observed no
Opportunistic salpingectomy should be considered in
women undergoing gynecologic procedures with access
increase in major perioperative complications or events.
to the peritoneal cavity, at hysterectomy, or for Blood loss, length of hospital stay, and readmission rates
permanent sterilization. For women at increased risk of were the same in patients who underwent opportunistic
ovarian carcinoma development, risk-reducing bilateral salpingectomy and in patients who underwent hysterec-
salpingo-oophorectomy remains standard of care. tomy alone or tubal ligation procedures. 18 Potential
Accruing data support the safety, acceptability, long-term hormonal consequences of opportunistic sal-
feasibility, and cost effectiveness of this procedure. pingectomy have been investigated through measurement
Low-grade serous ovarian cancer, a rare subtype of of ovarian sonographic parameters and hormonal assays;
ovarian cancer, is relatively chemotherapy resistant, data are reassuring but have relatively short follow-up.30-32
and current development focus is on targeted therapy Given that an earlier age at menopause has been associated
that is based on its distinct biology.
with increased mortality, it is imperative that this does not
Clear cell ovarian cancer is also a distinct pathological
and clinical entity of rare epithelial ovarian cancer. Its
offset any projected protective effect in ovarian cancer risk
carcinogenesis from endometriosis is now becoming reduction. Hysterectomy is known to affect ovarian reserve33,34
clearer, and research on molecular characteristics of and the relative impact of opportunistic salpingectomy per-
clear cell ovarian cancer has led to the new trials of formed with hysterectomy is likely small or immeasurable. To
target agents. answer this definitively we are studying our British Columbia
cohort of women who have undergone these procedures,

e248 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


PARADIGM SHIFT IN OVARIAN CANCER STRATEGY

FIGURE 1. Role of the Fallopian Tube in Ovarian Cancer

comparing onset of menopause (defined as cessation of ovarian cancer. The calculated number needed to treat to
menses for 1 year) in individuals who have undergone bilateral prevent a single occurrence of ovarian cancer was accept-
salpingectomy versus tubal ligation for permanent sterilization. able for both procedures.39
In addition, there is a planned randomized clinical trial in the
United Kingdom in women who will undergo bilateral sal-
pingectomy either at hysterectomy or as a stand-alone pro- Measuring the Impact of Opportunistic
cedure for contraception.35 Salpingectomy
Is the increased uptake of opportunistic salpingectomy Finally, and most importantly, will this change in surgical
responsible in terms of use of resources and health eco- paradigm translate to a decreased incidence of ovarian
nomics? Our group and others have shown that this pro- cancer? Proof of success of this initiative will be an observed
cedure can be undertaken by different surgical routes reduction in new occurrences of ovarian cancer, and we
(vaginal, minimally invasive, laparotomy)18,36-38 so it does anticipate there will be a shift in the distribution of histo-
not need to be limited to high-resource countries. Additional types of epithelial ovarian cancer in the population of
operating room time needed for opportunistic salpingec- women exposed to this procedure. The age of women un-
tomy with hysterectomy is approximately 16 minutes, and, dergoing this procedure as part of hysterectomy or for
in lieu of tubal ligation, the procedure requires approxi- permanent sterilization is younger than the age of onset of
mately 10 minutes,18 a time that arguably is of no clinical ovarian cancer in the general population, so we anticipate
impact to the majority of women undergoing these pro- that it will be at least 10 years and up to 20 years from the
cedures nor to the health systems where the procedures are start of our campaign before we will be able to discern a
being performed. Cost analysis modeling, which considers difference that would provide definitive support for this
perioperative risks, impact on ovarian cancer risk reduction, intervention. Pooling of our data with data from other
and morbidities associated with premature menopause geographic areas that have adopted this practice may help
secondary to oophorectomy, found that opportunistic sal- power the analysis and hasten results. Although nay-sayers
pingectomy with hysterectomy was less costly and more may be frustrated or unsatisfied with this lack of immediate
effective than hysterectomy alone, reduced the number of measurable impact, we hold to this long view and are very
ovarian cancer occurrences, and prolonged the average life encouraged by emerging evidence from other historical
expectancy. Opportunistic salpingectomy for sterilization cohorts that supports this intervention. In addition to the
would be considered more costly than tubal ligation in terms strong protective effect demonstrated in women who have
of operative time and complication risk; however, oppor- undergone tubal ligation (outlined previously), there are
tunistic salpingectomy was more effective at reducing risk of data from a small number of studies on women who have

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e249


FUJIWARA ET AL

undergone excisional tubal surgery (defined as complete LOW-GRADE SEROUS OVARIAN CANCER
salpingectomy, distal fimbriectomy, or partial salpingectomy). Current Clinical Practice
Researchers from the Rochester Epidemiology Project reported a The standard strategy for advanced epithelial ovarian cancer
64% reduction in the risk of ovarian cancer after excisional tubal remains the combination of surgery and platinum/taxane-
sterilization compared with those without sterilization or with based chemotherapy,44 which is primarily driven by activity
nonexcisional tubal sterilization (odds ratio [OR] 0.36; 95% CI, in unselected populations enrolled in clinical trials; the
0.131.02).40 Danish researchers, who used a national database, populations are predominantly composed of women with
reported that bilateral salpingectomy reduced risk for ovarian high-grade serous ovarian cancer. One of five histologic sub-
cancer by 42% (OR 0.58; 95% CI, 0.360.95).41 Most recently, a types of epithelial ovarian cancer, low-grade serous ovarian
retrospective population-based study that used Swedish health cancer is a rare subentity with a specific genomic landscape, as
registers reported that bilateral salpingectomy was associated evidenced by a different natural history and pattern of re-
with a 65% reduction in risk (hazard ratio 0.35; 95% CI, sponse to therapy.45 From available evidence, low-grade se-
0.170.73).42 Both the Danish and Swedish studies examined rous ovarian cancer may arise after an initial diagnosis of serous
retrospective data and included women who underwent sal- tumor of low malignant potential (Fig. 2) or de novo and does
pingectomies for pathologic reasons (e.g., hydrosalpinx, pelvic not seem to be part of the hereditary breast-ovarian cancer
inflammatory disease, ectopic pregnancy), which makes them syndrome related to the BRCA1/2 gene mutation.46
significantly different from the women undergoing opportunistic The dogma of cytotoxic chemotherapy for all epithelial
salpingectomy in British Columbia for the purposes of ovarian ovarian cancers is evolving on the basis of the recent clinical
cancer prophylaxis. We hypothesize that salpingectomy per- and molecular classification of the different subtypes of
formed for risk-reduction purposes may confer more protection epithelial ovarian cancer.47 To date, no prospective ran-
than those performed for other indications, because surgeons domized clinical trial data are available to provide guidance
will be more careful to remove the entire distal end of the about the optimal postoperative treatment of low-grade
fallopian tube. serous ovarian cancer.48 Although available data are mostly
Published data reassure us of the safety of the procedure and based on small retrospective, single-institution studies, che-
suggest that opportunistic salpingectomy should be easily within motherapy resistance is consistently reported in low-grade
the skill set of any pelvic/abdominal surgeon and can be per- serous ovarian cancer.48,49 From a single-institution database,
formed by multiple surgical routes; there is health economic 112 women with stages II to IV low-grade serous ovarian cancer
support, and the evidence of high uptake suggests feasibility. In who underwent primary surgery followed by platinum-based
addition, there are numerous nononcologic arguments for tubal chemotherapy were retrospectively identified. Of these, 42
removal, including prevention of hydrosalpinges, pyosalpinx, or patients underwent second-look surgery, and 2, 13, and 24
tubal prolapse that may require subsequent surgeries.43 However, had evidence of no residual disease, microscopically positive
as we wait to definitively measure the impact of this procedure on disease, or macroscopically positive disease, respectively.50
ovarian cancer risk, whether a woman undergoes opportunistic Similar findings were reported with low-grade primary peri-
salpingectomy will remain a decision between her and her toneal cancer, with high rates of persistent disease at the
treating physician, and the decision requires good judgment and completion of adjuvant chemotherapy.51 This relative che-
common sense. Our first dictum as physicians is to do no harm. motherapy resistance, possibly related to the nature of

FIGURE 2. Carcinogenesis Pathway in Low-Grade Serous Ovarian Cancer

Pictures courtesy of Dr. Patricia A. Shaw, University Health Network of Toronto.

e250 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


PARADIGM SHIFT IN OVARIAN CANCER STRATEGY

low-grade serous ovarian cancer, was also observed in a of the regimens resulted in a progression-free survival (PFS)
separate retrospective study that included patients treated duration of at least 6 months. Regimens that involved treat-
with neoadjuvant chemotherapy.52 Despite the receipt of a ment of estrogen receptorpositive/progesterone receptor
taxane and platinum combination in the majority of the 25 positive disease produced a longer median time to progression
patients with advanced low-grade serous ovarian cancer, only (8.9 months) than regimens that involved treatment of es-
one patient had a complete response (CR); an additional 21 trogen receptorpositive/progesterone receptor2negative
women had stable disease, but two experienced progression disease (time to progression, 6.2 months; p = .053). Given the
after neoadjuvant chemotherapy. Although CA-125 levels were relatively favorable adverse effect profile and demonstrated
reduced by greater than 50% with chemotherapy in half of the efficacy, hormonal therapy represents an interesting treatment
patients, radiologic response remained low. alternative.
Chemotherapy resistance also has been demonstrated in
the setting of recurrent disease. For example, in a retro-
spective study of 58 women with recurrent low-grade serous The Mitogen-Activated Protein Kinase Pathway
ovarian cancer who received 108 separate chemotherapy The mitogen-activated protein kinase (MAPK) pathway is
regimens,49 only four responses were seen (one CR and activated and appears to play a prominent role in the
three partial responses; overall response rate [ORR], 3.7%). pathogenesis of low-grade serous ovarian cancer.59 Ap-
The ORR was 4.9% for the platinum-sensitive cohort and proximately 20% to 40% of low-grade serous carcinomas
2.1% for the platinum-resistant cohort. Stable disease that have a KRAS mutation, whereas BRAF mutations are rare
was observed in 65 (60.2%) of 108 chemotherapy regimens (approximately 5%).60,61 KRAS is a frequent mutation de-
may have been related to tumor biology or may have been a tected in advanced low-grade serous ovarian cancer, and the
bias of assessmentmeasureable lesions can often appear BRAF V600E mutation is associated with serous borderline
stable by RECIST criteria on CT scans53and not a true tumors and early-stage low-grade serous ovarian cancer.62
therapeutic effect. As such, given the relative chemotherapy As a result, the V600E mutation is associated with a better
resistance, we should consider making clinical trials the new clinical outcome. In a study of 23 patients with an original
standard. Targeted biologically directed therapy is an in- diagnosis of serous tumor of low malignant potential who
teresting area of investigation in low-grade serous ovarian subsequently experienced recurrence with a diagnosis of
cancer, given the molecular characteristics of disease. low-grade serous ovarian cancer, patients with KRAS G12V
Compared with high-grade serous ovarian cancer, low-grade mutations experienced shorter survival times than those
serous ovarian cancer has lower expression of p53, WT1, with either KRAS G12D, wild-type, or rare KRAS variants
c-KIT, Ki-67, and MMP-954 but a higher expression of estrogen (hazard ratio 4.77; p = .023).63 Another retrospective study
receptor, progesterone receptor, and E-cadherin.55 showed the potential impact of mutational status; patients
with KRAS and BRAF mutations appeared to have better
overall survival than those with wild-type KRAS or BRAF.64
Hormonal Therapy Additional investigations are warranted to elucidate the role
Low-grade serous ovarian cancer often is diagnosed at a of the mutation type in low-grade serous ovarian cancer,
younger age (43 to 55 years), so a higher proportion of because this may have important clinical implications, such
patients with low-grade serous ovarian cancer are pre- as introduction of the analysis of BRAF/KRAS status in the
menopausal at diagnosis; as such, hormonal status may postdebulking setting to predict the risk of recurrence.65
be implicated in pathogenesis. 56 Indeed, among the five The MAPK cascade is triggered by the binding of a ligand
different subtypes of ovarian cancer, low-grade serous that ultimately leads to phosphorylation of ERK.66,67 Thus,
ovarian cancer has the higher proportion of hormone MEK is a good candidate for targeted therapy, and a number
receptorpositive (progesterone receptor and/or estrogen of MEK inhibitors (MEKi) have been developed.
receptorpositive) tumors.57 In this large retrospective The GOG-0239 open-label phase II study of patients with
study, strong progesterone but not estrogen receptor ex- recurrent low-grade serous ovarian cancer, by prospective
pression appeared to be associated with improved survival pathologic evaluation, received selumetinib (AZD6244)
after accounting for site, age, stage, and grade (p = .019 for a MEK1/2 inhibitor that targets the downstream effect of
progesterone and p = .78 for estrogen receptor). But this activating BRAF and KRAS mutationsat a dosage of 50 mg
association was not statistically significant after adjusting for twice daily.68 The majority of patients had received three or
residual disease in this subset of 64 patients with low-grade more prior chemotherapy regimens. Fifty-two women with
serous ovarian cancer (p = .27 and .90, respectively).57 The recurrent low-grade serous ovarian cancer were enrolled,
agents used as hormonal therapy, which are targeted and the ORR was 15% (one CR and seven partial responses).
therapies against the progesterone receptor and estrogen Another 65% of patients in the trial had stable disease, and
receptor, are mainly tamoxifen and aromatase inhibitors the median PFS was 11.0 months. The most common tox-
(anastrozole and letrozole). A retrospective study in 64 icities were gastrointestinal, dermatologic, and metabolic.
patients with recurrent low-grade serous ovarian cancer Three patients experienced grade 4 toxicitiesone each of
who received 89 separate hormonal regimens showed an cardiac, pain, and pulmonary toxicity. Mutational analysis
ORR of 9% (six CRs and two partial responses).58 In total, 61% was conducted on formalin-fixed, paraffin-embedded tumor

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e251


FUJIWARA ET AL

samples from 34 patients enrolled in this trial, and the A (VEGF-A), showed sustained responses of 15-, 15-, and
primary tumor accounted for 82% of the cases. In these 34 22-month durations.70 From a retrospective cohort of 17
cases, there were two BRAF mutations (6%) and 14 KRAS patients with low-grade serous ovarian cancer and serous
mutations (41%), and 15% of tumors had NRAS mutations. In borderline tumors, two patients were treated with single-
this study, there was no correlation between mutations agent bevacizumab, and the others were treated with a
of BRAF or KRAS and ORR; however, only hotspot muta- combination of bevacizumab and chemotherapy.71 Fifteen
tions were assessed. Tissue analysis of an extraordinary patients were evaluable for response; six had a partial re-
responder who experienced a complete, durable, and ongoing sponse, and five had stable disease that lasted 3 months or
(. 5 years) response to selumetinib identified a previously longer. The response rate for the low-grade serous ovarian
uncharacterized MAP2K1 deletion (Q56_V60del). Additional cancer group was 55%. Therefore, additional studies to
investigations support that this novel deletion is a driver evaluate the possible role of antiangiogenics in low-grade
alteration in this exceptional responder and serves as the serous ovarian cancer are warranted, potentially in com-
molecular basis for her dramatic, sustained response to the bination with MAPK inhibitors.
MEKi.69 This finding may explain why targeted genotyping of
only the most common hotspot alterations in BRAF and KRAS
Future Directions
failed to ascertain the molecular basis for a subset of the
The insulin-like growth factor (IGF) pathway is another
responses observed in the GOG-0239 trial and justify the
pathway overexpressed in low-grade serous ovarian cancer,
incorporation of broader profiling methods. After these
and related effectors, such as PI3K/Akt/mTOR, have been
promising results, several studies have been designed, such
also described to play a role in disease pathogenesis.72
as the ongoing GOG-281 trial. GOG-281 is a randomized
Activating mutations of PI3KCA are observed in approxi-
phase II study between investigator choice (of letrozole,
mately 40% of tumors, whereas inactivating PTEN mutations
tamoxifen, weekly paclitaxel, pegylated liposomal doxoru-
are present in 3% to 8% of tumors.73 AMG-479, a fully human
bicin, or weekly topotecan) and the MEKi agent trametinib
antiinsulin-like growth factor receptor type I monoclonal
for recurrent low-grade serous ovarian cancer. The study
antibody, had been assessed in frontline and recurrent
incorporates prospective pathology review, pretissue bi-
settings in ovarian cancer but not specifically in low-grade
opsy, and blood collection for correlative analyses, such as
serous ovarian cancer (NCT00718523 and NCT00719212).
next-generation sequencing and proteomics. The primary
OSI-906, a tyrosine kinase inhibitor of both IGF-1R and the
endpoint is PFS, the estimated enrolment is 250 patients,
insulin receptor was assessed in recurrent ovarian cancer
and crossover at disease progression is allowed.
(NCT00889382).
Several additional targeted studies are also ongoing in
On the basis of retrospective studies and ad hoc co-
this space. NCT01936363 was a randomized phase II trial
operative group studies, low-grade serous ovarian cancer is
for recurrent low-grade serous ovarian cancer that com-
described as relatively chemotherapy resistant, which has
pared pimasertib (MEKi) plus or minus SAR245409a
led to the investigation of novel therapies that are based on
phosphatidylinositol-4,5-biphosphate 3-kinase and mTOR.
the specific molecular pathways of low-grade serous ovarian
This randomized phase II trial started, and patients were
cancer. Although low-grade serous ovarian cancer is asso-
recruited, but the trial subsequently was stopped because of
ciated with superior overall survival compared with the
toxicity with the combination. No results have been pre-
other histologic subgroups of ovarian cancer, more than 70%
sented or published as yet. The MILO trial (NCT01849874)
of low-grade serous ovarian cancer patients experience
is a randomized, open-label phase III study between phy-
relapse and die of their disease. There is an urgent clinical
sician choice (paclitaxel, liposomal doxorubicin, topotecan)
need to develop additional trials of combination targeted
and binimetinib, or MEK162, for recurrent low-grade serous
agents that are tolerable for the patients and that do not
ovarian cancer. In addition, the RTM 1313 study is a ran-
significantly impinge upon quality of life. Because low-grade
domized phase II trial with patients with newly diagnosed
serous ovarian cancer is a rare entity, the cooperation be-
stages II to IV low-grade serous ovarian cancer that is in-
tween different institutes is essential alongside a common
vestigating trametinib/GSK 214170550 every 3 weeks for six
effort to reduce heterogeneity of grading and biases in
cycles versus standard adjuvant chemotherapy carboplatin/
treatment and response evaluation.65
paclitaxel for six cycles. Taken together, biomarkers that predict
MEKi activity and the identification of MEKi-independent
compensatory pathways are needed. CLEAR CELL OVARIAN CANCER: TIME FOR A NEW
PARADIGM?
Clear cell ovarian cancer is a unique entity of adenocarci-
Antiangiogenesis noma of the ovary. Sugiyama et al74 first reported that
The angiogenesis pathway may also be a therapeutic target advanced-stage clear cell ovarian cancer was less sensitive
in patients with low-grade serous ovarian cancer. An initial than the serous counterpart to conventional chemotherapy
report of three patients with recurrent low-grade serous and provoked the discussion about its uniqueness.74
ovarian cancer treated with bevacizumab, the monoclonal Recently, a marked ethnic difference in the incidence of
antibody against the vascular endothelial growth factor clear cell ovarian cancer has been recognized, although the

e252 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


PARADIGM SHIFT IN OVARIAN CANCER STRATEGY

reason is not clear. The incidence of clear cell ovarian cancer cytology could suggest microscopic implantation of clear cell
is less than 10% in Europe and North America.75 However, in ovarian cancer cells.
Japan, the prevalence of clear cell ovarian cancer is in- Another important recent observation is the incidence of
creasing, and now approximately 25% of epithelial ovarian thromboembolic complication in clear cell ovarian cancer. It
cancer is clear cell, according to the Japanese Society of has been reported to be higher than in other epithelial
Obstetrics and Gynecology tumor registry data from 2014.74 ovarian cancers (16.9%27.3% vs. 0%6.8%),77 and it is also
A hypothesis for the reason of increasing incidence of this associated with an elevated interleukin 6 level and with
disease in Japan is the increasing incidence of endometriosis. worse prognosis.80
An, association between clear cell ovarian cancer and
endometriosis has been reported (relative risk, 12.4).76 The Chemotherapy Resistance
risk increased significantly when the patients were di- Several studies that retrospectively analyzed the prognosis
agnosed at older ages (. 50), which suggests that the of patients in large randomized clinical trials have been
malignant change of endometriosis occurs near menopause reported in terms of difference of prognosis between
stage. Pathogenesis of clear cell ovarian cancer from en- pathologic subtypes.81,82 The survival was significantly
dometriosis will be discussed later. worse in clear cell than in the serous counterpart in ad-
vanced stages, which suggests that clear cell ovarian cancer
Clinical Features of Clear Cell Ovarian Cancer was resistant to conventional chemotherapy regimens. New
Most clear cell ovarian cancer tumors are unilateral at di- cytotoxic regimens, dose-dense paclitaxel regimen,83 and
agnosis, and most are diagnosed at an early stage.77 In a CPT-11 plus cisplatin78 did not demonstrate a benefit for
prospective randomized trial (JGOG 3017), the proportion of clear cell ovarian cancer. Chemotherapy administered in-
patients with stage I disease was 67%.78 The incidence of traperitoneally using cisplatin and paclitaxel improved the
lymph node metastasis was 9.1% in apparent stage IA tu- OS in optimally debulked stage III ovarian cancer, but a
mors, 7.1% in stage IC tumors, and was 10.8% in pT2 tu- survival benefit with intraperitoneal chemotherapy was not
mors.77 This stands in contrast with serous ovarian cancers, observed in clear cell ovarian cancer and mucinous ade-
which were associated with a higher incidence of lymph nocarcinoma.84 Therefore, finding new strategies for ad-
node metastasis than nonserous tumors.79 Although it is vanced or relapsed clear cell ovarian cancer is urgent.
controversial, evaluation of lymph node status by surgical Finding molecular pathologic characteristics of this disease
staging is recommended, because lymph node involvement to direct improvement of targeted therapies is critical.
in patients with clinical stage I clear cell ovarian cancer was
identified as a stronger prognostic factor.77 Pathogenesis of Clear Cell Ovarian Cancer From
Management of clear cell ovarian cancer with positive Endometriosis
peritoneal cytology or surgical rupture remains unclear. Investigators from Kyoto University have conducted a series
Disease progression was observed in 11% of stage IC of excellent studies to delineate the pathogenesis of clear
intraoperative rupture tumors but in only 3% of stage IA cell ovarian cancer from endometriosis (Fig. 3).
tumors.77 Progression-free survival of the patients who had They first analyzed the content of endometriotic cysts and
stage IC tumors with ascites/malignant washing or ovarian found that the cysts contained huge amounts of iron, oxi-
surface involvement was significantly worse than patients dative stress marker LPO, and 8-0HdG, a marker of DNA
who had stage IC disease with capsule rupture during sur- damage caused by oxidative stress. When the content of an
gery (p = .04),77 which implies that positive peritoneal endometriotic cyst or iron was added to immortalized

FIGURE 3. Hypothetical Carcinogenic Pathway of Clear Cell Ovarian Cancer From Endometriotic Cyst

Abbreviations: IL, interleukin; HNF1b, hepatocyte nuclear factor 1-beta; CCOC, clear cell ovarian cancer.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e253


FUJIWARA ET AL

ovarian surface epithelial cells, intracellular reactive oxygen may be important for progression in the stressful condition
species was elevated, and the contents of endometriotic cyst of endometriotic cysts and platinum resistance. Epigenetic
or iron increased DNA mutations. Therefore, researchers changes, gene mutations, and copy number alterations may
hypothesized that iron-mediated reactive oxygen species cause stabilization of clear cell ovarian cancerspecific gene
may cause DNA mutations and carcinogenesis.85 expression and biologic features, including chemotherapy
Next, researchers conducted a gene expression microarray resistance.
analysis and identified clear cellspecific genes, including
HNF1B, SOD2, ANXA4 HIF1A, IL6, and STAT3. They enriched
Molecular and Genomic Features of Clear Cell Ovarian
gene ontology terms related to oxidative stress and glucose
Cancer
metabolism. Hepatocyte nuclear factor (HNF) 1-beta binds
Compared with other histologic types of epithelial ovarian
to DNA as either a homodimer or a heterodimer with the
cancer, mutation in p53 is less frequent90 in clear cell ovarian
related protein HNF 1-alpha. HNF1 alpha and beta share
cancer, and mutation of BRCA1 and BRCA2 is also lower.91
a common biding motif. Genes having the HNF1 binding
Wilms tumor suppressor 1 gene (WT1) and the WT1-
motif in promoter regions are candidate downstream genes
antisense promoter were significantly methylated in clear
of HNF 1 beta. Interestingly, these genes were upregulated
cell ovarian cancer (88.2%) compared with serous adeno-
in immortalized ovarian surface epithelial cells by adding the
carcinoma (24%).92 PTEN mutation is also frequently ob-
content of endometriotic cysts or iron.86 Researchers also
served (27.3%) in this disease.93 Multidrug resistance
conducted a methylation DNA microarray analysis, which
protein 3 (MRP3)94 and HNF1B, which has antiapoptotic
revealed that clear cell is distinct from other subtypes in terms
effects, are highly expressed.95 More recently, frequent
of methylation profile. Estrogen receptor pathway genes were
alteration of ARID1A96 and PIK3CA97 has been reported.
hypermethylated and downregulated, whereas HNF1 pathway
Mabuchi et al showed that the Akt/mTOR pathway is
genes were hypermethylated and upregulated.87 Therefore,
thought to be the most important passage for tumor growth
clear cell ovarian cancerspecific gene expression seems to be
in clear cell ovarian cancer.98,99
stabilized via epigenetic mechanisms.
Recently, Uehara et al100 conducted genomic analyses on
The researchers also investigated the roles of HNF1B in
clear cell ovarian cancer and found that, by using micro-
metabolism of clear cell ovarian cancer cells and found that
array analysis, this disease could be classified into three
HNF1B increases glucose uptake by increasing expression
clusters: one that showed fewer alterations of PIK3CA and
of a glucose transporter, GLUT1.88 A subsequent metab-
ARID1A and a better prognosis compared with the two
olome analysis was done and revealed that upregulated
other clusters, which had more alterations.100 This work
HNF1B expression enhances anaerobic glucose metabolism.
suggests that the genomic pattern may be a strong
Known as the Warburg effect, the enhanced anaerobic
prognostic factor.
glucose metabolism has been reported to cause resistance
to oxidative stress. Investigators also analyzed the re-
lationship between HNF1B and oxidative stress in clear cell Recently Closed or Ongoing Clinical Trials and Future
ovarian cancer. Knockdown of HNF1B decreased the amount Directions
of glutathione, a redox substance. This was due to decreased As mentioned earlier, the Japanese Gynecologic Oncology
intracellular cystine, a substrate for the biosynthesis of Group conducted a randomized phase III trial to compare
glutathione, via the decreased expression of rBAT, a cystine conventional paclitaxel with carboplatin regimen versus an
transporter. Then, the investigators found that HNF1B irinotecan with cisplatin regimen for patients with newly
knockdown increased intracellular reactive oxygen species diagnosed stage I to IV clear cell ovarian cancer. This is the
and cytotoxicity by iron-induced oxidative stress. Further- first randomized trial specifically on this disease. Un-
more, in hypoxia, suppression of HNF1B increased sensi- fortunately, there was no survival benefit between the
tivity to cisplatin. Collectively, HNF1B in clear cell carcinoma regimens.78
causes resistance to oxidative stress and platinum.89 On the basis of the molecular profile pattern of clear cell
Finally, researchers conducted an exome sequencing ovarian cancer, the therapeutic target of interest involves
analysis of clear cell ovarian cancer. ARID1A was the top- the Akt/mTOR pathway. The GOG-268 study tested one of
mutated gene, and PIK3CA was the second one. Through an the mTOR inhibitors, temsirolimus, administered in com-
integrated analysis of gene mutations and copy number bination with paclitaxel and carboplatin for six cycles and
variations, researchers found that the KRAS-PI3K pathway, then given for 11 more cycles as a maintenance therapy.
SWI/SNF complex, and MYC-RB pathway were the most Although the results will be presented this year at the
frequently altered pathways (written communication with 2016 American Society of Clinical Oncology Annual Meeting,
N. Matsumura, February 2016). we await comparison of results in the United States and
The researchers hypothesize that iron-induced oxidative South Korea with those obtained in Japan. In addition, the
stress may cause DNA damage and mutations of PIK3CA and translational component of this trial is underway to evaluate
ARID1A, which may lead to carcinogenesis of clear cell the possible difference on molecular characteristics of ad-
ovarian cancer. HNF1B plays an important role in the vanced clear cell ovarian cancer between Japanese and non-
Warburg effect and in resistance to oxidative stress. This Japanese populations.

e254 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


PARADIGM SHIFT IN OVARIAN CANCER STRATEGY

Antiangiogenetic agents are also of interest as therapeutic of the disease coupled with a heightened awareness of the
targets for clear cell ovarian cancer. The GOG-254 study genomic variability of this cancer, with recognition that
evaluated the efficacy and safety of sunitinib and showed that ovarian cancer is not one disease but comprises many types
sunitinib was minimally active in the second- and third-line of histologies, each with its own genomic landscape. We
treatments of persistent or recurrent clear cell ovarian cancer. are now looking at opportunistic salpingectomy as a means
Another antiangiogenetic agent being tested is nintedanib, of prevention, which builds on the growing evidence that
and a Scottish group is conducting a randomized phase II study implicates the fallopian tube as the origin of high-grade
to compare nintedanib to other chemotherapeutic agents. serous ovarian cancer. In addition, we are seeing a sepa-
Finally, immunotherapy that uses checkpoint inhibitors is ration of treatment paradigms, particularly when it comes
of interest as a treatment strategy. One recent publication to the treatment of high-grade serous ovarian cancer
on an antibody to PD-1, nivolumab, showed promising signs versus low-grade serous ovarian cancer. We hope that
of efficacy for clear cell ovarian cancer.101 continued progress in translational research will yield
important findings that can be used therapeutically to
CONCLUSION redefine our approach to epithelial ovarian cancer, in-
The treatment of epithelial ovarian cancer has been in- cluding the rarer histologies, such as clear cell and low-
formed by an improved understanding of the pathogenesis grade serous cancers.

References
1. Buys SS, Partridge E, Black A, et al; PLCO Project Team. Effect of 12. Przybycin CG, Kurman RJ, Ronnett BM, et al. Are all pelvic (nonuterine)
screening on ovarian cancer mortality: the Prostate, Lung, Colorectal serous carcinomas of tubal origin? Am J Surg Pathol. 2010;34:1407-1416.
and Ovarian (PLCO) cancer screening randomized controlled trial. 13. Tone AA, Salvador S, Finlayson SJ, et al. The role of the fallopian tube in
JAMA. 2011;305:2295-2303. ovarian cancer. Clin Adv Hematol Oncol. 2012;10:296-306.
2. Kobayashi H, Yamada Y, Sado T, et al. A randomized study of screening 14. Kurman RJ, Shih IeM. Molecular pathogenesis and extraovarian origin
for ovarian cancer: a multicenter study in Japan. Int J Gynecol Cancer. of epithelial ovarian cancer: shifting the paradigm. Hum Pathol. 2011;
2008;18:414-420. 42:918-931.
3. Menon U, Gentry-Maharaj A, Hallett R, et al. Sensitivity and specificity 15. Sieh W, Salvador S, McGuire V, et al; Australian Cancer Study (Ovarian
of multimodal and ultrasound screening for ovarian cancer, and stage Cancer); Australian Ovarian Cancer Study Group; Ovarian Cancer Asso-
distribution of detected cancers: results of the prevalence screen of ciation Consortium. Tubal ligation and risk of ovarian cancer subtypes:
the UK Collaborative Trial of Ovarian Cancer Screening (UKCTOCS). a pooled analysis of case-control studies. Int J Epidemiol. 2013;42:579-589.
Lancet Oncol. 2009;10:327-340. 16. Rice MS, Murphy MA, Tworoger SS. Tubal ligation, hysterectomy and
4. Jacobs IJ, Menon U, Ryan A, et al. Ovarian cancer screening and ovarian cancer: a meta-analysis. J Ovarian Res. 2012;5:13.
mortality in the UK Collaborative Trial of Ovarian Cancer Screening 17. Narod SA, Sun P, Ghadirian P, et al. Tubal ligation and risk of ovarian
(UKCTOCS): a randomised controlled trial. Lancet. 2015;S0140-6736 cancer in carriers of BRCA1 or BRCA2 mutations: a case-control study.
(15)01224-6. Lancet. 2001;357:1467-1470.
5. Domchek SM, Friebel TM, Singer CF, et al. Association of risk-reducing 18. McAlpine JN, Hanley GE, Woo MM, et al. Opportunistic salpingec-
surgery in BRCA1 or BRCA2 mutation carriers with cancer risk and tomy: uptake, risks, and complications of a regional initiative for
mortality. JAMA. 2010;304:967-975. ovarian cancer prevention. Am J Obstet Gynecol. 2014;210:471 e1-11.
6. Marchetti C, De Felice F, Palaia I, et al. Risk-reducing salpingo- 19. The Society of Gynecologic Oncology of Canada (GOC). GOC statement
oophorectomy: a meta-analysis on impact on ovarian cancer risk regarding salpingectomy and ovarian cancer prevention, 2011. http://
and all cause mortality in BRCA1 and BRCA2 mutation carriers. BMC www.g-o-c.org/uploads/11sept15_gocevidentiarystatement_final_
Womens Health. 2014;14:150. en.pdf. Accessed March 12, 2016.
7. Finch A, Metcalfe KA, Chiang J, et al. The impact of prophylactic 20. Society of Gynecologic Oncology (SGO). SGO clinical practice state-
salpingo-oophorectomy on quality of life and psychological dis- ment: salpingectomy for ovarian cancer, 2013. https://www.sgo.org/
tress in women with a BRCA mutation. Psychooncology. 2013;22: clinical-practice/guidelines/sgo-clinical-practice-statement-salpingectomy-
212-219. for-ovarian-cancer-prevention/. Accessed March 12, 2016.
8. Finch AP, Lubinski J, Mller P, et al. Impact of oophorectomy on cancer 21. Committee on Gynecologic Practice. Committee opinion no. 620:
incidence and mortality in women with a BRCA1 or BRCA2 mutation. salpingectomy for ovarian cancer prevention. Obstet Gynecol. 2015;
J Clin Oncol. 2014;32:1547-1553. 125:279-281.
9. Parker WH, Broder MS, Chang E, et al. Ovarian conservation at the 22. Kapurubandara S, Qin V, Gurram D, et al. Opportunistic bilateral
time of hysterectomy and long-term health outcomes in the nurses salpingectomy during gynaecological surgery for benign disease:
health study. Obstet Gynecol. 2009;113:1027-1037. a survey of current Australian practice. Aust N Z J Obstet Gynaecol.
10. Parker WH, Feskanich D, Broder MS, et al. Long-term mortality associated 2015;55:606-611.
with oophorectomy compared with ovarian conservation in the nurses 23. Venturella R, Rocca M, Lico D, et al. Prophylactic bilateral sal-
health study. Obstet Gynecol. 2013;121:709-716. pingectomy for the prevention of ovarian cancers: what is happening
11. Kindelberger DW, Lee Y, Miron A, et al. Intraepithelial carcinoma of in Italy? Eur J Cancer Prev. Epub 2015 Aug 13.
the fimbria and pelvic serous carcinoma: evidence for a causal re- 24. Parker W. Oophorectomy versus salpingectomy: a convergence of
lationship. Am J Surg Pathol. 2007;31:161-169. ideas. Menopause. 2014;21:323-324.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e255


FUJIWARA ET AL

25. Reade CJ, Finlayson S, McAlpine J, et al. Risk-reducing salpingectomy 44. Jayson GC, Kohn EC, Kitchener HC, et al. Ovarian cancer. Lancet. 2014;
in Canada: a survey of obstetrician-gynaecologists. J Obstet Gynaecol 384:1376-1388.
Can. 2013;35:627-634. 45. Sung PL, Chang YH, Chao KC, et al; Task Force on Systematic Review
26. Gill SE, Mills BB. Physician opinions regarding elective bilateral sal- and Meta-analysis of Ovarian Cancer. Global distribution pattern of
pingectomy with hysterectomy and for sterilization. J Minim Invasive histological subtypes of epithelial ovarian cancer: a database analysis
Gynecol. 2013;20:517-521. and systematic review. Gynecol Oncol. 2014;133:147-154.
27. American College of Obstetritians and Gynecologists (ACOG). ACOG 46. Gershenson DM. Molecular targeting of low-grade serous and mu-
practice bulletin No. 89: elective and risk-reducing salpingo-oopho- cinous carcinomas of the ovary or peritoneum. Transl Cancer Res.
rectomy. Obstet Gynecol. 2008;111:231-241. 2015;4:29-39.
28. Chandrasekaran D, Menon U, Evans G, et al. Risk reducing sal- 47. Bookman MA, Gilks CB, Kohn EC, et al. Better therapeutic trials in
pingectomy and delayed oophorectomy in high-risk women: views of ovarian cancer. J Natl Cancer Inst. 2014;106:dju029.
cancer geneticists, genetic counselors and gynecological oncologists 48. Gourley C, Farley J, Provencher DM, et al. Gynecologic Cancer
in the UK. Fam Cancer. 2015;14:521-530. InterGroup (GCIG) consensus review for ovarian and primary
29. Kwon JS, Tinker A, Pansegrau G, et al. Prophylactic salpingectomy and peritoneal low-grade serous carcinomas. Int J Gynecol Cancer.
delayed oophorectomy as an alternative for BRCA mutation carriers. 2014;24(9 Suppl 3)S9-S13.
Obstet Gynecol. 2013;121:14-24. 49. Gershenson DM, Sun CC, Bodurka D, et al. Recurrent low-grade serous
30. Morelli M, Venturella R, Mocciaro R, et al. Prophylactic salpingectomy ovarian carcinoma is relatively chemoresistant. Gynecol Oncol. 2009;
in premenopausal low-risk women for ovarian cancer: primum non 114:48-52.
nocere. Gynecol Oncol. 2013;129:448-451. 50. Gershenson DM, Sun CC, Lu KH, et al. Clinical behavior of stage II-IV low-
31. Venturella R, Morelli M, Lico D, et al. Wide excision of soft tissues adjacent grade serous carcinoma of the ovary. Obstet Gynecol. 2006;108:361-368.
to the ovary and fallopian tube does not impair the ovarian reserve in 51. Schmeler KM, Sun CC, Malpica A, et al. Low-grade serous primary
women undergoing prophylactic bilateral salpingectomy: results from a peritoneal carcinoma. Gynecol Oncol. 2011;121:482-486.
randomized, controlled trial. Fertil Steril. 2015;104:1332-1339. 52. Schmeler KM, Sun CC, Bodurka DC, et al. Neoadjuvant chemotherapy
32. Findley AD, Siedhoff MT, Hobbs KA, et al. Short-term effects of sal- for low-grade serous carcinoma of the ovary or peritoneum. Gynecol
pingectomy during laparoscopic hysterectomy on ovarian reserve: Oncol. 2008;108:510-514.
a pilot randomized controlled trial. Fertil Steril. 2013;100:1704-1708. 53. Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation
33. Yuan H, Wang C, Wang D, et al. Comparing the effect of laparoscopic criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J
supracervical and total hysterectomy for uterine fibroids on ovarian Cancer. 2009;45:228-247.
reserve by assessing serum anti-mullerian hormone levels: a pro- 54. ONeill CJ, Deavers MT, Malpica A, et al. An immunohistochemical
spective cohort study. J Minim Invasive Gynecol. 2015;22:637-641. comparison between low-grade and high-grade ovarian serous
34. Wang HY, Quan S, Zhang RL, et al. Comparison of serum anti-mullerian carcinomas: significantly higher expression of p53, MIB1, BCL2,
hormone levels following hysterectomy and myomectomy for benign gy- HER-2/neu, and C-KIT in high-grade neoplasms. Am J Surg Pathol.
necological conditions. Eur J Obstet Gynecol Reprod Biol. 2013;171:368-371. 2005;29:1034-1041.
35. Manchanda R, Chandrasekaran D, Saridogan E, et al. Should oppor- 55. Wong KK, Lu KH, Malpica A, et al. Significantly greater expression of
tunistic bilateral salpingectomy (OBS) for prevention of ovarian cancer ER, PR, and ECAD in advanced-stage low-grade ovarian serous car-
be incorporated into routine care or offered in the context of a clinical cinoma as revealed by immunohistochemical analysis. Int J Gynecol
trial? Int J Gynecol Cancer. 2016;26:31-33. Pathol. 2007;26:404-409.
36. Robert M, Cenaiko D, Sepandj J, et al. Success and complications of 56. Plaxe SC. Epidemiology of low-grade serous ovarian cancer. Am J
salpingectomy at the time of vaginal hysterectomy. J Minim Invasive Obstet Gynecol. 2008;198:459 e1-8.
Gynecol. 2015;22:864-869. 57. Sieh W, Kobel M, Longacre TA, et al. Hormone-receptor expression
37. Vorwergk J, Radosa MP, Nicolaus K, et al. Prophylactic bilateral sal- and ovarian cancer survival: an Ovarian Tumor Tissue Analysis con-
pingectomy (PBS) to reduce ovarian cancer risk incorporated in sortium study. Lancet Oncol. 2013;14:853-862.
standard premenopausal hysterectomy: complications and re- 58. Gershenson DM, Sun CC, Iyer RB, et al. Hormonal therapy for recurrent
operation rate. J Cancer Res Clin Oncol. 2014;140:859-865. low-grade serous carcinoma of the ovary or peritoneum. Gynecol
38. Kamran MW, Vaughan D, Crosby D, et al. Opportunistic and interventional Oncol. 2012;125:661-666.
salpingectomy in women at risk: a strategy for preventing pelvic serous 59. Romero I, Sun CC, Wong KK, et al. Low-grade serous carcinoma: new
cancer (PSC). Eur J Obstet Gynecol Reprod Biol. 2013;170:251-254. concepts and emerging therapies. Gynecol Oncol. 2013;130:660-666.
39. Kwon JS, McAlpine JN, Hanley GE, et al. Costs and benefits of op- 60. Singer G, Oldt R III, Cohen Y, et al. Mutations in BRAF and KRAS
portunistic salpingectomy as an ovarian cancer prevention strategy. characterize the development of low-grade ovarian serous carcinoma.
Obstet Gynecol. 2014;125:338-345. J Natl Cancer Inst. 2003;95:484-486.
40. Lessard-Anderson CR, Handlogten KS, Molitor RJ, et al. Effect of tubal 61. Wong KK, Tsang YT, Deavers MT, et al. BRAF mutation is rare in
sterilization technique on risk of serous epithelial ovarian and primary advanced-stage low-grade ovarian serous carcinomas. Am J Pathol.
peritoneal carcinoma. Gynecol Oncol. 2014;135:423-427. 2010;177:1611-1617.
41. Madsen C, Baandrup L, Dehlendorff C, et al. Tubal ligation and sal- 62. Grisham RN, Iyer G, Garg K, et al. BRAF mutation is associated with
pingectomy and the risk of epithelial ovarian cancer and borderline early-stage disease and improved outcome in patients with low-grade
ovarian tumors: a nationwide case-control study. Acta Obstet Gynecol serous ovarian cancer. Cancer. 2013;119:548-554.
Scand. 2015;94:86-94. 63. Tsang YT, Deavers MT, Sun CC, et al. KRAS (but not BRAF) mutations in
42. Falconer H, Yin L, Gronberg H, et al. Ovarian cancer risk after sal- ovarian serous borderline tumour are associated with recurrent low-
pingectomy: a nationwide population-based study. J Natl Cancer Inst. grade serous carcinoma. J Pathol. 2013;231:449-456.
2015;107. 64. Gershenson DM, Sun CC, Wong KK. Impact of mutational status on
43. Dietl J, Wischhusen J, Hausler SF. The post-reproductive fallopian survival in low-grade serous carcinoma of the ovary or peritoneum. Br
tube: better removed? Hum Reprod. 2011;26:2918-2924. J Cancer. 2015;113:1254-1258.

e256 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


PARADIGM SHIFT IN OVARIAN CANCER STRATEGY

65. Della Pepa C, Tonini G, Santini D, et al. Low-grade serous ovarian cancer (JGOG 3016): a randomised, controlled, open-label trial. Lancet
carcinoma: from the molecular characterization to the best thera- Oncol. 2013;14:1020-1026.
peutic strategy. Cancer Treat Rev. 2015;41:136-143. 84. Tewari D, Java JJ, Salani R, et al. Long-term survival advantage and
66. Friday BB, Adjei AA. Advances in targeting the Ras/Raf/MEK/Erk prognostic factors associated with intraperitoneal chemotherapy
mitogen-activated protein kinase cascade with MEK inhibitors for treatment in advanced ovarian cancer: a gynecologic oncology group
cancer therapy. Clin Cancer Res. 2008;14:342-346. study. J Clin Oncol. 2015;33:1460-1466.
67. Jing J, Greshock J, Holbrook JD, et al. Comprehensive predictive 85. Yamaguchi K, Mandai M, Toyokuni S, et al. Contents of endometriotic
biomarker analysis for MEK inhibitor GSK1120212. Mol Cancer Ther. cysts, especially the high concentration of free iron, are a possible
2012;11:720-729. cause of carcinogenesis in the cysts through the iron-induced per-
68. Farley J, Brady WE, Vathipadiekal V, et al. Selumetinib in women with sistent oxidative stress. Clin Cancer Res. 2008;14:32-40.
recurrent low-grade serous carcinoma of the ovary or peritoneum: an 86. Yamaguchi K, Mandai M, Oura T, et al. Identification of an ovarian
open-label, single-arm, phase 2 study. Lancet Oncol. 2013;14:134-140. clear cell carcinoma gene signature that reflects inherent disease
69. Grisham RN, Sylvester BE, Won H, et al. Extreme outlier analysis biology and the carcinogenic processes. Oncogene. 2010;29:
identifies occult mitogen-activated protein kinase pathway mutations 1741-1752.
in patients with low-grade serous ovarian cancer. J Clin Oncol. 2015; 87. Yamaguchi K, Huang Z, Matsumura N, et al. Epigenetic determinants of
33:4099-4105. ovarian clear cell carcinoma biology. Int J Cancer. 2014;135:585-597.
70. Bidus MA, Webb JC, Seidman JD, et al. Sustained response to bev- 88. Okamoto T, Mandai M, Matsumura N, et al. Hepatocyte nuclear
acizumab in refractory well-differentiated ovarian neoplasms. factor-1b (HNF-1b) promotes glucose uptake and glycolytic activity in
Gynecol Oncol. 2006;102:5-7. ovarian clear cell carcinoma. Mol Carcinog. 2015;54:35-49.
71. Grisham RN, Iyer G, Sala E, et al. Bevacizumab shows activity in pa- 89. Amano Y, Mandai M, Yamaguchi K, et al. Metabolic alterations caused
tients with low-grade serous ovarian and primary peritoneal cancer. by HNF1b expression in ovarian clear cell carcinoma contribute to cell
Int J Gynecol Cancer. 2014;24:1010-1014. survival. Oncotarget. 2015;6:26002-26017.
72. Groen RS, Gershenson DM, Fader AN. Updates and emerging ther- 90. Ho ES, Lai CR, Hsieh YT, et al. p53 mutation is infrequent in clear cell
apies for rare epithelial ovarian cancers: one size no longer fits all. carcinoma of the ovary. Gynecol Oncol. 2001;80:189-193.
Gynecol Oncol. 2015;136:373-383. 91. Kobel M, Kalloger SE, Boyd N, et al. Ovarian carcinoma subtypes are
73. Angarita AM, Cholakian D, Fader AN. Low-grade serous carcinoma: different diseases: implications for biomarker studies. PLoS Med.
molecular features and contemporary treatment strategies. Expert 2008;5:e232.
Rev Anticancer Ther. 2015;15:893-899. 92. Kaneuchi M, Sasaki M, Tanaka Y, et al. WT1 and WT1-AS genes are
74. Sugiyama T, Kamura T, Kigawa J, et al. Clinical characteristics of clear inactivated by promoter methylation in ovarian clear cell adenocar-
cell carcinoma of the ovary: a distinct histologic type with poor cinoma. Cancer. 2005;104:1924-1930.
prognosis and resistance to platinum-based chemotherapy. Cancer. 93. Sato N, Tsunoda H, Nishida M, et al. Loss of heterozygosity on 10q23.3
2000;88:2584-2589. and mutation of the tumor suppressor gene PTEN in benign endo-
75. del Carmen MG, Birrer M, Schorge JO. Clear cell carcinoma of the metrial cyst of the ovary: possible sequence progression from benign
ovary: a review of the literature. Gynecol Oncol. 2012;126:481-490. endometrial cyst to endometrioid carcinoma and clear cell carcinoma
76. Kobayashi H, Sumimoto K, Moniwa N, et al. Risk of developing ovarian of the ovary. Cancer Res. 2000;60:7052-7056.
cancer among women with ovarian endometrioma: a cohort study in 94. Ohishi Y, Oda Y, Uchiumi T, et al. ATP-binding cassette superfamily
Shizuoka, Japan. Int J Gynecol Cancer. 2007;17:37-43. transporter gene expression in human primary ovarian carcinoma.
77. Takano M, Kikuchi Y, Yaegashi N, et al. Clear cell carcinoma of the Clin Cancer Res. 2002;8:3767-3775.
ovary: a retrospective multicentre experience of 254 patients with 95. Tsuchiya A, Sakamoto M, Yasuda J, et al. Expression profiling in ovarian
complete surgical staging. Br J Cancer. 2006;94:1369-1374. clear cell carcinoma: identification of hepatocyte nuclear factor-1 beta
78. Okamoto A, Sugiyama T, Hamano T, et al. Randomized phase III trial of as a molecular marker and a possible molecular target for therapy of
paclitaxel/carboplatin (PC) versus cisplatin/irinotecan (CPT-P) as first- ovarian clear cell carcinoma. Am J Pathol. 2003;163:2503-2512.
line chemotherapy in patients with clear cell carcinoma (CCC) of the 96. Wiegand KC, Shah SP, Al-Agha OM, et al. ARID1A mutations in
ovary: A Japanese Gynecologic Oncology Group (JGOG)/GCIG study. J endometriosis-associated ovarian carcinomas. N Engl J Med. 2010;
Clin Oncol. 2014;32:5s (suppl; abstr 5507). 363:1532-1543.
79. Takeshima N, Hirai Y, Umayahara K, et al. Lymph node metastasis in 97. Kuo KT, Mao tubal ligation, Jones S, et al. Frequent activating mu-
ovarian cancer: difference between serous and non-serous primary tations of PIK3CA in ovarian clear cell carcinoma. Am J Pathol. 2009;
tumors. Gynecol Oncol. 2005;99:427-431. 174:1597-1601.
80. Matsuo K, Hasegawa K, Yoshino K, et al. Venous thromboembolism, 98. Hisamatsu T, Mabuchi S, Matsumoto Y, et al. Potential role of mTORC2
interleukin-6 and survival outcomes in patients with advanced ovarian as a therapeutic target in clear cell carcinoma of the ovary. Mol Cancer
clear cell carcinoma. Eur J Cancer. 2015;51:1978-1988. Ther. 2013;12:1367-1377.
81. Winter WE III, Maxwell GL, Tian C, et al; Gynecologic Oncology Group 99. Mabuchi S, Kawase C, Altomare DA, et al. mTOR is a promising
Study. Prognostic factors for stage III epithelial ovarian cancer: a Gyne- therapeutic target both in cisplatin-sensitive and cisplatin-resistant
cologic Oncology Group Study. J Clin Oncol. 2007;25:3621-3627. clear cell carcinoma of the ovary. Clin Cancer Res. 2009;15:5404-5413.
82. Mackay HJ, Brady MF, Oza AM, et al; Gynecologic Cancer InterGroup. 100. Uehara Y, Oda K, Ikeda Y, et al. Integrated copy number and expression
Prognostic relevance of uncommon ovarian histology in women with stage analysis identifies profiles of whole-arm chromosomal alterations and
III/IV epithelial ovarian cancer. Int J Gynecol Cancer. 2010;20:945-952. subgroups with favorable outcome in ovarian clear cell carcinomas.
83. Katsumata N, Yasuda M, Isonishi S, et al; Japanese Gynecologic On- PLoS One. 2015;10:e0128066.
cology Group. Long-term results of dose-dense paclitaxel and car- 101. Hamanishi J, Mandai M, Ikeda T, et al. Safety and antitumor activity of
boplatin versus conventional paclitaxel and carboplatin for treatment anti-pd-1 antibody, nivolumab, in patients with platinum-resistant
of advanced epithelial ovarian, fallopian tube, or primary peritoneal ovarian cancer. J Clin Oncol. 2015;33:4015-4022.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e257


GYNECOLOGIC CANCER

Divide and Conquer: Epithelial


Gynecologic Cancers Beyond
BRCA

CHAIR
Elise C. Kohn, MD
National Cancer Institute at the National Institutes of Health
Bethesda, MD

SPEAKERS
Paul J. Goodfellow, PhD
The Ohio State University College of Medicine
Columbus, OH

Rebecca S. Kristeleit, MD, PhD


UCL Cancer Institute
London, United Kingdom
TARGETING DNA REPAIR DEFICIENCY IN GYNECOLOGIC CANCERS

Gynecologic Cancers: Emerging Novel Strategies for Targeting


DNA Repair Deficiency
Rebecca S. Kristeleit, MD, PhD, Rowan E. Miller, MD, PhD, and Elise C. Kohn, MD

OVERVIEW

The presence of a BRCA mutation, somatic or germline, is now established as a standard of care for selecting patients with
ovarian cancer for treatment with a PARP inhibitor. During the clinical development of the PARP inhibitor class of agents, a
subset of women without BRCA mutations were shown to respond to these drugs (termed BRCAness). It was hypothesized
that other genetic abnormalities causing a homologous recombinant deficiency (HRD) were sensitizing the BRCA wild-type
cancers to PARP inhibition. The molecular basis for these other causes of HRD are being defined. They include individual gene
defects (e.g., RAD51 mutation, CHEK2 mutation), homozygous somatic loss, and whole genome properties such as genomic
scarring. Testing this knowledge is possible when selecting patients to receive molecular therapy targeting DNA repair, not
only for patients with ovarian cancer but also endometrial and cervical cancers. The validity of HRD assays and multiple gene
sequencing panels to select a broader population of patients for treatment with PARP inhibitor therapy is under evaluation.
Other non-HRD targets for exploiting DNA repair defects in gynecologic cancers include mismatch repair (MMR), checkpoint
signaling, and nonhomologous end-joining (NHEJ) DNA repair. This article describes recent evidence supporting strategies in
addition to BRCA mutation for selecting patients for treatment with PARP inhibitor therapy. Additionally, the challenges and
opportunities of exploiting DNA repair pathways other than homologous recombination for molecular therapy in gyne-
cologic cancers is discussed.

O ur increased understanding of cancer biology coupled


with increasingly refined technology to examine the cancer
genome has offered a rich supply of opportunities applicable
double-strand breaks via the highly conserved homologous-
recombination repair (HRR) pathway.5 Although the syn-
thetic lethality between deleterious BRCA mutations and
to improving outcomes in patients with gynecologic cancer.1 PARP inhibition is well established,4,5 it is becoming increas-
We now recognize at least six major interactive pathways ingly evident that gynecologic tumors have other molecu-
involved in DNA damage and repair.2 The most recent ex- lar features, germline or somatic, which portend an HRD
ample of molecularly targeted drug success in patients with or BRCA-like susceptibility to platinums and DNA repair in-
ovarian cancer is the development of PARP inhibitors as ther- hibitors. Substantial efforts are underway to categorize the
apeutics. The PARP inhibitor olaparib was the first drug breadth of molecular causes of HRD. Individual genetic mu-
worldwide to be licensed in 2014 for a molecularly defined tations and whole genome features, expressed as genomic
population of patients with BRCA-mutated ovarian cancer. scarring, have been identified as HRD-causing and correlate
The rapidity and success of identifying this molecularly di- with potential responsiveness to DNA repair inhibitors.6-9 In
rected and defined therapeutic has catalyzed the gynecologic addition, defects in other DNA repair pathways, such as the
cancer community to further explore the application of DNA MMR pathway (common in patients with endometrial cancer)
repair inhibitors as a class and question other ways DNA repair and cell cycle checkpoint proteins, cause potential vulnera-
defects might be harnessed for novel treatment approaches. bilities that offer therapeutic possibilities (Fig. 1).10-13 Going
Targeting tumors with defective DNA repair exploits the beyond BRCA-targeting in gynecologic cancers highlights the
molecular differences between tumor and normal cells. This potential for expanded therapeutic strategies. The pheno-
mechanism is the basis for tumor-specific cell death induced typic and genotypic consequences of HRD are a particular
by PARP inhibitors in patients with BRCA-mutated ovarian vulnerability in patients with epithelial ovarian cancer, and
cancer.3,4 BRCA1 and BRCA2 are essential for maintaining new insight shows how other events in endometrial, cervical,
genomic stability through the error-free repair of DNA and ovarian cancers also may yield a HRD phenotype. An

From the Department of Medical Oncology, University College London Hospital, London, United Kingdom; UCL Cancer Institute, University College London, London, United Kingdom;
Clinical Investigations Branch, Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Rebecca S. Kristeleit, MD, PhD, UCL Cancer Institute, 72 Huntley St., London WC1E 6BT United Kingdom; email: r.kristeleit@ucl.ac.uk.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e259


KRISTELEIT, MILLER, AND KOHN

immediate challenge is learning how best to navigate and FIGURE 1. Representation of the Main DNA Repair
apply the new information at both the scientific and clinical Pathways and Interaction
levels, where genetically defined treatment decisions are now
being made with patients.

HOMOLOGOUS-RECOMBINATION REPAIR AND


BRCA1/2
DNA is constantly subjected to damage by environmental
exposures and endogenous activities such as DNA replica-
tion and cellular free radical generation. These cause a variety
of DNA lesions, including base modifications, double-strand
breaks, and single-strand breaks.14 DNA repair is critical to
maintain genomic integrity by allowing cells to progress
through the cell cycle and complete replication without
errors.15 Homologous-recombination repair is the principle
mechanism by which double-strand breaks are repaired.
The BRCA1/2 genes, along with other genes in the Fanconi
anemia (FA) pathway, encode essential proteins for this
process. Homologous-recombination repair is a conserva-
tive form of DNA repair that restores the DNA to its original
sequence using the homologous normal DNA template dur-
ing S and G2/M phases of the cell cycle. When either BRCA1
or BRCA2 are defective, homologous recombination is dys-
functional and double-strand break repair proceeds using
error-prone nonconservative repair mechanisms such as NHEJ
and single-strand repair.5 Nonhomologous end-joining does
This figure represents the six main DNA repair pathways and their close but
not use a DNA template and occurs in G0 or G1, propagating discrete interaction. Targetable proteins associated with each pathway for which
error rather than repairing it.16 Any two free DNA break ends pharmacological agents exist appear in red.
Abbreviations: MMR, mismatch repair; BER, base excision repair; NHEJ,
are directly ligated during repair of double-strand breaks by nonhomologous end-joining; HRR, homologous recombination repair; NER,
NHEJ. nucleotide excision repair; TLJ, translesional joining.
A variety of mechanisms exist for repairing single-strand
breaks. These include base excision repair, nucleotide ex- PARP. PARP senses and binds to DNA-break sites, which
cision repair, and MMR, processes that are modulated by results in catalytic activation and the recruitment of other
components of the DNA repair complex.17 If a cell fails to
repair single-strand breaks before attempting replication, a
KEY POINTS double-strand break will then form.
Inherited mutations in the tumor suppressor genes BRCA1
The synthetic lethality and validity of using BRCA1/2 and BRCA2 account for the majority of familial ovarian
mutation as a biomarker predictive of response to PARP cancers.18 The BRCA1 and BRCA2 protein products function
inhibition has been confirmed in clinical trials of patients in multiple cellular pathways, including cell cycle regulation
with ovarian cancer. and maintenance of genome integrity.19 Cells with defective
Homologous recombinant deficiency (HRD) represents a HRR must rely on alternative pathways for DNA repair to
key vulnerability in patients with high-grade ovarian
survive, thereby providing potential therapeutic targets.
cancer and possibly other gynecologic cancers, which
can be exploited using PARP inhibitors.
Patients with epithelial ovarian cancer and germline or so-
Methods to identify HRD cancers and broaden the matic BRCA1 or BRCA2 mutations demonstrate impaired
applicability of DNA repair inhibitors are being ability to repair double-strand breaks through HRR, which
evaluated in the clinic. likely explains the increased sensitivity to platinum and the
Nonhomologous recombinant DNA repair pathways, potentially more favorable outcome compared with patients
such as mismatch repair and nonhomologous who are wild-type.20,21
end-joining, represent important targets for novel
therapeutic strategies in gynecologic cancer.
Targeting cell cycle checkpoint proteins such as CHK1,
HOMOLOGOUS-RECOMBINATION REPAIR
CHK2, and WEE-1 that regulate DNA damage and INHIBITION
repair is a promising therapeutic approach, particularly PARP Inhibition
in molecular subsets such as p53 mutant and PARP inhibitors were developed for BRCA1/2 mutant epi-
ARID1A-mutated gynecologic cancer. thelial ovarian cancer following observation that BRCA1/2
mutations greatly increased the in vitro sensitivity to PARP

e260 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


TARGETING DNA REPAIR DEFICIENCY IN GYNECOLOGIC CANCERS

inhibition, exploiting a concept known as synthetic lethal- studies have demonstrated that deficiency in these genes
ity.3,4 Synthetic lethality arises when a combination of de- and in other HRR-associated proteins, such as ATM, CHEK1,
fects in two or more genes or proteins leads to cell death, CHEK2 and CDK12, also confer sensitivity to DNA damage
whereas a single defect is compatible with cell viability. and DNA repair inhibition.31,34-36 These have been found
BRCA1/2 defective cells are dependent on non-HR DNA in sporadic epithelial ovarian cancer and other cancers in
repair and they are sensitive to any induction in double- which they appear to function to create a BRCA mutation-
strand breaks. PARP inhibition produces stalled replication like phenocopy.37 Understanding other DNA damage mech-
forks, which increases the number of double-strand breaks anisms and potential targets across gynecologic cancers
and leads to genetic chaos and cell death by apoptosis or can further extend the success of DNA repair inhibitors,
senescence.2 The synthetic lethality between BRCA1/2 exemplified by PARP inhibitors.
mutations and PARP inhibition has been confirmed in The Cancer Genome Atlas (TCGA) identified mutations
clinical trials.22-25 Multiple PARP inhibitors, including olaparib now recognized to be related to the HRR pathway in ap-
(AZD2281), rucaparib (CO-338), veliparib (ABT888), and proximately 30% of high-grade serous ovarian cancers.38
niraparib (MK4827), are in clinical development either This included somatic mutations in BRCA1/2 (3%), ATM and
as single agents or in combination therapy for the ATR (2%), the FANC family (5%), and hypermethylation of
management of patients with epithelial ovarian cancer RAD51C (3%), as well as germline mutations in BRCA1 (9%)
(Table 1). or BRCA2 (8%). EMSY amplification (13%), which is proposed
Olaparib, (Lynparza/AZD2281) is the first licensed PARPi to inactive BRCA2, has not been validated in patients yet.
for the treatment of BRCA-mutated epithelial ovarian can- Pennington and colleagues used targeted capture and
cer.26,27 The initial olaparib phase I study provided clinical massively parallel genomic sequencing to examine germline
proof-of-concept of synthetic lethality between BRCA1/2 and somatic loss-of-function mutations in 30 genes, in-
mutant tumors and PARP inhibition. Of the expansion phase cluding 13 HRR genes in 390 epithelial ovarian cancers.36
patients, 40% attained either RECIST partial or complete re- Thirty-one percent of ovarian cancers had a deleterious
sponse, CA-125 responses by Gynecological Cancer Intergroup germline (24%) and/or somatic (9%) mutation in one or more
criteria, or both. Subsequent phase II studies evaluating olaparib of the 13 HRR genes, with similar incidence in serous (31%)
monotherapy in patients with relapsed epithelial ovarian cancer and nonserous ovarian cancers (28%, p = .06). The germline
have shown response rates of 31% to 41% in BRCA1/2-mutation or somatic HRR gene mutations predicted platinum sensi-
carriers and up to 21% in BRCA1/2 wild-type patients.28,29 tivity (p = .0002) and improved OS (p = .0006; Table 2). The
A phase II trial investigating olaparib maintenance therapy majority of germline and somatic HRR gene mutations were
following an initial response to platinum therapy showed a in BRCA1/2, and 26% occurred in other HRR genes. A similar
progression-free survival (PFS) extension from 4.3 months frequency of mutations was observed in patients with non-
with placebo to 11.2 months with olaparib (hazard ratio [HR] serous epithelial ovarian cancer but with a different spectrum
0.18; 95% CI, 0.100.31) in tumors harboring BRCA1/2 mu- of targeted genes. The functionality of these additional
tations. A benefit for olaparib maintenance in patients with mutations has been observed with rucaparib in patients
BRCA wild-type tumors was observed, although the magni- with both germline and somatic RAD51C mutations within
tude was smaller (7.4 vs. 5.5 months; HR 0.54, 95% CI, the ongoing phase II ARIEL2 study (NCT01891344).39 It is
0.340.85).24,30 No overall survival (OS) benefit has been now being recognized in epithelial ovarian cancer and in
observed at the current time of reporting, the reasons for other tumor types that the presence of HRD might be a
which are likely multifactorial. viable strategy for selection for DNA repair inhibitor trials.
For example, 88% (14 of 16) of men with metastatic
castration-resistant prostate cancer in the TO-PARP phase II
Defects in Non-BRCA Homologous-Recombination olaparib study with a somatic mutation in an HRR gene,
Repair Genes That Modulate Genomic Stability and including BRCA1, BRCA2, ATM, the FANC genes, and CHEK2,
May Promote Sensitivity to DNA Repair Inhibitors responded to olaparib compared with only two of 33 (6%) of
Genomic instability is an important therapeutic target in patients who were wild-type.40
gynecologic cancers, not just because of the advent of PARPi
but also because of the key roles of radiation and platinum
therapies in managing them. Platinum analogs induce intra- New Opportunities Leveraging Genomic Instability
and interstrand purine base cross-links (ICL), which form Recent translational data show BRCA1/2-mutated epithelial
covalent bonds and stress DNA repair. Repair of ICLs de- ovarian cancer has a greater immune infiltration.41,42 It has
pends on nucleotide excision repair and, secondarily, upon been suggested that these cancers may have sensitivity to
double-strand break formation.14 The marked sensitivity of immune checkpoint inhibitors targeting the PD-1/PD-L1
epithelial ovarian cancer to platinum agents is thought to pathway. It is hypothesized that PD-1/PD-L1 targeting agents
be related to the high frequency of underlying HRR defects. may preferentially benefit these patients because BRCA1/2-
Germline or homozygous somatic mutations in other mutated and other HRR-deficient tumors have higher numbers
members of the FA family, such as RAD51C, RAD51D, and of neoantigens.41 This hypothesis remains to be tested in clinical
BRIP1, increase susceptibility to ovarian cancer.31-33 In vitro trials.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e261


e262
TABLE 1. Phase II/III Trials of PARP Inhibitors in Ovarian Cancer
PARP Inhibitor NCT Trial Number Phase Study Type Combination Comparator Platinum Status Previous Lines of Treatment Inclusion Criteria
KRISTELEIT, MILLER, AND KOHN

Olaparib (AZD2281) NCT02392676 III Maintenance NA Placebo PS $ 2 platinum sBRCA, sHRR


NCT01844986 III Maintenance NA Placebo PS 1 platinum gBRCA
(SOLO1)
NCT02477644 III First-line Olaparib in combination Placebo NA Treatment nave HGSOC/
(PAOLA-1) treatment with platinum-taxane endometrioid,
and bevacizumab and as stage IIIB-IV
maintenance therapy
NCT02446600 III Combination Olaparib and cediranib Platinum-doublet PS Any number of HGSOC/
NRG GY004 chemotherapy platinum regimes endometrioid
or gBRCA and
any high-grade
histology
NCT02502266 III Combination Olaparib and cediranib Chemotherapy, olaparib PR # 2 in platR setting HGSOC/

2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


NRG GY005 alone or cediranib endometrioid
alone or gBRCA and
any high-grade
histology
ICON9 III First relapse, Olaparib with cediranib as Cediranib and placebo PS 1 platinum HGSOC
maintenance maintenance therapy as maintenance
following platinum-based therapy
chemotherapy with cediranib
Rucaparib (CO338) NCT01482715 II Maintenance NA NA PS $ 3 chemotherapy Any BRCA
NCT01968213 III Maintenance Placebo Placebo PS $ 2 platinum HGSOC or
(ARIEL3) endometrioid
Veliparib (ABT-888) NCT02470585 III First-line Veliparib in combination with Placebo NA Treatment nave HGSOC, stage III-IV
Treatment carboplatin and paclitaxel
and as maintenance therapy
NCT01113957 II Combination Veliparib and temozolamide Liposomal PR $ 1 platinum and # 3 HGSOC
Doxorubicin cytotoxic regimes
Niraparib (MK4827) NCT01847274 III Maintenance NA Placebo PS $ 2 platinum HGSOC gBRCA and
any high-grade
histology
NCT02354586 II Single Agent NA NA PS $ 2 chemotherapy HGSOC
Talazoparib (BMN673) Single-agent and combination phase I studies in multiple tumor types
Abbreviations: PR, platinum resistant ovarian cancer; PS, platinum sensitive ovarian cancer; HGSOC, High-grade serous ovarian cancer.
TARGETING DNA REPAIR DEFICIENCY IN GYNECOLOGIC CANCERS

TABLE 2. Known Deleterious Homologous Recombinant Deficiency Gene Frequencies in Ovarian Cancer
HR-Path- Observed Frequency All Epithelial Observed Frequency High-Grade
way Gene Ovarian Cancer (%) Ovarian Cancer (%) References
RAD51C 0.412.9 1.9 Walsh et al8, Pennington et al36, Minion et al83, Cunningham et al84,
Song et al85
RAD51D 0.351.1 0.95 Pennington et al36, Cancer Genome Atlas Research Network38, Song
et al85
RAD51B 0.06 0.95 Cancer Genome Atlas Research Network38, Song et al85
RAD50 0.21.0 Walsh et al8, Minion et al83
RAD54L 0.5 Kristeleit et al86
ATM 0.80.86 0.321.0 Pennington et al36, Cancer Genome Atlas Research Network38,
Minion et al83
BRIP1 0.94.0 0.321.0 Walsh et al8, Pennington et al36, Cancer Genome Atlas Research
Network38, Ramus et al87
CHEK2 0.45.0 0.321.0 Walsh et al8, Pennington et al36, Cancer Genome Atlas Research
Network38, Minion et al83
FANCA 0.5 Kristeleit et al86
FANCI 0.5 Kristeleit et al86
NBN 0.21.0 0.631.0 Walsh et al8, Pennington et al36, Cancer Genome Atlas Research
Network38, Candido-dos-Reis71, Minion et al83
PALB2 0.22.0 0.63 Walsh et al8, Pennington et al36, Cancer Genome Atlas Research
Network38, Ramus et al87

The Need for Biomarkers: Identifying the Homologous LOH population (PFS 7.1 vs. 3.7 months; HR 0.61), although
Recombinant Deficiency Signature the benefit was not as great as in the BRCA1/2 mutant cohort
The ability to perform rapid whole-genome sequencing (PFS 9.4 months).39 Additional assays to identify structural
makes it feasible to classify cancers according to their un- changes associated with HRD include the large-scale tran-
derlying mutational spectrum. A series of in vitro studies has sitions assay quantifying chromosomal breaks of at least
confirmed the presence of large subchromosomal deletions 10 Mb45 and the telomeric alleic imbalance score,43 both of
and other genomic changes that cause allelic imbalance which correlate with alterations in BRCA1/2 and other HRR
and confer an HRD phenotype.6,43,44 This has yielded se- pathway genes in patients with ovarian cancer. Although each
veral assays or weighted signatures now being examined of these assays offers the exciting prospect of identifying a
as companion diagnostic predictive biomarkers. This HRR- subset of BRCA-like tumors, which respond to HRR-directed
deficient mutational signature or mutational scar in- therapy, the relevance of each assay requires prospective
dicates reliance on error-prone DNA repair pathways.9 validation in clinical trials. As more data emerge from post hoc
Array-based technology using single nucleotide poly- analyses of tumor samples from completed studies, it is likely
morphisms genotyping and comparative genomic hybrid- that HRD mutational signatures for consideration of DNA
ization has demonstrated that the genomes of high-grade repair inhibition therapy will become more established.
serous ovarian cancer harbor common loss of single parental
alleles. Wang and colleagues examined loss of heterozy-
gosity (LOH) and copy number changes in patients with high- DNA REPAIR INHIBITION BEYOND
grade serous ovarian cancer and divided patients into two HOMOLOGOUS-RECOMBINATION REPAIR
clusters of LOH high and LOH low.44 High-levels of LOH were GENES
associated with platinum sensitivity and improved PFS. Nature has recognized the need to maintain genomic
BRCA1/2-mutant tumors fell within the LOH high group; an integrity as demonstrated by at least six majorand
increased sensitivity to platinum was seen in the LOH high interactiveDNA repair pathways (Fig. 1). Dissection of HRR
group even after exclusion of these patients. An LOH-based has shown this interaction and identified new potential
score has been developed that is strongly associated with targets for therapeutic intervention. Likewise, dissection of
functional defects in BRCA1/2 and other genes implicated in developmental processes in patients with endometrial and
the HRR pathway.6 Prospective validation of a genomic scar colon cancers, and now cervical cancers, has led to recog-
LOH assay is ongoing within the ARIEL2 rucaparib PARP nition of other key DNA repair cancer risk genes. Some of
inhibitor phase II trial (NCT01891344) to dichotomize BRCA these risk genes are involved in other DNA repair or biologic
wild-type patients who benefit from rucaparib. Initial results pathways critical to cellular survival.
suggest increased activity for rucaparib within the BRCA-like Early results implicate deficiency in ARID1A, a key com-
population with high genomic LOH compared with the low ponent of the chromatin-remodeling complex, as sensitizing

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e263


KRISTELEIT, MILLER, AND KOHN

tumor cells to PARP inhibition in vitro and in animal No direct therapy exists to target MMR-deficient tumors;
models.46 ARID1A is a suppressor gene. It is recruited by the however, in vitro data suggest a number of potential di-
homologous-recombinant protein ATR, upstream of the cell rections, including inhibition of select DNA polymerases
cycle G2/M regulator, CHK1, to sites of DNA double-strand through an accumulation of oxidative DNA damage. Spe-
breaks where it facilitates efficient processing of double- cifically, MSH2 deficiency is synthetically lethal with in-
strand breaks and sustains DNA damage signaling. Muta- hibition of DNA polymerase b (POLB), the DNA polymerase
tions in ARID1A are common in clear cell and endometrioid that catalyzes nuclear base excision repair. MLH1 deficiency
epithelial ovarian cancer occurring in up to 57% and 30% is synthetically lethal with inhibition of DNA polymerase g
of cases, respectively.47,48 Loss of ARID1A expression is also (POLG), the only polymerase specific to mitochondrial DNA.62
common in patients with endometrioid endometrial cancer Methotrexate has been shown to be lethal to MSH2-deficient
(approximately 40%49), and similarly in patients with cer- cells through the accumulation of nuclear oxidative DNA
vical cancer. These results imply that DNA repair inhibitors damage,63 leading to an ongoing phase II trial in colorectal
must be tested more broadly in patients with gynecologic cancer (NCT00952016). Perhaps more promising is the
cancers. ability to target secondary mutations that arise as a result of
DNA repair pathways have cell cycle specificity.50 Ac- MMR deficiency believed to drive the tumorigenic pheno-
cordingly, many proteins of the cell cycle, especially within type. Secondary mutations in the double-strand break DNA
the G2/M checkpoint, also regulate DNA damage and repair. repair gene MRE11 are commonly associated with MMR-
CHK1/2 and WEE-1 kinases are examples.51-53 Agents tar- deficient colorectal cancer and lead to PARP inhibitor sen-
geting these kinases have minimal single-agent activity in sitivity in vitro.64 The role of PARP inhibition in patients with
the absence of p53, or more likely, p53 and second (so endometrial cancer is ready for assessing in clinical trial with
far undefined) mutations. Efficacy is much greater when accompanying tissue analysis for microsatellite instability,
examined in a defined p53 mutant background in which MRE11, and PTEN status.
the G1/S checkpoint is aberrant and also in combination. The TCGA endometrial cancer group61 identified a second
There is a subset of patients with ovarian cancer with wild- subgroup of endometrial cancers characterized by muta-
type BRCA1/2 function and cyclin E amplification and tions in POLE, a catalytic subunit DNA polymerase epsilon
overexpression.38,54-56 Emerging data indicate that block- involved in nuclear DNA replication and repair. Muta-
ade of the G1/S checkpoint, such as with pertinent CDK tions in POLE resulted in an ultramutated phenotype with a
inhibitors, may unmask an unexpected sensitivity to DNA mutation frequency approximately 10-fold greater than
repair inhibitors such as PARP inhibitors.57 Furthermore, seen in microsatellite instability tumors. The POLE-mutated
pharmacologic augmentation of hypoxia, such as in the use subgroup also had extraordinarily good survival. Very pre-
of cediranib, can reduce expression of many key homologous- liminary data suggest that the microsatellite instability/POLE
recombinant proteins. This may underpin the activity of the phenotype may result in marked increased frequency
cediranib/olaparib combination in women who have wild- in neoantigen production, rendering the tumors more
type BRCA.58 immunogenic and, therefore, vulnerable to immunother-
apy.65 Early preclinical data support the use of antiPD-
L1 therapy in POLE-hypermutated and microsatellite
Targeting Nonhomologous-Recombination Repair instabilitypositive endometrial cancers because they
DNA Repair Pathways have greater expression of PD-1, PD-L1, and tumor in-
Mismatch repair deficiency. Mismatch repair deficiency is a filtrating lymphocytes.66 Several clinical trials currently
single-strand DNA repair mechanism. It maintains genomic underway are evaluating the role of antiPD-1/PD-L1 ther-
integrity by correcting base substitution mismatches and apy in endometrial cancer either as a single agent (e.g.,
small insertion-deletion mismatches generated by errors in NCT02628067) or in combination with carboplatin and
base pairing during DNA replication. Mismatch repair de- paclitaxel (NCT02549209).
ficiency is critical to maintaining genomic stability. Failure
to recognize and repair DNA mismatches results in micro- Base excision repair. Many other potential targets have
satellite instability and a mutator phenotype, with mutation been identified in other DNA repair pathways. The potential
rates 100- to 1,000-fold higher than in MMR proficient therapeutic importance of DNA-PKcs in base excision repair
cells.13 Loss of one of the MMR proteins (MSH2, MSH6, was identified during the demonstration that PARP was not
MSH3, MLH1, and PMS2) is associated with an increased risk the base excision repair rate-limiting step.16 A normal role
of cancers, including endometrial, ovarian, colorectal, and of PARP is to maintain inactive DNA-PKcs, which keeps
gastric cancers.59 Germline mutations in MMR genes give NHEJ quiet. Agents now in clinical development are targeting
rise to Lynch syndrome, which is associated with a 60% and DNA-PKcs. DNA-PKcs has been implicated with adverse
25% lifetime risk of developing endometrial and colon outcome in patients with epithelial ovarian cancer, sug-
cancers, and ovarian cancer, respectively.59,60 Somatic loss gesting they may be a clinically relevant target in gynecologic
of MMR genes can occur either by mutation or methylation, cancer.67 TRC-102 is an experimental agent targeting base
such that MMR deficiency is associated with up to 30% of all excision repair that also can be considered for patients with
endometrial cancers.61 gynecologic cancers.

e264 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


TARGETING DNA REPAIR DEFICIENCY IN GYNECOLOGIC CANCERS

Cervical Cancer and DNA Repair Defects implications of the potential findings or the treatment plan
The role of defective DNA repair in cervical cancer is less well that the results should inform.74 Using a targeted panel of
established. HPV infection and its associated production of HRR genes may allow the identification of HRD cancers,
the oncoviral proteins E6 and E7 causes inactivation and notwithstanding a number of limitations. With the exception
degradation of the p53 and pRB tumor-suppressor genes. of BRCA1/2, each individual HRR gene defect is present at
This results in cell cycle dysfunction and altered DNA repair very low frequency in patients with ovarian cancer, and
capacity.68 Because of defective DNA damage response, the overall numbers of additional HRD patients identified
cervical tumor cells are increasingly dependent on residual by testing this way is small (Table 2).8,74,75 Homologous
repair pathways to cope with certain types of DNA damage. recombinant deficiency arises via heterogenous mecha-
In support of this, a correlation between response to DNA nisms, which include epigenetic changes, gene amplifica-
damaging therapy and activation of DNA repair pathways tions, and chromosomal translocations. Therefore, a suitable
has been noted in clinical series, albeit involving relatively and validated genomic platform is required to capture all
small numbers of patients. Patients treated with chemo- patients with potentially actionable HRR aberrations. A third
radiotherapy were found to have high expression of the concern is that not all genes are equally important in de-
nucleotide excision repair protein ERCC1 associated with termining therapeutic response, and the number of variants
a decreased PFS and worse OS 69 and activation of the of uncertain significance is massive, leaving many patients
FA/BRCA pathway correlated with treatment failure; con- and physicians without guidance.
versely, impaired NHEJ repair was related to increased OS in Addressing whether there is sufficient overall risk to
patients treated primarily with radiotherapy.70 These ob- mandate germline testing for all women across all 11 or
servations suggest that therapeutic exploitation of DNA more genes (panel testing) or to focus on validated bio-
repair pathways may be useful in potentiating chemo- marker(s) is critical. The latter would allow us to address the
radiotherapy, which is the mainstay of treatment for cervical more complicated question of the role of haplo-insufficiency
cancer. Numerous early phase trials incorporating modu- complementation in genomic instability of more than one
lators of DNA repair such as PARP, ATM and ATR inhibitors, of the homologous-recombinant genes or homologous-
and triapine in combination with standard chemotherapy or recombinant plus another DNA repair pathway gene. Fi-
radiotherapy currently are underway in patients with ad- nancial, personal, and family costs make navigating this new
vanced cervical cancer (NCT01281852, NCT02223923, information more complicated.
NCT02595879, NCT02466971).
Applying the New Information
BRCA1 or BRCA2 germline or homozygous somatic muta-
NAVIGATING THE NEW INFORMATION tions are now considered predictive biomarkers for plati-
Picture the cartoon: a patient with the balloon above her
num- and PARP inhibitorsensitivity.36 A woman with a
head full of question marks. The caption reads What do I
BRCA1 or BRCA2 mutation who progresses on a platinum-
do? The most difficult question for providers and patients is
based therapy often will respond subsequently. Data are
how to weigh progress and new agents in the context of
limited to guide application of the platinum-resistant
population data when addressing individual patient de-
moniker to such women. How to apply the growing data
cisions. As our understanding of the meaning and roles of
related to complex DNA repair pathways and new agents
HRD mutation and dysfunction progresses, we complicate
also is becoming more complex. The first question we need
informing patients.
to address urgently in the clinic is how to interpret loss of
We always start with whats simple. Germline deleterious
sensitivity to such agents as the patient moves along the
BRCA mutation in patients with ovarian cancer is the most
treatment line. Acquisition of secondary mutations in BRCA1
common and actionable finding that generally directs sim-
or BRCA2 may result in resumption of BRCA1 or BRCA2
ple decisions as described above. BRCA mutation or geno-
function.76,77 Estimates of frequency of these secondary
mic instability as a result of HRD, in general, allows greater
mutations range from single digits to nearly 40% of cases in
confidence in the use of DNA damaging agents, DNA repair
the small case numbers examined to date. Other potential
inhibitors, cell cycle inhibitors, and novel combinations that
molecular events include loss of 53bp1, a regulator of the
leverage these dysfunctional pathways. We know that BRCA-
poor fidelity NHEJ DNA repair pathway,78,79 and regula-
mutation patients with ovarian cancer are more responsive
tion of phosphoproteins, such as DNA-PKcs,16 and others in
and have better PFS, at least in the first decade after
the DNA repair pathways, such as ATM and ATR.80-82 The
diagnosis.1,71-73 Newer data of small, unpublished numbers
cause of true platinum resistance in these women is still a
suggest that any homozygous loss of BRCA1/2 may result in a
conundrum.
nearly identical phenotype to germline or somatic homo-
zygous loss, an HRD phenotype for which new directions
have been outlined. WHERE TO GO NEXT?
Germline evaluations of BRCA and other recently identi- DNA repair is a very complex series of parallel and interacting
fied HRD genes and broader gene panel testing are being pathways. There are several druggable targets within these
done for many patients, often without considering the pathways. Progress in understanding these pathways and

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e265


KRISTELEIT, MILLER, AND KOHN

TABLE 3. Defining the Decisions?


Event Implication in Cancer Opportunities
Germline Deleterious gBRCA Presumed homozygous deletion, especially breast and DNA damaging agent and PARP inhibitor
ovarian cancers susceptibility
Somatic BRCA Mutation Role of haplo-insufficiency is unclear Sensitivity requires homozygous loss of function
BRCA1 Promoter Methylation Does not appear to confer stable gene downregulation Unclear any benefit
in ovarian cancer
BRCA1/2 Downregulation Ambient micro-environmental events may affect protein Correlative and preclinical studies suggest this may
production or stability yield a sensitive phenotype
Cyclin E Overexpression Cell cycle dysregulation is associated with altered Targeting CDKs alone and in combination with DNA
balance in DNA damage repair repair inhibitors
Unknown and Wild-Type Work still to do Wide-open opportunities for patient care and
learning

identifying areas of commonality, such as the role of CHK1 other mechanisms. Many questions remain unanswered.
in both the G2/M cell cycle checkpoint and downstream of These include what determines the best predictor of re-
ATM and ATR in DNA repair, has led to new therapeutic sponse to PARP inhibition and how best to use them in
approaches. This is an example of the concept that targeting the management of epithelial ovarian cancer (i.e., as single
sites of biochemical convergence may yield more than the agents, in combination with chemotherapy, or as main-
sum of the inhibition of single independent targets. Che- tenance therapy)? Should we rechallenge with PARP in-
motherapy is targeted therapy. Most chemotherapies target hibitors for patients in which PARP-directed therapy has
DNA damage. We now add the therapeutic category of failed?
DNA repair inhibitors. How to exploit this new and growing Several trials in progress will likely help answer these
therapeutic category in the context of the treatment of all questions. It is becoming increasingly apparent that tar-
women with ovarian cancers is a challenge and a great geting defective DNA repair in patients with nonserous ep-
opportunity (Table 3). This growth provides great oppor- ithelial ovarian cancer, endometrial cancer, and cervical
tunities beyond BRCA. cancer also may have therapeutic potential. This could be
achieved either through the identification of non-HRR genes,
CONCLUSION AND FUTURE DIRECTIONS which may modulate HRR pathways, or by targeting alter-
Targeting defective DNA repair represents a viable treat- native DNA repair pathways such as MMR. Options likely will
ment option for patients with gynecologic malignancies. increase in tandem with our understanding as long as we
Defective HRR is a key vulnerability for patients with high- keep asking relevant questions. The overall aim is, as always,
grade serous epithelial ovarian cancer, which is present in up to increase benefit for our patients.
to 50% of patients. The use of PARP inhibitors allows the
exploitation of molecular differences between tumor nor- ACKNOWLEDGMENT
mal tissues. To maximize the benefit from PARP inhibition, it Rebecca Kristeleit, MD, PhD, is supported in part by the
is important to identify those tumors not only characterized UCH/UCL Biomedical Research Centre and UCL Experimental
by BRCA1/2 mutations but also those with HRD as a result of Cancer Medicine Centre.

References
1. Jayson GC, Kohn EC, Kitchener HC, et al. Ovarian cancer. Lancet. 2014; 6. Abkevich V, Timms KM, Hennessy BT, et al. Patterns of genomic loss
384:1376-1388. of heterozygosity predict homologous recombination repair defects in
2. Dietlein F, Reinhardt HC. Molecular pathways: exploiting tumor-specific epithelial ovarian cancer. Br J Cancer. 2012;107:1776-1782.
molecular defects in DNA repair pathways for precision cancer therapy. 7. Konstantinopoulos PA, Spentzos D, Karlan BY, et al. Gene expression
Clin Cancer Res. 2014;20:5882-5887. profile of BRCAness that correlates with responsiveness to chemo-
3. Bryant HE, Schultz N, Thomas HD, et al. Specific killing of BRCA2-deficient therapy and with outcome in patients with epithelial ovarian cancer.
tumours with inhibitors of poly(ADP-ribose) polymerase. Nature. 2005; J Clin Oncol. 2010;28:3555-3561.
434:913-917. 8. Walsh T, Casadei S, Lee MK, et al. Mutations in 12 genes for inherited
4. Farmer H, McCabe N, Lord CJ, et al. Targeting the DNA repair defect in ovarian, fallopian tube, and peritoneal carcinoma identified by massively
BRCA mutant cells as a therapeutic strategy. Nature. 2005;434:917-921. parallel sequencing. Proc Natl Acad Sci USA. 2011;108:18032-18037.
5. Prakash R, Zhang Y, Feng W, et al. Homologous recombination and 9. Watkins JA, Irshad S, Grigoriadis A, et al. Genomic scars as biomarkers of
human health: the roles of BRCA1, BRCA2, and associated proteins. Cold homologous recombination deficiency and drug response in breast and
Spring Harb Perspect Biol. 2015;7:a016600. ovarian cancers. Breast Cancer Res. 2014;16:211.

e266 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


TARGETING DNA REPAIR DEFICIENCY IN GYNECOLOGIC CANCERS

10. Do K, Doroshow JH, Kummar S. Wee1 kinase as a target for cancer a preplanned retrospective analysis of outcomes by BRCA status in a
therapy. Cell Cycle. 2013;12:3159-3164. randomised phase 2 trial. Lancet Oncol. 2014;15:852-861.
11. Kobayashi H, Shigetomi H, Yoshimoto C. Checkpoint kinase 1 inhibitors 31. Loveday C, Turnbull C, Ramsay E, et al; Breast Cancer Susceptibility
as targeted molecular agents for clear cell carcinoma of the ovary. Oncol Collaboration (UK). Germline mutations in RAD51D confer susceptibility
Lett. 2015;10:571-576. to ovarian cancer. Nat Genet. 2011;43:879-882.
12. Krajewska M, Fehrmann RS, Schoonen PM, et al. ATR inhibition pref- 32. Meindl A, Hellebrand H, Wiek C, et al. Germline mutations in breast
erentially targets homologous recombination-deficient tumor cells. and ovarian cancer pedigrees establish RAD51C as a human cancer
Oncogene. 2015;34:3474-3481. susceptibility gene. Nat Genet. 2010;42:410-414.
13. Martin SA, Lord CJ, Ashworth A. Therapeutic targeting of the DNA 33. Rafnar T, Gudbjartsson DF, Sulem P, et al. Mutations in BRIP1 confer
mismatch repair pathway. Clin Cancer Res. 2010;16:5107-5113. high risk of ovarian cancer. Nat Genet. 2011;43:1104-1107.
14. Hoeijmakers JH. Genome maintenance mechanisms for preventing 34. Bajrami I, Frankum JR, Konde A, et al. Genome-wide profiling of genetic
cancer. Nature. 2001;411:366-374. synthetic lethality identifies CDK12 as a novel determinant of PARP1/2
15. Bouwman P, Jonkers J. The effects of deregulated DNA damage sig- inhibitor sensitivity. Cancer Res. 2014;74:287-297.
naling on cancer chemotherapy response and resistance. Nat Rev 35. McCabe N, Turner NC, Lord CJ, et al. Deficiency in the repair of DNA
Cancer. 2012;12:587-598. damage by homologous recombination and sensitivity to poly(ADP-
16. Patel AG, Sarkaria JN, Kaufmann SH. Nonhomologous end joining drives ribose) polymerase inhibition. Cancer Res. 2006;66:8109-8115.
poly(ADP-ribose) polymerase (PARP) inhibitor lethality in homol- 36. Pennington KP, Walsh T, Harrell MI, et al. Germline and somatic mu-
ogous recombination-deficient cells. Proc Natl Acad Sci USA. 2011;108: tations in homologous recombination genes predict platinum response
3406-3411. and survival in ovarian, fallopian tube, and peritoneal carcinomas. Clin
17. Murai J, Huang SY, Das BB, et al. Trapping of PARP1 and PARP2 by clinical Cancer Res. 2014;20:764-775.
PARP inhibitors. Cancer Res. 2012;72:5588-5599. 37. Lord CJ, Ashworth A. BRCAness revisited. Nat Rev Cancer. 2016;16:
18. Tutt A, Ashworth A. The relationship between the roles of BRCA genes 110-120.
in DNA repair and cancer predisposition. Trends Mol Med. 2002;8: 38. Cancer Genome Atlas Research Network. Integrated genomic analyses
571-576. of ovarian carcinoma. Nature. 2011;474:609-615.
19. Gudmundsdottir K, Ashworth A. The roles of BRCA1 and BRCA2 and 39. McNeish IA, Oza AM, Coleman RL, et al. Results of ARIEL2: a phase 2 trial
associated proteins in the maintenance of genomic stability. Oncogene. to prospectively identify ovarian cancer patients likely to respond to
2006;25:5864-5874. rucaparib using tumor genetic analysis. J Clin Oncol. 2015;33s (suppl;
20. Cass I, Baldwin RL, Varkey T, et al. Improved survival in women with abstr 5508).
BRCA-associated ovarian carcinoma. Cancer. 2003;97:2187-2195. 40. Mateo J, Carreira S, Sandhu S, et al. DNA-repair defects and olaparib in
21. Tan DSP, Rothermundt C, Thomas K, et al. BRCAness syndrome in metastatic prostate cancer. N Engl J Med. 2015;373:1697-1708.
ovarian cancer: a case-control study describing the clinical features and 41. Birkbak NJ, Kochupurakkal B, Izarzugaza JM, et al. Tumor mutation
outcome of patients with epithelial ovarian cancer associated with burden forecasts outcome in ovarian cancer with BRCA1 or BRCA2
BRCA1 and BRCA2 mutations. J Clin Oncol. 2008;26:5530-5536. mutations. PLoS One. 2013;8:e80023.
22. Fong PC, Boss DS, Yap TA, et al. Inhibition of poly(ADP-ribose) poly- 42. Strickland K, Howitt BE, Rodig SJ, et al. Tumor infiltrating and peritu-
merase in tumors from BRCA mutation carriers. N Engl J Med. 2009;361: moral T cells and expression of PD-L1 in BRCA1/2-mutated high grade
123-134. serous ovarian cancers. J Clin Oncol. 2015;33s (suppl: abstr 5512).
23. Fong PC, Yap TA, Boss DS, et al. Poly(ADP)-ribose polymerase inhibition: 43. Birkbak NJ, Wang ZC, Kim JY, et al. Telomeric allelic imbalance indicates
frequent durable responses in BRCA carrier ovarian cancer correlating defective DNA repair and sensitivity to DNA-damaging agents. Cancer
with platinum-free interval. J Clin Oncol. 2010;28:2512-2519. Discov. 2012;2:366-375.
24. Ledermann J, Harter P, Gourley C, et al. Olaparib maintenance therapy 44. Wang ZC, Birkbak NJ, Culhane AC, et al; Australian Ovarian Cancer
in platinum-sensitive relapsed ovarian cancer. N Engl J Med. 2012;366: Study Group. Profiles of genomic instability in high-grade serous
1382-1392. ovarian cancer predict treatment outcome. Clin Cancer Res. 2012;18:
25. Lord CJ, Tutt AN, Ashworth A. Synthetic lethality and cancer therapy: 5806-5815.
lessons learned from the development of PARP inhibitors. Annu Rev 45. Popova T, Manie E, Rieunier G, et al. Ploidy and large-scale genomic
Med. 2015;66:455-470. instability consistently identify basal-like breast carcinomas with BRCA1/2
26. European Medicines Agency. 2015, EMA Approval of Olaparib. http://www. inactivation. Cancer Res. 2012;72:5454-5462.
ema.europa.eu/docs/en_GB/document_library/ EPAR_-_Summary_ 46. Shen J, Peng Y, Wei L, et al. ARID1A deficiency impairs the DNA damage
for_the_public/human/003726/WC500180153.pdf. Accessed February 1, checkpoint and sensitizes cells to PARP inhibitors. Cancer Discov. 2015;
2016. 5:752-767.
27. Kim G, Ison G, McKee AE, et al. FDA approval summary: olaparib 47. Jones S, Wang TL, Shih IeM, et al. Frequent mutations of chromatin
monotherapy in patients with deleterious germline BRCA-mutated remodeling gene ARID1A in ovarian clear cell carcinoma. Science. 2010;
advanced ovarian cancer treated with three or more lines of chemo- 330:228-231.
therapy. Clin Cancer Res. 2015;21:4257-4261. 48. Wiegand KC, Shah SP, Al-Agha OM, et al. ARID1A mutations in
28. Gelmon KA, Tischkowitz M, Mackay H, et al. Olaparib in patients with endometriosis-associated ovarian carcinomas. N Engl J Med. 2010;363:
recurrent high-grade serous or poorly differentiated ovarian carcinoma 1532-1543.
or triple-negative breast cancer: a phase 2, multicentre, open-label, 49. Wiegand KC, Lee AF, Al-Agha OM, et al. Loss of BAF250a (ARID1A) is
non-randomised study. Lancet Oncol. 2011;12:852-861. frequent in high-grade endometrial carcinomas. J Pathol. 2011;224:
29. Kaufman B, Shapira-Frommer R, Schmutzler RK, et al. Olaparib mono- 328-333.
therapy in patients with advanced cancer and a germline BRCA1/2 50. Morgan MA, Parsels LA, Maybaum J, et al. Improving the efficacy of
mutation. J Clin Oncol. 2015;33:244-250. chemoradiation with targeted agents. Cancer Discov. 2014;4:280-291.
30. Ledermann J, Harter P, Gourley C, et al. Olaparib maintenance therapy 51. Niida H, Nakanishi M. DNA damage checkpoints in mammals. Muta-
in patients with platinum-sensitive relapsed serous ovarian cancer: genesis. 2006;21:3-9.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e267


KRISTELEIT, MILLER, AND KOHN

52. Stathis A, Oza A. Targeting Wee1-like protein kinase to treat cancer. cervical cancer with intrinsic resistance and therapy failure. BMC
Drug News Perspect. 2010;23:425-429. Cancer. 2014;14:246.
53. Zhang Y, Hunter T. Roles of Chk1 in cell biology and cancer therapy. Int J 70. Wilson CR, Davidson SE, Margison GP, et al. Expression of Ku70 cor-
Cancer. 2014;134:1013-1023. relates with survival in carcinoma of the cervix. Br J Cancer. 2000;83:
54. Etemadmoghadam D, Au-Yeung G, Wall M, et al. Resistance to CDK2 1702-1706.
inhibitors is associated with selection of polyploid cells in CCNE1- 71. Candido-dos-Reis FJ, Song H, Goode EL, et al; for EMBRACE; kConFab
amplified ovarian cancer. Clin Cancer Res. 2013;19:5960-5971. Investigators; Australian Ovarian Cancer Study Group. Germline
55. Etemadmoghadam D, Weir BA, Au-Yeung G, et al; Australian Ovarian mutation in BRCA1 or BRCA2 and ten-year survival for women di-
Cancer Study Group. Synthetic lethality between CCNE1 amplification agnosed with epithelial ovarian cancer. Clin Cancer Res. 2015;21:
and loss of BRCA1. Proc Natl Acad Sci USA. 2013;110:19489-19494. 652-657.
56. Karst AM, Jones PM, Vena N, et al. Cyclin E1 deregulation occurs early 72. Jain RK. Normalization of tumor vasculature: an emerging concept in
in secretory cell transformation to promote formation of fallopian antiangiogenic therapy. Science. 2005;307:58-62.
tube-derived high-grade serous ovarian cancers. Cancer Res. 2014;74: 73. Kotsopoulos J, Rosen B, Fan I, et al. Ten-year survival after epithelial
1141-1152. ovarian cancer is not associated with BRCA mutation status. Gynecol
57. Alagpulinsa DA, Ayyadevara S, Yaccoby S, et al. A cyclin-dependent Oncol. 2016;140:42-47.
kinase inhibitor, dinaciclib, impairs homologous recombination and 74. Norquist BM, Pennington KP, Agnew KJ, et al. Characteristics of women
sensitizes multiple myeloma cells to PARP inhibition. Mol Cancer Ther. with ovarian carcinoma who have BRCA1 and BRCA2 mutations not
2016;15:241-250. identified by clinical testing. Gynecol Oncol. 2013;128:483-487.
58. Liu JF, Barry WT, Birrer M, et al. Combination cediranib and olaparib 75. Norquist BM, Harrell MI, Brady MF, et al. Inherited mutations in women
versus olaparib alone for women with recurrent platinum-sensitive with ovarian carcinoma. JAMA Oncol. Epub 2015 Dec 30.
ovarian cancer: a randomised phase 2 study. Lancet Oncol. 2014;15: 76. Edwards SL, Brough R, Lord CJ, et al. Resistance to therapy caused by
1207-1214. intragenic deletion in BRCA2. Nature. 2008;451:1111-1115.
59. Guillotin D, Martin SA. Exploiting DNA mismatch repair deficiency as a 77. Norquist B, Wurz KA, Pennil CC, et al. Secondary somatic mutations
therapeutic strategy. Exp Cell Res. 2014;329:110-115. restoring BRCA1/2 predict chemotherapy resistance in hereditary
60. Aarnio M, Mecklin JP, Aaltonen LA, et al. Life-time risk of different ovarian carcinomas. J Clin Oncol. 2011;29:3008-3015.
cancers in hereditary non-polyposis colorectal cancer (HNPCC) syn- 78. Bunting SF, Callen E, Wong N, et al. 53BP1 inhibits homologous re-
drome. Int J Cancer. 1995;64:430-433. combination in Brca1-deficient cells by blocking resection of DNA breaks.
61. Kandoth C, Schultz N, Cherniack AD, et al; Cancer Genome Atlas Re- Cell. 2010;141:243-254.
search Network. Integrated genomic characterization of endometrial 79. Pennington KP, Wickramanayake A, Norquist BM, et al. 53BP1 ex-
carcinoma. Nature. 2013;497:67-73. pression in sporadic and inherited ovarian carcinoma: relationship
62. Martin SA, McCabe N, Mullarkey M, et al. DNA polymerases as potential to genetic status and clinical outcomes. Gynecol Oncol. 2013;128:
therapeutic targets for cancers deficient in the DNA mismatch repair 493-499.
proteins MSH2 or MLH1. Cancer Cell. 2010;17:235-248. 80. Guleria A, Chandna S. ATM kinase: much more than a DNA damage
63. Martin SA, McCarthy A, Barber LJ, et al. Methotrexate induces oxidative responsive protein. DNA Repair (Amst). Epub 2015 Dec 29.
DNA damage and is selectively lethal to tumour cells with defects in the 81. Khanna A. DNA damage in cancer therapeutics: a boon or a curse?
DNA mismatch repair gene MSH2. EMBO Mol Med. 2009;1:323-337. Cancer Res. 2015;75:2133-2138.
64. Vilar E, Bartnik CM, Stenzel SL, et al. MRE11 deficiency increases 82. Roos WP, Thomas AD, Kaina B. DNA damage and the balance between
sensitivity to poly(ADP-ribose) polymerase inhibition in microsatellite survival and death in cancer biology. Nat Rev Cancer. 2016;16:20-33.
unstable colorectal cancers. Cancer Res. 2011;71:2632-2642. 83. Minion LE, Dolinsky JS, Chase DM, et al. Hereditary predisposition to
65. Alexandrov LB, Nik-Zainal S, Wedge DC, et al; Australian Pancreatic ovarian cancer, looking beyond BRCA1/BRCA2. Gynecol Oncol. 2015;
Cancer Genome Initiative; ICGC Breast Cancer Consortium; ICGC MMML- 137:86-92.
Seq Consortium; ICGC PedBrain. Signatures of mutational processes in 84. Cunningham JM, Cicek MS, Larson NB, et al. Clinical characteristics of
human cancer. Nature. 2013;500:415-421. ovarian cancer classified by BRCA1, BRCA2, and RAD51C status. Sci Rep.
66. Howitt BE, Sholl LM, Ritterhouse L, et al. Association of POLE-mutated 2014;4:4026.
and MSI endometrial cancers with an elevated number of tumor- 85. Song H, Dicks E, Ramus SJ, et al. Contribution of germline mutations in
infiltrating and peritumoral lymphocytes and higher expression of the RAD51B, RAD51C, and RAD51D genes to ovarian cancer in the
PD-L1. J Clin Oncol. 2015;33s (suppl: abstr 5511). population. J Clin Oncol. 2015;33:2901-2907.
67. Abdel-Fatah TM, Arora A, Moseley P, et al. ATM, ATR and DNA-PKcs 86. Kristeleit R, Swisher E, Oza A, et al. Final results of ARIEL2 (Part 1):
expressions correlate to adverse clinical outcomes in epithelial ovarian a phase 2 trial to prospectively identify ovarian cancer (OC) responders
cancers. BBA Clin. 2014;2:10-17. to rucaparib using tumor genetic analysis. Eur J Cancer. 2015;51s (suppl;
68. Duensing S, Munger K. The human papillomavirus type 16 E6 and S531).
E7 oncoproteins independently induce numerical and structural chro- 87. Ramus SJ, Song H, Dicks E, et al; AOCS Study Group; Ovarian Cancer
mosome instability. Cancer Res. 2002;62:7075-7082. Association Consortium. Germline mutations in the BRIP1, BARD1,
69. Balacescu O, Balacescu L, Tudoran O, et al. Gene expression profiling PALB2, and NBN genes in women with ovarian cancer. J Natl Cancer Inst.
reveals activation of the FA/BRCA pathway in advanced squamous 2015;107:11.

e268 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


GYNECOLOGIC CANCER

Intraperitoneal Chemotherapy for


Ovarian Cancer: Trials and
Tribulations

CHAIR
Helen J. Mackay, MD
Princess Margaret Cancer Centre
Toronto, ON

SPEAKERS
Charlie Gourley, PhD, FRCPE
University of Edinburgh Cancer Research UK Centre
Edinburgh, United Kingdom

Joan L. Walker, MD
The University of Oklahoma Health Sciences Center
Oklahoma City, OK
INTRAPERITONEAL CHEMOTHERAPY IN THE TREATMENT OF OVARIAN CANCER

Update on Intraperitoneal Chemotherapy for the Treatment


of Epithelial Ovarian Cancer
Charlie Gourley, BSC, MBChB, PhD, FRCP, Joan L. Walker, MD, and Helen J. Mackay, BSc, MBCh, MD, MRCP

OVERVIEW

Surgical treatment and chemotherapy administration in women with epithelial ovarian cancer is more controversial today
than at any point in the last 3 decades. The use of chemotherapy administered intraperitoneally has been particularly
contentious. Three large randomized phase III studies, multiple meta-analyses, and now real-world data have demonstrated
substantial outcome benefit for the use of chemotherapy administered intraperitoneally versus intravenously for first-line
postoperative treatment of optimally debulked advanced ovarian cancer. Unfortunately, for each of these randomized
studies, there was scope to either criticize the design or otherwise refute adoption of this route of administration. As a result,
the uptake has been variable in North America, although in Europe it has been practically nonexistent. Reasons for this
include unquestionable additional toxicity, more inconvenience, and extra cost. However, 10-year follow up of these studies
demonstrates unprecedented survival in the intraperitoneal arm (median survival 110 months in patients with completely
debulked stage III), raising the possibility that by combining maximal debulking surgery with postoperative intraperitoneal
chemotherapy it may be possible to bring about a step change in the outcomes for these patients. In this review, we discuss
the rationale for administering chemotherapy intraperitoneally, the merits of the main randomized clinical trials, the
evidence regarding optimal regimes, issues of toxicity, port considerations, and reasons for lack of universal adoption. We
also explore potential clinical and biologic factors that may be useful for patient selection in the future.

W e have witnessed improvements in epithelial ovarian


cancer survival over the last 3 decades without seeing
significant improvements in disease-specific mortality rates
that they represent pathologic markers of what are essen-
tially discrete disease entities. These differ in terms of their
tissue of origin, stage of presentation, driver molecular
(Fig. 1). Epithelial ovarian cancer remains the leading cause mutations, sensitivity to chemotherapy, and prognosis
of death from gynecologic malignancy in North America with (Fig. 2). Ultimately, it is very likely that these histologic
the majority of women presenting with stage III or IV dis- subtypes will require different treatment strategies.6
ease.1,2 Advances in genetic testing, counseling, and pre- In clinical practice, only a few risk factors remain that can
vention with risk-reducing salpingo-oophorectomy (and be modified based on the decisions of patients and their
salpingectomy) have the potential to produce further physicians. The surgical decision making and chemotherapy
modest decreases in ovarian cancer mortality in the future.3 administration choices, particularly for women with stage III
However, we currently do not have a reliable population or IV epithelial ovarian cancer, can potentially affect survival.
screening test for epithelial ovarian cancer, and, given the Informed selection of the best treatment (including clinical
often nonspecific symptoms with which epithelial ovarian trial options) for any individual patient is most likely to be
cancer presents, it is likely the majority of patients will achieved with enthusiastically committed multidisciplinary
continue to present with late-stage disease. Therefore, teams working in high-volume institutions.7,8 Discussions
optimizing treatment is critical if we are to improve outcome. around chemotherapy administered intraperitoneally for
Retrospective analyses suggest that overall survival (OS) is individual patients require this type of environment to allow
associated with younger age, good performance status, the patient to make an informed choice about care.
lower stage of disease, and lower comorbidity scores.4,5 This article summarizes the history and role of chemo-
Although histologic subtype and tumor grade previously therapy administered intraperitoneally in epithelial ovarian
were regarded as simply prognostic, it has now become clear cancer, focuses on the practical choices that patients and

From the Edinburgh Cancer Research Centre, Medical Research Council, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom; Stephenson
Cancer Center, University of Oklahoma, Health Sciences Center, Oklahoma City, OK; Faculty of Medicine, University of Toronto, Sunnybrook Odette Cancer Centre, Toronto, Canada.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Helen J. Mackay, BSc, MBCh, MD, MRCP, University of Toronto, Sunnybrook Odette Cancer Centre, 2075 Bayview Ave., Toronto, ON M4N 3M5, Canada; email:
helen.mackay@sunnybrook.ca.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 143


GOURLEY, WALKER, AND MACKAY

FIGURE 1. Increase in Overall Survival Over Time for minimizes systemic toxicity, therefore, it is an attractive
Women Diagnosed With Ovarian Cancer therapeutic approach.9 Advantages of intraperitoneal ad-
ministration include high intraperitoneal concentration of
the drug, as well as a longer half-life of the drug in the
peritoneal cavity, compared with that observed with ad-
ministration intravenously alone. For cisplatin, historically,
the most extensively studied agent administered by the
intraperitoneal route in epithelial ovarian cancer translates
into a 10- to 20-fold greater exposure over that which is
achievable with the intravenously administrated route.10-12
Furthermore, preclinical studies suggested that cisplatin is
capable of penetrating small volume tumors (13 mm).
Hence, the hypothesis arose that the maximum benefit from
administration of the drug intraperitoneally was likely to be
demonstrated in patients with microscopic or low-volume
macroscopic disease.13 However, our understanding of why
the intraperitoneal chemotherapy route is more effective
may be oversimplified, and it has been challenged. Mea-
surement of drug in peritoneal fluid probably does not
represent actual tumor drug penetration in patients.14 There
clinicians face (with an emphasis on emerging data), and may be added barriers in tumor implants, notably disordered
stresses the importance of building skilled multidisciplinary capillary architecture, fibrosis, and adhesions. Furthermore,
teams for treating women undergoing intraperitoneal dose intensification of platinum administered intravenously
treatment. Finally, we explore how emerging data on epi- has failed to show a benefit in multiple randomized stud-
thelial ovarian cancer biology may be able to guide the ies.15,16 However, these dose intensification studies did not
future of intraperitoneal therapy in this disease. select neither patients with high-grade serous ovarian
cancer nor the subgroups with disease that we know to be
RATIONALE FOR INTRAPERITONEAL most sensitive to platinum (those exhibiting homologous
CHEMOTHERAPY recombination deficiency, discussed below). As such, the
The peritoneal cavity is the principle site of spread and impact in the dose-dense arms of these studies (which were
recurrence in women with epithelial ovarian cancer. Ad- twice the density of the control arm at most) in any patients
ministration of chemotherapy intraperitoneally is a means of with disease sensitive to this approach would be diluted out
increasing the dose intensity delivered to the tumor and by the majority of patients whose tumor biology would make
them unlikely to benefit from this approach. Although the
intraperitoneal studies were similarly unselected for
immunohistologic or molecular subtype, the local dose in-
KEY POINTS tensification in the intraperitoneal arm is potentially an
order of magnitude higher than in the intravenously ad-
Patients with epithelial ovarian cancer should be treated ministrated arm. Under these circumstances, it may be
by high-volume multidisciplinary teams.
possible that the signal would be evident even without
Randomized phase III trial, meta-analysis, and real-
world data support the benefit of chemotherapy
preselection on the basis of histology or biology.
administered intraperitoneally in the treatment of Other factors such as drug recirculation following peri-
select groups of women with epithelial ovarian cancer toneal absorption may play a role in efficacy. Despite the fact
following up-front optimal cytoreductive surgery. that chemotherapy administered intraperitoneally has been
The optimal intraperitoneal/intravenous chemotherapy studied for decades, we are still not fully aware of the key
regimen has yet to be defined, and emerging data from biologic factors that determine its success.15,16 Moving
the randomized studies GOG 252, OV21/PETROC, and forward, a greater understanding of how tumor biology and
JGOG iPocc will help clarify whether cisplatin the immune and micro-environments are affected by
administered intraperitoneally can be replaced by treatment administered intraperitoneally may help us un-
carboplatin administered intraperitoneally. derstand how this treatment can best be deployed and
Discussion of chemotherapy administered
combined with the newer generation of targeted agents.
intraperitoneally to treat women diagnosed with
ovarian cancer is essential.
Understanding the biology underlying the success of
chemotherapy administered intraperitoneally is a INTRAPERITONEAL CHEMOTHERAPY TRIALS IN
priority; initial data suggest exploration of tumors WOMEN WITH EPITHELIAL OVARIAN CANCER
deficient in DNA repair are of particular interest. Intraperitoneal chemotherapy is not a recent concept in the
treatment of epithelial ovarian cancer; it was originally used

144 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


INTRAPERITONEAL CHEMOTHERAPY IN THE TREATMENT OF OVARIAN CANCER

FIGURE 2. Schema for GOG 252 select patients following primary cytoreductive surgery
(Table 1).17-23 GOG 172, a randomized phase III study of
cisplatin administered intraperitoneally combined with
both delivery of paclitaxel intraperitoneally and intravenously,
published in 2006, demonstrated a 16-month improvement
in median OS over intravenous administration of the same
drugs alone.18 This prompted the National Cancer Institute
(NCI) to issue a rare clinical announcement regarding the
clinical utility of cisplatin-based chemotherapy adminis-
tered intraperitoneally in the treatment of patients with
small volume (, 1 cm), advanced-stage (stage III) epithelial
ovarian cancer following an attempt at maximal cytore-
ductive surgery. On average, intraperitoneal/intravenous
chemotherapy was associated with a 21.6% decrease in
risk of death (hazard ratio (HR) 0.78; 95% CI, 0.690.89,
the original clinical announcement can be viewed at ctep.
cancer.gov).
An update published in 2015 with a median follow-up
of 10.7 years showed that women who underwent
in the 1950s to control ascites. Interest in chemotherapy intraperitoneal/intravenous chemotherapy in GOG 172
administered intraperitoneally as a strategy for reducing continued to derive benefit with a median survival of
the risk of disease recurrence and prolonging survival 61.8 months (95% CI, 55.569.5 months) compared with
emerged approximately 30 years ago. This resulted in a 51.4 months (95% CI, 4658.2 months) for chemotherapy
number of randomized phase III studies that demonstrated administered intravenously alone.24 The recent Cochrane
an improvement in survival for the combination of delivery Review, restricted to newly diagnosed patients receiving
of chemotherapy intraperitoneally and intravenously over treatment after primary cytoreductive surgery, accepted
chemotherapy administrated intravenously alone for data from eight randomized studies on 2,026 women and

TABLE 1. Summary of Randomized Clinical Trials of Intraperitoneal Chemotherapy for Up-Front Primary
Cytoreductive Surgery

No. of
Study/Reference Control Regimen Experimental Regimen Eligible Patients Patients
Kirmani et al20 Cisplatin 100 mg/m2 IV cyclophosphamide Cisplatin 200 mg/m2 IP; etoposide 350 mg/m2 IP Stage IIC-IV 62
600 mg/m2
Every 3 weeks x 6 Every 4 weeks x 6
SWOG 8501/ Cisplatin 100 mg/m2 IV; cyclophosphamide Cisplatin 100 mg/m2 IP; cyclophosphamide Stage III, 546
GOG 10417 600 mg/m2 IV 600 mg/m2 IV # 2 cm residual
Every 3 weeks x 6 Every 3 weeks x 6
Polyzos et al22 Carboplatin 350 mg/m2 IV; cyclophosphamide Carboplatin 350 mg/m2 IP; cyclophosphamide Stage III 90
600 mg/m2 IV 600 mg/ m2 IV
Every 3 weeks x 6 Every 3 weeks x 6
GONO19 Cisplatin 50 mg/m2 IV; cyclophosphamide Cisplatin 50 mg/m2 IP; cyclophosphamide Stage II-IV, 113
600 mg/m2 IV; epidoxorubicin 60 mg/m2 IV 600 mg/ m2 IV; epidoxorubicin 60 mg/m2 IV , 2 cm residual
Every 4 weeks x 6 Every 4 weeks x 6
GOG 114/ Cisplatin 75 mg/m2 IV; paclitaxel Carboplatin (AUC 9) IV every 28 days x 2; Stage III, 462
SWOG 922721 135 mg/m2 (24-hr) IV cisplatin 100 mg/m2 IP; paclitaxel # 1 cm residual
135 mg/m2 (24-hr) IV
Every 3 weeks x 6 Every 3 weeks x 6
Yen et al23 Cisplatin 50 mg/m2 IV; cyclophosphamide Cisplatin 100 mg/m2 IP; cyclophosphamide Stage III, 118
50 mg/m2 IV; epidoroxorubin/ 500 mg/m2 IV; epidoxorubicin/ # 1 cm residual
doxorubicin 50 mg/m2 IV doxorubicin 50 mg/m2 IV
Every 3 weeks x 6 Every 3 weeks x 6
GOG 17218 Cisplatin 75 mg/m2 IV; paclitaxel Paclitaxel 135 mg/m2 (24-hr) IV; cisplatin Stage III, 415
135 mg/m2 (24-hr) IV 100 mg/m2 IP; paclitaxel 60 mg/m2 IP on day 8 # 1 cm residual
Every 3 weeks x 6 Every 3 weeks x 6
Abbreviations: IP, intraperitoneally; IV, intravenously.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 145


GOURLEY, WALKER, AND MACKAY

concluded that women experienced increased survival if intensities between the arms (GOG 114 and GOG 172)
they received intraperitoneal/intravenous chemotherapy were excluded from the analysis of a Cochrane systematic
(HR 0.81; 95% CI, 0.700.9) and that intraperitoneal/ review in 2011, the survival benefit remained in favor of
intravenous chemotherapy also prolonged the disease- the intraperitoneal route of administration.25 In the real-
free interval (five studies, 1,311 women; HR 0.78; 95% CI, world analysis by Wright et al, 43% of patients received
0.70.86),25 thus potentially affecting quality of life going modified intraperitoneal/intravenous regimens over time
forward. (i.e., regimens differing from trial-specified protocols). De-
Wright et al recently reported data on the real-world spite these modifications, women receiving intraperitoneal/
uptake of intraperitoneal/intravenous chemotherapy in a intravenous chemotherapy continued to derive benefit over
prospective cohort of 823 women with stage III optimally those who only received treatment intravenously (3-year OS
cytoreduced epithelial ovarian cancer treated in six National 81% vs. 71%; HR 0.68; 95% CI, 0.470.99).8
Comprehensive Cancer Network (NCCN) institutions.8 De- Undoubtedly, delivery of intraperitoneal/intravenous
spite the trial-based evidence and although adoption of chemotherapy requires increased resources in terms of
intraperitoneal/intravenous chemotherapy increased be- both space and time to deliver compared with therapy
tween 2007 and 2008, it plateaued with fewer than 50% of administrated intravenously. Because of placement of
eligible patients receiving intraperitoneal/intravenous catheters and regional delivery of drug, intraperitoneal/
treatment. They also observed marked variation in up- intravenous chemotherapy is potentially associated with
take between institutions from 4% to 67%, suggesting greater toxicity, including catheter-related complications,
underutilization of this effective treatment approach in gastrointestinal toxicity, pain, and infection. These have
this subgroup of women. been reported across trials and supported in meta-ana-
lyses.25,29 However, real-world reports suggest many of
these potential issues can be overcome with time and de-
WHY HAS INTRAPERITONEAL/INTRAVENOUS velopment of expertise.30,31 Furthermore, randomized data
CHEMOTHERAPY NOT BEEN UNIVERSALLY and the NCI alert support the use of cisplatin administered
ADOPTED? intraperitoneally.17-21,23 Cisplatin is known to be more toxic
Despite a proven survival benefit, clearly intraperitoneal/ than the standard of care carboplatin, which is administered
intravenous chemotherapy has not been universally adop- intravenously for epithelial ovarian cancer.32 To date, we do
ted for the treatment of epithelial ovarian cancer. The not have randomized trial data to determine whether car-
reasons behind this are numerous. The publication of each boplatin administered intraperitoneally is equivalent to
positive intraperitoneal/intravenous randomized study has cisplatin administered intraperitoneally in terms of its im-
been met with considerable debate over the interpretation pact on survival. However, preclinical and some clinical data
of the trial data.26 Arguments around the validity of the suggest that it might be equivalent in efficacy and less
conclusions drawn from GOG 172 have included statistical toxic than cisplatin administered intraperitoneally.33 Results
analysis queries (intention-to-treat analysis, number of from GOG 252 (NCT00951496) and OV21/PETROC (NCT00993655)
patients lost to follow-up), and questions over second-line are awaited. These trials include direct comparison of
treatment and scheduling effects.26 The most relevant regimens, including cisplatin and carboplatin administered
criticism of the pivotal intraperitoneal studies is the in- intraperitoneally.
equality of dose intensity between the treatment arms in As yet, we have not arrived at the optimal intraperitoneal/
both GOG11421 and GOG17218 (Table 1). Given the fact that intravenous chemotherapy regimen, which balances efficacy
paclitaxel administered intravenously weekly has been with toxicity and quality of life. Finally, effective and safe
shown to be superior to paclitaxel administered in- delivery of intraperitoneal/intravenous chemotherapy re-
travenously tri-weekly in one large randomized phase III quires the multidisciplinary expertise of a skilled team, which
study,27 it could be argued that the administration of simply may not be available in smaller-volume centers.
an additional dose of paclitaxel on day 8 in the Armstrong Therefore, consideration for referral and management in
study was solely responsible for the benefit demonstrated. high-volume centers is appropriate. Even with all the con-
However, a second randomized phase III study of weekly cerns that exist, it is clear that women who are appropriate
versus triweekly paclitaxel administered intravenously did for intraperitoneal/intravenous chemotherapy should be at
not show superiority.28 In addition, the Alberts (GOG104) least offered this as an option, and clinicians should be
study was a clean comparison of the same doses of cisplatin considering how best to make it available to them.31
administered intravenously or intraperitoneally, which
demonstrated substantial progression-free and OS advan-
tages,17 but it was overlooked in many areas of the world SURGICAL CONSIDERATIONS FOR
because paclitaxel came to prominence and the comparator INTRAPERITONEAL CHEMOTHERAPY
arm in GOG104 was regarded as outdated. It does, however, The goal of ovarian cancer surgery whenever it is performed
serve as a useful proof of principle regarding the advantages is no gross residual disease or R0, as defined by Chi and
that can be attributed purely to the route of administration. Bristow.7,34 This has been shown to result in improved
In addition, when the two studies with unequal dose progression-free survival (PFS) and OS. Intraoperative

146 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


INTRAPERITONEAL CHEMOTHERAPY IN THE TREATMENT OF OVARIAN CANCER

treatment decisions that have been demonstrated to in- receiving neoadjuvant treatment. Neoadjuvant chemo-
fluence patient survival and maximize surgical effort must therapy is usually platinum-based and administered in-
include a willingness to perform diaphragm resection, travenously; there is no role for intraperitoneal/intravenous
splenectomy, bowel resections, and thorough peritoneal therapy in the preoperative patient. A recent study by Rosen
and retroperitoneal resections of tumor. This requires ex- et al40 showed that the long-term survival for patients
perience and potentially a team of surgeons.35 Although an undergoing primary cytoreduction was far superior to those
R0 resection improves OS, it remains somewhat unclear receiving neoadjuvant chemotherapy and delayed primary
whether microscopic versus visible disease has an impact on surgery (9% vs. 41%, p , .0001). Although selection bias may
intraperitoneal chemotherapy effectiveness, as the allow- account for some of this difference, the percentage of long-
able residual volume at the end of surgery differed in term survivors is strikingly low in neoadjuvant chemother-
the randomized trials (, 2 or , 1 cm). Many have made apy studies.41 Patients undergoing optimal cytoreductive
the assumption that patients with microscopic disease surgery following administration of neoadjuvant chemo-
would derive the greatest benefit from intraperitoneal/ therapy were not included in the previous intraperitoneal/
intravenous chemotherapy. However, in a subgroup analysis intravenous randomized trials. Theoretically, they may
of the GOG172 patient population, the 64% of women in derive a similar level of benefit to women undergoing
GOG172 who had macroscopic (gross) residual disease less up-front cytoreductive surgery from intraperitoneal/
than or equal to 1 cm (which was the upper limit allowed by intravenous chemotherapy delivery. The combination of
study eligibility) had a significant improvement in OS (HR intraperitoneal/intravenous chemotherapy following neo-
0.75; 95% CI, 0.620.92).24 Further exploration of the effect adjuvant chemotherapy and optimal cytoreductive surgery
on larger-volume residual disease should emerge from GOG- is being studied in the Gynecologic Cancer Intergroup study
0252 and the Japanese iPocc trial (NCT01506856), both of OV21/PETROC. Women who had initial (clinical/imaging)
which enrolled a proportion of patients with larger-volume stage IIB-IV (intravenously based on the presence of pleu-
residual disease. ral effusion alone) epithelial ovarian cancer and who had
Landrum looked into the effects of lymphadenectomy and received three or four cycles of platinum-based neoadjuvant
nodal metastasis on the benefit of administering chemo- chemotherapy before definitive optimal cytoreductive
therapy intraperitoneally using data from both GOG 114 and surgery (# 1 cm residual disease) were eligible for this trial.
GOG 172. In these studies, despite undergoing cytoreductive Women were enrolled either intra- or postoperatively. This
surgery to less than 1 cm, only 59% of women had lymph study has now closed and the primary analysis is expected
nodes sampled or excised. Of the 254 women who had lymph shortly.
node evaluation, intraperitoneal benefit in terms of PFS and
OS was independent of nodal status. This suggests that
chemotherapy administered intraperitoneally may be equally INTRAPERITONEAL PORT PLACEMENT
effective for patients with both intraperitoneal and retro- Essential to the delivery of chemotherapy administered in-
peritoneal disease. Interestingly, the patients without lym- traperitoneally is the placement of an intraperitoneal port.
phadenectomy did worse than patients with metastatic Consideration of port placement should occur at the time of
tumor removed from their lymph nodes, although the de- surgery when the port can be placed under direct visuali-
cision not to perform the lymphadenectomy may have been zation by the surgeon. This is the most time-efficient ap-
secondary to some poor prognostic factor perceived by the proach and prevents delays in the initiation of chemotherapy
surgeon. An important long-term quality of life finding by postoperatively. Patients should be treated as soon as they
Landrum was a decrease in recurrence in the abdominal cavity have resumed a normal diet and bowel function and are
after intraperitoneal chemotherapy. Thus intraperitoneal/ ambulatory at home, which should occur within 21 days
intravenous treatment may spare patients from suffering of the primary surgery. Patients electing to receive in-
from ascites and an inability to eat when they recur.4 traperitoneal chemotherapy who do not already have
A key and controversial area around surgical decision peritoneal catheters can have devices implanted by inter-
making in epithelial ovarian cancer is the choice of neo- ventional radiologists or surgeons familiar with laparoscopic
adjuvant chemotherapy followed by a definitive cytore- techniques using the right upper quadrant entry techniques.
ductive surgical attempt versus primary cytoreductive Minilaparotomy in the right lower quadrant is also generally
surgery. Initially, neoadjuvant chemotherapy was consid- successful if resection of the terminal ileum or right colon did
ered only for those women who were medically unfit for not occur. Careful review of the cytoreduction operative
aggressive surgery or for women with a high tumor burden report can improve outcomes with intraperitoneal port
(especially those with stage IV disease).36,37 In recent years, placement to avoid complications.42,43
the use of neoadjuvant chemotherapy has gained in pop- The preferred location for port placement is the right
ularity. This followed the publication of two studies: the lower costal grill on the midclavicular line, below the location
European Organization for Research and Treatment of where the breast or the bra may be uncomfortable in
Cancer Gynecologic Cancer Group (EORTC GCG) 5597138 and a standing position. This site allows for posterior rigid
the CHORUS study,39 which demonstrated equivalence in support that facilitates access. Ports also are installed over
outcome with some reduction in morbidity for patients the left side or right lower quadrant for convenience.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 147


GOURLEY, WALKER, AND MACKAY

Failures of intraperitoneal catheters can be corrected in- three bowel obstructions (7%) were observed.50 Further
frequently. Infected catheters should be removed and not safety data on the combination of bevacizumab and
replaced. Blocked catheters can be replaced if the patient intraperitoneal/intravenous therapy will be available from
has free intraperitoneal space remaining between bowel GOG 252. GOG 262 is a study that compared carboplatin/
loops and has not had peritonitis as a complication of having paclitaxel administered intravenously every 3 weeks with
undergone surgery or having received chemotherapy. Ac- carboplatin administered intravenously on day 1 with
cess problems attributable to a rotated port can be easily weekly dosing of paclitaxel intravenously. Bevacizumab
corrected. Most patients, however, resume their chemo- was allowed as an (nonrandomized) option in both arms of
therapy with treatment administered intravenously alone the trial. This study demonstrated that dose-dense pacli-
when catheter complications occur. taxel improve PFS over the dosing of paclitaxel every
3 weeks, but this was only true in women not receiving
bevacizumab. The addition of bevacizumab appeared to
INTRAPERITONEAL CHEMOTHERAPY: WHICH eliminate the beneficial effects of dose-dense pacli-
REGIMEN? taxel.47 In GOG 252, bevacizumab is included on cycles
The 2013 update of the NCCN Guidelines suggested an in- 222, and the study was designed with the assumption
traperitoneal regimen based on the experimental arm of GOG that there would be no interaction between the various
172: cisplatin administered intraperitoneally 75 to 100 mg/m2 chemotherapy arms and bevacizumab, which would hide
and a 3-hour infusion of paclitaxel intravenously on day 1 with the effects of the individual chemotherapy regimens. The
60 mg/mg2 delivered intraperitoneally on day 8. The reduced surprising results of GOG 262 raise concern that the ad-
dose of cisplatin administered intraperitoneally (75 mg/m2) dition of bevacizumab to all patients on GOG 252 may
was included because of concerns over the toxicity of 100 mg/ have obscured the effects of the individual chemotherapy
m2 with patients who did not complete the planned six cycles regimens alone. Insufficient data exist at this time to rec-
of intraperitoneal/intravenous therapy postsurgery. This ommend bevacizumab in combination with intraperitoneal/
dosing is also reflected in the experimental arms of both GOG intravenous chemotherapy as a standard-of-care option.
252 and OV21/PETROC.44 Results for GOG 252 are expected to be reported at the
Given the toxicity associated with cisplatin and the Society of Gynecologic Oncology meeting in March 2016.
emerging nonrandomized data on carboplatin administered Ultimately, the factors that patients and families should be
intraperitoneally, some centers have adopted intraperitoneal empowered to consider when making decisions about ad-
carboplatin-based regimens.45,46 Randomized data to better juvant treatment will include convenience, potential toxic-
inform the selection of an intraperitoneal/intravenous regi- ities, and quality of life in addition to the data on efficacy.
men will be available soon from GOG 252 and OV21/PETROC.
Given that GOG 172 demonstrated an OS benefit in an
intention-to-treat analysis when the median number of PATIENT SELECTION FOR INTRAPERITONEAL
cycles delivered was three (although the studies to date have CHEMOTHERAPY
looked at six cycles of chemotherapy), a question remains The last consideration is whether there are individual
as to whether six is the optimal number of intraperitoneal patient findings that should be factored into decisions
treatments.18 regarding whether chemotherapy should be administered
When discussing chemotherapy options for patients intraperitoneally/intravenously or just intravenously to pa-
whose disease is optimally cytoreduced, clinicians face the tients. It is now clear that at the immunohistochemic level,
challenge of discussing three basic choices: (1) carboplatin epithelial ovarian cancer consists of at least five different
and paclitaxel administered intravenously every 3 weeks diseases.51-53 Although previous randomized studies of
(GOG 158, GOG 182), (2) carboplatin administered in- intraperitoneal/intravenous chemotherapy versus in-
travenously every 3 weeks with paclitaxel administered travenous chemotherapy did not select or stratify on the basis
intravenously weekly (JGOG-3016, GOG 262), or (3) an of histologic subtype, it is now apparent that subtypes such as
intraperitoneal/intravenous regimen.32,42,47,48 The subset of low-grade serous, low-grade mucinous, and clear cell are less
women with stage III optimally cytoreduced disease in ICON sensitive to chemotherapy than high-grade serous or high-
7 did not derive benefit from the addition of bevacizumab to grade endometrioid ovarian cancer. The low likelihood of a
carboplatin and paclitaxel administered intravenously every considerable chemotherapy dose-response relationship in
3 weeks.49 Data from OV21/PETROC, GOG 252, and JGOG these less-sensitive subtypes makes it difficult to justify
iPocc (NCT01506856) studies are eagerly awaited because subjecting these patients to the additional toxicity of in-
they will provide information on the comparison of a dose- traperitoneal chemotherapy. Rather, the main subgroup of
dense intravenous regimen with intraperitoneal/intravenous patients who should be considered for intraperitoneal ad-
chemotherapy. This will address the question of dose density ministration of chemotherapy are those with high-grade
that remains from the GOG 172 data. The role of bevacizumab serous ovarian cancer (patients with high-grade endome-
in combination with intraperitoneal/intravenous chemo- trioid cancer are an under-researched subgroup), but what
therapy is being explored in GOG 252. The tolerability of the little evidence there is suggests they could be considered
regimen was established in a previous phase II trial, although similar to high-grade serous from a biologic perspective).

148 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


INTRAPERITONEAL CHEMOTHERAPY IN THE TREATMENT OF OVARIAN CANCER

Currently, given the strong evidence of benefit from GOG urgency using material from randomized studies of in-
11421 and GOG17218 studies, there is insufficient evidence to traperitoneal chemotherapy to demonstrate whether par-
support any further patient selection based on biologic ticular molecular subgroups have more to gain from
subtype in patients with optimally cytoreduced disease who intraperitoneal treatment. It is already clear that some
have good performance status. However, the finding that molecular subgroups have a high incidence of primary
patients from the GOG 172 study with low-tumor BRCA1 platinum resistance (Fig. 3; homologous recombination
expression (as assessed by immunohistochemistry) appeared proficient),55,56 and these patients may not benefit from
to benefit more from intraperitoneal/intravenous che- intraperitoneal therapy; instead, perhaps they should be
motherapy than intravenous chemotherapy (median sur- considered for dose-dense paclitaxel and carboplatin ad-
vival 84 compared with 48 months, p = .0002) than those ministered intravenously, or trials of other novel therapies
with normal BRCA1 expression (median survival 58 months in combination with their first-line chemotherapy, to op-
for intraperitoneal chemotherapy compared with 50 months timize survival and avoid unnecessary toxicity.
for intravenous chemotherapy) suggests that there are Although cure is a word that most ovarian cancer on-
molecular subgroups that may benefit more from the in- cologists try to avoid, this must be our aim. The survival that
traperitoneal route of delivery.54 The main molecular has been demonstrated in patients with completely
characteristics underlying high-grade serous ovarian can- cytoreduced disease and who received intraperitoneal
cer have recently been uncovered (Fig. 3).51,55 Given the chemotherapy in the GOG172 study is a sea change in terms
strong preclinical and clinical data supporting a high level of of what we expect in this disease. It reinforces the argument
platinum sensitivity in BRCA1- and BRCA2-deficient ovarian that in fit patients, maximal cytoreductive surgery followed
cancer cells, it is not surprising that patients with low by intraperitoneal chemotherapy is the treatment likely to
BRCA1 protein expression may derive particular benefit produce the best possible outcome. However, the morbidity
from administration of chemotherapy intraperitoneally. induced by both these treatments makes it even more im-
The question is whether this benefit applies only to tumors perative that we are able to define whether it is only patients
that carry germline or somatic BRCA1 mutations, or whether with a particular biology (e.g., BRCA mutations or homolo-
those patients with epigenetic BRCA1 inactivation, BRCA2 gous recombination deficiency) that benefit. For these pa-
mutations, PTEN loss, EMSY amplification, or mutations in tients, full-on surgical/intraperitoneal chemotherapy with
other homologous recombination deficiency genes also de- or without PARP maintenance inhibitors (depending on the
rive benefit. Although BRCA1 immunohistochemistry analysis outcome of currently running first-line PARP inhibitor
is notoriously inaccurate, the technology now exists to per- studies) could be adopted. For the patients with different
form this other sequencing and copy number characterization biology, alternative tailored approaches could be sought.
on archival formalin-fixed, paraffin-embedded material. This
retrospective analysis should be performed as a matter of
CONCLUSION
Large randomized phase III studies have been ubiquitously
positive, producing some of the best survival data ever seen
FIGURE 3. Molecular Subgroups of High-Grade Serous
in the treatment of ovarian cancer. However, all of these
Epithelial Ovarian Cancer
studies have had issues, either with the intravenous che-
motherapy (control) arm being perceived as no longer con-
temporary or the intraperitoneal (test) arm having higher
dose intensity than the control arm and, therefore, not
being a trial solely of route of administration. These apparent
shortcomings combined with undoubted toxicity of the ap-
proach have led to inconsistent uptake in North America and
Australia and negligible uptake in Europe. This seems a shame
given that the benefits in terms of outcome appear so
marked. Indeed, the benefits are well in excess of those
demonstrated in the first-line bevacizumab studies that led to
licensing and reimbursement of this agent across Europe.
Further work is required to improve the toxicity from
chemotherapy administered intraperitoneally and to iden-
tify the best regimen and determine the extent to which the
benefits witnessed also can be produced by dose-dense
approaches or the use of targeted agents. These latter
two questions may be answered to some extent by the
GOG252 study. Perhaps the most important question that
requires to be answered is exactly which molecular sub-
Abbreviations: HRD, homologous recombination deficient. groups of ovarian cancer patients benefit most from

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 149


GOURLEY, WALKER, AND MACKAY

intraperitoneal treatment. If this can be established, these intraperitoneal chemotherapy approach. The question is,
patients may benefit greatly from an aggressive surgical and will this be enough to change practice?

References
1. National Cancer Institute. SEER Stat Fact Sheets: Ovary Cancer. http:// cancer: a randomized trial of the Gruppo Oncologico Nord-Ovest.
seer.cancer.gov/statfacts/html/ovary.html. Accessed February 5, 2016. Gynecol Oncol. 2000;76:157-162.
2. Cannistra SA. Cancer of the ovary. N Engl J Med. 2004;351:2519-2529. 20. Kirmani S, Braly PS, McClay EF, et al. A comparison of intravenous versus
3. McCann GA, Eisenhauer EL. Hereditary cancer syndromes with high risk intraperitoneal chemotherapy for the initial treatment of ovarian
of endometrial and ovarian cancer: surgical options for personalized cancer. Gynecol Oncol. 1994;54:338-344.
care. J Surg Oncol. 2015;111:118-124. 21. Markman M, Bundy BN, Alberts DS, et al. Phase III trial of standard-dose
4. Landrum LM, Java J, Mathews CA, et al. Prognostic factors for stage III intravenous cisplatin plus paclitaxel versus moderately high-dose car-
epithelial ovarian cancer treated with intraperitoneal chemotherapy: boplatin followed by intravenous paclitaxel and intraperitoneal cisplatin
a Gynecologic Oncology Group study. Gynecol Oncol. 2013;130:12-18. in small-volume stage III ovarian carcinoma: an intergroup study of the
5. Winter WE III, Maxwell GL, Tian C, et al; Gynecologic Oncology Group Gynecologic Oncology Group, Southwestern Oncology Group, and
Study. Prognostic factors for stage III epithelial ovarian cancer: a Gy- Eastern Cooperative Oncology Group. J Clin Oncol. 2001;19:1001-1007.
necologic Oncology Group Study. J Clin Oncol. 2007;25:3621-3627. 22. Polyzos A, Tsavaris N, Kosmas C, et al. A comparative study of in-
6. Banerjee S, Kaye SB. New strategies in the treatment of ovarian cancer: traperitoneal carboplatin versus intravenous carboplatin with in-
current clinical perspectives and future potential. Clin Cancer Res. 2013; travenous cyclophosphamide in both arms as initial chemotherapy for
19:961-968. stage III ovarian cancer. Oncology. 1999;56:291-296.
7. Bristow RE, Tomacruz RS, Armstrong DK, et al. Survival effect of maximal 23. Yen MS, Juang CM, Lai CR, et al. Intraperitoneal cisplatin-based che-
cytoreductive surgery for advanced ovarian carcinoma during the motherapy vs. intravenous cisplatin-based chemotherapy for stage III
platinum era: a meta-analysis. J Clin Oncol. 2002;20:1248-1259. optimally cytoreduced epithelial ovarian cancer. Int J Gynaecol Obstet.
8. Wright AA, Cronin A, Milne DE, et al. Use and effectiveness of in- 2001;72:55-60.
traperitoneal chemotherapy for treatment of ovarian cancer. J Clin 24. Tewari D, Java JJ, Salani R, et al. Long-term survival advantage and
Oncol. 2015;33:2841-2847. prognostic factors associated with intraperitoneal chemotherapy
9. Dedrick RL, Myers CE, Bungay PM, et al. Pharmacokinetic rationale for treatment in advanced ovarian cancer: a Gynecologic Oncology Group
peritoneal drug administration in the treatment of ovarian cancer. study. J Clin Oncol. 2015;33:1460-1466.
Cancer Treat Rep. 1978;62:1-11. 25. Jaaback K, Johnson N, Lawrie TA. Intraperitoneal chemotherapy for the
10. Fujiwara K, Armstrong D, Morgan M, et al. Principles and practice of initial management of primary epithelial ovarian cancer. Cochrane
intraperitoneal chemotherapy for ovarian cancer. Int J Gynecol Cancer. Database Syst Rev. 2016;1:CD005340.
2007;17:1-20. 26. Gore M, du Bois A, Vergote I. Intraperitoneal chemotherapy in ovarian
11. Howell SB. Pharmacologic principles of intraperitoneal chemotherapy cancer remains experimental. J Clin Oncol. 2006;24:4528-4530.
for the treatment of ovarian cancer. Int J Gynecol Cancer. 2008;18(suppl 27. Katsumata N, Yasuda M, Takahashi F, et al; Japanese Gynecologic
1):20-25. Oncology Group. Dose-dense paclitaxel once a week in combination
12. Markman M. Intraperitoneal chemotherapy. Semin Oncol. 1991;18: with carboplatin every 3 weeks for advanced ovarian cancer: a phase 3,
248-254. open-label, randomised controlled trial. Lancet. 2009;374:1331-1338.
13. Markman M. Intraperitoneal drug delivery of antineoplastics. Drugs. 28. Pignata S, Scambia G, Katsaros D, et al; Multicentre Italian Trials in
2001;61:1057-1065. Ovarian cancer (MITO-7); Groupe dInvestigateurs Nationaux pour
14. Royer B, Jullien V, Guardiola E, et al. Population pharmacokinetics and lEtude des Cancers Ovariens et du sein (GINECO); Mario Negri Gy-
dosing recommendations for cisplatin during intraperitoneal per- necologic Oncology (MaNGO); European Network of Gynaecological
operative administration: development of a limited sampling strategy Oncological Trial Groups (ENGOT-OV-10); Gynecologic Cancer In-
for toxicity risk assessment. Clin Pharmacokinet. 2009;48:169-180. terGroup (GCIG) Investigators. Carboplatin plus paclitaxel once a week
15. Bookman MA, Brady MF. Intraperitoneal chemotherapy: long-term versus every 3 weeks in patients with advanced ovarian cancer (MITO-
outcomes revive a long-running debate. J Clin Oncol. 2015;33: 7): a randomised, multicentre, open-label, phase 3 trial. Lancet Oncol.
1424-1426. 2014;15:396-405.
16. Kaye SB, Paul J, Cassidy J, et al; Scottish Gynecology Cancer Trials Group. 29. Elit L, Oliver TK, Covens A, et al. Intraperitoneal chemotherapy in the
Mature results of a randomized trial of two doses of cisplatin for the first-line treatment of women with stage III epithelial ovarian cancer:
treatment of ovarian cancer. J Clin Oncol. 1996;14:2113-2119. a systematic review with metaanalyses. Cancer. 2007;109:692-702.
17. Alberts DS, Liu PY, Hannigan EV, et al. Intraperitoneal cisplatin plus 30. Chin SN, Pinto V, Rosen B, et al. Evaluation of an intraperitoneal
intravenous cyclophosphamide versus intravenous cisplatin plus in- chemotherapy program implemented at the Princess Margaret Hospital
travenous cyclophosphamide for stage III ovarian cancer. N Engl J Med. for patients with epithelial ovarian carcinoma. Gynecol Oncol. 2009;
1996;335:1950-1955. 112:450-454.
18. Armstrong DK, Bundy B, Wenzel L, et al; Gynecologic Oncology Group. 31. Markman M. Chemotherapy: Limited use of the intraperitoneal route
Intraperitoneal cisplatin and paclitaxel in ovarian cancer. N Engl J Med. for ovarian cancer-why? Nat Rev Clin Oncol. 2015;12:628-630.
2006;354:34-43. 32. Ozols RF, Bundy BN, Greer BE, et al; Gynecologic Oncology Group. Phase
19. Gadducci A, Carnino F, Chiara S, et al. Intraperitoneal versus intravenous III trial of carboplatin and paclitaxel compared with cisplatin and
cisplatin in combination with intravenous cyclophosphamide and paclitaxel in patients with optimally resected stage III ovarian cancer:
epidoxorubicin in optimally cytoreduced advanced epithelial ovarian a Gynecologic Oncology Group study. J Clin Oncol. 2003;21:3194-3200.

150 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


INTRAPERITONEAL CHEMOTHERAPY IN THE TREATMENT OF OVARIAN CANCER

33. Jandial DD, Messer K, Farshchi-Heydari S, et al. Tumor platinum con- for advanced-stage epithelial ovarian cancer. Gynecol Oncol. 2016;140:
centration following intraperitoneal administration of cisplatin versus 36-41.
carboplatin in an ovarian cancer model. Gynecol Oncol. 2009;115: 46. Fujiwara K, Markman M, Morgan M, et al. Intraperitoneal carboplatin-
362-366. based chemotherapy for epithelial ovarian cancer. Gynecol Oncol. 2005;
34. Chi DS, Eisenhauer EL, Zivanovic O, et al. Improved progression-free and 97:10-15.
overall survival in advanced ovarian cancer as a result of a change in 47. Chan J, Brady M, Penson R, et al. Phase III trial of every-3-weeks
surgical paradigm. Gynecol Oncol. 2009;114:26-31. paclitaxel vs. dose dense weekly paclitaxel with carboplatin +/2
35. Aletti GD, Dowdy SC, Gostout BS, et al. Aggressive surgical effort and bevacizumab In epithelial ovarian, peritoneal, fallopian tube cancer:
improved survival in advanced-stage ovarian cancer. Obstet Gynecol. GOG 262 (NCT01167712). Int J Gynecol Cancer. 2013;23:8(suppl 1, abstr
2006;107:77-85. 8).
36. Ansquer Y, Leblanc E, Clough K, et al. Neoadjuvant chemotherapy for 48. Katsumata N, Yasuda M, Isonishi S, et al; Japanese Gynecologic On-
unresectable ovarian carcinoma: a French multicenter study. Cancer. cology Group. Long-term results of dose-dense paclitaxel and carbo-
2001;91:2329-2334. platin versus conventional paclitaxel and carboplatin for treatment of
37. Schwartz PE, Rutherford TJ, Chambers JT, et al. Neoadjuvant chemo- advanced epithelial ovarian, fallopian tube, or primary peritoneal
therapy for advanced ovarian cancer: long-term survival. Gynecol Oncol. cancer (JGOG 3016): a randomised, controlled, open-label trial. Lancet
1999;72:93-99. Oncol. 2013;14:1020-1026.
38. Vergote I, Trope CG, Amant F, et al; European Organization for Research 49. Oza AM, Cook AD, Pfisterer J, et al; ICON7 Trial Investigators. Standard
and Treatment of Cancer-Gynaecological Cancer Group; NCIC Clinical chemotherapy with or without bevacizumab for women with newly
Trials Group. Neoadjuvant chemotherapy or primary surgery in stage diagnosed ovarian cancer (ICON7): overall survival results of a phase 3
IIIC or IV ovarian cancer. N Engl J Med. 2010;363:943-953. randomised trial. Lancet Oncol. 2015;16:928-936.
39. Kehoe S, Hook J, Nankivell M, et al. Primary chemotherapy versus 50. Konner JA, Grabon DM, Gerst SR, et al. Phase II study of intraperitoneal
primary surgery for newly diagnosed advanced ovarian cancer (CHO- paclitaxel plus cisplatin and intravenous paclitaxel plus bevacizumab as
RUS): an open-label, randomised, controlled, non-inferiority trial. adjuvant treatment of optimal stage II/III epithelial ovarian cancer. J Clin
Lancet. 2015;386:249-257. Oncol. 2011;29:4662-4668.
40. Rosen B, Laframboise S, Ferguson S, et al. The impacts of neoadjuvant 51. Bowtell DD, Bohm S, Ahmed AA, et al. Rethinking ovarian cancer II:
chemotherapy and of debulking surgery on survival from advanced reducing mortality from high-grade serous ovarian cancer. Nat Rev
ovarian cancer. Gynecol Oncol. 2014;134:462-467. Cancer. 2015;15:668-679.
41. Colombo PE, Labaki M, Fabbro M, et al. Impact of neoadjuvant che- 52. Kobel M, Kalloger SE, Boyd N, et al. Ovarian carcinoma subtypes are
motherapy cycles prior to interval surgery in patients with advanced different diseases: implications for biomarker studies. PLoS Med. 2008;
epithelial ovarian cancer. Gynecol Oncol. 2014;135:223-230. 5:e232.
42. Bookman MA, Brady MF, McGuire WP, et al. Evaluation of new 53. Vaughan S, Coward JI, Bast RC Jr, et al. Rethinking ovarian cancer:
platinum-based treatment regimens in advanced-stage ovarian cancer: recommendations for improving outcomes. Nat Rev Cancer. 2011;11:
a phase III trial of the Gynecologic Cancer Intergroup. J Clin Oncol. 2009; 719-725.
27:1419-1425. 54. Lesnock JL, Darcy KM, Tian C, et al. BRCA1 expression and improved
43. Walker JL, Armstrong DK, Huang HQ, et al. Intraperitoneal catheter survival in ovarian cancer patients treated with intraperitoneal cisplatin
outcomes in a phase III trial of intravenous versus intraperitoneal and paclitaxel: a Gynecologic Oncology Group Study. Br J Cancer. 2013;
chemotherapy in optimal stage III ovarian and primary peritoneal 108:1231-1237.
cancer: a Gynecologic Oncology Group Study. Gynecol Oncol. 2006;100: 55. Patch AM, Christie EL, Etemadmoghadam D, et al; Australian Ovarian
27-32. Cancer Study Group. Whole-genome characterization of chemo-
44. Morgan RJ Jr, Alvarez RD, Armstrong DK, et al; National Comprehensive resistant ovarian cancer. Nature. 2015;521:489-494.
Cancer Networks. Ovarian cancer, version 2.2013. J Natl Compr Canc 56. Etemadmoghadam D, George J, Cowin PA, et al; Australian Ovarian
Netw. 2013;11:1199-1209. Cancer Study Group. Amplicon-dependent CCNE1 expression is critical
45. Bouchard-Fortier G, Rosen B, Vyarvelska I, et al. A comparison of the for clonogenic survival after cisplatin treatment and is correlated with
toxicity and tolerability of two intraperitoneal chemotherapy regimens 20q11 gain in ovarian cancer. PLoS One. 2010;5:e15498.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 151


GYNECOLOGIC CANCER

Neoadjuvant Chemotherapy:
Location, Location, Location

CHAIR
Alexandra Leary, MD, PhD
Gustave Roussy Cancer Center
Villejuif, France

SPEAKERS
Dennis Chi, MD
Memorial Sloan Kettering Cancer Center
New York, NY

Sean Kehoe, MD
University of Birmingham
Birmingham, United Kingdom
KEEPING THE PEACE AND BALANCING THE BIOLOGY

Primary Surgery or Neoadjuvant Chemotherapy in Advanced


Ovarian Cancer: The Debate Continues
Alexandra Leary, MD, PhD, Renee Cowan, MD, MPH, Dennis Chi, MD, Sean Kehoe, MD, DCH, FRCOG, and
Matthew Nankivell, MSc

OVERVIEW

Primary debulking surgery (PDS) followed by platinum-based chemotherapy has been the cornerstone of treatment for
advanced ovarian cancer for decades. Primary debulking surgery has been repeatedly identified as one of the key factors in
improving survival in patients with advanced ovarian cancer, especially when minimal or no residual disease is left behind.
Achieving these results sometimes requires extensive abdominal and pelvic surgical procedures and consultation with other
surgical teams. Some clinicians who propose a primary chemotherapy approach reported an increased likelihood of leaving
no macroscopic disease after surgery and improved patient-reported outcomes and quality-of-life (QOL) measures. Given
the ongoing debate regarding the relative benefit of PDS versus neoadjuvant chemotherapy (NACT), tumor biology may aid
in patient selection for each approach. Neoadjuvant chemotherapy offers the opportunity for in vivo chemosensitivity
testing. Studies are needed to determine the best way to evaluate the impact of NACT in each individual patient with
advanced ovarian cancer. Indeed, the biggest utility of NACT may be in research, where this approach provides the op-
portunity for the investigation of predictive markers, mechanisms of resistance, and a forum to test novel therapies.

D espite the fact that most patients with ovarian cancer


have stage III or IV disease, advances in surgical and
medical therapies over the last 4 decades have significantly
There are some in the field of gynecologic oncology,
however, who propose that inherent tumor biology is more
important than surgical resection and question the value of
improved the 5-year and overall survival (OS) for women extensive cytoreductive surgeries as the first step of the
diagnosed with ovarian cancer.1,2 What used to be a death advanced ovarian cancer treatment plan. Within the last
sentence can now be considered a chronic disease in many decade, this issue has been widely debated. In 2008, Vergote
cases.3 However, ovarian cancer is still the leading gyne- presented the data of the first randomized control phase III
cologic malignancy cause of cancer-related deaths in high- trial comparing PDS versus NACT followed by interval
income countries.4 Furthermore, there is much work to be debulking surgery (IDS). The trial enrolled more than
done in efforts to improve not only OS and progression-free 600 women with bulky stage III or IV advanced ovarian
survival (PFS), but also QOL. cancer and was conducted by the European Organisation
According to the National Comprehensive Cancer Net- for Research and Treatment of CancerGynecologic Cancer
work, the current primary treatment of advanced epithelial Group (EORTC-GCG) and the National Cancer Institute of
ovarian cancer is optimal cytoreductive surgery followed by Canada (NCIC) Clinical Trials Group. They found no major
six to eight cycles of dual-agent platinum- and taxane- difference in the PFS and OS between the two treatment
based chemotherapy.5 Cytoreductive surgery (also called arms and concluded that NACT with IDS was not inferior
debulking) was initially proposed by Meigs in 1934.6 Forty and possibly safer than the current standard of care,
years later, Griffiths showed that survival depended on the PDS.11,12 The recently published trial on primary chemo-
maximum diameter of residual disease left after cytore- therapy versus primary surgery for newly diagnosed ad-
ductive surgery.7 Primary debulking surgery has since been vanced ovarian cancer (CHORUS) supported the EORTC/NCIC
repeatedly demonstrated as one of the key factors in im- results. In the CHORUS trial, more than 550 women with
proving survival and the cornerstone of ovarian cancer advanced ovarian cancer were randomly selected to receive
treatment by many studies.8-10 primary chemotherapy versus primary surgery, with similar

From the Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY; School of Cancer Sciences, University of Birmingham, Birmingham, United Kingdom; St. Peters
College, National Cancer Intelligence Network, Public Health England, National Health Service, Birmingham, United Kingdom; School of Cancer Sciences, University of Birmingham,
Birmingham, United Kingdom; Medical Research Council Clinical Trials Unit, University College London, London, United Kingdom; Gustave Roussy Cancer Centre, Translational Research
Laboratory, Gustave Roussy Cancer Centre, Villejuif, France.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Alexandra Leary, MD, PhD, Gustave Roussy Cancer Center, 114 Rue Edouard Vaillant, Villejuif, France; email: leary.alexandra@gmail.com.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 153


LEARY ET AL

conclusions that NACT/IDS yielded comparable survival surgical procedures as needed during their PDS. This ap-
results and decreased surgical morbidity when compared proach also increases the rate of optimal cytoreduction,
with PDS.13 without significantly increasing postoperative morbidity.
In 2009, Chi et al demonstrated a change in the surgical
THE ARGUMENT FOR PRIMARY DEBULKING paradigm at Memorial Sloan Kettering Cancer Center
SURGERY (MSKCC) that incorporated splenectomy, distal pancrea-
It Is Possible tectomy, cholecystectomy, liver wedge resection, dis-
It is well accepted that patients who undergo suboptimal section of the porta hepatis, and diaphragm resection/
tumor cytoreductive surgeries are at risk for surgical mor- peritonectomy when necessary to achieve optimal PDS.
bidity without deriving the survival benefit of the debulking This paradigm change led to an improvement in optimal
procedure.14,15 However, when a patient is treated with the primary cytoreduction from 46% to 80%. The rate of CGR
appropriate surgery by an experienced surgeon or team of also more than doubled.19 Multiple U.S. and international
surgeons at an experienced hospital, optimal and even com- studies have supported these findings, demonstrating not
plete gross resection (CGR) at the time of PDS is achievable only the feasibility, but also the safety, of this approach in
in a vast majority of cases. Reported optimal cytoreduction expert centers, with the morbidity, mortality, and time to
rates in the literature range from 15% to greater than start of chemotherapy not being statistically different
85%.10,16 Many experts agree that this variation is not from that of pelvic limited surgery.20-24 The complexity of
caused by a difference in geography, case presentation, the surgery alone is not a predictor of PFS or OS. When
and/or case selection, but is highly dependent on the surgical cancer in these patients is optimally debulked to less than
training, expertise, commitment, and resources of the pri- 1 cm of residual disease, they are afforded the same
mary operating surgeon, team, and institution. Moreover, survival rates as patients who required only standard
greater than 40% of patients with advanced ovarian cancer pelvic surgery.25-27
have bulky upper abdominal disease that is difficult to Because the surgeries can be more complex, preoperative
resect, which is cited by many gynecologic oncologists as preparation is crucial, sometimes requiring consultation
the most common reason for a suboptimal outcome at the with general surgeons, surgical oncologists, or hepatobiliary
time of PDS.17,18 surgeons to assist in the event that disease encountered
To address, the widespread nature of advanced ovarian at PDS is difficult or impossible for the gynecologic oncol-
cancer at presentation, two strategies have emerged. One ogist to remove. This can sometimes be difficult at medical
strategy, NACT for three or more cycles followed by the centers with less experienced practitioners. Recent analyses
cytoreductive procedure, IDS, followed by the completion of have shown that optimal cytoreductive surgery rates are
up to six to eight total cycles of chemotherapy, has been higher among more experienced surgeons at higher-volume
proposed as a viable method to decrease preoperative hospitals, with gynecologic oncologists at expert centers
disease burden, increase the rates of optimal cytoreduction, attaining optimal resection rates of greater than 75%.10,28,29
and lower postoperative morbidity. The second strategy has This is in stark contrast to the 41% to 42% optimal cytoreduction
been to expand the surgical armamentarium of gynecologic rates reported in the PDS arms of the EORTC/NCIC and
oncology surgeons to include extensive upper abdominal CHORUS PDS versus NACT/IDS trials (Table 1).12,13 More-
over, the median operating times of 165 and 120 minutes,
respectively, in the PDS arms of the two studies seems to be
inadequate for a complete surgical evaluation and attempt
KEY POINTS at achieving optimal or no gross residual disease status.12,13
When a patient is treated with the appropriate surgery
These findings question whether the PDS attempted in
by an experienced surgeon or team of surgeons at an these two trials were what one would see in more expe-
experienced hospital, optimal and even complete gross rienced, expert centers. Indeed, it is the duty of the op-
resection at the time of PDS is achievable in a vast erating physician to give the patient the best chance
majority of cases. at achieving CGR, even if that means additional train-
With the median overall survival of 66 months for ing, multispecialty consultation, or referral to another
patients with stage III, optimally debulked ovarian surgeon.
cancer treated with intraperitoneal chemotherapy on
GOG 172 serves as the gold standard for quality care,
which all future studies should attempt to attain. It Decreases Chemoresistance
NACT may result in better patient-reported outcomes
There are innate benefits to PDS. Large bulky tumors are
and QOL measures with decreased morbidity when
compared with PDS.
often necrotic and hypoxic. Their poor blood supply does not
The biggest utility of NACT may be in research, where often lend itself to maximal intravenous or intraperitoneal
this approach provides the opportunity for the chemotherapy delivery. Comparatively, smaller tumors are
investigation of predictive markers, mechanisms of well perfused and easier to target.30 At the time of diag-
resistance, and a forum to test novel therapies. nosis and the initiation of treatment, both chemotherapy-
sensitive and chemotherapy-resistant cells are present in a

154 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


KEEPING THE PEACE AND BALANCING THE BIOLOGY

TABLE 1. Randomized Trials of PDS Versus NACT/IDS FIGURE 1. MSKCC Primary Cytoreduction OS and CGR
Rates
EORTC/ EORTC/ CHORUS CHORUS
NCIC PDS NCIC NACT PDS NACT
Number of 361 357 255 219
Patients
Residual 1 cm 42% 81% 41% 73%
No Gross Residual 18% 45% 17% 39%
PFS (months) 12 12 11 12
OS (months) 29 30 23 24
Abbreviations: IDS, interval debulking surgery; EORTC, European Organization for
Research and Treatment of Cancer; NCIC, National Cancer Institute of Canada; PDS,
primary debulking surgery; NACT, neoadjuvant chemotherapy; PFS, progression-free
survival; OS, overall survival.

patient. Primary surgery decreases the tumor burdens of


both of these cell lines and decreases the quantity of cells
that can spontaneously mutate to drug-resistant pheno-
types.31 Thus, surgical debulking aids in overcoming negative
tumor biology.32 Approximately 25% of patients have
platinum-resistant disease at the time of their first relapse,
and almost all patients who have disease recurrence will
eventually become chemotherapy-resistant.33 By initiating
chemical debulking first with the NACT approach, the tumor
cells have more time to build increased resistance. Breaking Abbreviations: MSKCC, Memorial Sloan Kettering Cancer Center; OS, overall
survival; CGR, complete gross resection; mos, months. CGR, complete gross
up the chemotherapy by introducing IDS in the middle of the resection; PDS, primary debulking surgery.
six to eight cycles of chemotherapy may also have this effect.
of any medical intervention. Though not an integral part
It Yields Better Survival Outcomes of standard care, QOL measures are regularly used in
The median OS of 66 months in stage III ovarian cancer prospective clinical trials. Increasingly popular is the use
patients who underwent optimal debulking and were of patient-reported outcomes (PROs), in which QOL mea-
treated with intraperitoneal chemotherapy on GOG 172 was sures serve as an inherent element in addition to reported
stated to be the longest median survival ever reported in a adverse sequelae.38 The increasing importance of QOL is
randomized phase III ovarian cancer GOG clinical trial.34 accentuated by data demonstrating that it is also a predictive
These results serve as a gold standard for quality care, which variable for survival.39 Furthermore, a recent cost-efficacy
future studies should try to attain. Although the PFS and OS analysis of the GOG 218 trial determining the benefit of the
for the NACT arms of the EORTC/NCIC and CHORUS trials are addition of bevacizumab to platinum-based chemotherapy
consistent with those described in other NACT studies, the revealed that the incorporation of QOL outcomes made a
overall OS rates of 23 to 30 months in all four arms of these meaningful contribution to the results, rendering the ad-
studies, and, in fact, in essentially all studies advocating dition of bevacizumab even less favorable compared with
the NACT/IDS approach, are very low (Table 1).12,13,35 In the Markov model, which excluded QOL outcomes.40 In-
contrast, a retrospective review of an identical sample cluding QOL outcomes in noninferiority studies as a measure
population of patients treated during the same time period of the morbidity and overall impact of an intervention is
as the EORTC/NCIC trial at MSKCC, demonstrated a median undoubtedly of major importance.
OS rate of 50 months for all patients treated with PDS.36 A Although both surgery and chemotherapy are essential
recent publication from the same institution reported a interventions in treating patients with advanced ovarian
median survival of 72 months for all patients who had cancer, there is a relative paucity of trials addressing the
undergone PDS regardless of residual disease status (in- role of surgery. The surgically based, randomized pro-
cluding those who had both optimal and suboptimal spective studies encompass the effect of IDS, the role
cytoreduction; Fig. 1).37 of secondary cytoreductive surgery at the time of re-
currence, the value of lymphadenectomy in debulking, and
CHEMOTHERAPY FIRST: QUICKER delayed PDS.12,13,30,41-43 The EORTC/NCIC and CHORUS
IMPROVEMENT IN QUALITY OF LIFE? studies are the only two reported prospective randomized
Quality of Life Is an Essential Parameter trials addressing the use of NACT in ovarian cancer. In
QOL is an important parameter in the evaluation of ther- both trials, similar QOL assessment tools were used: the
apeutic interventions and is currently the main mecha- EORTC QLQ-C30 questionnaire version 3, which consists
nism to derive patients perspective regarding the impact of global health status/QOL multifunctional scales and

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 155


LEARY ET AL

symptom scales combined with a scale regarding financial An Evaluation of Quality of Life in CHORUS
difficulties; and the ovarian cancerspecific QOL (QLQ- The CHORUS trial measured similar QOL parameters within
Ov28).44 These are well-validated and reliable question- the same time frame as the EORTC study. CHORUS reported
naires used frequently in ovarian cancer studies. The an equivalent survival pattern in each arm, but with lower
assessments were administered before patient randomi- median survival rates at 22 months for PDS and 23 months
zation at the end of the third and the sixth cycles of for NACT.13 Notably, the median age of women enrolled was
chemotherapy and at 6- and 12-month follow-up visits. In older, at age 65, and 19% had a performance status (PS)
both studies, the end of the third cycle of chemotherapy score of 2 or 3. The older age and poorer PS probably
was the presurgical assessment in the NACT arm of the contributed, in part, to the lower median survival patterns.
studies. For the purposes of this article, the QOL data were collected
from all possible cases and not confined to the centers with
An Evaluation of Quality of Life in EORTC 55971 better QOL data collection rates. There was sufficient data
The EORTC 55971 trial included QOL analysis from more available from 102 patients in the PDS arm and 117 in the
than 400 women from the institutions with best com- NACT arm. The Global QOL remained unchanged in both
pliance for QOL data collection. This provided sufficient arms, with mean scores of 51.5 (95% CI, 45.856.2) at
numbers to permit clinically meaningful differences to be baseline to 69.2 (95% CI, 56.164.3) at 3 months, and 53.6
detected.44 Good compliance was defined as institutions (95% CI, 48.658.7) to 58.6 (95% CI, 54.562.8; p = .58,
that had at least 50% compliance at baseline and 35% respectively). Again, accepting a 10-point change in the mean
compliance on average during follow-up over all enrolled score as being clinically significant, improvements were noted
patients. The QLQ-C30 scales and single-item questions in nausea/vomiting and dyspnea at 3 months compared with
were linearly transformed to a 0 to 100 scale and analyzed baseline in the PDS arm. In the same time frame, diarrhea
according to the procedures recommended by the EORTC symptoms improved in the NACT arm. In both arms, pain and
QOL group. A higher score on the functional and global/QOL appetite scores improved, whereas abdominal/gastric
scales indicated better QOL, whereas a higher score on the symptoms and peripheral neuropathy worsened. Not sur-
symptoms scale indicated poorer QOL. At least a 10-point prisingly, chemotherapy-associated side effects worsened
alteration from baseline was necessary to indicate a ma- in the NACT arm. These were the only parameters with
jor change in QOL. The analysis was also repeated changes; however, they were not significant. However, if
with stratification factors at randomization including the data are graphically demonstrated, as can be seen in
country, method of tumor biopsy, International Federation Fig. 2, over the 6-month period, the trend is that the NACT
of Gynecology and Obstetrics stage, and largest tumor arm showed an improving trend compared with the
diameter. PDS arm.
There were no differences noted in the QOL functioning Hence, is NACT a quicker route to a better QOL compared
or symptom scales between the two arms except for with the primary surgery approach? Both prospective ran-
fatigue, pain, and dyspnea. These changes were deemed domized trials seem to indicate that there are no major
clinically relevant because there was a 10-point change statistical differences in QOL between PDS and NACT.
from baseline. The level of fatigue improved in the PDS However, it may be argued that the trends are of more
arm at the third cycle of chemotherapy and persisted to importance than the exact numbers. Combined with other
12 months, whereas in the NACT arm, the same im-
provement occurred, though was noted only at 6 months
(i.e., no changes during treatment, and persisted to
12 months). The pain scale also improved in both arms to FIGURE 2. CHORUS Global Quality of Life Evolution
subsequent points of measurement. Regarding dyspnea, Over Time
an improvement was noted at the sixth chemotherapy
cycle and at 6 months in the NACT arm, but not the PDS
arm. Though these differences were noted, the overall
conclusion was that QOL did not differ between the treatment
strategies. However, there was an unusual and notable
finding. There was a significant survival difference between
institutions with good compliance collecting QOL measures
versus those who were less compliant: the median OS was
32 months versus 23 months (p = .0006). Additionally, the
optimal debulking rates were nearly twice that in selected
versus excluded institutions at 40% and 20%, respectively
(p = .001). Although there may be other explanations and
possible confounding factors, QOL data collection compliance
may also be an indicator of an institutions excellence. Abbreviation: QOL, quality of life.

156 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


KEEPING THE PEACE AND BALANCING THE BIOLOGY

morbidity data, one could favor the neoadjuvant approach in to determine whether these genomic markers may in-
this patient population. fluence the therapeutic sequence for patients with ad-
vanced HGSOC.
BIOLOGIC CONSIDERATIONS REGARDING THE
THERAPEUTIC SEQUENCE
Evidence for NACT as a Driver of Chemotherapy
Histologic and Genomic Factors to Select Patients
Resistance
One argument proposed in support of NACT is to reduce
As previously mentioned, a risk of the neoadjuvant approach
tumor bulk and increase the chance of a complete and
is that treating a greater volume of a heterogeneous tumor
potentially less morbid surgical resection. This makes
with NACT may be driving platinum resistance.58 Retro-
biologic sense in chemotherapy-sensitive high-grade se-
spective studies have shown that NACT was associated
rous ovarian cancer (HGSOC), where response rates to
with a higher risk of platinum-resistant relapse, even when
first-line platinum-based chemotherapy approach 75%.
IDS resulted in complete macroscopic resection.59,60 In the
However, rare subtypes such as low-grade serous ovarian
case of platinum-sensitive progression, lower response rates
cancer (LGSOC) and mucinous ovarian cancer (mOC) are
to a platinum combination have been described after NACT,
much less responsive to standard first-line platinum-
suggesting that NACT may be compromising responsiveness
based therapy (Table 2).45-54 A small study on LGSOC
to subsequent therapy.59,61 Although this remains a hypo-
reported an objective response rate of 3.5% to platinum-
thetical risk, these studies are limited by their retrospective
based NACT, whereas only 20% of patients with advanced
nature and inherent biases that justified the decision to offer
mucinous ovarian cancer recruited to a randomized trial
NACT in the first place, which likely explain these patients
showed an objective response to a first-line platinum
poor outcomes. Similarly, studies evaluating whether the
combination.45,50 Although numbers of patients included
number of cycles of NACT influenced outcome have shown
in these reports were small, these data raise important
worse survival with more than three or four cycles of NACT
questions regarding the value of NACT in rare subtypes of
even when complete macroscopic resection was achieved at
epithelial ovarian cancer. A major concern with NACT is
IDS.62,63 Again, because of the retrospective nature of these
the risk of progression and loss of opportunity for patients
studies, the poor outcome for patients receiving more than
who might go from being candidates for a morbid, but
four cycles may simply reflect inherent differences in tumor
feasible, PDS to not being candidates for surgery at all. In
biology and chemotherapy responsiveness rather than a
this regard, tumor biology should clearly influence the
deleterious impact of more cycles of NACT. In fact, a more
therapeutic sequence and one could propose a histologically
recent meta-analysis of 21 studies failed to show that the
driven algorithm for patients with bulky epithelial ovarian
number of NACT cycles affected survival.64 The only avail-
cancer. Recent comprehensive genomic studies in HGSOC
able prospective data come from the two randomized
have started to identify candidate molecular predictors
controlled trials of PDS versus NACT, and these failed to
of platinum sensitivity (BRCA1/2 mutations or alterations
show a difference in PFS or OS, making it unlikely that NACT
in other homologous recombination genes) or resistance
is a major driver of platinum resistance.12,24
(CCNE1 amplifications).55-57 Future studies will be crucial
In vivo evaluation of chemotherapy responsiveness during
TABLE 2. Response to First-Line Platinum-Based NACT. As opposed to the adjuvant setting in which patients
Chemotherapy in Rare Subtype of Epithelial Ovarian are treated with no measurable disease, an advantage of
Cancer NACT is that it could offer the opportunity for an in vivo
evaluation of chemotherapy responsiveness. Careful mon-
No. of Patients itoring of tumor response during chemotherapy could
With Evaluable Activity
Disease (oRR) Reference(s) identify nonresponders early and justify treatment change in
an effort to improve the efficacy of NACT and long-term
LGSOC
patient outcomes. Some prospective studies in locally ad-
First-Line 24 , 5% Schmeler et al 45
Carboplatin- vanced breast cancer have started to suggest a role for
Paclitaxel response-adapted NACT where the neoadjuvant protocol is
CCC modified according to early tumor response. One trial
First-Line 4 studies, 2368 22%41% Kita et al, Sugyama showed an improvement in PFS and OS among patients with
Platinum- et al, Ho et al, and locally advanced breast cancer randomly selected to an
Based Takano et al 46-49 adaptive NACT strategy with a chemotherapy switch in
mOC case of poor response compared with standard NACT.65
First-Line 5 studies, 950 13%60% Hess et al, Alexandre Whether adaptive NACT could be beneficial in ovarian
Platinum et al, Pectasides cancer has never been investigated and may be limited by
Based et al, Gore et al, and
Shimada et al 50-54 the fact that measuring on-treatment response to NACT is
Abbreviations: oRR, objective response rate; LGSOC, low-grade serous ovarian cancer;
difficult in ovarian cancer.66 Neither improvement in CT
CCC, clear cell ovarian cancer; mOC, mucinous ovarian cancer. imaging nor biologic response by CA-125 are reliable for

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 157


LEARY ET AL

NACT response evaluation. 67 Other technologies such as allow for optimization of postoperative management, con-
metabolic imaging by fluorodeoxyglucose-PET or circulat- sideration of maintenance treatment, or participation in
ing cell-free tumor DNA may offer early-response in- clinical trials, with the aim of improving long-term outcome.
formation during NACT for advanced ovarian cancer. The question remains whether the degree of pathologic re-
However, to date it is unclear how this information would be sponse is prognostic, or whether the only valid surrogate for
clinically useful. A small subset of patients (15%) with HGSOC OS is pCR strictly defined as no residual cancer cells within the
are refractory and progress clinically and biologically during entire surgical specimen. Studies in breast cancer have shown
NACT. The only value of identifying these early non- that complete response in both primary site and lymph nodes
responders would be if this could inform clinical decision (occurring in 18% of patients with NACT) is required to
making. Unfortunately, ovarian cancer does not have an predict a major improvement in OS,78 the biologic explanation
effective alternative to standard neoadjuvant carboplatin being that pCR is the evidence that other micrometastatic
and paclitaxel. deposits may have also been eradicated by NACT. In-
terestingly, the correlation between pathologic response and
In vivo evaluation of chemotherapy responsiveness OS was the strongest for triple-negative breast cancer, a
at IDS. Complete resection of all macroscopic disease at PDS highly proliferative subtype that shares genetic homology
has been confirmed as a critical prognostic factor32; how- with HGSOC.
ever, its value after NACT is less clear because visualization of
residual disease may be less reliable after chemotherapy.68
Studies have sought to evaluate whether the degree of
The Neoadjuvant Setting as a Tool to Evaluate
pathologic response to NACT could provide prognostic
New Regimens
The neoadjuvant design may be useful to test new therapies
information (Table 3).69-76 A number of studies have
in a faster way and with fewer patients than is required for
confirmed the prognostic value of pathologic complete
large adjuvant trials. Given the strong correlation between
response (pCR) after NACT. Among patients treated with
pCR and OS in breast cancer, new drugs are being evaluated
NACT and no residual disease at IDS, Ferron et al showed that
in randomized neoadjuvant trials in which the primary
pCR with no viable tumor cells in the surgical specimen
endpoint is an improvement in pCR at surgery.79 pCR rates
occurred in 14% of patients and was predictive of PFS, but
are somewhat lower in ovarian cancer than in other tumor
the degree of histologic response was not.69 A larger study
types (6% to 12%), but randomized studies designed to test
(322 patients) confirmed pCR (defined as no residual tumor
the benefit of adding a new agent to standard chemo-
cells in the surgical specimen) as predictive of both PFS and
therapy, where the primary objective would be a major
OS, whereas a good response not amounting to pCR was
improvement in pCR, could provide a useful signal-finding
not prognostic.70 Several groups investigated composite
strategy for new drugs. The advantages of this approach
pathologic response scores, incorporating an assessment
are a translational research opportunity to identify pre-
of viable tumor cells, fibrosis, inflammatory changes, or
dictive biomarkers and obtaining a rapid answer because
macrocytic infiltration, and have shown conflicting re-
results are available within a few months of the last patient
sults using heterogeneous definitions for pathologic re-
who was randomly placed. In addition, the neoadjuvant trial
sponse (Table 3).71-75,77 A more recent study suggested that a
design allows the testing of a novel agent in treatment-naive
three-tier histopathologic chemotherapy response score was
patients, as opposed to metastatic studies, where prior
reproducible and prognostically meaningful for patients with
exposure to multiple lines of chemotherapy may distort drug
HGSOC treated with NACT, regardless of debulking status.76
activity. The U.S. Food and Drug Administration has recently
Complete or near-complete pathologic response (defined as
issued a statement that they will strongly consider evidence
a chemotherapy response score [CRS] of 3) was associated
from randomized controlled studies using pCR as an end-
with significantly improved PFS (18 vs. 12 months, p , .001)
point to allow accelerated approval of novel therapies.80
and decreased risk of platinum-resistant relapse (odds ratio
Although this guidance was designed for trials conducted in
[OR] 0.08; p , .001) compared with those tumors showing
breast cancer, using pCR as an endpoint has the potential to
less in vivo chemosensitivity (CRS scores of 1 and 2).
help address unmet needs in high-risk populations in a far
Although the authors concluded that the three-tier scoring
shorter time frame than would be required via the con-
system may be useful, in fact, outcomes were only different
ventional approach to drug development.79
when considering CRS 3 versus CRS 1/2, making it more of a
two-tier system. In addition, CRS 3 included near-complete
responses defined as residual viable cells, or nodules larger Translational Research Opportunities
than 2 mm, and did not predict OS. Although the pathologic Despite exquisite sensitivity to first-line platinum-based
response score may need refining and will require pro- chemotherapy, HGSOC almost invariably relapses. Yet the
spective validation, taken together these data suggest that mechanisms underpinning primary or acquired chemo-
an evaluation of in vivo response to NACT at the time of IDS therapy resistance are poorly understood (Fig. 3). HGSOC
could offer useful prognostic information and may inform is characterized by extreme genomic instability and
postoperative management. The identification of a che- intratumoral heterogeneity (ITH).81 Inherent genetic in-
motherapy response score to risk-stratify patients would stability and ITH provide the ideal setting for adaptation and

158 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


KEEPING THE PEACE AND BALANCING THE BIOLOGY

TABLE 3. Chemotherapy Pathologic Scores Evaluated in Retrospective Studies


Categories of Pathological Response No. of
Reference Score Patients Correlation of Pathological Score and Outcome
Ferron et al69 Group 1: No residual tumor cells 8 3-year PFS: 63% (p 5 .02 vs. groups 2/3)
(58 patients)
Group 2: Persistent disease with marked 14 3-year PFS: 12% (not significant)
changes
Group 3: Persistence of at least one site 36 3-year PFS: 19% (not significant)
with no chemotherapy effect
Petrillo et al70 pCR: No residual tumor cells 21 PFS: 36 months (p 5 .001 vs. micro/macroPR); OS: 72 months (p 5 .018 vs.
(322 patients) micro/macroPR)
MicroPR: Residual tumor foci # 3 mm 104 PFS microPR: 16 months vs. 13 months for macro PR (not significant); OS microPR:
38 months vs. 29 months for macroPR (not significant)
MacroPR . 3-mm foci 197
Le et al71 Score* 1-8: Some chemotherapy effect NA OS: Scores 1-8 vs. score 0 (73 months vs. 39 months; p 5 .14)
(62 patients)
Score 0: No chemotherapy effect NA
Le et al72 Marked/moderate necrosis 36 Improved PFS; HR 0.51; p 5 .048
(73 patients)
Little to no necrosis 37
Le et al73 Chemotherapy effect on omental 58 OS: 84 months
(66 patients) deposits
No omental response 8 OS: 31 months
Muraji et al74 Composite score** G2/3 72 Improved OS; HR 0.61; p 5 .03
(124 patients)
Composite score** G0/1 52
Sassen et al75 No residual tumor cells or foci , 5 mm 7 OS: 46 months; p 5 .02
(49 patients)
Tumor foci . 5 mm 42 OS: 27 months
Bohm et al76 CRS 3: No residual tumor cells or 19 Improved PFS: CRS 3 vs. CRS 1/2, (18 months vs. 12 months;
(71 patients) foci , 2 mm p , .001); OS: CRS 3 (45 months) vs. CRS 1/2 (28 months) not significant
CRS 2: Appreciable response 47
CRS 1: No or minimal response 5
Abbreviations: PFS, progression-free survival; OS, overall survival; IDS, interval debulking surgery; NA, not available; pCR, pathologic complete response; CRS, chemotherapy response
score; HR, hazard ratio; microPR, microscopic pathologic response; macroPR, macroscopic pathologic response.
*Score (0-8) of necrosis, fibrosis, macrophage infiltration, tumor inflammation.
**Composite score 5 degree of disappearance of tumor cells, displacement by necrosis and fibrosis, and inflammatory changes graded from 0 to 3, with G0 being no chemotherapy
effect and G3 being no viable tumor cells.

treatment escape and have been shown to drive the heterogeneous may be relatively uninformative. However,
accelerated acquisition of multidrug resistance.82 Evidence identifying chromosomal alterations or somatic muta-
in HGSOC cell lines suggests that relapse is not caused by tional events that occur at low frequency at baseline and
linear acquisition of genetic alterations, but rather to are selected for during therapy could uncover new ther-
treatment-induced selection and expansion of intrinsically apeutic strategies to overcome platinum resistance. Given
resistant clones.83 Most genomic studies to date in HGSOC the intrinsic chemotherapy sensitivity of the majority of
have focused on comprehensive analyses of the primary HGSOC, platinum-based chemotherapy will likely always
tumor, but evidence is emerging that a comparison be- provide an essential component of the treatment strat-
tween matched tumor samples from diagnosis and IDS can egy; however, profiling the post-NACT tumor may identify
reveal the mechanisms accounting for the acquisition of actionable targets and provide the rationale for sub-
resistance in individual patients. For example, an enrich- sequent clinical trials testing novel agents as maintenance
ment of ALDH1-positive cells in post-NACT samples has therapy in an effort to eradicate the minimal residual dis-
been demonstrated in some patients and shown to cor- ease post-chemotherapy.
relate with poor survival post-NACT, suggesting that a
cancer stem cell subpopulation may account for chemo-
therapy resistance.84 Studies among patients with germline FUTURE DIRECTIONS AND CONCLUSION
BRCA mutations treated with NACT have shown that, al- The discussed considerations truly underscore the need
though initial response rates to platinum are high, a subset for further research on the most appropriate sequence of
shows restoration of BRCA heterozygosity in the post-NACT care for women with advanced ovarian cancer. Despite
sample, suggesting a rapid expansion of subclones without years of debate and two randomized controlled trials,
somatic LOH for the BRCA wild-type allele during the question of whether surgery or chemotherapy should
treatment.85 be the primary treatment strategy has yet to be an-
Profiling HGSOC at diagnosis when the bulk of dis- swered. Survival and QOL-related measures are both
ease is chemotherapy-sensitive and potentially highly of paramount importance because they pertain to

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 159


LEARY ET AL

FIGURE 3. The Neoadjuvant Setting: A Translational Research Opportunity

(A) Fifteen percent of HGSOC cases have refractory disease and progress on NACT. Prognosis is poor and studies are needed to characterize the profile of these primary
resistant tumors and develop effective alternatives to carboplatin and paclitaxel. However, HGSOC is very chemotherapy-sensitive and most show an initial response to
NACT and may progress to IDS. (B) A small subset (5% to 12%) show pCR with no residual tumor cells after NACT. Prognosis is excellent and studies should focus on
finding therapies that increase the proportion of patients achieving pCR. (C) Most HGSOC demonstrate initial sensitivity but despite complete debulking will relapse.
Identification of molecular alterations selectively enriched in these post-treatment tumors may uncover mechanisms of acquired resistance and identify new therapeutic
targets that could eradicate minimal residual disease.
Abbreviations: HGSOC, high-grade serous ovarian cancer; NACT, neoadjuvant chemotherapy; IDS, interval debulking surgery; pCR, pathologic complete response.

overall outcomes. Although we seek to answer these role for NACT in the laboratory and in clinical research.
questions, excellent clinical judgment is necessary to care- Much effort must be made to determine novel ap-
fully and aptly select the appropriate patient for each proaches to the management of this complicated and
treatment regimen. Furthermore, there is a definitive ever-evolving disease.

References
1. Holschneider CH, Berek JS. Ovarian cancer: epidemiology, biology, and 10. Bristow RE, Tomacruz RS, Armstrong DK, et al. Survival effect of maxi-
prognostic factors. Semin Surg Oncol. 2000;19:3-10. mal cytoreductive surgery for advanced ovarian carcinoma during the
2. Lowe KA, Chia VM, Taylor A, et al. An international assessment of platinum era: a meta-analysis. J Clin Oncol. 2002;20:1248-1259.
ovarian cancer incidence and mortality. Gynecol Oncol. 2013;130: 11. Vergote I, Trope CG, Amant F, et al. EORTC-GCG/NCIC-CTG randomised
107-114. trial comparing primary debulking surgery with neoadjuvant chemo-
3. Armstrong DK. Relapsed ovarian cancer: challenges and management therapy in stage IIIC-IV ovarian, fallopian tube and peritoneal cancer
strategies for a chronic disease. Oncologist. 2002;7(suppl 5):20-28. (OVCA). Presented at: 12th Biennial Meeting of the International Gy-
4. Torre LA, Bray F, Siegel RL, et al. Global cancer statistics, 2012. CA Cancer necologic Cancer Society; October 2008; Bangkok, Thailand.
J Clin. 2015;65:87-108. 12. Vergote I, Trope CG, Amant F, et al; European Organization for Research
5. Morgan RJ Jr, Alvarez RD, Armstrong DK, et al; National Comprehensive and Treatment of Cancer-Gynaecological Cancer Group; NCIC Clinical
Cancer Network. Epithelial ovarian cancer. J Natl Compr Canc Netw. Trials Group. Neoadjuvant chemotherapy or primary surgery in stage
2011;9:82-113. IIIC or IV ovarian cancer. N Engl J Med. 2010;363:943-953.
6. Meigs JV, Greenough RB. Tumors of the female pelvic organs. Am J Med 13. Kehoe S, et al. Chemotherapy or upfront surgery for newly diagnosed
Sci. 1935;189:430. advanced ovarian cancer: Results from the MRC CHORUS trial. J Clin
7. Griffiths CT. Surgical resection of tumor bulk in the primary treatment of Oncol. 2013;31:15s (suppl; abstr 5500).
ovarian carcinoma. Natl Cancer Inst Monogr. 1975;42:101-104. 14. Bristow RE, Duska LR, Lambrou NC, et al. A model for predicting surgical
8. Eisenkop SM, Friedman RL, Wang H-J. Complete cytoreductive surgery outcome in patients with advanced ovarian carcinoma using computed
is feasible and maximizes survival in patients with advanced epithelial tomography. Cancer. 2000;89:1532-1540.
ovarian cancer: a prospective study. Gynecol Oncol. 1998;69:103-108. 15. Hoskins WJ, McGuire WP, Brady MF, et al. The effect of diameter of
9. Allen DG, Heintz AP, Touw FW. A meta-analysis of residual disease and largest residual disease on survival after primary cytoreductive surgery
survival in stage III and IV carcinoma of the ovary. Eur J Gynaecol Oncol. in patients with suboptimal residual epithelial ovarian carcinoma. Am J
1995;16:349-356. Obstet Gynecol. 1994;170:974-979, discussion 979-980.

160 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


KEEPING THE PEACE AND BALANCING THE BIOLOGY

16. Dauplat J, Le Bouedec G, Pomel C, Scherer C. Cytoreductive surgery for 34. Armstrong DK, Bundy B, Wenzel L, et al; Gynecologic Oncology Group.
advanced stages of ovarian cancer. Semin Surg Oncol. 2000;19:42-48. Intraperitoneal cisplatin and paclitaxel in ovarian cancer. N Engl J Med.
17. Zivanovic O, Eisenhauer EL, Zhou Q, et al. The impact of bulky upper 2006;354:34-43.
abdominal disease cephalad to the greater omentum on surgical 35. Bristow RE, Chi DS. Platinum-based neoadjuvant chemotherapy and
outcome for stage IIIC epithelial ovarian, fallopian tube, and primary interval surgical cytoreduction for advanced ovarian cancer: a meta-
peritoneal cancer. Gynecol Oncol. 2008;108:287-292. analysis. Gynecol Oncol. 2006;103:1070-1076.
18. Eisenkop SM, Spirtos NM. What are the current surgical objectives, 36. Chi DS, Musa F, Dao F, et al. An analysis of patients with bulky advanced
strategies, and technical capabilities of gynecologic oncologists treating stage ovarian, tubal, and peritoneal carcinoma treated with primary
advanced epithelial ovarian cancer? Gynecol Oncol. 2001;82:489-497. debulking surgery (PDS) during an identical time period as the ran-
19. Chi DS, Eisenhauer EL, Zivanovic O, et al. Improved progression-free and domized EORTC-NCIC trial of PDS vs neoadjuvant chemotherapy
overall survival in advanced ovarian cancer as a result of a change in
(NACT). Gynecol Oncol. 2012;124:10-14.
surgical paradigm. Gynecol Oncol. 2009;114:26-31.
37. Mueller JJ, Zhou QC, Iasonos A, et al. Neoadjuvant chemotherapy and
20. Angioli R, Plotti F, Aloisi A, et al. Does extensive upper abdomen surgery
primary debulking surgery utilization for advanced-stage ovarian cancer
during primary cytoreduction impact on long-term quality of life? Int J
at a comprehensive cancer center. Gynecol Oncol. 2016;140:436-442.
Gynecol Cancer. 2013;23:442-447.
38. Soo Hoo S, Marriott N, Houlton A, et al. Patient-reported outcomes
21. Barlin JN, Long KC, Tanner EJ, et al. Optimal (#1 cm) but visible residual
after extensive (ultraradical) surgery for ovarian cancer: results from a
disease: is extensive debulking warranted? Gynecol Oncol. 2013;130:
prospective longitudinal feasibility study. Int J Gynecol Cancer. 2015;25:
284-288.
1599-1607.
22. Chi DS, Zivanovic O, Levinson KL, et al. The incidence of major com-
39. Carey MS, Bacon M, Tu D, et al. The prognostic effects of performance
plications after the performance of extensive upper abdominal surgical
procedures during primary cytoreduction of advanced ovarian, tubal, status and quality of life scores on progression-free survival and overall
and peritoneal carcinomas. Gynecol Oncol. 2010;119:38-42. survival in advanced ovarian cancer. Gynecol Oncol. 2008;108:100-105.
23. Fanfani F, Fagotti A, Gallotta V, et al. Upper abdominal surgery in 40. Cohn DE, Barnett JC, Wenzel L, et al. A cost-utility analysis of NRG
advanced and recurrent ovarian cancer: role of diaphragmatic surgery. Oncology/Gynecologic Oncology Group Protocol 218: incorporating
Gynecol Oncol. 2010;116:497-501. prospectively collected quality-of-life scores in an economic model of
24. Kehoe SM, Eisenhauer EL, Chi DS. Upper abdominal surgical procedures: treatment of ovarian cancer. Gynecol Oncol. 2015;136:293-299.
liver mobilization and diaphragm peritonectomy/resection, splenectomy, 41. Rose PG, Nerenstone S, Brady MF, et al; Gynecologic Oncology Group.
and distal pancreatectomy. Gynecol Oncol. 2008;111:S51-S55. Secondary surgical cytoreduction for advanced ovarian carcinoma.
25. Aletti GD, Dowdy SC, Gostout BS, et al. Aggressive surgical effort and N Engl J Med. 2004;351:2489-2497.
improved survival in advanced-stage ovarian cancer. Obstet Gynecol. 42. Maggioni A, Benedetti Panici P, DellAnna T, et al. Randomised study of
2006;107:77-85. systematic lymphadenectomy in patients with epithelial ovarian cancer
26. Eisenhauer EL, Abu-Rustum NR, Sonoda Y, et al. The addition of macroscopically confined to the pelvis. Br J Cancer. 2006;95:699-704.
extensive upper abdominal surgery to achieve optimal cytoreduction 43. Gynecologic Cancer Intergroup. GCIG Clinical Trials By Lead Cooperative
improves survival in patients with stages IIIC-IV epithelial ovarian Group. http://www.gcig.igcs.org/ClinicalTrials.html. Accessed February
cancer. Gynecol Oncol. 2006;103:1083-1090. 1, 2016.
27. Horowitz N, Miller A, Rungruang B, et al. Reply to G.D. Aletti et al. 44. Greimel E, Kristensen GB, van der Burg ME, et al; European Organization
J Clin Oncol. 2015;33:3521-3522. for Research and Treatment of Cancer - Gynaecological Cancer Group
28. Vernooij F, Heintz AP, Coebergh JW, et al. Specialized and high-volume and NCIC Clinical Trials Group. Quality of life of advanced ovarian cancer
care leads to better outcomes of ovarian cancer treatment in the patients in the randomized phase III study comparing primary debulking
Netherlands. Gynecol Oncol. 2009;112:455-461. surgery versus neo-adjuvant chemotherapy. Gynecol Oncol. 2013;131:
29. Long B, Chang J, Ziogas A, et al. Impact of race, socioeconomic status, 437-444.
and the health care system on the treatment of advanced-stage ovarian 45. Schmeler KM, Sun CC, Bodurka DC, et al. Neoadjuvant chemotherapy
cancer in California. Am J Obstet Gynecol. 2015;212:468.e1-468.e9.
for low-grade serous carcinoma of the ovary or peritoneum. Gynecol
30. van der Burg ME, van Lent M, Buyse M, et al; Gynecological Cancer
Oncol. 2008;108:510-514.
Cooperative Group of the European Organization for Research and
46. Takano M, Kikuchi Y, Yaegashi N, et al. Clear cell carcinoma of the ovary:
Treatment of Cancer. The effect of debulking surgery after induction
a retrospective multicentre experience of 254 patients with complete
chemotherapy on the prognosis in advanced epithelial ovarian cancer.
surgical staging. Br J Cancer. 2006;94:1369-1374.
N Engl J Med. 1995;332:629-634.
47. Kita T, Kikuchi Y, Kudoh K, et al. Exploratory study of effective che-
31. Goldie JH, Coldman AJ. The genetic origin of drug resistance in neo-
motherapy to clear cell carcinoma of the ovary. Oncol Rep. 2000;7:
plasms: implications for systemic therapy. Cancer Res. 1984;44:
327-331.
3643-3653.
48. Sugiyama T, Kamura T, Kigawa J, et al. Clinical characteristics of clear cell
32. du Bois A, Reuss A, Pujade-Lauraine E, et al. Role of surgical outcome as
prognostic factor in advanced epithelial ovarian cancer: a combined carcinoma of the ovary: a distinct histologic type with poor prognosis and
exploratory analysis of 3 prospectively randomized phase 3 mul- resistance to platinum-based chemotherapy. Cancer. 2000;88:2584-2589.
ticenter trials: by the Arbeitsgemeinschaft Gynaekologische Onkologie 49. Ho CM, Huang YJ, Chen TC, et al. Pure-type clear cell carcinoma of the
Studiengruppe Ovarialkarzinom (AGO-OVAR) and the Groupe ovary as a distinct histological type and improved survival in patients
dInvestigateurs Nationaux Pour les Etudes des Cancers de lOvaire treated with paclitaxel-platinum-based chemotherapy in pure-type
(GINECO). Cancer. 2009;115:1234-1244. advanced disease. Gynecol Oncol. 2004;94:197-203.
33. Pujade-Lauraine E, Hilpert F, Weber B, et al. Bevacizumab combined 50. Gore ME, Hackshaw A, Brady WE, Penson RT, et al. Multicenter trial of
with chemotherapy for platinum-resistant recurrent ovarian cancer: carboplatin/paclitaxel versus oxaliplatin/capecitabine, each with/without
The AURELIA open-label randomized phase III trial. J Clin Oncol. 2014; bevacizumab, as first line chemotherapy for patients with mucinous
32:1302-1308. epithelial ovarian cancer. J Clin Oncol. 2015;33 (suppl; abstr 5528).

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 161


LEARY ET AL

51. Hess V, AHern R, Nasiri N, et al. Mucinous epithelial ovarian cancer: 69. Ferron JG, Uzan C, Rey A, et al. Histological response is not a prognostic
a separate entity requiring specific treatment. J Clin Oncol. 2004;22: factor after neoadjuvant chemotherapy in advanced-stage ovarian
1040-1044. cancer with no residual disease. Eur J Obstet Gynecol Reprod Biol. 2009;
52. Alexandre J, Ray-Coquard I, Selle F, et al; GINECO. Mucinous advanced 147:101-105.
epithelial ovarian carcinoma: clinical presentation and sensitivity to 70. Petrillo M, Zannoni GF, Tortorella L, et al. Prognostic role and predictors
platinum-paclitaxel-based chemotherapy, the GINECO experience. Ann of complete pathologic response to neoadjuvant chemotherapy in
Oncol. 2010;21:2377-2381. primary unresectable ovarian cancer. AM J Obstet Gynecol. 2014; 211:
53. Pectasides D, Fountzilas G, Aravantinos G, et al. Advanced stage mu- 632.e1-8.
cinous epithelial ovarian cancer: the Hellenic Cooperative Oncology 71. Le T, Williams K, Senterman M, et al. Histopathologic assessment of
Group experience. Gynecol Oncol. 2005;97:436-441. chemotherapy effects in epithelial ovarian cancer patients treated with
54. Shimada M, Kigawa J, Ohishi Y, et al. Clinicopathological characteristics neoadjuvant chemotherapy and delayed primary surgical debulking.
of mucinous adenocarcinoma of the ovary. Gynecol Oncol. 2009;113: Gynecol Oncol. 2007;106:160-163.
331-334. 72. Le T, Shahriari P, Hopkins L, et al. Prognostic significance of tumor
55. Tan DS, Rothermundt C, Thomas K, et al. BRCAness syndrome in necrosis in ovarian cancer patients treated with neoadjuvant chemo-
ovarian cancer: a case-control study describing the clinical features and therapy and interval surgical debulking. Int J Gynecol Cancer. 2006;16:
outcome of patients with epithelial ovarian cancer associated with 986-990.
BRCA1 and BRCA2 mutations. J Clin Oncol. 2008;26:5530-5536. 73. Le T, Williams K, Senterman M, et al. Omental chemotherapy effects as a
56. Dann RB, DeLoia JA, Timms KM, et al. BRCA1/2 mutations and ex- prognostic factor in ovarian cancer patients treated with neoadjuvant
pression: response to platinum chemotherapy in patients with chemotherapy and delayed primary surgical debulking. Ann Surg Oncol.
advanced stage epithelial ovarian cancer. Gynecol Oncol. 2012;125: 2007;14:2649-2653.
677-682. 74. Muraji M, Sudo T, Iwasaki S, et al. Histopathology predicts clinical
57. Patch AM, Christie EL, Etemadmoghadam D, et al; Australian Ovarian outcome in advanced epithelial ovarian cancer patients treated with
neoadjuvant chemotherapy and debulking surgery. Gynecol Oncol.
Cancer Study Group. Whole-genome characterization of chemoresistant
2013;131:531-534.
ovarian cancer. Nature. 2015;521:489-494.
75. Sassen S, Schmalfeldt B, Avril N, et al. Histopathologic assessment of
58. Goldie JH, Coldman AJ. Quantitative model for multiple levels of drug
tumor regression after neoadjuvant chemotherapy in advanced-stage
resistance in clinical tumors. Cancer Treat Rep. 1983;67:923-931.
ovarian cancer. Hum Pathol. 2007;38:926-934.
59. Rauh-Hain JA, Nitschmann CC, Worley MJ Jr, et al. Platinum resistance
76. Bohm S, Faruqi A, Said I, et al. Chemotherapy response score: devel-
after neoadjuvant chemotherapy compared to primary surgery in
opment and validation of a system to quantify histopathologic response
patients with advanced epithelial ovarian carcinoma. Gynecol Oncol.
to neoadjuvant chemotherapy in tubo-ovarian high-grade serous car-
2013;129:63-68.
cinoma. J Clin Oncol. 2015;33:2457-2463.
60. Petrillo M, Ferrandina G, Fagotti A, et al. Timing and pattern of re-
77. Le T, Alshaikh G, Hopkins L, et al. Prognostic significance of post-
currence in ovarian cancer patients with high tumor dissemination
operative morbidities in patients with advanced epithelial ovarian
treated with primary debulking surgery versus neoadjuvant chemo-
cancer treated with neoadjuvant chemotherapy and delayed primary
therapy. Ann Surg Oncol. 2013;20:3955-3960.
surgical debulking. Ann Surg Oncol. 2006;13:1711-1716.
61. da Costa AA, Valadares CV, Baiocchi G, et al. Neoadjuvant chemo-
78. Cortazar P, Zhang L, Untch M, et al. Pathological complete response and
therapy followed by interval debulking surgery and the risk of platinum
long-term clinical benefit in breast cancer: the CTNeoBC pooled
resistance in epithelial ovarian cancer. Ann Surg Oncol. 2015;22(suppl
analysis. Lancet. 2014;384:164-172.
3):971-978.
79. Prowell TM, Pazdur R. Pathological complete response and accelerated
62. Bristow RE, Nugent AC, Zahurak ML, et al. Impact of surgeon specialty
drug approval in early breast cancer. N Engl J Med. 2012;366:
on ovarian-conserving surgery in young females with an adnexal mass. 2438-2441.
J Adolesc Health. 2006;39:411-416. 80. U.S. Food and Drug Administration. Guidance for Industry: Pathological
63. Colombo PE, Labaki M, Fabbro M, et al. Impact of neoadjuvant che- Complete Response in Neoadjuvant Treatment of High-Risk Early-Stage
motherapy cycles prior to interval surgery in patients with advanced Breast Cancer: Use as an Endpoint to Support accelerated Approval. http://
epithelial ovarian cancer. Gynecol Oncol. 2014;135:223-230. www.fda.gov/downloads/drugs/guidancecomplianceregulatoryinformation/
64. Kang S, Nam BH. Does neoadjuvant chemotherapy increase optimal guidances/ucm305501.pdf. Accessed March 21, 2016.
cytoreduction rate in advanced ovarian cancer? Meta-analysis of 21 81. Choi YP, Shim HS, Gao MQ, et al. Molecular portraits of intratumoral
studies. Ann Surg Oncol. 2009;16:2315-2320. heterogeneity in human ovarian cancer. Cancer Lett. 2011;307:62-71.
65. von Minckwitz G, Blohmer JU, Costa SD, et al. Response-guided neo- 82. Duesberg P, Stindl R, Hehlmann R. Explaining the high mutation rates of
adjuvant chemotherapy for breast cancer. J Clin Oncol. 2013;31: cancer cells to drug and multidrug resistance by chromosome reas-
3623-3630. sortments that are catalyzed by aneuploidy. Proc Natl Acad Sci USA.
66. Bellati F, Gasparri ML, Caccetta J, et al. Response criteria can be mis- 2000;97:14295-14300.
leading when drawing conclusion regarding neoadjuvant chemother- 83. Cooke SL, Ng CK, Melnyk N, et al. Genomic analysis of genetic het-
apy in advanced ovarian cancer. J Surg Oncol. 2012;106:529, author erogeneity and evolution in high-grade serous ovarian carcinoma.
reply 527-528. Oncogene. 2010;29:4905-4913.
67. Menczer J, Usviatzov I, Ben-Shem E, et al. Neoadjuvant chemotherapy in 84. Ayub TH, Keyver-Paik MD, Debald M, et al. Accumulation of ALDH1-
ovarian, primary peritoneal and tubal carcinoma: can imaging results positive cells after neoadjuvant chemotherapy predicts treatment re-
prior to interval debulking predict survival? J Gynecol Oncol. 2011;22: sistance and prognosticates poor outcome in ovarian cancer. Oncotarget.
183-187. 2015;6:16437-16448.
68. Hynninen J, Lavonius M, Oksa S, et al. Is perioperative visual estimation 85. Gorodnova TV, Sokolenko AP, Ivantsov AO, et al. High response rates to
of intra-abdominal tumor spread reliable in ovarian cancer surgery after neoadjuvant platinum-based therapy in ovarian cancer patients car-
neoadjuvant chemotherapy? Gynecol Oncol. 2013;128:229-232. rying germ-line BRCA mutation. Cancer Lett. 2015;369:363-367.

162 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


GYNECOLOGIC CANCER

Practical Challenges in
Endometrial Cancer

CHAIR
Linda Duska, MD
University of Virginia Health System
Charlottesville, VA

SPEAKERS
Armin Shahrokni, MD, MPH
Memorial Sloan Kettering Cancer Center
New York, NY

Melanie Powell, MD
Barts Health NHS Trust
London, United Kingdom
DUSKA, SHAHROKNI, AND POWELL

Treatment of Older Women With Endometrial Cancer:


Improving Outcomes With Personalized Care
Linda Duska, MD, MPH, Armin Shahrokni, MD, MPH, and Melanie Powell, MD, FRCR, FRCP

OVERVIEW

Endometrial cancer is the most common gynecologic cancer, and with a median age of 62 at diagnosis, it affects a significant
number of older women. With increasing age and obesity rates in the worlds population, there is an anticipated concomitant
increase in older women with endometrial cancer. Older women are more likely to die of endometrial cancer compared with
younger patients. Reasons for this include more aggressive tumor biology, less favorable clinicopathologic features, and
more advanced disease. Other factors, however, such as reluctance to offer surgical treatment to the older patient and
increased complications of treatment are likely to be important. Management of endometrial cancer requires multidis-
ciplinary care (surgery, radiation therapy, and systemic therapy). For each treatment, the feasibility (related to technical
aspect of the procedure/treatment), side effects and safety (related to older-patient factors), and the overall benefit as it
pertains to older women with endometrial cancer should be assessed carefully with a multidisciplinary approach. Despite
the importance of these issues, the data are limited to answer these issues with clarity. In this article, we will review each
treatment modality for older women with endometrial cancer. We will introduce the components of comprehensive
geriatric assessment and their practical implication for older women with cancer in general and older women with en-
dometrial cancer specifically.

E ndometrial cancer is the most common gynecologic


cancer in the United States, with 54,870 cases expected in
2016.1 It is also a cancer of older women, with a median age
in older women, including: more advanced stage at di-
agnosis, higher tumor grade, more aggressive tumor biology,
and a selection bias by providers for less aggressive surgery
at diagnosis of 62.1 As the worlds population ages, we can and less adjuvant therapy.
expect to see more cases of endometrial cancer in older The standard-of-care treatment of most patients with
women. In particular, the oldest old, or those age 80 or endometrial cancer is surgical and begins with hysterec-
older, is the segment of the population that will see the tomy, including removal of the uterus, cervix, and bilateral
greatest growth in the next 2 decades. In the United States, fallopian tubes and ovaries. Although the role of complete
the number of residents over age 80 is expected to increase lymphadenectomy (removal of bilateral pelvic and para-
from 3.3% in 2000 to 5.5% in 2030. At the global level, the aortic retroperitoneal lymph nodes) remains controver-
average annual growth rate of persons age 80 or older is sial, most U.S. gynecologic oncologists follow the so-called
twice as high as the growth rate of the population over age Mayo criteria with respect to staging and remove lymph
60 (3.8% vs. 1.9%).2 Obesity is also a considerable risk factor nodes for high-grade and/or deeply myoinvasive tumors.4
for developing endometrial cancer, and obesity rates are Surgery alone may be curative for early-stage, low-grade
rising worldwide, with an associated increase in endometrial disease, with radiation and/or chemotherapy recommended
cancer rates.3 for adjuvant therapy depending on stage of disease and
Forty percent of women diagnosed with endometrial histology.
cancer are age 65 or older (24%, age 6574; 12%, age 7584; In the older woman, the management of endometrial
and 4%, age . 84).1 Notably, most deaths (67%) from en- cancer may require a more multidisciplinary approach.
dometrial cancer occur in women over 65, with a median age Older women are more likely to have high-grade disease,
at death of 70 (28%, age 6574; 24%, age 7584; and 15%, poor histology, and advanced disease; require adjuvant
age . 84). There are a number of explanations that have treatment; and experience disease recurrence. Gayar et al5
been postulated for the increased cancer-specific death rate considered 121 patients age 75 and older with apparent

From the Division of Gynecologic Oncology, Obstetrics and Gynecology, University of Virginia School of Medicine, Charlottesville, VA; Memorial Sloan Kettering Cancer Center, Weill
Cornell Medicine, New York, NY; Department of Clinical Oncology, Barts Health NHS Trust, St. Bartholomews Hospital, London, United Kingdom.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Linda Duska, MD, MPH, University of Virginia School of Medicine, P.O. Box 800712, Charlottesville, VA 22903; email: lduska@virginia.edu.

2016 by American Society of Clinical Oncology.

164 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


MULTIDISCIPLINARY CARE OF OLDER PATIENTS WITH ENDOMETRIAL CANCER

early-stage disease, specifically considering tumor re- We will discuss the holistic approach toward older patients
currence and survival. When compared with patients with cancer (as a part of person-centered care), the role and
younger than age 75, the older patients had higher Fed- outcome of surgery in older patients with endometrial
eration Internationale de Gynecologie et dObstetrique cancer, radiation therapy considerations in these patients,
grade, higher stage tumors, more deep myometrial invasion, and, finally, how CGA can be a useful tool for risk stratifi-
and more lower uterine segment involvement. Tumor cation, management, and predicting outcomes of older
recurrence rates were significantly higher in the older patients with cancer across the cancer care continuum.
patients (15% vs. 7%) and 5-year disease-specific survival Although a holistic approach such as the one described
was 91% versus 96%. Interestingly, in this study, age alone below would likely benefit all patients considering cancer
was not found to be a specific independent predictor of care, it will clearly benefit the geriatric patient who may
recurrence or disease-specific survival. However, other suffer untoward effects of an overly aggressive treatment
studies have demonstrated that cancer-specific mortal- approach.
ity is higher in older women.6 In the robotic study from
Vaknin et al7 (in which all patients underwent hysterec-
tomy and pelvic node dissection), the elderly group of
HOLISTIC APPROACH TO OLDER PATIENTS WITH
patients (age 70 and older) had a higher rate of advanced
CANCER
Person-centered care is a novel model of care that places
disease (39% vs. 18.7%; p = .04); of the eight patients in
emphasis on the patient as a whole rather than just a
the series who had positive nodes, six were older than
biomedical approach.8 Some of the major principles and
age 80.
values of person-centered care include caring for the whole
The limited available data suggest that age alone should
person through a holistic approach, developing an individ-
not be a sole decision factor in prescribing treatment,
ualized model of care, and understanding the patients
surgical and otherwise, for older women with endometrial
psychologic, social, and cultural complexity.9 To reach these
cancer. Instead, older patients with cancer should be
goals, person-centered care proposes to develop and en-
assessed for their fitness to receive any type of endometrial
hance the connection between medical care and supportive
cancer treatments. For older patients with (or without)
services and to provide multidisciplinary and team-based
cancer, a comprehensive geriatric assessment (CGA) can
care.
evaluate their fitness for treatment in much more detail. The
Older patients with cancer will benefit significantly from
model proposes that older patients (regardless of their age)
person-centered care as a result of the increased complexity
who are fit should be offered standard-of-care treatment of
of their care, their unmet needs, and the aging-related is-
their endometrial cancer, whereas those who are frail re-
sues. The approach begins with CGA. Over the past 2 de-
quire more extensive discussion about the risks of standard
cades, investigators have developed CGA specific to the
cancer treatment. Involvement of the patients primary care
cancer setting. Although there is no one standard instrument
provider and/or geriatrician may assist in obtaining a better
to perform CGA,10 it is important for cancer care providers to
understanding of both her overall fitness for treatment and
be familiar with the components of CGA (Fig. 1) and some of
her goals of care.
the more common instruments, the likelihood of patients
In this article, we will discuss the approach to the older
having abnormal findings, the practical implications of such
woman with endometrial cancer, taking into account the
findings, and the proper solutions (Table 1).
complexities of caring for a geriatric population with cancer.

SURGERY IN THE OLDER WOMAN WITH


ENDOMETRIAL CANCER: SAFETY AND
KEY POINTS FEASIBILITY
The goals of any medical or surgical intervention in the older
Endometrial cancer is the most common gynecologic age group should be to maintain or maximize the potential
cancer among older patients with cancer. life span, maximize independent function, relieve suffering,
Older women with endometrial cancer are more likely and maintain dignity. Although most modern series suggest
to be diagnosed with poor prognosis disease (higher that surgery is feasible in the older woman, it is not without
stage or high-risk histology) and more likely to die of morbidity. The older patient with endometrial cancer,
disease compared with younger patients. variably defined with respect to age parameters, appears to
Older patients with endometrial cancer are less likely to suffer more postoperative morbidity than her younger
be offered surgical therapy, retroperitoneal node
counterpart, regardless of surgical approach. There is also
dissection, and adjuvant therapy.
Older patients should not be denied treatment (surgery
morbidity from general anesthesia, required for all mini-
or adjuvant therapy) based on age alone. mally invasive procedures (but potentially avoidable in the
Involving geriatricians and/or performing vaginal approach, which may be performed in some cases
comprehensive geriatric assessment may aid in under regional anesthesia). Physiologic changes associated
treatment decisionmaking. with increasing age must be considered, including reduced
ability to increase cardiac output, decreased capacity of the

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 165


DUSKA, SHAHROKNI, AND POWELL

FIGURE 1. Components of a Comprehensive Geriatric Assessment

respiratory system with reduced lung elastic recoil and in- rate was 13.1%, with the older group more likely to have
creased chest wall stiffness, decreased functional reserve, postoperative complications (23.1% vs. 8.5%). There were
and skin changes that may impact wound healing. These age- no postoperative deaths. In a logistic regression model, age
related physiologic changes along with the medical 75 or older, chronic lung disease, and the performance of
comorbidities that accompany aging must be carefully lymphadenectomy (discussed further below) correlated
considered when planning a surgical procedure. with higher probability of perioperative morbidity. The
In the relatively recent past, surgery for endometrial authors concluded that older patients should be offered
cancer was traditionally performed via laparotomy, allowing surgical treatment given the survival advantage associated
for complete removal of the uterus, cervix, and bilateral with surgery.
tubes and ovaries along with a complete retroperitoneal Other authors have also demonstrated the safety of
lymphadenectomy. With improvement in optics and hysterectomy with or without lymphadenectomy in the
instrumentation, a minimally invasive approach became older population of patients. These studies are discussed
more acceptable, and in fact became standard of care with further below. It is notable, however, that in most of these
the publication of LAP2, a Gynecology Oncology Group studies, the perioperative complication rate for the older
(GOG) study that compared oncologic outcomes between group of patients was higher than that of the younger group.
laparotomy and laparoscopy in women with endometrial Perhaps more importantly, by their retrospective nature and
cancer.11 The approval of the robotic system in the United study design, these studies only report those women who
States in 2005, with its increased magnification, three- were felt to be good surgical candidates; they do not provide
dimensional optics, and wristed instruments, resulted in information regarding patients who were not offered sur-
an increased percentage of endometrial cancer procedures gery as a primary approach to cancer treatment.
performed via a minimally invasive approach. Finally, the
American Congress of Obstetrics and Gynecology has en- Laparoscopy
dorsed the vaginal approach as the optimal approach for Laparoscopy is the preferred surgical technique to lapa-
benign hysterectomy; although this method does not always rotomy for endometrial cancer for multiple reasons, in-
allow removal of the tubes and ovaries and never allows cluding shorter hospital stay, shorter recovery period, less
assessment of the retroperitoneal lymph nodes, it may be an discomfort (and therefore less use of postoperative narcotic
appropriate alternative for a patient who is too frail for an medications), and potentially improved quality of life. In
abdominal operation and general anesthesia. contrast to these benefits, laparoscopy requires longer
Several retrospective studies have addressed the safety of operating room (OR) time and steep Trendelenburg posi-
surgery for endometrial cancer in the older woman. DeMarzi tioning, aspects of surgery that may be particularly difficult
et al12 analyzed operability, perioperative morbidity, and for the older patient.
mortality in 124 women age 65 and older who were un- The LAP2 study was the first randomized study in the
dergoing either open laparotomy or vaginal approach for United States to compare open laparotomy to laparoscopy
hysterectomy. For statistical purposes, the authors divided for the surgical treatment of endometrial cancer and
the patients into younger than age 75 and 75 or older. Not was a major impetus for the transition to minimally invasive
surprisingly, the vaginal approach was used more often in surgery for endometrial cancer in the United States.11 The
the older patient group. Overall perioperative complication study was sponsored by the GOG, and patients were enrolled

166 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


MULTIDISCIPLINARY CARE OF OLDER PATIENTS WITH ENDOMETRIAL CANCER

TABLE 1. Instruments and Practice Implication of Comprehensive Geriatric Assessment Components


Likelihood of
Component Instrument Description of Instrument Abnormality Practical Use Solution
Functional Activities of Daily Assesses patients ability to do bathing, dressing, 15%20% Could lead to malnutrition, dif- Assessing caregiver ability to
Activity Living grooming, feeding, walking, and bladder and ficulty to present to health take care of patients
bowel control care provider office needs
Involving social worker for
further support
Instrumental Activi- Assesses patients ability for telephone use, doing Up to 50%52 Could lead to malnutrition, lack Assessing caregiver ability to
ties of Daily Living laundry, shopping, preparing meals, doing of compliance with medica- take care of patients
housework, handling own medications, han- tions, difficulty to present to needs
dling money and finances, and transportation health care provider office
Involving social worker for
further support
Falls Number of falls in the past 6 or 12 months Up to 25%50 May lead to bone fracture, Referral to physical therapy
leading to interruption in Further exploration of the
cancer care context of falls
May need to avoid neurotoxic Assessment of patients
chemotherapy agents if falls medication list for
are a result of neuropathy sedatives
Use of assistive Not applicable Up to 25%53 May be suggestive of patients Referral to physical therapy
device history of falls, imbalance, or
fear of falling
Sensory deficit Observation, physical exam Hearing deficit Could lead to decline in quality of Referral to audiologist and/or
(hearing, vision) $ 33%54 life and cognitive function; ophthalmologist
patient may not hear the
medical instructions properly
May need to avoid ototoxic
chemotherapy agents
Emotional Geriatric Depression It has 30-, 15-, and four-item versions 10%65%57 Patients with depression could Explore the reasons for
Well-being Scale55,56 be at higher risk for receiving depression
(Depression) nondefinitive treatment and
worse overall survival58,59
Hospital Anxiety and Seven items related to depression and seven It increases the risk of being Refer to psychologist/
Depression items related to anxiety nonadherent to medical psychiatrist
Scale62 treatment60
Patient Health It has nine-, four-, and two-item versions Could be suggestive to multiple If depression is result of lack
Questionnaire63,64 other geriatrics deficits61 of proper social support,
refer to social worker
Emotional Distress Patient rates his/her distress level over the past About 40%66 It is usually suggestive of unmet More and better patient-
Well-being Thermometer65 two weeks from 010. Score of $ 4 is sug- needs66 physician communication
(Distress) gestive of high level of distress. could lower the distress
level67
Referral to social worker,
mental health service, and/
or chaplaincy service68
Social Support Medical Outcome Available in 19-, 12-, and four-item versions The exact likeli- Could be associated with more Referral to social worker
Study-Social Sup- hood of ab- psychosomatic symptoms72,73 might be needed
The 19- and four-item versions measure four
port Survey6971 normality is
social support domains: emotional, tangible,
unknown
affectionate, and positive social interaction.
Multidimensional 12-item questionnaire that categorizes patients Could be associated with worse Empower the limited but
Scale of Perceived social support into support he/she receives overall survival74,75 available social support for
76
Social Support from family, friends, and significant other the patient
Nutritional Mini-Nutritional Available in six-item questionnaire with the score Up to 65% could It could be multifactorial (dis- Referral to nutritionist
Status Assessment77 of 014 be at risk for ease progression, fatigue, al-
malnutrition78 tered taste, inability to
prepare meals, etc.)
Score of 811 is suggestive of being at risk for Patients with malnutrition have Explore other reasons for
malnutrition, and score of # 7 is suggestive of worse overall survival79 malnutrition besides can-
malnutrition. The longer version includes cer progression and/or
additional questions and the score ranges from side effects from
030. Score of 1723.5 is suggestive of being chemotherapy
at risk for malnutrition, and score of # 17 is
suggestive of being malnourished.

Continued

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 167


DUSKA, SHAHROKNI, AND POWELL

TABLE 1. Instruments and Practice Implication of Comprehensive Geriatric Assessment Components (Contd)
Likelihood of
Component Instrument Description of Instrument Abnormality Practical Use Solution
Polypharmacy/ Beers criteria80 List of medications that could harmful for older Could be as Could lead to prolonged Referral to pharmacist
Potentially patients. The recommendations for harmful high as 50%81 hospitalization82
Inappropriate medications are categorized into avoid, and
Medication use with caution.
Polypharmacy Taking $ 5 medications is considered Could lead to drug-drug inter- Review medication list
polypharmacy83 action, noncompliance, and
increase in cost51
Taking . 10 is considered excessive Discontinue nonessential
polypharmacy47 medications, especially in
patients with limited life
expectancy
Check for drug-drug
interaction
Cognitive Mini-Cog84 Short instrument that takes about 2 to 3 minutes. About 20%,78 Patients could be at higher risk Referral to geriatrician, neu-
Status It includes three-word recall and clock but it varies by for delirium during hospital rologist, psychiatrist, or
drawing. age group stay43,85 neuropsychologist
Mini-Mental Status Administration of the test takes longer. It as-
Examination sesses cognitive domains of orientation, reg-
(MMSE)86 istration, attention and calculation, recall and
language.
Montreal Cognitive Administration of the test takes longer than
Assessment87 MMSE. It assesses visuospatial, naming,
memory, attention, language, abstraction,
delayed recall, and orientation of patients.
Blessed Orientation- Takes 10 minutes to administer. It has 26 ques-
Memory-Concen- tions and tests orientation, long-term mem-
tration test88 ory, recall, and concentration.
Comorbidity Charlson Comorbid- It is a method of categorizing comorbidities (a Varies by age Impacts overall survival90 Referral to primary care pro-
ity Index89 total of 22 conditions) based on the Interna- vider/geriatrician for opti-
tional Classification of Diseases, which can mizing comorbid condition
predict the 10-year mortality management
Adult Comorbidity It is a validated chart-based comorbidity It may negatively impact com-
Evaluation 2793 instrument pliance with cancer
treatment91
Each individual comorbid condi-
tion may have negative out-
come on survival92

from multiple sites across the United States. Notably, entry however, that in the study from Bogani et al,15 lymphade-
into LAP2 required a laparoscopic lymphadenectomy as part nectomy was not generally performed in the oldest patients.
of the surgical staging procedure. Thirty-one percent of In contrast, in the retrospective review of women age 65 and
patients in this study were age 70 or older, and 6.6% were older from Oklahoma, a more aggressive approach to
80 or older. Conversion to laparotomy occurred in 25.8% lymphadenectomy was undertaken. Sixty-seven women
of patients. Failure to successfully complete laparoscopy underwent either laparoscopic hysterectomy or lapa-
was greater with increasing age (odds ratio 1.27; 95% CI, roscopically assisted vaginal hysterectomy with lymphade-
1.141.42 for a 10-year increase in age), but the reasons for nectomy. Laparoscopy was successful in 78% percent of
this finding are not clear. Additionally, operating time was cases, and a complete node dissection was successfully
significantly longer in the laparoscopy arm (204 vs. 103 performed in all patients. Of note, the laparoscopic group
minutes). The longer operating time is significant in the older had a longer OR time than the comparison laparotomy group
age group, in part because longer duration of anesthesia may (236 vs. 148 minutes). Despite the longer OR time, the
be associated with postoperative delirium, particularly in authors note there was no subsequent increase in morbidity
patients with a pre-existing cognitive defect. There was and mortality.16
statistically significantly better quality of life across many
parameters in the laparoscopy arm at 6 weeks, but age was Robotic Surgery
not specifically considered as a factor for quality of life.13 Robotic surgery, approved in the United States in 2005,
Multiple single-institution studies have suggested that a provides advantages over traditional laparoscopy, but there
laparoscopic approach is safe in the older patient. Two are also patient-related drawbacks: in particular, once the
Italian studies found that laparoscopy was safe and feasible robot is docked, the patient must remain in steep Tren-
and associated with lower blood loss, shorter hospital stay, delenburg position. Prolonged Trendelenburg poten-
and lower complication rates. 14,15 It should be noted, tially increases the potential risk of blindness in patients

168 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


MULTIDISCIPLINARY CARE OF OLDER PATIENTS WITH ENDOMETRIAL CANCER

suffering from moderate- or high-pressure glaucoma17 and cancer-specific death when cancer-directed surgery was
may particularly impact the older patient with respect to undertaken within each age category, adjusted for radiation
ability to ventilate and cardiac output. therapy, histology, and stage. Rauh-Hain et al23 performed a
Despite these concerns, several series suggest that ro- similar analysis using the national cancer database, although
botic surgery is safe and effective in the older patient, albeit this group only considered women with advanced stage
with a possible increased perioperative morbidity.1820 The (stage III or IV) disease and also demonstrated that elderly
series from Guy et al, 18 for example, demonstrated in- women were less likely to be treated with surgery. Patients
creased rates of perioperative surgical and medical com- younger than age 55 had surgery more frequently compared
plications in the older age group with robotic surgery when with patients age 75 to 84 (97.2% compared with 95.8%;
compared with younger patients with the same procedure. p , .001) and age 85 or older (97.2% compared with 94.8%;
Specifically, older patients had higher rates of periop- p , .001) and a higher rate of lymph node dissection (78.7%
erative surgical (8.3% vs. 5.2%; p , .001) and medical compared with 70.5%, p , .001; and 78.7% compared with
(12.3% vs. 6.7%; p , .001) complications, longer length of 59.5%, p , .001, respectively). In a multivariate analysis,
stay (2.00 vs. 1.67 days; p , .001), and lower rates of which took into account medical comorbidities as well as
discharge to home (88.8% vs. 96.8%; p , .001). As age adjuvant therapy, older women also had a higher risk of
increased, perioperative surgical and medical complica- death when compared with women younger than age 55
tions also increased in a linear fashion. In contrast, in the (women age 7584, HR 2.38; 95% CI, 2.142.65; women age
series of 41 women 70 and older from the series by Vaknin 85 or older, HR 3.16; 95% CI, 2.763.61).
et al,7 older and younger patients had the same rate of In contrast to the population-based studies above, most
postoperative complications. It is clear, however, that the single-institution studies from tertiary care centers, re-
robotic approach is preferable to the open approach in the ported in large part by gynecologic oncologists, suggest that
older population. When compared directly to laparotomy, all patients should be offered surgery, regardless of age.
the robotic approach was preferable for women age 70 or These types of retrospective reviews represent a selection
older, with a decrease in complication rates, surgical blood bias of sorts and by study design do not account for those
loss, and hospital stay.19 women who were never offered surgery. They do suggest,
however, that gynecologic oncologists may be more likely to
Lymphadenectomy offer surgery to older women with endometrial cancer than
The role of lymphadenectomy in endometrial cancer con- other providers.
tinues to be one of some controversy, particularly in the It is also important to note that database studies by their
older patient. There is no doubt that adding lymph node very nature cannot provide us with data regarding medical
dissection will increase operating time as well as the po- comorbidities affecting the elderly or the desires of the
tential for perioperative morbidity. In the series by de Marzi patient and her family regarding goals of care. In the study by
et al,12 performance of lymphadenectomy significantly in- de Marzi et al,12 for example, three women in their late 80s
creased the rate of postoperative complications, but other declined surgery; this patient decision to decline an offered
studies suggest that lymphadenectomy is safe in the older surgery would not be captured in a population-based da-
patient.16,20 Although all patients in the study by Scribner tabase study. Nevertheless, the studies point to an alarming
et al16 were staged, this practice led to a high conversion rate trend, confirming the national bias toward not offering
to laparotomy and increased OR time. Other studies have surgery to the older patient with endometrial cancer, re-
confirmed that staging procedures in elderly patients are gardless of fitness.
associated with a significantly longer operating time.21 Both population-based studies and institutional studies
Further studies are needed to weigh the benefits of lym- suggest an age bias regarding lymph node dissection. In the
phadenectomy on overall survival versus the potential SEER study from Wright et al,6 older women were less likely
complications in this group of patients. to have lymph node dissection performed when compared
with women age 65 to 69 (43.5% for women age 6569,
Surgery for the Older Patient With Cancer: Is There a 37.5% for age 8084, and 24.8% for women 85 and older).
Bias? Lowery et al20 reported similar results in their SEER analysis,
Population database studies suggest that there is a selection with lymph node dissection performed in 41% of the age 80
bias with respect to performing surgery for older patients, or older population. Institution-based studies from Europe
both for hysterectomy and lymph node dissection. With reveal similar results. For example, in the study by de Marzi
respect to hysterectomy specifically, two different SEER et al,12 lymphadenectomy was performed more often in
analyses demonstrated that older patients with endometrial younger patients (84.5% in women younger than 75 vs.
cancer were less likely to have a hysterectomy.6,22 In the 15.5% in women 75 and older). Bogani et al15 noted that we
study from Ahmed et al,22 a survival analysis was done within usually omit execution of retroperitoneal staging in this
each age group to compare the endometrial cancer-specific group. In contrast, single-institution studies from the
survival rate of those who received cancer-directed surgery United States report higher node staging rates in older
(hysterectomy at a minimum) to those who did not. They patients. This aggressive approach to lymphadenectomy,
found a significant reduction in the hazard ratio (HR) for however, is not without morbidity.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 169


DUSKA, SHAHROKNI, AND POWELL

In summary, surgery should be considered as a first step increased risk of second malignancy cannot be ignored
for an older patient, if that patient is deemed medically fit. A and is of particular significance when even older patients
minimally invasive approach appears to be safe and offers may be expected to have a life expectancy exceeding
the best postoperative recovery despite longer operating 10 years.
times. The role for complete lymphadenectomy is still A newer technique that provides similar highly conformal
controversial, particularly in the population of older women, radiation fields uses a continuous rotation of the radiation
and should be individualized; however, the plans for adju- source through 360 degrees. The advantage of this arc
vant therapy should also be considered when planning the technique is twofold. First, the speed of delivery is much
surgical procedure. faster compared with IMRT. A typical pelvic field of two full
arcs might be delivered in less than 5 minutes. This is more
comfortable for patients, particularly the elderly, who
RADIOTHERAPY FOR ENDOMETRIAL CANCER IN sometimes find it difficult to hold a full bladder. It may
THE OLDER PATIENT thereby reduce the potential for inaccuracy as a result of
Radiotherapy remains an essential component of the movement and also allow faster throughput in the working
treatment of both early and locally advanced endometrial day. Second, the number of monitor units required to deliver
cancer. Over the past decade, data from large randomized arc treatment is approximately two-thirds that of IMRT,
controlled trials have established the place of radiation in leading to a lower total dose delivered to the patient.
adjuvant treatment of uterine cancers. Risk groups based on Pelvic insufficiency fractures occur when the elastic re-
clinicopathological factors have been devised to identify sistance of bone is reduced and unable to withstand the
those patients who might benefit most from additional stress of weight bearing and are most frequent in older
treatment (Table 2). In the older patient, in whom age itself women with postmenopausal osteoporosis. Pelvic radiation
is a risk factor, the use of pelvic radiotherapy needs to be is also a recognized cause of pelvic insufficiency fractures
considered carefully, balancing the advantage of high local and, although not always symptomatic, may give rise to
control rates with potential toxicity and lack of survival significant pain. VMAT and IMRT, with careful planning to
benefit.24,25 avoid areas such as the sacrum, may reduce the incidence of
Although initially slow to integrate new three-dimensional fractures, which is estimated to occur in up to 50% of pa-
radiotherapy planning techniques into routine treatment, tients who undergo pelvic radiotherapy.29,30
the gynecology oncology community has now embraced the The integration of three-dimensional imaging into radio-
technology, and intensity modulated radiotherapy (IMRT), therapy and the use of intensity modulated radiation to
volumetric modulated arc therapy (VMAT), and image- produce radiation treatment plans that maximize dose to
guided radiotherapy are in widespread use. IMRT uses tumor while minimizing dose to normal tissues has been a
multiple beams divided into smaller beamlets of varying major step forward in radiotherapy. But it has also revealed
dose intensity, allowing more accurate shaping of the de- the uncertainties that exist in affirming that we are actually
livered dose to the target. This technique has been shown in treating what was intended. With shaping of fields and the
theoretical planning studies to reduce the dose to nearby steep dose gradients around target, it has become more
organs such as bowel and bladder by up to 60%26 and important than ever to avoid a geographical miss of the
translates to a reduction in both acute and late radiation target either by not correctly identifying the target or
toxicity.27,28 The reduction in toxicity is likely to be especially through internal organ movement. Delivering this highly
beneficial in an elderly population who have a higher in- focused radiotherapy places the onus on the radiation on-
cidence of underlying bowel and bladder problems with cologist to ensure accurate delineation of the radiation
urgency and incontinence often present. target. Courses and online training aids have been de-
One of the downsides of IMRT is the high number of veloped, and atlases, comprising serial axial CT or MRIs with
monitor units needed to deliver the plan compared with anatomy clearly labeled, are now widely available to help the
old-fashioned conventional treatment (estimated to be oncologist.31,32 In particular, variation in bladder filling and
two to three times higher), which leads to an increase in rectal dimensions, as a result of either gas or feces, may
lower dose radiation to other parts of the body. The have a significant impact on the position of the vagina and, if
long-term effect of IMRT is not known, but the potential present, the uterus and cervix. In elderly patients in whom

TABLE 2. Suggested Radiotherapy by Risk Group

Low Risk Intermediate Risk High Risk


IA G1, G2, 1B G1 without LVSI 1A G3, IBG1 or 2, with LVSI 1A G3 LVSI, IB G3, II, III
No treatment94 Vault brachytherapy95 Pelvic radiotherapy94,96
Risk of relapse is 10% Brachytherapy will decease vaginal recurrence rate to 2%
Survival exceeds 95%
Abbreviation: LVSI, lymphovascular space invasion.

170 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


MULTIDISCIPLINARY CARE OF OLDER PATIENTS WITH ENDOMETRIAL CANCER

constipation and or bladder weakness is often an issue, THE UTILITY OF COMPREHENSIVE GERIATRIC
careful consideration is needed as to how best to ensure ASSESSMENT ACROSS ENDOMETRIAL CANCER
consistency in rectal and bladder dimensions on a day-to-day CONTINUUM
basis. Daily use of enemas, which is advocated by many, is An approach toward whole care for the older patient with
not always appropriate for older patients who may find it cancer is critical for delivering best quality of care. Data on
physically challenging to administer the enema and manage older patients with endometrial cancer are just emerging;
the result. For all patients undergoing pelvic radiotherapy, however, data on older patients with various cancers in
whether old or young, choosing an appropriate margin different phases of their treatment prove the importance
around the clinical target volume has become as critical to of such an approach. Although health care providers rely
success as actually delineating the original target and must on standard methods of assessing functional status (e.g.,
be sufficient to avoid missing the target but not so large as to Karnofsky performance status or American Society of An-
lose the advantage of reducing the volume of normal tissue esthesiologists classification), CGA has been shown to be
irradiated. complementary to standard assessment.38 Shared decision-
In the past, simple kilovoltage imaging was the mainstay of making among patients, families, and health care providers
treatment verification. Its major limitation is that it only may be impacted in up to 40% of cases as a result of new
displays bony anatomy. Although this is helpful in providing a information through performing CGA. Comprehensive ge-
rapid check that the patient is correctly aligned, it gives no
riatric assessment can predict surgical complications, length
indication as to how the target is positioned relative to the
of stay, and hospital disposition following surgery, hospital
radiation field. More sophisticated on-board CT imaging
readmission, and overall survival.3941 Older patients with
features almost all modern linear accelerators, allowing real-
cancer with cognitive deficits could be especially at high risk
time visualization of not just bony landmarks but also other
for delirium, which can lead to prolonged hospital stay and
internal structures and even tumors.
possibly poorer overall survival.40,42,43 Despite these data,
Many studies have been published looking at movement
only 6.4% of surgeons use CGA in their preoperative eval-
of pelvic organs during a course of radiotherapy and
uation, and only one out of three surgeons collaborates with
guidelines suggested on suitable margins to account for day-
geriatricians to optimize care of older patients with cancer
to-day change in bladder and rectal filling.33,34 These mar-
preoperatively.44
gins, which are based on population data, are necessarily
Various models of collaborative care between geriatricians
generous, as allowance must be made for the largest move-
and/or geriatric-allied health care professional and surgical
ment possible. So, although they are a practical way of ensur-
ing target coverage, they potentially lead to larger treatment services have been proposed.45 Although the effectiveness
volumes and in particular more normal tissue likely to be and sustainability of these models must be further exam-
irradiated.35 ined, the importance of performing CGA in the preoperative
Daily individualized image-guided adaptive radiotherapy is evaluation and acting proactively based on findings by in-
the best way to ensure both accuracy of delivery and re- corporating different supportive services (e.g., physical
duction of total volume irradiated. This has been made therapy, nutrition) is clear. As a result of these findings, the
possible with the ready availability of cone beam CT scanning American College of Surgeons and the American Geriatrics
on linear accelerators, allowing a CT scan of the radiation Society have issued a best practice guideline to incorporate
field to be obtained in just a few minutes immediately before geriatric assessment in the preoperative assessment of older
radiation that can then be merged and compared with the patients undergoing surgery to fully assess older patients for
original planning scan. Daily variation in organ movement their fitness to undergo surgery.46
can be partly accounted for by creating a so-called library of For older patients with recurrent or metastatic endome-
plans using differing margins around the target. Each day a trial cancer in need of chemotherapy, CGA has significant
plan can be selected that is the best fit. This has been used value. Two major chemotherapy toxicity calculators (Cancer
with some success in bladder cancer radiotherapy.36,37 and Aging Research Group47 and the Chemotherapy Risk
Taking a daily cone beam CT with real-time review and Assessment for High-Age Patients48) have developed based
selecting a suitable plan on every patient requires time and on the concept of CGA. Although these two calculators can
highly trained individuals. Most busy radiotherapy depart- be useful for oncologists, patients, and their families to
ments are short of one or both and cannot offer this approach understand the risk of chemotherapy toxicity in older pa-
for all. tients with cancer, the exact intervention based on this
Older age confers an additional risk factor in uterine finding is not clear. One approach would be to modify the
cancer, and careful consideration should be given to offering chemotherapy regimen for a patient at high risk for de-
adjuvant treatment to this group. Modern radiation tech- veloping chemotherapy toxicity; however, another ap-
niques have given us treatment that is better tolerated with proach could be to involve and empower the primary care
improved delivery with significantly less severe acute and provider/geriatrician to monitor these patients more in-
late toxicity. These techniques combined with good medical, tensively for early and accurate detection of chemotherapy
nursing, and psychosocial support should mean that age toxicity and acting promptly and adequately to manage
need not necessarily be a barrier to radiation. these toxicities. Although these calculators are useful to give a

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 171


DUSKA, SHAHROKNI, AND POWELL

broad overview of patients risk for chemotherapy toxicities, it to a pharmacist and/or primary care provider/geriatrician to
is important to note any geriatrics deficit could be associated reassess the patients medication list and to discontinue
with adverse outcomes and needs proper intervention.47,49 medications that are not absolutely necessarily could in-
Moreover, CGA can help with selecting the proper che- crease the compliance rate of taking hormonal therapies for
motherapy agent. For example, selecting a neurotoxic endometrial cancer. Patients with impaired cognitive status
chemotherapy agent for treatment of metastatic endome- with no reliable social support may not be the best candi-
trial cancer when the patient has fallen multiple times as a dates for oral treatments. Like preoperative care, life ex-
result of neuropathy secondary to poorly controlled di- pectancy of older patients with recurrent or metastatic
abetes may not be in the best interest of an older patient endometrial cancer should be assessed, and the impact of
with metastatic endometrial cancer. The need for in- cancer treatment on both longevity and quality of life should
corporating CGA in daily oncology practice becomes much be discussed with the patient and his/her family.
more important when one study showed that only 10% of In summary, for older patients with cancer in general and
patients who reported falls during CGA (performed as a in older patients with endometrial cancer in particular, a
research tool) had a documented history of falls in their whole-patient approach should be part of standard of care.
medical chart by medical oncologists.50 Comprehensive geriatric assessment is a useful tool for such
For older patients with endometrial cancer who require an approach, and deficits found through CGA can be managed
oral hormone therapy, the issue of polypharmacy needs to by involving various supportive services as well as empow-
be addressed, as polypharmacy could be associated with ering primary care providers/geriatricians in comanagement
lower compliance rate and drug-drug interaction.51 Referral of these patients.

References
1. National Cancer Institute. SEER Stat Fact Sheets: Endometrial Cancer. http:// 13. Kornblith AB, Huang HQ, Walker JL, et al. Quality of life of patients
seer.cancer.gov/statfacts/html/corp.html. Accessed January 22, 2016. with endometrial cancer undergoing laparoscopic international
2. United Nations. World Populating Ageing 1950-2050. http://www.un.org/ federation of gynecology and obstetrics staging compared with
esa/population/publications/worldageing19502050/pdf/90chapteriv.pdf. laparotomy: a Gynecologic Oncology Group study. J Clin Oncol.
Accessed January 22, 2016. 2009;27:5337-5342.
3. Arnold M, Karim-Kos HE, Coebergh JW, et al. Recent trends in incidence 14. Ciavattini A, Di Giuseppe J, Cecchi S, et al. Gynecologic laparoscopy
of five common cancers in 26 European countries since 1988: Analysis of in patients aged 65 or more: feasibility and safety in the presence of
the European Cancer Observatory. Eur J Cancer. 2015;51:1164-1187. increased comorbidity. Eur J Obstet Gynecol Reprod Biol. 2014;175:
4. Mariani A, Webb MJ, Keeney GL, et al. Low-risk corpus cancer: is 49-53.
lymphadenectomy or radiotherapy necessary? Am J Obstet Gynecol. 15. Bogani G, Cromi A, Uccella S, et al. Perioperative and long-term out-
2000;182:1506-1519. comes of laparoscopic, open abdominal, and vaginal surgery for en-
5. Gayar OH, Robbins JR, Parikh K, et al. Hysterectomy for uterine ade- dometrial cancer in patients aged 80 years or older. Int J Gynecol Cancer.
nocarcinoma in the elderly: tumor characteristics, and long-term 2014;24:894-900.
outcome. Gynecol Oncol. 2011;123:71-75. 16. Scribner DR Jr, Walker JL, Johnson GA, et al. Surgical management of
6. Wright JD, Lewin SN, Barrena Medel NI, et al. Endometrial cancer in the early-stage endometrial cancer in the elderly: is laparoscopy feasible?
oldest old: Tumor characteristics, patterns of care, and outcome. Gynecol Oncol. 2001;83:563-568.
Gynecol Oncol. 2011;122:69-74. 17. Rupp-Montpetit K, Moody ML. Visual loss as a complication of non-
7. Vaknin Z, Perri T, Lau S, et al. Outcome and quality of life in a ophthalmic surgery: a review of the literature. Insight. 2005;30:10-17.
prospective cohort of the first 100 robotic surgeries for endometrial 18. Guy MS, Sheeder J, Behbakht K, et al. Comparative outcomes in older
cancer, with focus on elderly patients. Int J Gynecol Cancer. 2010;20: and younger women undergoing laparotomy or robotic surgical staging
1367-1373. for endometrial cancer. Am J Obstet Gynecol. Epub 2015 Nov 17.
8. Laine C, Davidoff F. Patient-centered medicine. A professional evolu- 19. Lavoue V, Zeng X, Lau S, et al. Impact of robotics on the outcome of
tion. JAMA. 1996;275:152-156. elderly patients with endometrial cancer. Gynecol Oncol. 2014;133:
9. Kogan AC, Wilber K, Mosqueda L. Person-centered care for older adults 556-562.
with chronic conditions and functional impairment: a systematic lit- 20. Lowery WJ, Gehrig PA, Ko E, et al. Surgical staging for endometrial
erature review. J Am Geriatr Soc. 2016;64:e1-e7. cancer in the elderly - is there a role for lymphadenectomy? Gynecol
10. Wildiers H, Heeren P, Puts M, et al. International Society of Geriatric Oncol. 2012;126:12-15.
Oncology consensus on geriatric assessment in older patients with 21. Giannice R, Susini T, Ferrandina G, et al. Systematic pelvic and aortic
cancer. J Clin Oncol. 2014;32:2595-2603. lymphadenectomy in elderly gynecologic oncologic patients. Cancer.
11. Walker JL, Piedmonte MR, Spirtos NM, et al. Laparoscopy compared 2001;92:2562-2568.
with laparotomy for comprehensive surgical staging of uterine cancer: 22. Ahmed A, Zamba G, DeGeest K, et al. The impact of surgery on survival of
Gynecologic Oncology Group Study LAP2. J Clin Oncol. 2009;27: elderly women with endometrial cancer in the SEER program from
5331-5336. 1992-2002. Gynecol Oncol. 2008;111:35-40.
12. De Marzi P, Ottolina J, Mangili G, et al. Surgical treatment of elderly 23. Rauh-Hain JA, Pepin KJ, Meyer LA, et al. Management for elderly women
patients with endometrial cancer ($ 65 years). J Geriatr Oncol. 2013;4: with advanced-stage, high-grade endometrial cancer. Obstet Gynecol.
368-373. 2015;126:1198-1206.

172 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


MULTIDISCIPLINARY CARE OF OLDER PATIENTS WITH ENDOMETRIAL CANCER

24. Creutzberg CL, van Putten WL, Koper PC, et al. Surgery and post- 41. Badgwell B, Stanley J, Chang GJ, et al. Comprehensive geriatric as-
operative radiotherapy versus surgery alone for patients with stage-1 sessment of risk factors associated with adverse outcomes and resource
endometrial carcinoma: multicentre randomised trial. PORTEC Study utilization in cancer patients undergoing abdominal surgery. J Surg
Group. Post Operative Radiation Therapy in Endometrial Carcinoma. Oncol. 2013;108:182-186.
Lancet. 2000;355:1404-1411. 42. Robinson TN, Raeburn CD, Tran ZV, et al. Postoperative delirium in the
25. Keys HM, Roberts JA, Brunetto VL, et al; Gynecologic Oncology elderly: risk factors and outcomes. Ann Surg. 2009;249:173-178.
Group. A phase III trial of surgery with or without adjunctive ex- 43. Korc-Grodzicki B, Sun SW, Zhou Q, et al. Geriatric assessment as a
ternal pelvic radiation therapy in intermediate risk endometrial predictor of delirium and other outcomes in elderly patients with
adenocarcinoma: a Gynecologic Oncology Group study. Gynecol cancer. Ann Surg. 2015;261:1085-1090.
Oncol. 2004;92:744-751. 44. Ghignone F, van Leeuwen BL, Montroni I, et al. The assessment and
26. Taylor A, Powell ME. Conformal and intensity-modulated radiotherapy management of older cancer patients: A SIOG surgical task force survey
for cervical cancer. Clin Oncol (R Coll Radiol). 2008;20:417-425. on surgeons attitudes. Eur J Surg Oncol. 2016;42:297-302. Epub 2015
27. Mundt AJ, Mell LK, Roeske JC. Preliminary analysis of chronic gastroin- Dec 17.
testinal toxicity in gynecology patients treated with intensity-modulated 45. Dale W, Chow S, Sajid S. Socioeconomic considerations and shared-
whole pelvic radiation therapy. Int J Radiat Oncol Biol Phys. 2003;56: care models of cancer care for older adults. Clin Geriatr Med. 2016;32:
1354-1360. 35-44.
28. Mundt AJ, Lujan AE, Rotmensch J, et al. Intensity-modulated whole 46. Chow WB, Rosenthal RA, Merkow RP, et al; American College of Sur-
pelvic radiotherapy in women with gynecologic malignancies. Int J geons National Surgical Quality Improvement Program; American
Radiat Oncol Biol Phys. 2002;52:1330-1337. Geriatrics Society. Optimal preoperative assessment of the geriatric
29. Shih KK, Folkert MR, Kollmeier MA, et al. Pelvic insufficiency fractures surgical patient: a best practices guideline from the American College of
in patients with cervical and endometrial cancer treated with post- Surgeons National Surgical Quality Improvement Program and the
operative pelvic radiation. Gynecol Oncol. 2013;128:540-543. American Geriatrics Society. J Am Coll Surg. 2012;215:453-466.
30. Kwon JW, Huh SJ, Yoon YC, et al. Pelvic bone complications after ra- 47. Hurria A, Togawa K, Mohile SG, et al. Predicting chemotherapy toxicity
diation therapy of uterine cervical cancer: evaluation with MRI. AJR Am J in older adults with cancer: a prospective multicenter study. J Clin
Roentgenol. 2008;191:987-994. Oncol. 2011;29:3457-3465.
31. Small W Jr, Mell LK, Anderson P, et al. Consensus guidelines for de- 48. Extermann M, Boler I, Reich RR, et al. Predicting the risk of che-
lineation of clinical target volume for intensity-modulated pelvic ra- motherapy toxicity in older patients: the Chemotherapy Risk As-
diotherapy in postoperative treatment of endometrial and cervical sessment Scale for High-Age Patients (CRASH) score. Cancer. 2012;
cancer. Int J Radiat Oncol Biol Phys. 2008;71:428-434. 118:3377-3386.
32. Taylor A, Rockall AG, Powell ME. An atlas of the pelvic lymph node 49. Soubeyran P, Fonck M, Blanc-Bisson C, et al. Predictors of early death
regions to aid radiotherapy target volume definition. Clin Oncol (R Coll risk in older patients treated with first-line chemotherapy for cancer.
Radiol). 2007;19:542-550. J Clin Oncol. 2012;30:1829-1834.
33. Chan P, Dinniwell R, Haider MA, et al. Inter- and intrafractional tumor 50. Guerard EJ, Deal AM, Williams GR, et al. Falls in older adults with cancer:
and organ movement in patients with cervical cancer undergoing ra- evaluation by oncology providers. J Oncol Pract. 2015;11:470-474. Epub
diotherapy: a cinematic-MRI point-of-interest study. Int J Radiat Oncol 2015 Jul 14.
Biol Phys. 2008;70:1507-1515. 51. Balducci L, Goetz-Parten D, Steinman MA. Polypharmacy and
34. Taylor A, Powell ME. An assessment of interfractional uterine the management of the older cancer patient. Ann Oncol. 2013;
and cervical motion: implications for radiotherapy target volume 24:vii36-vii40.
definition in gynaecological cancer. Radiother Oncol. 2008;88: 52. Milla n-Calenti JC, Tubo J, Pita-Ferna ndez S, et al. Prevalence of
250-257. functional disability in activities of daily living (ADL), instrumental
35. Bondar ML, Hoogeman MS, Mens JW, et al. Individualized nonadap- activities of daily living (IADL) and associated factors, as predictors
tive and online-adaptive intensity-modulated radiotherapy treatment of morbidity and mortality. Arch Gerontol Geriatrics. 2010;50:
strategies for cervical cancer patients based on pretreatment acquired 306-310.
variable bladder filling computed tomography scans. Int J Radiat Oncol 53. Gell NM, Wallace RB, LaCroix AZ, et al. Mobility device use in older
Biol Phys. 2012;83:1617-1623. adults and incidence of falls and worry about falling: findings from the
36. Lutkenhaus LJ, Visser J, de Jong R, et al. Evaluation of delivered dose 2011-2012 national health and aging trends study. J Am Geriatr Soc.
for a clinical daily adaptive plan selection strategy for bladder cancer 2015;63:853-859.
radiotherapy. Radiother Oncol. 2015;116:51-56. 54. Walling AD, Dickson GM. Hearing loss in older adults. Am Fam Physician.
37. Foroudi F, Pham D, Rolfo A, et al. The outcome of a multi-centre 2012;85:1150-1156.
feasibility study of online adaptive radiotherapy for muscle-invasive 55. Burke WJ, Roccaforte WH, Wengel SP. The short form of the Geriatric
bladder cancer TROG 10.01 BOLART. Radiother Oncol. 2014;111: Depression Scale: a comparison with the 30-Item Form. J Geriatr
316-320. Psychiatry Neurol. 1991;4:173-178.
38. Repetto L, Fratino L, Audisio RA, et al. Comprehensive geriatric as- 56. Almeida OP, Almeida SA. Short versions of the geriatric depression
sessment adds information to Eastern Cooperative Oncology Group scale: a study of their validity for the diagnosis of a major depressive
performance status in elderly cancer patients: an Italian Group for episode according to ICD-10 and DSM-IV. Int J Geriatr Psychiatry. 1999;
Geriatric Oncology Study. J Clin Oncol. 2002;20:494-502. 14:858-865.
39. Schmidt M, Neuner B, Kindler A, et al. Prediction of long-term mortality 57. Weiss Wiesel TR, Nelson CJ, Tew WP, et al; Cancer Aging Research Group
by preoperative health-related quality-of-life in elderly onco-surgical (CARG). The relationship between age, anxiety, and depression in older
patients. PLoS One. 2014;9:e85456. adults with cancer. Psychooncology. 2015;24:712-717.
40. Large MC, Reichard C, Williams JT, et al. Incidence, risk factors, and 58. Goodwin JS, Zhang DD, Ostir GV. Effect of depression on diagnosis,
complications of postoperative delirium in elderly patients undergoing treatment, and survival of older women with breast cancer. J Am Geriatr
radical cystectomy. Urology. 2013;81:123-128. Soc. 2004;52:106-111.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 173


DUSKA, SHAHROKNI, AND POWELL

59. Prasad SM, Eggener SE, Lipsitz SR, et al. Effect of depression on di- 79. Aaldriks AA, van der Geest LGM, Giltay EJ, et al. Frailty and mal-
agnosis, treatment, and mortality of men with clinically localized nutrition predictive of mortality risk in older patients with advanced
prostate cancer. J Clin Oncol. 2014;32:2471-2478. colorectal cancer receiving chemotherapy. J Geriatr Oncol. 2013;4:
60. DiMatteo MR, Lepper HS, Croghan TW. Depression is a risk factor for 218-226.
noncompliance with medical treatment: meta-analysis of the effects of 80. American Geriatrics Society 2015 Beers Criteria Update Expert Panel.
anxiety and depression on patient adherence. Arch Intern Med. 2000; American Geriatrics Society 2015 Updated Beers Criteria for Potentially
160:2101-2107. Inappropriate Medication Use in Older Adults. J Am Geriatr Soc. 2015;
61. Canoui-Poitrine F, Reinald N, Laurent M, et al; ELCAPA Study Group. 63:2227-2246.
Geriatric assessment findings independently associated with clinical 81. Nightingale G, Hajjar E, Swartz K, et al. Evaluation of a pharmacist-
depression in 1092 older patients with cancer: the ELCAPA Cohort led medication assessment used to identify prevalence of and as-
Study. Psychooncology. 2016;25:104-111. sociations with polypharmacy and potentially inappropriate medication
62. Herrmann C. International experiences with the Hospital Anxiety and use among ambulatory senior adults with cancer. J Clin Oncol. 2015;33:
Depression Scalea review of validation data and clinical results. 1453-1459.
J Psychosom Res. 1997;42:17-41. 82. Park JW, Roh J-L, Lee SW, et al. Effect of polypharmacy and potentially
63. Kroenke K, Spitzer RL, Williams JB. The Patient Health Questionnaire-2: inappropriate medications on treatment and posttreatment courses in
validity of a two-item depression screener. Med Care. 2003;41: elderly patients with head and neck cancer. J Cancer Res Clin Oncol.
1284-1292. Epub 2016 Jan 7.
64. Kroenke K, Spitzer RL, Williams JB. The PHQ-9: validity of a brief de- 83. Turner JP, Jamsen KM, Shakib S, et al. Polypharmacy cut-points in older
pression severity measure. J Gen Intern Med. 2001;16:606-613. people with cancer: how many medications are too many? Support Care
65. Zwahlen D, Hagenbuch N, Carley MI, et al. Screening cancer patients Cancer. Epub 2015 Oct 9.
families with the distress thermometer (DT): a validation study. Psy- 84. Borson S, Scanlan J, Brush M, et al. The mini-cog: a cognitive vital signs
chooncology. 2008;17:959-966. measure for dementia screening in multi-lingual elderly. Int J Geriatr
66. Hurria A, Li D, Hansen K, et al. Distress in older patients with cancer. Psychiatry. 2000;15:1021-1027.
J Clin Oncol. 2009;27:4346-4351. 85. Korc-Grodzicki B, Root JC, Alici Y. Prevention of post-operative delirium
67. Zachariae R, Pedersen CG, Jensen AB, et al. Association of perceived in older patients with cancer undergoing surgery. J Geriatr Oncol. 2015;
physician communication style with patient satisfaction, distress, 6:60-69.
cancer-related self-efficacy, and perceived control over the disease. Br J 86. Folstein MF, Folstein SE, McHugh PR. Mini-mental state. A practical
Cancer. 2003;88:658-665. method for grading the cognitive state of patients for the clinician.
68. Holland JC, Andersen B, Breitbart WS, et al. Distress management. J Natl J Psychiatr Res. 1975;12:189-198.
Compr Canc Netw. 2010;8:448-485. 87. Nasreddine ZS, Phillips NA, Bedirian V, et al. The Montreal Cognitive
69. Moser A, Stuck AE, Silliman RA, et al. The eight-item modified Medical Assessment, MoCA: a brief screening tool for mild cognitive impair-
Outcomes Study Social Support Survey: psychometric evaluation ment. J Am Geriatr Soc. 2005;53:695-699.
showed excellent performance. J Clin Epidemiol. 2012;65:1107-1116. 88. Katzman R, Brown T, Fuld P, et al. Validation of a short Orientation-
70. Gjesfjeld CD, Greeno CG, Kim KH. A confirmatory factor analysis of an Memory-Concentration Test of cognitive impairment. Am J Psychiatry.
abbreviated social support instrument - The MOS-SSS. Res Soc Work 1983;140:734-739.
Pract. 2008;18:231-237. 89. Charlson M, Szatrowski TP, Peterson J, et al. Validation of a combined
71. Jyrkka J, Enlund H, Korhonen MJ, et al. Polypharmacy status as an comorbidity index. J Clin Epidemiol. 1994;47:1245-1251.
indicator of mortality in an elderly population. Drugs Aging. 2009;26: 90. Bje CR, Dalton SO, Grnborg TK, et al. The impact of comorbidity on
1039-1048. outcome in 12 623 Danish head and neck cancer patients: a pop-
72. Hughes S, Jaremka LM, Alfano CM, et al. Social support predicts in- ulation based study from the DAHANCA database. Acta Oncol. 2013;52:
flammation, pain, and depressive symptoms: Longitudinal relationships 285-293.
among breast cancer survivors. Psychoneuroendocrinology. 2014;42:38-44. 91. Sgaard M, Thomsen RW, Bossen KS, et al. The impact of comorbidity on
73. Eom C-S, Shin DW, Kim SY, et al. Impact of perceived social support on cancer survival: a review. Clin Epidemiol. 2013;5:3-29.
the mental health and health-related quality of life in cancer patients: 92. Currie CJ, Poole CD, Jenkins-Jones S, et al. Mortality after incident
results from a nationwide, multicenter survey in South Korea. Psy- cancer in people with and without type 2 diabetes: impact of metformin
chooncology. 2013;22:1283-1290. on survival. Diabetes Care. 2012;35:299-304.
74. Kroenke CH, Quesenberry C, Kwan ML, et al. Social networks, social 93. Bang D, Piccirillo J, Littenberg B, et al. The Adult Comorbidity Evaluation-
support, and burden in relationships, and mortality after breast cancer 27 (ACE-27) test: a new comorbidity index for patients with cancer.
diagnosis in the Life After Breast Cancer Epidemiology (LACE) study. ASCO Annual Meeting Proceedings. 2000; abstr 1701.
Breast Cancer Res Treat. 2013;137:261-271. 94. Kong A, Johnson N, Kitchener HC, et al. Adjuvant radiotherapy for stage I
75. Steptoe A, Shankar A, Demakakos P, et al. Social isolation, loneliness, endometrial cancer: an updated Cochrane systematic review and meta-
and all-cause mortality in older men and women. Proc Natl Acad Sci. analysis. J Natl Cancer Inst. 2012;104:1625-1634.
2013;110:5797-5801. 95. Nout RA, Smit VT, Putter H, et al; PORTEC Study Group. Vaginal
76. Zimet GD, Dahlem NW, Zimet SG, et al. The multidimensional scale of brachytherapy versus pelvic external beam radiotherapy for pa-
perceived social support. J Pers Assess. 1988;52:30-41. tients with endometrial cancer of high-intermediate risk (PORTEC-2):
77. Cereda E. Mini nutritional assessment. Curr Opin Clin Nutr Metab Care. an open-label, non-inferiority, randomised trial. Lancet. 2010;375:
2012;15:29-41. 816-823.
78. Bellera CA, Rainfray M, Mathoulin-Pelissier S, et al. Screening older 96. Maggi R, Lissoni A, Spina F, et al. Adjuvant chemotherapy vs radio-
cancer patients: first evaluation of the G-8 geriatric screening tool. Ann therapy in high-risk endometrial carcinoma: results of a randomised
Oncol. 2012;23:2166-2172. trial. Br J Cancer. 2006;95:266-271.

174 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


GYNECOLOGIC CANCER

Symptom Management for the


Patient with Gynecologic Cancer

CHAIR
Linda Van Le, MD
The University of North Carolina at Chapel Hill
Chapel Hill, NC

SPEAKERS
Deborah Mayer, RN, PhD, AOCN, FAAN
University of North Carolina at Chapel Hill School of Medicine
Chapel Hill, NC

Mary McCormack, BSc, MSc, PhD, MBBS, FRCR


University College Hospital
London, United Kingdom
VAN LE AND MCCORMACK

Enhancing Care of the Survivor of Gynecologic Cancer:


Managing the Menopause and Radiation Toxicity
Linda Van Le, MD, and Mary McCormack, BSc, MSc, PhD, MBBS, FRCR

OVERVIEW

It is expected that there will be 290,000 cases of gynecologic cancers in 2016. Of these cancers, 60,000 will be endometrial and
22,000 will be ovarianthe two most common gynecologic cancers. Endometrial and ovarian cancers occur in menopausal
women with mean ages of 60 and 63, respectively. The majority of endometrial cancers are early stage, and 5-year survival is
considered good at upwards of 75%. For ovarian cancer, while survival rates have improved, the 5-year survival rate for the
most common stage (stage III) is 40%. Thus, a substantial number of patients with gynecologic cancer are menopausal, and
a significant number of patients are survivors, particularly of endometrial cancers. It will be important for survivors of
gynecologic cancers to receive care tailored to their needs as women and to mitigate gender-specific side effects of their
cancer treatment.

A ccording to the National Cancer Institute, an individual is


considered a cancer survivor from the time of diagnosis
through the balance of his or her life. The American Society
Vasomotor symptoms (hot flashes) occur in up to 80% of
postmenopausal women. A hot flash is characterized by
development of heat in the torso and face that disseminates
of Clinical Oncology (ASCO) endorses the importance of to the rest of the body. These last 2 to 4 minutes and are
survivorship care, broadly defined as care tailored to the associated with sweating, followed by chills. Hot flashes can
survivor.1 ASCO defines four components of survivorship continue for years after menopause (mean 7 years), with a
care: prevention and detection of new cancers and recurrent minority of women continuing to have hot flashes beyond
cancer, surveillance for recurrence or new other primary 10 years.
cancers, intervention for long-term and late effects from cancer A second major area of discomfort for menopausal women
treatment, and coordination between specialists and primary is GSM, which includes vulvovaginal atrophy and urinary
care providers to optimize survivor care.2 In the United tract dysfunction. In the absence of circulating estrogens,
Kingdom, the National Cancer Survivorship Initiative, an ini- the vulva and vagina epithelium lose elasticity, and the
tiative of the 2007 cancer reform strategy, was created to epithelium thins. Symptoms associated with this include
support those living with and beyond cancer. In this article, burning, pain, dyspareunia, development of vulvar fissures,
we address two important aspects of survivorship care for and introital narrowing. Women may be chronically un-
women with gynecologic cancer: management of menopause comfortable or uncomfortable at the time of intercourse.
needs and care for the patient who has received radiation. Sexual activity and satisfaction may be diminished. Urinary
symptoms include dysuria and urinary frequency, and re-
MANAGEMENT OF MENOPAUSE FOR PATIENTS current urinary tract infections may develop in patients
WITH CANCER resulting from decreased integrity of the bladder epithelium.
Menopause is the cessation of menstrual periods that occurs Genitourinary symptoms of menopause may occur without
at a median age of 52. Menopause occurs because of atresia systemic vasomotor symptoms.
and dissolution of the original number of millions of oocytes Other symptoms associated with menopause include
found at birth. For 5% of women, menopause may occur sleep disturbances (occurring independent of hot flashes),
after the age of 55 or between the ages of 40 and 45. depression, sexual dysfunction, joint pain, and cognitive
Menopause is officially declared after 12 months without changes attributable to anxiety and depression. Physiologic
menses. The two most common symptom groups are va- changes associated with menopause include bone loss and in-
somotor symptoms (VMS) and genitourinary symptoms of creased risk of cardiovascular disease. The data supporting de-
menopause (GSM).3 velopment of dementia, arthritis, changes in body configuration,

From the Division of Gynecologic Oncology, University of North Carolina School of Medicine, Chapel Hill, NC; University College Hospital, London, United Kingdom.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Linda Van Le, MD, The University of North Carolina at Chapel Hill, Campus Box 7572, P.O. Box B103, Chapel Hill, NC 27599-7572; email: lvl@med.unc.edu.

2016 by American Society of Clinical Oncology.

e270 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


SURVIVOR CARE FOR THE PATIENT WITH GYNECOLOGIC CANCER

skin changes, and balance have not been definitively shown study of this group, however, it is extrapolated that hormone
to be attributable to menopause. There are no indications replacement is similarly safe for the younger patient. Risks of
for hormone treatment of prevention or protection of other combined estrogen and progestin replacement include a
health risks such as cardiovascular disease or colon cancer. slightly increased risk of breast cancer, coronary artery
For patients with cancer, menopause can be accelerated disease, stroke, and venous thromboembolism (VTE). When
as a result of cancer treatment, including chemotherapy, estrogen alone is administered, only an increased risk of
radiation therapy, and surgery. Various chemotherapy drugs thromboembolic events is observed without risk of car-
potentially induce ovarian failure, either temporarily or diovascular disease or breast cancer.5,6
permanently. A prime example is the alkylating drug cy- There is a wide variety of choices for treatment of VMS.
clophosphamide. The breast cancer regimen cyclophos- Oral replacement is associated with higher levels of circu-
phamide, methotrexate, and fluorouracil is known to induce lating estrogen metabolites than estrogens administered via
amenorrhea in 70% of patients. Younger patients may re- other routes. Transdermal and cutaneous (gel or spray)
cover ovarian function, whereas older patients are less likely formulations are also available and felt to be as effective.
to do so. Pelvic radiation therapy is particularly toxic to the There is a benefit to a transdermal approach, which delivers
ovaries. Although uncommon, young patients with ovarian estrogen directly to the systemic circulation and bypasses
cancers and some patients with breast cancer will require liver processing (first-pass effect); the transdermal formu-
oophorectomy for successful treatment and will experience lations do not result in a procoagulant affect and also do not
menopause prematurely. affect binding globulins, triglycerides, and C-reactive protein.
Patients with a uterus require addition of a progestin to de-
crease the risk of endometrial neoplasia. Progestins are also
Treatment available for transdermal, vaginal, or intrauterine delivery.
Vasomotor symptoms can cause considerable discomfort Duavee is a conjugated estrogen combined with bazedoxifene, a
and affect quality of life. For mild symptoms, lifestyle selective estrogen receptor modulator (SERM). This drug pre-
changes may suffice. Patients can wear light nightgowns, use vents hot flashes and bone loss. Bazedoxifene counteracts the
lighter bedding and cooling fans, drink cool water, and turn stimulatory effect of estrogen on endometrium and is not
down the thermostat at night. For patients with moderate to expected to stimulate endometrial hyperplasia. Its effect on
severe VMS, estrogen replacement is the most effective breast and uterine cancers is unknown, and, as such, use of
treatment. Hot flashes decrease by 75% compared Duavee is contraindicated for these patients. The overall rec-
with 30%50% seen with administration of a placebo. For ommendation for estrogen or estrogen/progestin treatment is
patients without a uterus, estrogen treatment alone will to use the lowest dose possible, and there is evidence that this is
suffice. For patients with a uterus, addition of a progestin is effective for treatment of VMS.
recommended. Currently, it is recommended that hormones There are many hormone replacement regimens for the
be offered to the immediately menopausal or younger patient with VMS, and these can be delivered in many
women requesting treatment of symptom relief. The En- different ways. Some commonly prescribed low-dose regi-
docrine Society recommends treating symptomatic women mens include: (1) 0.25 mg patch of Climara every week and
if they have experienced menopause within 10 years or are 200 mg of Prometrium (micronized progesterone) orally
younger than age 60, and are without health concerns such every day, (2) ClimaraPro patch applied weekly, and (3)
as cardiovascular disease or stroke.4 For patients with 0.05 mg of estradiol taken orally every day and 2.5 mg of
cancer, there will occasionally be younger patients in need of medroxyprogesterone acetate taken orally every day (a generic
treatment. The Womens Health Initiative did not include inexpensive combination of Estrace and Provera).
For some patients, a nonhormonal approach may be pre-
ferred. Use of selective serotonin reuptake inhibitors (SSRIs)
KEY POINTS or serotonin and norepinephrine reuptake inhibitors is asso-
ciated with a 25%70% decrease in VMS. Paroxetine mesylate
Vasomotor symptoms and genitourinary symptoms of is the only U.S. Food and Drug Administrationapproved SSRI
menopause affect up to 80% of postmenopausal women for treatment of hot flashes. Gabapentin has been shown to
and diminish quality of life. improve hot flashes that predominate at night. Pregabalin,
Patients with hormone-dependent cancers should be venlafaxine, and desvenlafaxine are also effective for treat-
offered nonhormonal treatment first. ment of hot flashes. The clonidine transdermal patch is an-
Gastrointestinal symptoms are the most common side other nonhormonal option for treatment of hot flashes.
effect of pelvic radiotherapy, with up to 50% of patients
Phytoestrogens such as the isoflavone genistein and daidezein
experiencing some form of late toxicity.
Urologic adverse events are more modest; however,
found in soy products are often suggested as an alternative
they increase over time. to treatment, as are herbal remedies; however, there is no
The most common side effects resulting from evidence to support their efficacy in treatment of VMS.
pelvic radiation therapy are pelvic insufficiency Treatment of GSM includes estrogen. Some of these
fractures. symptoms may improve among patients already taking
hormone replacement for VMS. For the patient with only

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e271


VAN LE AND MCCORMACK

GSM, a vaginal formulation should be prescribed. A con- with nonhormonal treatment, low-dose vaginal estrogens (pill,
tinuum of treatment exists for treatment of vaginal symp- ring, or cream) that are not associated with increased systemic
toms ranging from use of nonhormonal vaginal moisturizers estrogen levels may be considered. For patients taking ta-
and lubricants to vaginal estrogens. Moisturizers can be used moxifen, low-dose vaginal estrogens can be offered; any
regularly to decrease vaginal atrophy symptoms. Lubricants minimal increase in systemic estrogen resulting from vaginal
are used in addition to moisturizers during intercourse to treatment is blocked by tamoxifen. For patients taking aro-
alleviate local symptoms of irritation and pain. Neither of matase inhibitors, estrogen administration is not recom-
these improves urinary symptoms, however. If the patient mended, and these patients should be offered nonhormonal
requires escalation of treatment, vaginal estrogens are treatment. See the Sidebar for treatment options for GSM.
available. It is best to use the lowest effective dose to Patient with endometrial cancer also require thoughtful
minimize systemic absorption. Options for treatment of management of menopause. The GOG 137 trial, published in
vaginal symptoms include use of a low-dose vaginal ring, 2006, studied 1,236 patients with stage I and II endometrial
which can be placed every 3 months. Another low-dose cancer in a randomized control trial in which hormone re-
choice is use of vaginal estradiol tablets (10 mg twice a week placement was offered to patients with endometrial can-
vaginally). Both of these preparations have not been ob- cer.9 This study was closed early because of enrollment
served to increase systemic levels of circulating estrogen. issues and was underpowered when it closed. It did conclude
Estradiol creams and higher dose estradiol tablets increase that there was no notable difference in recurrence or death
systemic levels of estrogen and require progestin supple- between the treated and control patients. A 2014 meta-
mentation if a uterus is in place. A new SERM, ospemifene, analysis of hormone replacement for patients with endo-
was approved in 2013 for treatment of vulvovaginal atro- metrial cancer concluded that there was no increased risk of
phy.7 Patients with cancer were excluded from enrollment in recurrence for these patients.10 However, there are no data
ospemifene studies, therefore, there is no safety data for its regarding hormone replacement for patients with advanced
use for patients with cancer. Side effects of ospemifene disease. If estrogen treatment is chosen, the National
include increase of VTE and increase in VSM. See the Sidebar Comprehensive Cancer Network recommends waiting 6
for hormonal and nonhormonal treatment options. 12 months before starting estrogen treatment.
Granulosa cell tumors are uncommon and often hormone-
dependent. There are no data regarding the safety of hor-
Nonhormone-Dependent Gynecologic Cancers
mone treatment in this group, and these patients should be
There is no contraindication for hormone treatment of men-
offered nonhormonal management.
opausal symptoms for patients with cervical or vulvar cancers.
If estrogens are indicated for patients with VTE, a
A recent randomized controlled trial of hormone replacement
transdermal approach should be offered. Administration
among patients with ovarian cancer showed no increase in
of paroxetine for patients taking tamoxifen should be
recurrence; relapse-free survival and overall survival appeared
prescribed with caution. Paroxetine inhibits the cytochrome
to be improved in the hormone-treated group. For younger
450-enzyme system and can decrease the efficacy of tamoxifen.
patients with germ cell tumors, hormone treatment is advo-
Female patients with cancer may experience premature
cated. There are no data regarding safety of hormone treat-
menopause because of necessary cancer therapeutics or
ment for patients with sarcomas or carcinosarcomas.
undergo spontaneous menopause by virtue of their age.
Other than breast cancers, it is uncommon for non-
Quality of life has been shown to improve when symptomatic
gynecologic cancers to be hormone dependent. Unless there
patients are treated. There are many nonhormonal formu-
are contraindications to estrogen treatment, these cancers
lations, and it may be best to offer these first. However, some
can be treated as in general menopausal patients.
patients may continue to be symptomatic from VSM, and
discussion of hormone treatment should be undertaken after
Hormone-Dependent Cancers taking into consideration the patients cancer status. It is
Patients with breast cancer requiring menopausal symptom widely acknowledged that definitive trials are lacking re-
management constitute a unique patient group. In 2003, the garding treatment of hormone-dependent cancers. Given the
HABITS trial and Stockholm trial, both randomized trials increase in the female survivor population, management of
studying menopausal estrogen replacement among patients menopause will require additional study in a timely fashion.
with breast cancer, were stopped prematurely because of
the observation of an increased risk of recurrent cancer in
the treatment arms.8 Long-term follow-up of both has not SURVIVING THE CURE: MANAGING THE LATE
observed a difference in survival, and the Stockholm trial did EFFECTS OF RADIOTHERAPY
not observe an increase in recurrence in the treatment arm. The late effects of radiotherapy (RT) are defined as those
Nonetheless, because there has been no rigorous trial clearly that occur months to years after completion of a course of
demonstrating safety of hormone replacement in this group, RT. Such effects have become more important as the
nonhormonal treatment of VMS is recommended for pa- number of long-term survivors increase. Pelvic RT is an
tients with breast cancer. For GSM, vaginal moisturizers and effective treatment of many gynecologic cancers but often
lubricants are recommended. If symptoms do not resolve comes at a price. The extent of the problem is difficult to

e272 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


SURVIVOR CARE FOR THE PATIENT WITH GYNECOLOGIC CANCER

gauge accurately, as these data have generally not been diarrhea, flatulence, frequency, urgency, incontinence, mal-
collected prospectively in large randomized trials. Further- absorption, and pain. It is essential that the clinician take an
more, patients may present to different specialists with accurate and detailed history of the symptoms together with
symptoms that are not immediately recognized as late ef- establishing the precise details of all prior cancer therapies.
fects of RT, therefore contributing to the general under- An initial assessment of the patient should include evalu-
reporting. The National Survivorship Initiative in the United ation to exclude anemia and thyroid abnormalities, and
Kingdom11 identified four key needs of survivors of cancer: cross-sectional imaging should be ordered to exclude cancer
A personalized survivorship care plan
Support to self-manage their condition where appropriate
recurrence prior to referral to a gastroenterologist. An al-
gorithm detailing an approach to the management of GI
Provision of information on long-term effects of living
with and beyond cancer
radiation side effects, developed by colleagues at the Royal
Marsden Hospital in London, was published in a recent
Access to specialist medical care for complications that
occur after cancer
Nature Reviews Gastroenterology & Hepatology review and
is presented in Fig. 1.19 Altered consistency of bowel habit,
There are wide variations (up to 23%) in the reported frequency, urgency, and rectal bleeding are the most
prevalence of late toxicity following pelvic RT in women with commonly identified symptoms of late GI toxicity following
gynecologic cancers.12 The generally accepted prevalence pelvic RT. Patients usually must undergo sigmoidoscopy/
rate is approximately 10%.1315 It is likely that the classifi- colonoscopy for evaluation.20 The management of these
cation of the late effects together with underreporting will symptoms is complex and will depend upon the results of the
contribute to some of this variation. various investigations. Strategies include antibiotics to treat
small intestinal bacterial overgrowth, use of loperamide and
stoolbulking agents, and biofeedback to manage frequency
Gastrointestinal Toxicity and urgency. Rectal bleeding from radiation-induced tel-
Gastrointestinal (GI) symptoms are the most common side angiectasia is common, and, in the majority of patients, it is
effect of pelvic RT, with up to 50% of patients experienc- mild and requires no specific therapy. However, rectal
ing some form of late toxicity.16,17 A review of our in- bleeding leading to anemia can have a considerable impact
stitutional data showed that the mean time to presentation on the patients quality of life. Options for management
of bowel symptoms was 8 and 10 months for women often vary with resources and expertise but may include use
following treatment of cervical and endometrial cancers, of argon plasma coagulation embolization or hyperbaric oxygen
respectively.18 Chronic GI symptoms include rectal bleeding, therapy (HBO).11 A randomized controlled trial was undertaken
to assess the role of HBO in the treatment of chronic refractory
radiation proctitis. The trial, when analyzed according to in-
FIGURE 1. Modified Algorithm for Approach to tention to treat, demonstrated a notable improvement in the
Treating Patients With Gastrointestinal Symptoms Late Effects Normal Tissue Task Force-Subjective, Objective,
After Radiation Therapy Management, Analytic score for patients treated with HBO. The
quality of life bowel bother subscale was also significantly
improved for patients who received HBO.21 Bowel obstruction,
perforation, and fistulas are uncommon, but more serious late
effects of RT are more frequently seen in survivors of cervical,
rather than endometrial, cancer.
Small intestinal bowel obstruction (SIBO) is estimated to
occur in 9% of patients after RT for gynecologic malignan-
cies.22 However, the rates have been shown to be influenced
by both prior laparotomy and body weight.23 Management
of SIBO depends largely on the frequency and severity of
symptoms, with some patients experiencing only intermittent
episodes of SIBO that can be managed conservatively. In others,
the symptoms persist and progress to total obstruction over
weeks or months, and such patients require surgical
intervention.24 The type of procedure performed will vary
according to both patient factors and surgical expertise. In
expert hands, resection of the injured bowel with anastomosis is
the preferred procedure permitting resumption of enteral
nutrition postoperatively.25 Ogino et al26 reported late rectal
grade 3/4 complication rates of 6.8% and 8.1%, respectively,
Abbreviations: QOL, quality of life; APC, argon plasma coagulation. among patients following radical pelvic RT for cervical cancer.
Adapted from Hauer-Jensen M, Denham JW, Andreyev HJ. Radiation
enteropathypathogenesis, treatment and prevention. Nat Rev Gastroenterol
This was predominantly rectal bleeding and rectovaginal fis-
Hepatol. 2014;11:470-479. tulae, although they did not differentiate further between the

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e273


VAN LE AND MCCORMACK

were urethral strictures and radiation cystitis. In contrast,


SIDEBAR. Nonhormonal and Hormonal two large trials of postoperative RT in endometrial can-
Treatment for Genitourinary Symptoms of cers28,29 reported no grade 3 to 4 urinary adverse effects
Menopause with a median follow-up of 5268 months. This finding may
be the result of the relatively short follow-up in these
Vaginal Moisturizers
Fresh Start studies. The management of ureteral strictures may involve
K-Y Silk-E ureteral dilatation and intermittent self-catheterization,
K-Y Liquibeads whereas mild cystitis may be managed by dietary modifi-
Moist Again cations. The management of severe urologic adverse events
Replens is complex and beyond the scope of this article. In cases of
Vaginal Lubricants treatment for refractory chronic radiation injury, there may
Astroglide be a role for HBO in symptom management.30
FemGlide The impact of pelvic RT on bone health is increasingly
K-Y Jelly recognized and can manifest in a number of ways. The most
Summers Eve
common events are pelvic insufficiency fractures. These may
Vaginal Estrogen
be asymptomatic and identified only on MRI/CT/PET, or
Tablet: Estradiol (Vagifem) 10 mg daily first week, then two
times per week vaginally patients may also present with pelvic pain and reduced
Ring: Estradiol (Estring) 7.5 mg, insert every 90 days mobility. The median time to occurrence is 620 months after
Selective Estrogen Receptor Modulator: Ospemifeme radiation, and the majority is managed conservatively.31,32
(Osphema), 60 mg orally daily Radiation treatment of gynecologic malignancies can be as-
sociated with considerable adverse effects, leading to a detri-
mental effect on quality of life. Therefore, the risk benefits should
two. The fistula risk is less than 3% at 20 years, but pelvic surgery always be carefully explored, and patients should be monitored
before or after RT has been shown to almost double this risk.23 long term to facilitate early detection of serious adverse events
and subsequent prompt referral to an appropriate specialist.
Urologic Complications
Grade 1 and 2 urologic adverse events are relatively com- CONCLUSION
mon; however, the risk of major urinary tract complications Cancer survival rates are increasing. This increase is the
is more modest. Unlike GI complications, which usually result of improvements in screening, management of side
present within 2 years, the risk of urologic complications effects, and the availability of new treatments. The goal of
increases over time. A retrospective review of the compli- survivorship care is to improve overall health and quality of
cations following radical RT for early-stage cervical cancer life after cancer treatment. Women surviving gynecologic
concluded that the actuarial risk of grade 3/4 urologic cancer have unique needs. Understanding and addressing
complications was 11.1% at 10 years, 13% at 15 years, and these needs will help these patients celebrate and enjoy
14.4% at 20 years.23,27 The most common adverse events their survivorship and improve their quality of life.

References
1. McCabe MS, Bhatia S, Oeffinger KC, et al. American Society of Clinical women with hysterectomy: the Womens Health Initiative randomized
Oncology statement: achieving high-quality cancer survivorship care. controlled trial. JAMA. 2004;291:1701-1712.
J Clin Oncol. 2013;31:631-640. 7. Simon JA, Kingsberg SA, Shumel B, et al. Efficacy and safety of flibanserin
2. American Society of Clinical Oncology. Key Components of Survivorship Care. in postmenopausal women with hypoactive sexual desire disorder:
http://www.asco.org/practice-research/key-components-survivorship-care. results of the SNOWDROP trial. Menopause. 2014;21:633-640.
Accessed February 23, 2016. 8. Fahlen M, Fornander T, Johansson H, et al. Hormone replacement
3. ACOG Practice Bulletin No. 141: management of menopausal symp- therapy after breast cancer: 10 year follow up of the Stockholm
toms. Obstet Gynecol. 2014;123:202-216. randomised trial. Eur J Cancer. 2013;49:52-59.
4. Stuenkel CA, Davis SR, Gompel A, et al. Treatment of symptoms of the 9. Barakat RR, Bundy BN, Spirtos NM, et al; Gynecologic Oncology Group
menopause: an endocrine society clinical practice guideline. J Clin Study. Randomized double-blind trial of estrogen replacement therapy
Endocrinol Metab. 2015;100:3975-4011. versus placebo in stage I or II endometrial cancer: a Gynecologic On-
5. Rossouw JE, Anderson GL, Prentice RL, et al; Writing Group for the cology Group Study. J Clin Oncol. 2006;24:587-592.
Womens Health Initiative Investigators. Risks and benefits of estrogen 10. Shim SH, Lee SJ, Kim SN. Effects of hormone replacement therapy on the
plus progestin in healthy postmenopausal women: principal results rate of recurrence in endometrial cancer survivors: a meta-analysis. Eur
from the Womens Health Initiative randomized controlled trial. JAMA. J Cancer. 2014;50:1628-1637.
2002;288:321-333. 11. Andreyev HJ, Davidson SE, Gillespie C, et al; British Society of Gas-
6. Anderson GL, Limacher M, Assaf AR, et al; Womens Health Initiative Steering troenterology; Association of Colo-Proctology of Great Britain and
Committee. Effects of conjugated equine estrogen in postmenopausal Ireland; Association of Upper Gastrointestinal Surgeons; Faculty of

e274 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


SURVIVOR CARE FOR THE PATIENT WITH GYNECOLOGIC CANCER

Clinical Oncology Section of the Royal College of Radiologists. Practice 22. Stanic S, Mayadev JS. Tolerance of the small bowel to therapeutic ir-
guidance on the management of acute and chronic gastrointestinal radiation: a focus on late toxicity in patients receiving para-aortic nodal
problems arising as a result of treatment for cancer. Gut. 2012;61: irradiation for gynecologic malignancies. Int J Gynecol Cancer. 2013;23:
179-192. 592-597.
12. Kirwan JM, Symonds P, Green JA, et al. A systematic review of acute 23. Eifel PJ, Levenback C, Wharton JT, et al. Time course and incidence of
and late toxicity of concomitant chemoradiation for cervical cancer. late complications in patients treated with radiation therapy for FIGO
Radiother Oncol. 2003;68:217-226. stage IB carcinoma of the uterine cervix. Int J Radiat Oncol Biol Phys.
13. Eifel PJ, Jhingran A, Bodurka DC, et al. Correlation of smoking history and 1995;32:1289-1300.
other patient characteristics with major complications of pelvic radi- 24. Tsai MS, Liang JT. Surgery is justified in patients with bowel obstruction
ation therapy for cervical cancer. J Clin Oncol. 2002;20:3651-3657. due to radiation therapy. J Gastrointest Surg. 2006;10:575-582.
14. Vale CL, Tierney JF, Davidson SE, et al. Substantial improvement in UK 25. Onodera H, Nagayama S, Mori A, et al. Reappraisal of surgical treatment
cervical cancer survival with chemoradiotherapy: results of a Royal for radiation enteritis. World J Surg. 2005;29:459-463.
College of Radiologists audit. Clin Oncol (R Coll Radiol). 2010;22: 26. Ogino I, Kitamura T, Okamoto N, et al. Late rectal complication following
590-601. high dose rate intracavitary brachytherapy in cancer of the cervix. Int J
15. Guth U, Hadwin RJ, Schotzau A, et al. Clinical outcomes and patterns of Radiat Oncol Biol Phys. 1995;31:725-734.
severe late toxicity in the era of chemo-radiation for cervical cancer. 27. McIntyre JF, Eifel PJ, Levenback C, et al. Ureteral stricture as a late
Arch Gynecol Obstet. 2012;285:1703-1711. complication of radiotherapy for stage IB carcinoma of the uterine
16. Theis VS, Sripadam R, Ramani V, et al. Chronic radiation enteritis. Clin cervix. Cancer. 1995;75:836-843.
Oncol (R Coll Radiol). 2010;22:70-83. 28. Cruetzberg CL, van Putten WL, Koper PC, et al. Surgery and post-
17. Andreyev J. Gastrointestinal symptoms after pelvic radiotherapy: a new operative radiotherapy versus surgery alone for patients with stage-1
understanding to improve management of symptomatic patients. endometrial carcinoma; multicentre randomised trial. PORTEC Study
Lancet Oncol. 2007;8:1007-1017. Group. Post Operative Radiation Therapy in Endometrial Carcinoma.
18. Kuku S, Fragkos C, McCormack M, et al. Radiation-induced bowel injury: Lancet. 2000;355:1404-1411.
the impact of radiotherapy on survivorship after treatment for 29. Keys HM, Roberts JA, Brunetto VL, et al; Gynecologic Oncology Group. A
gynaecological cancers. Br J Cancer. 2013;109:1504-1512. phase III trial of surgery with or without adjunctive external pelvic
19. Hauer-Jensen M, Denham JW, Andreyev HJ. Radiation enteropathy radiation therapy in intermediate risk endometrial adenocarcinoma:
pathogenesis, treatment and prevention. Nat Rev Gastroenterol Hep- a Gynecologic Oncology Group study. Gynecol Oncol. 2004;92:744-751.
atol. 2014;11:470-479. 30. Craighead P, Shea-Budgell MA, Nation J, et al. Hyperbaric oxygen
20. Gami B, Harrington K, Blake P, et al. How patients manage gastroin- therapy for late radiation tissue injury in gynecologic malignancies. Curr
testinal symptoms after pelvic radiotherapy. Aliment Pharmacol Ther. Oncol. 2011;18:220-227.
2003;18:987-994. 31. Schmeler KM, Jhingran A, Iyer RB, et al. Pelvic fractures after radiotherapy
21. Clarke RE, Tenorio LM, Hussey JR, et al. Hyperbaric oxygen treatment of for cervical cancer: implications for survivors. Cancer. 2010;116:625-630.
chronic refractory radiation proctitis: a randomized and controlled 32. Park SH, Kim JC, Lee JE, et al. Pelvic insufficiency fracture after ra-
double-blind crossover trial with long-term follow-up. Int J Radiat Oncol diotherapy in patients with cervical cancer in the era of PET/CT. Radiat
Biol Phys. 2008;72:134-143. Oncol J. 2011;29:269-276.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e275


HEAD AND NECK CANCER

Best of the Rest: Top Abstracts


on Head and Neck Cancer From
20152016 Oncology Meetings

CHAIR
Sue S. Yom, MD, PhD
University of California, San Francisco
San Francisco, CA

SPEAKERS
Andreas Dietz, MD
University of Leipzig
Leipzig, Germany

Apar K. Ganti, MD
University of Nebraska Medical Center
Omaha, NE
YOM, GANTI, AND DIETZ

Whats New in Head and Neck Cancer: Key Findings in


20152016 From ECCO/ESMO, ASTRO, and the
Multidisciplinary Head and Neck Cancer Symposium
Sue S. Yom, MD, PhD, Apar K. Ganti, MD, MS, FACP, and Andreas Dietz, MD

OVERVIEW

Scientific investigation is extremely active in the treatment, management, and optimization of therapies for patients with
head and neck cancer. These issues have undergone recent rapid evolution in response to a changing epidemiology based on
an increasing proportion of HPV-associated oropharyngeal cancer with advances in multimodality technologies to improve
outcomes and reduce toxicity. Choices of definitive treatment of various anatomic subsites are being refined, balancing the
relative indications and advantages of surgery or chemoradiation-based strategies. The major potential influence of HPV-
associated etiology on therapy selection, prognostic factors, response to treatment, survival outcomes, and post-treatment
surveillance has created a robust and distinct field of scientific inquiry around this patient subset. Meanwhile, for patient subsets
where prognosis remains poor, therapeutic intensification is being explored, and for recurrent/metastatic disease, improved
selection for salvage and novel systemic therapies are under development. For all patients with head and neck cancer, upholding
principles of equity and access to the highly specialized care that results in optimal outcomes should be the goal.

T his article summarizes selected findings in head and neck


cancer research that were presented at several major
meetings over the past year. The European CanCer Organi-
concurrent chemoradiation therapy. Radical surgery was
performed on 35.29% and 37.63% of patients with stage III
and stage IVA oropharyngeal cancer, respectively, and 54.52%
zation (ECCO) and the European Society for Medical Oncology and 48.85% of patients with stage III and stage IVA hypo-
(ESMO) held a joint meeting, the European Cancer Congress, pharyngeal cancer, respectively. Primary surgery was asso-
from September 2529, 2015, in Vienna. The 57th Annual ciated with better overall cancer survival in most subset
Meeting of the American Society for Radiation Oncology analyses, but the potential effect of selection for surgery in
(ASTRO) took place in San Antonio, Texas, from October this population-based database could not be discounted.
1821, 2015. The 2016 Multidisciplinary Head and Neck Surgical treatment of advanced head and neck cancer is a
Cancer Symposium, a joint conference of the American Head well-recognized weapon that is very often followed by ra-
and Neck Society, American Society of Clinical Oncology, and diotherapy with or without chemotherapy, but in some cases,
American Society of Radiation Oncology, took place in surgery can be avoided and radiotherapy or chemoradiotherapy
Scottsdale, Arizona, from February 1820, 2016. is an appropriate standard of care. In many centers, the choice
of primary modality continues to be heavily influenced by local
SURGERY AS A PRIMARY TREATMENT factors such as experience or availability. These data further
MODALITY underscore the need for prospective research to define the
At the 2015 European Cancer Congress, Cheng et al pre- relative roles of surgery and radiotherapy within curative
sented an analysis of data from Taiwan national health treatment paradigms for these subsites.
insurance claims and the Taiwan cancer registry database, The importance of the prognostic features identified at
including data from 1,698 patients with oropharyngeal and surgical pathology was highlighted by a presentation from
1,619 patients with hypopharyngeal stage III/IVA cancer researchers in India. This group measured tumor thickness
diagnosed between 2004 and 2009.1 The researchers com- using intraoral ultrasound (IOUS) and correlated it with the
pared outcomes among those who had surgery and those who risk of nodal metastasis in early N0 oral cancer. A prospective
did not; in both groups, patients may or may not have received study was carried out for patients with cT1-2N0 oral cavity

From the Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA; Department of Internal Medicine, University of Nebraska Medical
Center, Omaha, NE; Department of Head Medicine and Oral Health, University of Leipzig, Leipzig, Germany.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Sue S. Yom, MD, PhD, University of California, San Francisco, 1600 Divisadero St., San Francisco, CA 94115; email: yoms@radonc.ucsf.edu.

2016 by American Society of Clinical Oncology.

176 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


WHATS NEW IN HEAD AND NECK CANCER

cancer in a high-volume head and neck surgical center from understand the underlying genetic basis for the differential
2013 to 2015. A cutoff level of greater than 8.5-mm tumor outcomes.5 They conducted targeted next-generation se-
thickness on IOUS was derived (area under the curve, 0.81; quencing for 66 patients with HPV-positive OPSCC (40
95% CI, 0.660.97; p = .005) that predicted lymph node had more than 10 pack years of smoking and 26 had less
metastasis with a sensitivity and specificity of 88% and 60%, than 10 pack years). The group with the lower smoking
respectively. Tumor thickness measuring less than 8 mm history had better outcomes. TP53, CDKN2A, KRAS, and
and greater than 9 mm on IOUS developed 3.2% and 25.9% NOTCH1 mutations were seen almost exclusively in the
lymph node metastasis, respectively (p = .01). This cutoff high-smoking group, whereas HLA-A mutations were more
recommendation could be valuable in designing clinical trials common in the low-smoking group.
based on indications for elective neck dissection in early oral In a more clinical vein, Lukens et al reviewed a series of 174
cancer.2 patients with HPV-positive OPSCC who had been surgically
Meanwhile, a group from Belgium took a different ap- treated with transoral robotic surgery and neck dissection
proach to this problem, testing the clinical feasibility of followed by adjuvant chemoradiotherapy. The focus was to
sentinel lymph node identification in a prospective phase II characterize the influence of lymph node characteristics
study. The goal of the study was to reduce the electively on the risk of distant metastasis. The follow-up was
radiated volumes of the neck. Among 21 patients with cN0 38 months. The probability of distant metastasis increased
oral cavity, oropharynx, larynx, or hypopharynx squamous proportionately when patients had four or more involved
cell carcinomas, 99mTc nanocolloid injection was followed nodes. The risk of distant metastasis was 14% for four or
with single-photon emission CT (SPECT).3 An average of 2.7 more nodes, 18% for five or more nodes, 22% for more than
sentinel nodes were detected per patient. Only the neck six nodes, and 28% for more than seven nodes. On univariate
levels containing the four hottest draining nodes were analysis, macroscopic/gross extracapsular extension (ECE) pre-
electively radiated. At a median follow-up of 14 months, dicted distant metastasis, with distant metastasis developing in
there were two fatal local relapses but no regional relapses. 20% of patients wtih ECE versus 4.2% of patients with micro-
scopic ECE, but on multivariate analysis, macroscopic/gross ECE
HPV-ASSOCIATED OROPHARYNGEAL was not significant (p = .84).6
SQUAMOUS CELL CARCINOMA
HPV-positive oropharyngeal squamous cell carcinoma (OPSCC)
is a distinct subtype of head and neck squamous cell carcinoma BIOLOGIC AND RADIOLOGIC PROGNOSTIC
(HNSCC) that has a significantly better prognosis compared FEATURES
with HPV-negative OPSCC.4 However, it appears that smoking Although various clinical factors, especially smoking, are
history may be used to stratify HPV-positive OPSCC into more considered prognostic, there is a continued search for other
distinct prognostic groups. Zevallos et al presented their study predictors of outcome that may be more biologically specific.
of the molecular profile of HPV-positive OPSCC, an attempt to At ASTRO 2015, Khwaja et al presented a well-conducted gene-
expression profiling study of HPV-positive OPSCC.7 Patients
among whom distant metastasis developed had an E6 ex-
pression level that was twofold higher. In the validation
KEY POINTS cohort, the authors were able to identify a cutoff level of E6
expression (7.3, by receiver operating curve analysis) that was
Surgery and chemoradiation-based strategies are correlated to a fivefold higher risk of distant metastasis. The
considered appropriate up-front modalities for
only minor inconsistency in this study was that the patients
definitive treatment, based on anatomic considerations,
the balance of toxicities, and morbidity.
were treated with either chemoradiation or surgery followed
For HPVassociated oropharyngeal cancers, by postoperative radiation with or without chemotherapy.
de-intensification of radiation therapy is under active From a different angle, radiographic predictors have been
exploration in an attempt to reduce the long-term an ongoing area of investigation that has yet to yield a clear
toxicities of treatment. standard pretreatment measurement tool, perhaps because
Post-treatment surveillance remains a relatively assessment tools should be specific for the type of treatment
undefined area, although physical examination, direct to be received. For example, a study from Australia analyzed
visualization, and patient-reported symptoms are the possible predictive value of fluorodeoxyglucose (FDG)-
recognized as key indices to follow. positron emission tomography (PETCT) after two cycles of
In the salvage setting, improved selection of patients for taxane/platinum/fluorouracil (5-FU) induction in correlation
surgical salvage may produce better outcomes, and
with outcome. Although a high overall survival (OS) at 89.5%
early results of reirradiation with novel technologies
such as proton beam therapy are promising.
was achieved, the FDG-PET/CTbased volumetric measures
For recurrent/metastatic disease, a number of were not predictive for locoregional control (impact of
combinations of novel targeted therapies and/or changes of SUVmax not reported).8
immunomodulatory therapies will move forward in the More detailed data concerning off-target FDG-PET pa-
near future. rameters for outcome prognosis after chemoradiotherapy
were reported from Germany. Early occurrence of mucositis

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 177


YOM, GANTI, AND DIETZ

higher than grade 1 was associated with better locoregional Thirty HPV-positive and seven HPV-negative patients had a
control (p = .012) and OS (p = .017). Occurrence of dysphagia good response, as defined before. The 2-year progression-
higher than level 2, but not mucositis, at the end of treatment free survival (PFS) and OS rates were 93.1% and 92.1% for
was associated with favorable locoregional control (p = .031) good response, HPV-positive OPSCC and 74.0% and 95.2% for
and OS (p = .008). Besides error-prone and observer-biased NR HPV-positive OPSCC, respectively.
determination of mucositis by clinical examination, the It has been an ongoing active controversy as to whether
evaluation of mucosa by PET during treatment showed a de-intensification with the use of cetuximab could risk
superior correlation with patients outcome.9 compromising outcomes. A new abstract on this subject was
Lastly, MAASTRO investigators reported a new radionomics presented at ASTRO 2015 but is unlikely to settle the debate.15
tool that has prognostic and predictive implications after In this study, the investigators found that patients who had
chemoradiotherapy. Five radiomic features of primary tumor cetuximab given concurrently with radiotherapy had higher
regions were evaluated. Strong correlations with OS were rates of distant metastases than those who had received
described by combining special features. This noninvasive chemoradiotherapy with a platinum agent. Maturing results of
new morphometric tool deserves further evaluation.10 the major study addressing this question, RTOG 1016, which
tested cetuximab versus cisplatin in HPV-positive oropha-
ryngeal squamous cell carcinoma, are eagerly awaited.
DE-INTENSIFIED CHEMORADIOTHERAPY FOR
HPV-POSITIVE OROPHARYNGEAL SQUAMOUS INTENSIFICATION FOR HPV-NEGATVE
CELL CARCINOMA SQUAMOUS CELL CARCINOMA
Given the excellent prognosis of HPV-positive OPSCC, efforts
Although the good prognosis of patients with HPV-positive
are underway to attempt de-escalation of therapy without
OPSCC has led to trials in therapeutic de-intensification, for
affecting outcomes. In a phase II trial that was reported at
patients with HPV-negative HNSCC who had significantly
ASTRO and the Multidisciplinary Head and Neck Cancer
worse outcomes, systemic therapy intensification may be
Symposium, Chera et al included 43 patients with T0-3, N0-
warranted. In an abstract presented at the Multidisciplinary
N2c, and M0 HPV-positive OPSCC with a minimal or remote
Head and Neck Cancer Symposium, Melotek et al investigated
smoking history.11,12 The treatment regimen was 60 Gy of
the addition of cetuximab to induction chemotherapy and
intensity-modulated radiation therapy with weekly cisplatin
concurrent chemoradiation for patients with locoregionally
(30 mg/m2). All patients then underwent a biopsy of the
advanced HNSCC.16 Patients were randomly selected to receive
primary site and a neck dissection of pretreatment positive
two cycles of weekly cetuximab, paclitaxel, carboplatin, and
lymph node regions, irrespective of response. The patho-
either cetuximab, 5-FU, hydroxyurea, and 1.5 Gy twice-daily
logic complete response rate was 86%. The patients without
radiotherapy every other week to 75 Gy (CFHX) or cetuximab,
pathologic complete response had only microscopic foci of
cisplatin, and accelerated radiotherapy with delayed concom-
residual disease. All patients were alive without recurrence
itant boost to 72 Gy in 42 fractions (CPX). Fifty-six patients with
after a median follow-up of 21.3 months. The incidence of
HPV-negative HNSCC (32 receiving CFHX and 24 CPX) were
acute Common Terminology Criteria for Adverse Events grade
included in the trial. The 2-year PFS was 75.0% for CFHX and
3/4 toxicity was as follows: mucositis 34%, pain 5%, nausea
70.8% for CPX. The 5-year PFS and OS for the HPV-negative
18%, vomiting 5%, dysphagia 39%, and xerostomia 2%. A
cohort were 65.9% and 72.5%, respectively, suggesting a
feeding tube was required temporarily for 39% of patients.
possible role for cetuximab for this group of patients.
Likewise, in another attempt to decrease late toxicity
presented at both ASTRO13 and the Multidisciplinary Head
and Neck Cancer Symposium,14 Melotek et al incorporated SURVEILLANCE
a response-adapted volume de-escalation approach using The optimal frequency of follow-up imaging after definitive
response-to-induction chemotherapy as a guide to decrease curative-intent therapy has not been defined. The National
the extent of radiation volume in a nonselected population of Comprehensive Cancer Network guidelines do not recom-
patients with locally advanced HNSCC. The regimen was mend routine imaging in the absence of symptoms, except
comprised of two cycles of chemotherapy (cisplatin 75 mg/m2, chest CT scans as recommended for lung cancer screening (in
paclitaxel 175 mg/m2, both on day 1, and weekly cetuximab high-risk patients who meet the criteria). At the Multidisciplinary
with or without everolimus). Patients with good response, Head and Neck Cancer Symposium, Frakes et al retrospectively
defined as a 50% reduction in the sum of gross tumor diameters, reviewed the charts of 246 patients with HPV-positive OPSCC
received concurrent chemoradiation (paclitaxel, 5-FU, hydroxy- who were treated with definitive radiotherapy or chemo-
urea, and 1.5 Gy twice-daily radiotherapy every other week). radiotherapy.17 Local control was achieved for 239 patients,
The radiation dose was as follows: 75 Gy with the planning with a 3-year local control rate of 97.8%. All local failures
target volume (PTV1) encompassing exclusively gross disease. were detected by direct visualization or flexible laryngos-
Patients with less than 50% response (NR) were treated with copy. The 3-year regional control rate was 95.3%. Risk
volumes encompassing PTV1 and the next nodal station at factors for regional failure included patients who had in-
risk (PTV2) to 45 Gy, followed by a sequential boost to PTV1 to volvement of five or more nodes or level 4 lymph nodes. The
75 Gy. Of the 94 patients in this study, 63% were HPV-positive. majority of regional recurrences (89%) were detected either

178 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


WHATS NEW IN HEAD AND NECK CANCER

by symptoms or 3-month post-treatment PET/CT. Twenty- chemoradiotherapy.21 The duration, incidence, severity, and
one patients had distant metastases. Factors predicting for onset of severe oral mucositis seemed to be markedly improved
distant metastases included lymph nodes larger than 6 cm, compared with historical controls. The efficacy of GC4419 was
bilateral lymphadenopathy, five or more nodes, or level 4 duration dependent with maximum benefit seen among
lymph node involvement. Of these 21 patients, 71% were patients who received 6 to 7 weeks of treatment. The drug
identified either from symptoms or on the 3-month post- was well tolerated, with the main side effects being grade 3
treatment imaging. These results suggest that if the 3-month gastroenteritis and nausea/vomiting (one patient each). In
post-treatment PET/CT is negative, no further routine imaging addition, peri-infusional facial tingling was attributable to
is necessary. The results also reiterate the importance of a good GC4419. The dose levels chosen for a future randomized
history and physical examination, including direct visualization. controlled trial were 30 and 90 mg/day.
Regarding the early toxicity of systemic therapy, the first
safety report of the Italian INTERCEPTOR trial (randomized,
POST-TREATMENT NECK DISSECTION multicenter phase III study comparing chemoradiotherapy
The previously standard practice of planned neck dissection
vs. induction chemoradiotherapy followed by radiotherapy
after radiation-based definitive treatment has waned.
and cetuximab) was presented at ECCO/ESMO, with 228
As presented at the European Cancer Congress, 147 patients
patients accrued thus far. The first 170 were considered in
with locoregionally advanced stage IVA (52%) or IVB (46%)
the present analysis (85 patients in each arm). Overall, the
HNSCC from Portugal were treated with induction taxane/
only significant difference between the two arms was grade 3 to
platinum/5-FU chemotherapy followed by chemoradiotherapy
4 neutropenia (p = .017), and the trial will continue. The excess of
from July 2008 to March 2014. Fifty-three patients (37%)
neutropenia was caused entirely by the induction phase.22
underwent neck dissection because of persistence of dis-
Late toxicity is likewise a concern, especially if consider-
ease (4/53, 7.5%), suspicious imaging (40/53, 75.5%), or pre-
ation is given to further intensification of standard chemo-
vious bulky disease (9/53, 17%). A majority of the positive neck
radiotherapy therapies. Twelve-year follow-up data from
dissections were N2c, instead of N3. There was no meaningful
Belgium at ECCO/ESMO compared two nonrandomized
difference in OS between patients who did not have neck
chemoradiotherapy cohorts with a platinum-based induction
dissection and those who had a negative neck dissection.
regimen followed by chemoradiotherapy. Induction was
The authors conclude that prophylactic neck dissection did
associated with a significantly longer time to distant metas-
not seem to improve OS.18
tases. A little over half (51.6%) of the induction patients sur-
Adding to the increasing conservatism around post-
vived beyond 5 years, whereas 5-year survival in the CCRT
radiotherapy neck dissection, Mehanna et al presented a
group was 29.6%. Second HNSCC primaries and other malig-
late-breaking abstract examining differences in the quality of
nancies accounted for 47% of all deaths beyond 5 years. The
life and functional outcomes of treatment between HPV-
most important late local toxicities among surviving patients in
positive and HPV-negative patients receiving primary che-
both groups were dysphagia and xerostomia.23
moradiotherapy, using patient data from the PET-NECK trial
The radiation oncology community has debated for some
previously presented at ASCO 2015.19 In this trial, the use
time whether proton beam therapy has the ability to im-
of PET surveillance reduced the rate of post-treatment neck
prove patient-reported outcomes compared with current
dissection to 20%, without affecting OS. The new presentation
state-of-the-art photon-based techniques. Ahn et al presented
reported that p16-positive subjects have major decreases in
a series of 95 patients, of whom 57 (60%) were treated with
quality-of-life scores during the acute phase of treatment and
photon-based volumetric arc therapy (VMAT) and 45 (45%)
may require additional support during that period, but that
were treated with proton pencil beam scanning (PBS). Fifty-two
they recover better than p16-negative subjects, who have
(55%) of the patients were irradiated after transoral robotic
lower quality of life in the longer term and may need more long-
surgery and 43 (45%) were treated definitively, with or without
term support.
concurrent chemotherapy. At the 6-month endpoint, using the
Performance Scale Status instrument, there was no overall
ACUTE AND LATE TOXICITY OF difference in diet, speech, or public eating. There was no overall
CHEMORADIATION difference in gastrostomy usage. Data from the the EORTC
As seen in the PET-NECK trial, toxicity remains an acute questionnaires showed there was no difference in global health,
and long-term concern for patients receiving chemoradiation. physical function, or sensory domains. However, patients who
Oral mucositis is one of the major acute adverse events as- underwent VMAT lost 5.5% of baseline weight compared with
sociated with head and neck radiotherapy, with or without 3.4% for those who underwent PBS. Patients undergoing VMAT
chemotherapy, occurring in more than 90% of patients. The also used more pain medication. In particular, among patients
incidence of severe mucositis (grade 3-4) is 66%,20 yet there receiving concurrent cisplatin, PBS was significantly associated
are no currently approved agents to decrease the burden with a higher global health score, less pain, lower painkiller use,
associated with this toxicity. and lower percutaneous endoscopic gastrostomy use.24 Given
In this vein, Anderson et al presented a study of 46 the inherent confounding factors that may be in play, further
patients who received increasing doses of GC4419, a su- study of these complex questions is needed and obtaining
peroxide dismutase mimetic, and concurrent cisplatin-based higher-level evidence should be a priority.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 179


YOM, GANTI, AND DIETZ

SINONASAL TUMORS More studies are needed to elucidate the role of proton beam
Induction chemotherapy remains an area of controversy and radiation therapy in HNSCC.
active ongoing interest in the treatment of sinonasal can-
cers. Response to induction chemotherapy before surgical
treatment of locally advanced epithelial sinonasal cancer is MANAGEMENT OF RECURRENT/METASTATIC
recognized as a prognostic factor for outcome. Bossi et al from DISEASE
Italy identified 63 patients with different types of advanced The LUX-Head and Neck-1 (LHN) trial demonstrated that
epithelial sinonasal cancer who received platinum-based in- afatinib was associated with an improved PFS compared with
duction chemotherapy, either associated with 5-FU and methotrexate (2.6 vs. 1.7 months; p = .03).28 In an attempt to
lederfolin or 5-FU and docetaxel. For neuroendocrine predict which subsets of patients would most benefit from
cancers, cisplatin and etoposide alternating with ifosfamide afatinib therapy, Cohen et al evaluated the association of
and doxorubicin were used. Thirty-four patients had a ERBB-related biomarkers and p16 on the antitumor activity of
recurrence (28 at the locoregional level and six at distant afatinib and methotrexate.29 They noted that afatinib had a
sites); 14 patients received salvage surgery (12 on primary higher response rate in EGFR-directed therapynaive patients
tumor and two on neck nodes), but only two of those with p16-negative tumors (27.5% vs. 4.8%) and patients with
remained free of disease (one patient on T and one on N level). p16-negative and EGFR-amplified tumors (15.5% vs. 0%). In
With a median follow-up of 45 months, 5-year OS and disease- contrast, no responses were observed among patients with
free survival rates were 58% and 40%, respectively. Only p16-positive disease, suggesting that EGFR targeting does not
the response to induction chemotherapy retained prog- have a role in HPV-positive HNSCC.
nostic value for OS (p = .0017).25 Because of the rarity and Adkins et al conducted a phase I study of cetuximab and a
heterogeneity of these tumors, the current treatment options suspension formulation of pazopanib among 22 patients with
remain unsatisfactory, and enrollment into international trials incurable HNSCC.30 Both cetuximab-naive and cetuximab-
is needed to make progress. refractory patients were eligible. The regimen was well toler-
ated and the maximally tolerated dose was not reached for
pazopanib at the maximum dose level. Six patients achieved a
LOCALLY RECURRENT DISEASE MANAGEMENT partial response (27%), with an additional 11 (50%) patients
It is generally accepted that surgery has a major role for the achieving stable disease. Three of the six responses were seen
successful treatment of patients with locally recurrent HNSCC. A among patients with cetuximab- and platinum-resistant dis-
retrospective analysis from Belgium had the goal of improving ease, suggesting that this combination may be worthy of future
the selection of patients for salvage surgery as well as identifying study.
the major negative prognostic features in surgical pathology. On After failure of platinum, anti-EGFR, and taxanes, patients
multivariate analysis, oropharyngeal primary site (p , .001), with recurrent/metastatic HNSCC are considered refractory,
positive resection margin (p = .002), extracapsular spread and methotrexate is the general option with palliative care
(p = .003), and major postoperative complications (p , .001) intent. Final results from the phase II French trial UNICANCER
were independent negative prognostic factors for relapse- ORL03 showed that cabazitaxel met the primary endpoint,
free survival. These data underline the poor outcome of producing a 27.6% 6-week disease control rate with ac-
recurrent oropharyngeal HNSCC and the need for highly ceptable tolerability among heavily pretreated patients
reflective indications for salvage surgery in this group.26 with refractory recurrent/metastatic HNSCC.31 Among pa-
For patients who are not determined to be surgical can- tients with recurrent/metastatic HNSCC refractory to prior
didates, reirradiation remains a generally unsatisfying enter- surgery, radiotherapy, or chemotherapy, preliminary phase II
prise. There is a wave of emerging data about the role of proton data of first-line paclitaxel combined with panitumumab
beam radiation therapy for reirradiation of recurrent HNSCC. In showed clinical benefit (median PFS: 7.5 months; 95% CI,
one retrospective analysis of an ongoing prospective data 4.98.3; and median OS: 9.9 months; 95% CI, 7.916.3).
registry presented at the Multidisciplinary Head and Neck This was a comparable efficacy to the standard-of-care
Cancer Symposium, Chang et al reported results for 90 pa- EXTREME regimen, but with a worse (unacceptable) safety
tients who received reirridation with proton beam radiation profile (15% fatal adverse events).32
therapy between 2011 and 2014.27 The actuarial 6-month So what can be currently expected for disease that is
locoregional control and OS were 86.5% and 84.1%, re- refractory to first-line cetuximab and platinum? Retro-
spectively. There was one death as a result of disease pro- spective Swiss data in a cohort of 117 patients described the
gression. Grade 3 acute toxicity included mucositis (10.0%), outcome after second-line regimens (monotherapies, mostly
dermatitis (3.3%), dysphagia (2.2%), and esophagitis (1.1%). In with methotrexate [55%] or other agents [43%]; only one
addition, there were three (5.3%) skin toxicities grade 3 or patient received platinum combination chemotherapy).
worse: a neck ulcer, a chronic neck wound managed with More than half (54%) of the patients did not reach the
hyperbaric oxygen treatments, and a pneumocephalus from a point of second-line treatment. The response rate (partial
skull base defect. There was one death caused by treatment- response, complete response) in 49 patients was 8%, with
related bleeding. Short follow-up limits the conclusions that 33% showing stable disease. Median PFS was 81 days
can be drawn, although the low rates of toxicity are attractive. (95% CI, 5292) and OS was 184 days (95% CI, 115220).

180 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


WHATS NEW IN HEAD AND NECK CANCER

In conclusion, second-line options after first-line cetux- and age, as well as clinical outcome data such as time to
imab containing regimens are strongly limited.33 local failure, PFS, and OS, did not correlate with PD-L1
expression at either of the cutoffs. Nonetheless, the high
PD-L1 expression rate will likely stimulate further studies
IMMUNE MODULATION IN SQUAMOUS CELL of checkpoint inhibitors in this subsite.38 Interestingly, plasma
CARCINOMA OF THE HEAD AND NECK Epstein-Barr virus DNA may correlate with response to pem-
The advent of immunotherapy has heralded a new era in the brolizumab treatment in metastatic nasopharyngeal carcinoma.
treatment paradigm for many malignancies, including HNSCC.
Multiple trials are underway evaluating the role of im-
munotherapies including PD-1 inhibitors in metastatic HNSCC. CARE DELIVERY AND ECONOMICS OF
Preclinical data suggest that radiotherapy may lead to anti- SQUAMOUS CELL CARCINOMA OF THE HEAD
tumor immune responses. In an attempt to identify immu- AND NECK
nologic changes seen during radiotherapy, Sridharan et al With the current emphasis on equitable care delivery to all
prospectively obtained blood samples from 16 consecutive patients, it is important to understand disparities in the care
patients with HNSCC undergoing curative-intent radiotherapy of patients with HNSCC. Churilla et al sought to evaluate
(with or without platinum-based chemotherapy) at the the association between health insurance status and clinical
beginning and end of therapy.34 The investigators found an features at presentation, treatment received, and outcomes
increase in the circulating CD8-positive T-effector cells, among patients with HNSCC.39 Using the SEER database,
CD4/PD-1positive cells, CD8/LAG3-positive cells, and regula- they analyzed 53,848 patients with squamous cell carcinoma
tory T cells (Treg). There was an increase in PD-L1 levels, which of the oral cavity, oropharynx, and larynx. They found that
mirrored increases in CD8-positive T cells over the course of a larger proportion of Medicaid (72.9%) and uninsured
therapy. Levels of CXCL10 decreased during treatment, patients (75.1%) had stage III or IV disease, compared with
whereas those of CXCL16 increased. These findings dem- insured patients (60.1%; p , .001). After adjusting for
onstrate the complex immunologic effects of chemoradiotherapy clinical and demographic factors, uninsured patients were
and suggest mechanisms for potential synergy among che- less likely to receive cancer-directed surgery (odds ratio
motherapy, radiotherapy, and immunotherapy in HNSCC, [OR] 0.86; 95% CI, 0.770.97), whereas Medicaid and un-
which may be exploited in future trials. insured patients were less likely to receive radiotherapy
Evaluation of these specific immunologic responses may (Medicaid: OR 0.77; 95% CI, 0.720.81; uninsured: OR
enable future optimization of the use of this novel class of 0.68; 95% CI, 0.620.75). On multivariate analysis, patients
agents. In a similar experiment, Ferris et al found that a with Medicaid (hazard ratio [HR] 1.55; 95% CI, 1.491.62)
combination of the Toll-like receptor 8 agonist motolimod or uninsured status (HR 1.48; 95% CI, 1.381.58) had
and chemotherapy resulted in a decrease in the pheno- inferior OS compared with insured patients.
typic markers of suppression in Treg cells (CTLA-4, CD73, Disparities may exist across multiple facets and at multiple
and membrane-bound transforming growth factor-b) in tumor levels across the cancer journey. It is recognized that opti-
and peripheral blood.35 In contrast, in nonresponders to mal cancer-related decision making requires very strong
chemotherapy alone, they noted an induction of Treg cells. collaborative work by experienced multidisciplinary teams,
Further data relevant to subgroups from KEYNOTE 012 and HNSCC outcomes have been previously reported to be
have provided greater insight into the efficacy of the antiPD-1 better in centers treating a high number of these patients.
antibody, pembrolizumab. In the subgroup analyses, the An abstract presented at ASTRO 2015 analyzed the impact of
overall response rate appeared higher for patients who radiation oncology provider volume on the clinical outcomes
received fewer than two prior therapies (31/97; 32.0%; two of patients.40 They found that for each additional patient with
complete reponses vs. 29 partial responses) compared with head and neck cancer treated per year, there was a significant
those who had received more than two prior therapies (10/63; reduction in the risk of gastrostomy tube placement (0.8%), the
16.0%; 10 partial responses). The overall response rate for rate of aspiration pneumonia (0.8%), and the incidence of a
subjects who were previously exposed to both platinum secondary head and neck surgery (1.0%). Among patients
and cetuximab therapy was 19.1% (18/94; 18 partial re- who received intensity-modulated radiation therapy, each
sponses). Pembrolizumab appeared to provide the most additional patient with head and neck cancer treated per
benefit for patients with recurrent/metastatic HNSCC who year by the radiation oncologist resulted in a decreased risk
have had smaller, less heavily pretreated tumors.36 of cancer-related death of 0.8%. Addressing disparities in
As explorations of immunotherapy continue, new indica- health care may involve not just providing availability of
tions may emerge. The prevalence of PD-L1 on tumor- and basic services but focusing on access to high-level expertise
infiltrating immune cells as well as other characteristics for these complex conditions.
of immune biology were investigated among 161 patients Finally, another controversial issue revolves around the
with nasopharyngeal carcinoma.37 PD-L1 expression was relative costs of up-front surgery versus nonsurgical therapy.
detected at greater than 1% and greater than 5% cutoff of There have been several conflicting reports on this question.
induction chemotherapy in 75% and 17% of tumors, re- Pinheiro and Kramer retrospectively reviewed the records of
spectively. Baseline clinical characteristics of stage, sex, 72 patients with T1-T3 OPSCC, of whom 42 were treated

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 181


YOM, GANTI, AND DIETZ

with a surgical approach (22 received adjuvant treatment), group ($38,462 vs. $83,222; p = .03). The cost of surgery
and 30 were treated nonsurgically.41 In this small analysis, followed by adjuvant chemoradiation was similar to that
cost was defined as the reimbursement for all charges in a associated with primary chemoradiation ($84,598 and
6-month episode of treatment after a positive biopsy. The $83,222, respectively; p = .95). In this analysis, the oppor-
authors found that the cost was significantly lower for those tunity for cost savings was seen in patients who did not
treated with surgery only compared with the nonsurgical require adjuvant therapy.

References
1. Cheng CT, Terng SD, Lin CY, et al. 2804 Primary surgery for advanced 15. Weller MA, Ward MC, Berriochoa CA, et al. Cetuximab-based bioradiation
oropharyngeal and hypopharyngeal cancers: a nationwide study in therapy is associated with higher rates of distant metastases than platinum-
Taiwan. Eur J Cancer. 2015;51:S559. based chemoradiation therapy in human papillomaviruspositive oro-
2. Vishnoi JR, Gupta S, Kumar V, et al. 2847 Importance of tumor thickness pharyngeal cancer. Int J Radiat Oncol Biol Phys. 2015, 93:S76-S77.
using intraoral ultrasound in predicting nodal metastasis in early oral 16. Melotek JM, Haraf DJ, Blair EA, et al. Final results of a randomized phase
cancer. Eur J Cancer. 2015;51:S573. 2 trial investigating the addition of cetuximab to induction chemo-
3. Longton E, Lawson G, Installe J, et al. Individualized super selective therapy and accelerated or hyperfractionated chemoradiation therapy
nodal radiation therapy for clinically negative lymph nodes (cN0) in for locoregionally advanced head and neck cancer: HPV-negative subset
head and neck squamous cell carcinoma patients based on sentinel analysis. Int J Radiat Oncol Biol Phys. 2016; 95:868.
node(s) identification: a prospective phase 2 study of SPECT/CT-guided 17. Frakes JM, Naghavi AO, Strom T, et al. Detection of recurrence in human
volume de-escalation. Int J Radiat Oncol Biol Phys. 93:S71-S72. papillomavirus-associated oropharynx squamous cell carcinoma. Int J
4. Panwar A, Batra R, Lydiatt WM, et al. Human papilloma virus positive Radiat Oncol Biol Phys. 2016; 94:866-867.
oropharyngeal squamous cell carcinoma: a growing epidemic. Cancer 18. Pires da Silva I, Mouta J, Winckler P, et al. 2818 The role of neck dis-
Treat Rev. 2014;40:215-219. section (ND) after neoadjuvant chemotherapy (CT) with docetaxel,
5. Zevallos JP, Yim E, Brennan P, et al. Molecular profile of human pap- cisplatin and 5-fluorouracil (TPF) followed by concomitant chemo-
illomavirus positive oropharyngeal squamous cell carcinoma stratified radiation (CR) with cisplatin for patients with locoregionally advanced
by smoking status. Int J Radiat Oncol Biol Phys. 2016, 94:864. squamous cell carcinomas of the head and neck (SCCHN). Eur J Cancer.
6. Lukens JN, Tangsriwong K, Mitra N, et al. Pathological factors predicting 2015;51:S563-S564.
the risk of distant failure for human papillomaviruspositive oro- 19. Mehanna H, Wong WL, McConkey CC, et al. 11LBA Differences in the
pharyngeal squamous cell carcinoma (OPSCC). Int J Radiat Oncol Biol quality of life (QoL) and functional outcomes of treatment between
Phys. 2015; 93:S77. HPV associated (HPV+) and HPV- patients receiving primary chemo-
7. Khwaja SS, Baker C, Haynes W, et al. High E6 gene expression predicts radiotherapy in PET-NECK - a multi-centre randomized phase III con-
for distant metastasis and cancer-specific survival in patients with HPV- trolled trial (RCT) comparing PETCT guided active surveillance with
positive oropharyngeal squamous cell carcinoma. Int J Radiat Oncol Biol planned neck dissection (ND) for locally advanced (N2/N3) nodal
Phys. 2015; 93:S75-S76. metastases (LANM) in patients with head and neck squamous cell
8. Schuler T, Nahar K, Chan D, et al. 2828 Towards predicting tumour cancer (HNC) treated with primary radical chemoradiotherapy (CRT).
response in patients with locally advanced head and neck cancer Eur J Cancer. 2015;51:S715.
treated with neoadjuvant chemotherapy. Eur J Cancer. 2015;51:S567. 20. Elting LS, Cooksley CD, Chambers MS, et al. Risk, outcomes, and costs of
9. Zschaeck S, Lock S, Leger S, et al. 2836 Off-target FDG-PET parameters radiation-induced oral mucositis among patients with head-and-neck
have prognostic value in head and neck squamous cell carcinomas malignancies. Int J Radiat Oncol Biol Phys. 2007;68:1110-1120.
undergoing primary radiochemotherapy and can be used to generate 21. Anderson CM, Allen BG, Sun W, et al. Phase 1b/2a trial of superoxide
radiobiological hypotheses. Eur J Cancer. 2015;51:S570. (SO) dismutase (SOD) mimetic GC4419 to reduce chemoradiation
10. Ou D, Levy A, Blanchard P, et al. 2876 Radiomics feature quantification therapyeInduced oral mucositis (OM) in patients with oral cavity or
in patients with locally advanced head and neck carcinomas treated oropharyngeal carcinoma (OCC). Int J Radiat Oncol Biol Phys. 2016; 94:
with chemoradiotherapy. Eur J Cancer. 2015;51:S582. 869-870.
11. Chera BS, Amdur RJ, Tepper JE, et al. A prospective phase II trial of dein- 22. Merlano M, Vecchio S, Bacigalupo A, et al. 2821 The phase III study
tensified chemoradiation therapy for low risk HPV associated oropharyngeal INTERCEPTOR in locally advanced head and neck cancer (LA-HNC).
squamous cell carcinoma. Int J Radiat Oncol Biol Phys. 2015; 93:S2. Preliminary safety report. Eur J Cancer. 2015;51:S560.
12. Chera BS, Amdur RJ, Tepper JE, et al. Phase 2 trial of deintensified 23. Dewaele E, Vermorken J, Verschueren C, et al. 2817 12-year follow-up
chemoradiation therapy for low-risk human papillomavirus-associated (FU) data and late local toxicity of two cohorts of patients with
oropharyngeal squamous cell carcinoma. Int J Radiat Oncol Biol Phys. locoregionally advanced squamous cell carcinoma of the head and neck
2015; 93:976-985. (LA-SCCHN) treated with concomitant chemoradiation (CCRT) with or
13. Melotek JM, Villaflor VM, Brisson RJ, et al. Response-adapted volume without induction chemotherapy (ICT). Eur J Cancer. 2015;51:S563.
Deescalation (RAVD) in locally advanced head and neck squamous 24. Ahn P, Sharma S, Zhou O, et al. A comparative quality of life cohort of
cell cancer (LA-HNSCC): toxicity and quality of life (QOL) analyses. oropharyngeal squamous cell (OPSCC) patients treated with volumetric
Int J Radiat Oncol Biol Phys. 2015; 93:E335. modulated radiation therapy (VMAT) versus proton pencil beam
14. Melotek JM, Villaflor VM, Karrison TG, et al. Response-adapted volume scanning (PBS). Int J Radiat Oncol Biol Phys. 2015; 93:S71.
de-escalation (RAVD) in locally advanced head and neck cancer: efficacy 25. Bossi P, Pala L, Orlandi E, et al. 2827 Role of induction chemotherapy in
and human papillomavirus-positive subgroup analysis. Int J Radiat the multimodal management of locally advanced epithelial sinonasal
Oncol Biol Phys. 2016; 94:865. cancer. Eur J Cancer. 2015;51:S567.

182 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


WHATS NEW IN HEAD AND NECK CANCER

26. Hamoir M, Schmitz S, Holvoet E, et al. 2848 Salvage surgery in recur- with recurrent/metastatic (RM) head and neck squamous cell cancer
rent head and neck squamous cell carcinoma. Oncologic outcome and (HNSCC): a retrospective analysis. Eur J Cancer. 2015;51:S578.
prognostic factors. Eur J Cancer. 2015;51:S573. 34. Sridharan V, Margalit DN, Curreri SA, et al. Systemic immunologic ef-
27. Chang JHC, Romesser PB, Scher ED, et al. Proton beam reirradiation for fects of definitive radiation in head and neck cancer. Int J Radiat Oncol
recurrent head and neck cancer: multi-institutional report on feasibility Biol Phys. 2016;94:864.
and early outcomes. Int J Radiat Oncol Biol Phys. 2016;94:865. 35. Ferris RL, Kansy BA, Gibson SP, et al. A phase 1b study of neoadjuvant
28. Machiels JP, Haddad RI, Fayette J, et al; LUX-H&N 1 investigators. Afatinib immune biomarker modulation with cetuximab and motolimod in head
versus methotrexate as second-line treatment in patients with recurrent or and neck cancer (HNC). Int J Radiat Oncol Biol Phys. 2016;94:867-868.
metastatic squamous-cell carcinoma of the head and neck progressing on or 36. Chow L, Mehra R, Haddad R, et al. 2866 Antitumor activity of the anti-
after platinum-based therapy (LUX-Head & Neck 1): an open-label, ran- PD-1 antibody pembrolizumab in subgroups of patients with recurrent/
domised phase 3 trial. Lancet Oncol. 2015;16:583-594. metastatic head and neck squamous cell carcinoma (R/M HNSCC):
29. Cohen E, Licitra L, Burtness B, et al. Neck squamous cell carcinoma (R/M exploratory analyses from KEYNOTE-012. Eur J Cancer. 2015;51:S579.
HNSCC) patients (Pts) treated with afatinib versus methotrexate (MTX): 37. Kao HF, Hsu C, Huang HC, et al. 2860 Correlation between plasma
LUX-Head & Neck 1 (LHN1) Int J Radiat Oncol Biol Phys. 2016;94:868- Epstein-Barr virus DNA and clinical response to pembrolizumab in
869. patients with advanced or metastatic nasopharyngeal carcinoma. Eur J
30. Adkins D, Ley J, Wildes T, et al. A phase 1 trial of pazopanib added to Cancer. 2015;51:S576.
cetuximab in patients with incurable head and neck squamous cell 38. Chan OSH, Kowanetz M, Ng WT, et al. 2877 Characterization of PD-L1
carcinoma (HNSCC). Int J Radiat Oncol Biol Phys. 2016;94:868. expression and immune cell infiltration in nasopharyngeal cancer (NPC).
31. Fayette J, Guigay J, Letourneau C, et al. 2800 Cabazitaxel in patients with Eur J Cancer. 2015;51:S582.
refractory recurrent or metastatic (R/M) squamous cell carcinoma of 39. Churilla TM, Egleston B, Dong Y, et al. The impact of health insurance
the head and neck (SCCHN): final results of phase II trial UNICANCER status on the presentation, local management, and outcomes of
ORL03. Eur J Cancer. 2015;51:S557. patients with head and neck cancer in the United States. Int J Radiat
32. Del Barco Morillo E, Mesia R, Adansa Klain JC, et al. 2802 Phase II study Oncol Biol Phys. 2016;94:870.
of first-line paclitaxel (PTX) with panitumumab (P) in patients with 40. Boero IJ, Xu B, Mell LK, et al. Impact of radiation oncologist provider
metastatic or recurrent head and neck cancer: TTCC-2009-03 study. Eur volume on clinical outcomes in head and neck cancer. Int J Radiat Oncol
J Cancer. 2015;51:S558. Biol Phys. 2015; 93:S127.
33. Siano M, Resteghini C, Cau MC, et al. 2865 Outcome of systemic 41. Pinheiro AD, Kramar RW. Determinants of cost in the treatment of T1-T3
treatments after first line platinum and cetuximab treatment in patients oropharynx cancer. Int J Radiat Oncol Biol Phys. 2016;94:866.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 183


HEAD AND NECK CANCER

Integrating Immune Checkpoint


Inhibitors and Targeted Agents
With Surgery and Radiotherapy
for Patients With Head and Neck
Cancer

CHAIR
David M. Brizel, MD
Duke University Medical Center
Durham, NC

SPEAKERS
Andrew G. Sikora, MD, PhD
Baylor College of Medicine
Houston, TX

Nooshin Hashemi Sadraei, MD


University of Cincinnati
Cincinnati, OH
IMMUNOTHERAPY FOR HEAD AND NECK CANCER

Immunotherapy and Checkpoint Inhibitors in Recurrent and


Metastatic Head and Neck Cancer
Nooshin Hashemi Sadraei, MD, Andrew G. Sikora, MD, PhD, and David M. Brizel, MD

OVERVIEW

Immune surveillance is well recognized as an important mechanism to prevent development or progression of head and
neck cancers. Head and neck cancer cells can escape the immune system through multiple mechanisms including de-
velopment of tolerance in T cells and inhibition of T-cellrelated pathways, generally referred to as checkpoint inhibitors.
This article highlights advances in immuno-oncology treatment approaches in recurrent and metastatic head and neck
squamous cell carcinoma. Clinical trials are discussed in detail, with an emphasis on response dynamics, oncologic efficacy,
safety, and tolerability of checkpoint inhibitors. In addition, developing concepts and ongoing studies in this setting are also
reviewed.

T umor cells release antigens, which are captured by an-


tigen presenting cells and subsequently presented to
T cells. Activated T cells then produce a cytotoxic response
thoroughly understood). PD-L1 can also activate CD80,
which, as previously discussed, is a known ligand for CTLA-4.
These preclinical findings have led to a new generation of
resulting in cancer cell death. Memory T cells that are clinical trials and novel treatments based on single and dual
formed subsequent to these events can result in a durable inhibition of CTLA-4 and PD-1, and in combination with
antitumor response. However, T-cellrelated pathways can cytotoxic and targeted therapies.
cause inhibition of these immune responses and result in
tumor cells evading the immune system.1 These pathways
are generally referred to as checkpoints, which are re-
IMMUNE CHECKPOINT INHIBITORS IN
sponsible for preventing a chronic autoimmune or in-
RECURRENT/METASTATIC HEAD AND NECK
flammatory status. Two of the most commonly discussed
SQUAMOUS CELL CARCINOMA
The majority of immune therapy in head and neck cancer has
checkpoint inhibitory mechanisms are CTLA-4 and PD-1/PD-L1,
so far focused on removal of the negative regulatory im-
which act at earlier and later stages of the immune re-
mune pathways. Many ongoing studies are investigating
sponse to tumors.1,2 CTLA-4 is present on the surface of
the role of immune checkpoint inhibitors in recurrent/
CD4 and CD8 cells and, through binding to CD80 and CD86,
metastatic head and neck cancer, including single agent
can transmit an inhibitory signal to T cells. CD28 has a
and combination of checkpoint inhibitors as dual-inhibition,
stimulatory effect on T cells and competes with CTLA-4 for
combination with immunomodulators, vaccines, chemotherapy,
the same ligands (CD80 and CD86); however, CTLA-4 has
biologics, and targeted agents, as well as radiation. Most of
greater affinity and is able to overcome effects of stimu-
these studies have not yet reported their results.
latory CD28.
PD-L1 can be found on tumor cells, as well as immune cells
and antigen presenting cells. PD-1 is primarily found on the PD-1 Inhibitor
surface of T cells. PD-L1 is the main ligand for PD-1, which has PD-1 inhibition has been the most extensively studied im-
been associated with poor prognosis in many cancer types. munotherapeutic strategy in head and neck squamous
PD-1 is expressed on T cells upon activation and results in cell carcinoma, particularly with the agent pembrolizumab
negative regulatory effects on immune function. (formerly MK-3475). This agent has shown promising effi-
PD-1 and PD-L1 both interact with other ligands, too. Most cacy compared with historical data and is thought to be well
importantly, PD-1/PD-L2 activation can have similar effects tolerated (KEYNOTE 012; NCT01848834).3 In a large phase IB
as PD-1/PD-L1 engagement (although PD-L2 function is less study of pembrolizumab for patients with PD-L1positive

From the Division of Medical Oncology, University of Cincinnati College of Medicine, Cincinnati, OH; Department of OtolaryngologyHead and Neck Surgery, Baylor College of Medicine,
Houston, TX; Department of Radiation Oncology, Duke University Cancer Center, Durham, NC.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Nooshin Hashemi Sadraei, MD, The Vontz Center for Molecular Studies, University of Cincinnati, 3125 Eden Ave., Cincinnati, OH 45267; email: hashemnn@
ucmail.uc.edu.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e277


HASHEMI SADRAEI, SIKORA, AND BRIZEL

recurrent/metastatic head and neck squamous cell carci- cohort of patients was 9.6 months (6.6 months to not
noma, the overall response rate was approximately 20%, reached) with a 1-year estimated survival of 47%.5 This is a
which was similar among patients who were HPV-positive very promising improvement compared with best historical
or HPV-negative.4 PD-L1 positivity in this study was defined survival outcome data among patients with recurrent/
as at least 1% or more stromal or tumor expression of PD-L1 metastatic disease (median survival, 10.1 months with
by immunohistochemistry. Based on this definition, 78% of chemotherapy doublet-cetuximab triplet regimen and
screened patients were considered positive and eligible for 7.4 months with chemotherapy doublet therapy).6
the study. Among patients with PD-L1 positivity, those with Overall drug-related adverse events were 61%, with fa-
higher PD-L1 expression demonstrated a much higher re- tigue (17%) being the most common, followed by endo-
sponse compared with those with a lower PD-L1 expression crinopathies (hypothyroidism), low appetite, pyrexia, and a
(50% vs. 11%, respectively).4 variety of skin-related toxicities, each reported in 5% to 8%
Median progression-free (PFS) and overall survival (OS) of patients. Compared with the use of other checkpoint
were favorable compared with historical cohorts of pa- inhibitors, mostly in melanoma, no colitis or pneumonitis of
tients with heavily pretreated head and neck squamous any grades were observed, and high-grade toxicities (grade
cell carcinoma; the median PFS was 9 months (range, 3 and above) were seen in 12% of cases.4,5
8 to 20 months) and the median OS was 12.6 months. Many randomized studies subsequently investigated the
Unlike response rates, which did not seem to be affected role of PD-1 inhibitors in recurrent and metastatic disease.
by HPV status, patients who were HPV-positive, as pre- A randomized study of pembrolizumab compared with
dicted, showed longer PFS and OS compared with pa- investigators choice chemotherapy (KEYNOTE-040) for
tients who were HPV-negative (median PFS, 17 vs. 8 months, patients with platinum-refractory disease is nearing com-
respectively; median OS, not reached vs. 9 months, pletion of accrual (NCT02252042),7 and another trial is a
respectively). three-arm randomized study in the first-line recurrent and
Subsequently, in a combined update of two cohorts of metastatic setting (KEYNOTE-048) that will evaluate stan-
patients with recurrent/metastatic head and neck squamous dard platinum/5-fluorouracil/cetuximab compared with
cell carcinoma that was heavily pretreated, pembrolizumab pembrolizumab alone or combination of chemotherapy
resulted in a response rate of 24% and an additional 25% plus pembrolizumab (NCT02358031).8
with stable disease,5 which was slightly improved compared More recently, CheckMate 141, a study of nivolumab,
with earlier reports.4 The median survival in the combined another PD-1 inhibitor, was stopped early due to a survival
advantage observed among patients who were treated with
nivolumab over investigators choice of therapy (cetuximab,
methotrexate, or docetaxel) in a second-line, recurrent/
KEY POINTS metastatic setting. Details of the results have not yet
been released (NCT02105636). 9
Immune surveillance is an important mechanism in
prevention of cancer development and inhibition of
growth of tumors and metastasis. Checkpoint pathways PD-L1 Inhibitor
prevent immune response through multiple methods Durvalumab (formerly MEDI4736) is an antiPD-L1 mono-
including CTLA-4 and PD-1/PD-L1 pathways. clonal antibody that was evaluated in a large global, mul-
A variety of immune checkpoint inhibitors (PD-1/PD-L1 ticenter, multiarm phase I/II trial in multiple disease
and CTLA-4 inhibitors) have been studied in recurrent
sites, including head and neck squamous cell carcinoma
and metastatic head and neck cancers with encouraging
efficacy and toxicity data.
(NCT01693562/CD-ON-MEDI4736-1108).10 This large trial
Although many of these treatments are being used with included components of dose escalation and dose explo-
encouraging results in a variety of different ration to define optimum biologic dose. Dose cohorts
malignancies, there appears to be some differences in started at 0.1 mg/kg and escalated in five consecutive co-
patterns of response and toxicities, which may be horts to 10 mg/kg, which was then taken to the expansion
disease- or population-specific, to some extent. phase as the selected dose. Although the overall rate of
Studies are ongoing to identify reliable biomarkers. drug-related adverse events was high (46%), 10 mg/kg of
To date, despite common use of PD-L1 as a biomarker, durvalumab every 2 weeks resulted in grade 3 or 4 drug-
consensus in detection methods, definition of positivity, related adverse events (determined by investigators) in only
and its use as a prognostic or predictive biomarker is 7% of patients with head and neck cancer, a rate that was
lacking, despite common use of PD-L1 as a biomarker.
similar to other malignancies.11 Among all types of cancer,
In addition to checkpoint inhibition, a variety of other
immune therapy strategies are under investigation in
fatigue, gastrointestinal symptoms, endocrinopathies,
head and neck cancers, some of which include dyspnea, peripheral neuropathy, and a variety skin rashes
combination strategies, positive costimulation, and were reported as more common drug-related adverse
targeting HPV-specific antigens in treatment of events. Drug-related colitis did not occur, and drug-related
HPV-related disease. pneumonitis (grade 1 to 2 only) was only reported in 3%
of patients with head and neck cancer. There were no

e278 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


IMMUNOTHERAPY FOR HEAD AND NECK CANCER

drug-related discontinuations, and, overall, durvalumab was chemotherapy. This study is powered to evaluate the coprimay
well-tolerated among patients with head and neck cancer and endpoints of PFS and OS (EAGLE; NCT02369874).17
was similar to responses observed in other cancers.12 Among
eight tumor types, response was seen as early as 5 weeks and PD-L1 Inhibitor in Combination With PD-1 Inhibitor
was very durable (56 weeks) in some cases. Overall response Some ongoing studies in advanced solid tumors are evaluating
rate was 10%, which was generally higher among pa- dual PD-1 (MEDI0680)/PD-L1 (durvalumab) inhibition.18 These
tients who were PD-L1positive (22% vs. 5%), and, once re- studies are currently in earlier phases of dose escalation and
sponse was achieved, duration of response was comparable expansion (NCT02118337).19
among patients who were PD-L1positive and PD-L1negative.11
Patients demonstrated a similar pattern: the response rate ADDITIONAL IMMUNE THERAPY STRATEGIES
was slightly higher among patients who were PD-L1 Positive Costimulation
positive versus PD-L1negative (18% vs. 8%, respectively) Another strategy to augment immune system recognition
with a respectable duration of response (16 to over 55 weeks). of cancer cells has focused on enhancement of positive
The overall response rate among patients with head and neck stimulatory pathways through cytokines and monoclonal
cancer was 11%, and the disease control rate (response plus antibodies.
stable disease) at 6 months was 15% (18% for PD-L1positive Preclinical data suggest that Toll-like receptor 8 (TLR8)
vs. 11% for PD-L1negative).12 activation subsequently activates monocytes, natural killer
These findings were very encouraging from a safety and cells, and myeloid-derived dendritic cells and can potentially
tolerability standpoint and showed promising improvement have synergistic effect with chemotherapy and increase the
in response and duration of disease control, particularly in effectiveness of monoclonal antibody treatment.20
the PD-L1positive population. A randomized study of platinum/5-fluorouracil/cetuximab
plus a TLR8 agonist (VTX-2337) or placebo recently com-
pleted accrual in first-line recurrent/metastatic head and
PD-L1 Inhibitor in Combination With CTLA-4 Inhibitor neck squamous cell carcinoma (NCT01836029).21 Many
A new series of combination studies in head and neck other products are being investigated as single agent or in
squamous cell carcinoma were initiated based on additive or combination, including the CD137 agonist, urelumab
synergistic effects of drug combinations observed in pre- (NCT02110082, NCT02253992),22,23 and Ox40 monoclonal
clinical studies or other diseases. The PD-1 (nivolumab) and antibodies (in other malignancies).
CTLA-4 inhibitor (ipilimumab) combination was shown to
be superior to CTLA inhibition alone among patients with
metastatic malignant melanoma and superior to either EGFR Monoclonal Antibody
CTLA-4 or PD-1 inhibition alone among patients who were Cetuximab is a mouse-human chimeric monoclonal antibody
PD-L1negative, resulting in significantly prolonged PFS.13 against EGFR, a well-recognized target in head and neck
Other emerging data support distinct mechanisms and a squamous cell carcinoma.24 Cetuximab has shown efficacy
synergistic effect of CTLA-4 and PD-L1 inhibition on immune in recurrent and metastatic head and neck squamous cell
regulation, which results in improved tumor response in carcinoma and in combination with radiation in the curative
mouse models and for patients.14 setting.25,26 Several studies have demonstrated immuno-
Multiple ongoing studies are currently evaluating the role modulatory effects with EGFR monoclonal antibodies
of dual checkpoint inhibition in head and neck squamous through multiple mechanisms including tumor-antigen
cell carcinoma. Some of these studies are directed toward specific T-cell activation27,28 as a result of T-cell activation
PD-L1negative disease and others are stratified based on through interaction with antigen presenting cells. Ongoing
PD-L1 status. studies are investigating cetuximab combination with
Patients with recurrent/metastatic head and neck squa- different checkpoint inhibitors, for example nivolumab
mous cell carcinoma whose disease progresses during (NCT02124850),29 ipilimumab (NCT01860430),30 and ure-
platinum therapy will be offered PD-L1 inhibitor alone (those lumab (NCT02110082).22
who are PD-L1positive; 10 mg/kg durvalumab every
2 weeks; HAWK; NCT02207530)15 or those who are PD- Therapeutic HPV Vaccines
L1negative will be randomly assigned to one of three arms Given the encouraging results with HPV-specific vaccine
to receive combination PD-L1 and CTLA-4 inhibition prevention studies,31 many ongoing trials are focused on
(durvalumab and tremelimumab), PD-L1 inhibitor alone treatment of already established HPV-related head and neck
(durvalumab), or CTLA-4 inhibitor alone (tremelimumab; cancer. Subcutaneous injection of HPV-16 peptide vaccine
CONDOR; NCT02319044).16 The primary endpoint in both for patients with recurrent/metastatic head and neck
studies is overall response rate. A separate large phase III squamous cell carcinoma was well tolerated and able to
study will randomly assign patients after PD-L1 expression stimulate cellular and humoral immune response.32 Other
based stratification to receive PD-L1 inhibitor alone (durva- studies have used bacterial or viral vaccine-base to deliver
lumab), dual PD-L1/CTLA-4 inhibitor therapy (durvalu- tumor-specific antigen and to use the dual advantage of bacterial/
mab and tremelimumab), or standard-of-care single-agent viral-induced immunogenicity as well as tumor-specific

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e279


HASHEMI SADRAEI, SIKORA, AND BRIZEL

antigens like E6 and E7, which have a well-established role in the Keynote 012 pembrolizumab study, none of the
in HPV-related malignancies.33 132 reported patients had a RECIST-defined radiographic
Many such studies are ongoing or in the early develop- progression of disease prior to response).38 These data
ment phase and some include multiple HPV-related malig- suggest that, in comparison with patients with melanoma
nancies (NCT02526316, NCT00019110).34,35 receiving a CTLA-4 inhibitor, clinicians must have a lower
threshold for recognizing increase in size of disease as
true progression, especially if accompanied by worsening
PREDICTORS OF RESPONSE TO CHECKPOINT symptoms.38,40
INHIBITORS
A post hoc analysis of nearly 200 patients treated with
pembrolizumab showed that patients who were less heavily
retreated (two or less prior lines of therapy) had higher re-
PROGRESS AND CHALLENGES
Immune-modulating treatments appear to have a clear
sponse rates compared with heavily pretreated patients (32%
clinical benefit for patients with head and neck squamous
vs. 16%, respectively). Patients who had smaller volumes
cell carcinoma, as shown by multiple clinical studies in the
of disease had improved response compared with those
recurrent/metastatic setting. This benefit seems to be in-
with larger volumes of disease (36% vs. 14%, respectively).5
dependent of previous treatment, even being observed
Small-volume disease was defined in one study as being
among patients who have received multiple lines of therapy
less than the median of the sum of the size of the lesions,
and who have been assumed to have exhausted all options.
where the median was 96 mm (range, 10 to 664 mm).5
Although patients with poor performance status and mul-
Patients with higher PD-L1 expression seem to derive a
tiple comorbidities have been excluded from these early
greater benefit as demonstrated by a much higher response
trials, the safety and toxicity profiles of these drugs have
compared with those with a lower PD-L1 expression when
been very promising, suggesting applicability to a wider
treated with pembrolizumab (50% vs. 11%, respectively)4 or
population of patients compared with many cytotoxic
durvalumab (22% PD-L1positive vs. 5% PD-L1negative).12
chemotherapeutic agents.
Interestingly, unlike many prior chemotherapy studies,
Many questions remain to be answered. As far as design of
patients with HPV-negative and HPV-positive disease did not
future studies, perhaps most importantly, we must address
demonstrate different response rates when treated with
whether the primary benefit should be measured by response or
PD-1 inhibition (25% HPV-negative vs. 24% HPV-positive).5
survival. Although, among patients who have been heavily
pretreated, response rates of 10% to 20% are considered re-
RESPONSE EVALUATION spectable (compared with the 3% to 13% historical response rates
Many recent studies have acknowledged differences in time among previously treated recurrent/metastatic patients25,41-43),
to initiation of response, as well as patterns of response it is the duration of response and the improvement in OS that
between immune therapy and cytotoxic chemotherapy. clearly differentiates these drugs from chemotherapy (median
Although chemotherapy and targeted therapy directly exert time to progression, 2 to 4 months; median OS, 6 to 7 months
their effect on cancer cells and can result in a more rapid in chemotherapy studies of patients who were previously
development of response, radiographic responses to im- treated25,41,43). The use of response as a primary endpoint for
mune therapy develop more slowly because of the nature of some studies may therefore underestimate the actual benefit.40
its indirect effect on cancer cells through activation or It also appears that the Immune-Related Response Criteria
disinhibition of the immune system. The original CTLA-4 (irRECIST), which was originally developed based on response
inhibitor studies in melanoma (with ipilimumab) showed a evaluation for patients with melanoma who were undergoing
rather slow time to initiation of response (3 to 4 months)36,37; CTLA-4 inhibition, may not be the most accurate method of
however, the more recent PD-1 (pembrolizumab) and PD-L1 measuring response (and hence benefit) across all checkpoint
(durvalumab) inhibitors demonstrate a shorter time to re- inhibitors and all types of disease.39,40,44 The search for a re-
sponse, as early as 5 weeks,12 with a median time to response liable biomarker to predict clinic benefit continues. The
of 9 weeks (range, 7 to 18 weeks).5,38 prognostic value of PD-1 and PD-L1 expression has varied
One of the challenges in response assessment and de- across studies, potentially because of the difficulty of accurately
termination of progression of disease with such treatments quantifying expression levels and determining appropriate
has been a concern for tumor-flare, a phenomenon that has cutoff levels for antigen positivity. An additional confounder
been attributed to initial T-cell infiltration into the tumor, is that upregulation of PD-1 and PD-L1 can be induced by
causing increase in the size of lesions. This finding has been interferon-gamma, one hallmark of a strong intratumoral T-cell
mostly reported among patients with melanoma who were response.45-47 In fact, several studies across different cancer
treated with CTLA-4 inhibition.37,39 Interestingly, unlike types have shown positive association of PD-L1 expression with
patients with melanoma undergoing treatment with ipili- survival in head and neck squamous cell carcinoma48,49 and
mumab, there seem to be very few patients with head and other cancers.50-52 This seemingly paradoxical association is
neck cancer who experience initial radiographic progression consistent with upregulation of PD-1/PD-L1 in response to host
of disease during treatment with PD-1 or PD-L1 inhibitors immune pressure and suggests that these molecules may serve
who later show response to these treatments (for example, as a potential biomarker of vigorous antitumor immunity.

e280 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


IMMUNOTHERAPY FOR HEAD AND NECK CANCER

As the field of biomarker research is evolving, an additional unrecognized population of long-term survivors with these
candidate biomarker of response, consistent with PD-1/PD- treatments. We must improve our understanding of the role
L1 axis upregulation induced by antitumor immune of PD-L1 positivity and other clinical and biologic predictive
response, is the presence of so-called T-cell inflamed biomarkers in patient selection and whether a specific pa-
phenotype gene expression signatures,53 which has been tient population benefits more from single-agent versus
suggested to predict benefit from antiPD-1 in patients combination therapy. Other important issues include finding
with head and neck squamous cell carcinoma.54 the best combination treatment (with other immuno-oncology
products, chemotherapy, targeted therapy, or radiation), se-
CONCLUSION quencing approach, and duration of treatment and whether
In summary, even in the absence of a RECIST- or irRECIST- these treatments should be introduced earlier in more curative
defined response, there appears to be a heretofore settings.

References
1. Pardoll DM. The blockade of immune checkpoints in cancer immu- 14. Twyman-Saint Victor C, Rech AJ, Maity A, et al. Radiation and dual
notherapy. Nat Rev Cancer. 2012;12:252-264. checkpoint blockade activate non-redundant immune mechanisms in
2. Shekarian T, Valsesia-Wittmann S, Caux C, et al. Paradigm shift in cancer. Nature. 2015;520:373-377.
oncology: targeting the immune system rather than cancer cells. 15. NCT02207530. Phase II Study of MEDI4736 Monotherapy in Treatment
Mutagenesis. 2015;30:205-211. of Recurrent or Metastatic Squamous Cell Carcinoma of the Head
3. NCT01848834. Study of Pembrolizumab (MK-3475) in Participants and Neck. https://clinicaltrials.gov/ct2/show/NCT02207530. Accessed
With Advanced Solid Tumors (MK-3475-012/KEYNOTE-012). https:// March 18, 2016.
clinicaltrials.gov/ct2/show/NCT01848834. Accessed March 18, 2016. 16. NCT02319044. Phase II Study of MEDI4736, Tremelimumab, and
4. Chow LQ, Burtness B, Weiss J, et al. A phase Ib study of pembrolizumab MEDI4736 in Combination w/ Tremelimumab Squamous Cell Carci-
(pembro; MK-3475) in patients (pts) with human papilloma virus (HPV)- noma of the Head and Neck. https://clinicaltrials.gov/ct2/show/
positive and negative head and neck cancer (HNC). Ann Oncol. 2014;25: NCT02319044. Accessed March 18, 2016.
v1-v41 (suppl 5; abstr LBA31). 17. NCT02369874. Study of MEDI4736 Monotherapy and in Combination
5. Chow L, Mehra R, Haddad R, et al. Antitumor activity of the anti-PD-1 With Tremelimumab Versus Standard of Care Therapy in Patients With
antibody pembrolizumab in subgroups of patients with recurrent/ Head and Neck Cancer. https://clinicaltrials.gov/ct2/show/NCT02369874.
metastatic head and neck squamous cell carcinoma (R/M HNSCC): Accessed March 18, 2016.
exploratory analyses from KEYNOTE-012. Eur J Cancer. 2015;51:S579 18. Hamid O, Chow LQM, Tavakkoli F, et al. Phase I, open-label study of
(suppl; abstr 2866). MEDI0680, an anti-programmed cell death-1 (PD-1) antibody, in
6. Vermorken JB, Mesia R, Rivera F, et al. Platinum-based chemotherapy combination with MEDI4736, an anti-programmed cell death ligand-1
plus cetuximab in head and neck cancer. N Engl J Med. 2008;359: (PD-L1) antibody, in patients with advanced malignancies. J Clin Oncol.
1116-1127. 2015;33 (suppl; abstr TPS3087).
7. NCT02252042. Pembrolizumab (MK-3475) Versus Standard Treatment 19. NCT02118337. A Phase 1, Open-label Study to Evaluate the Safety and
for Recurrent or Metastatic Head and Neck Cancer (MK-3475-040/ Tolerability of MEDI0680 (AMP-514) in Combination With MEDI4736 in
KEYNOTE-040). https://clinicaltrials.gov/ct2/show/NCT02252042. Subjects With Advanced Malignancies. https://clinicaltrials.gov/ct2/
Accessed March 18, 2016. show/NCT02118337. Accessed March 18, 2016.
8. NCT02358031. A Study of Pembrolizumab (MK-3475) for First Line 20. Lu H, Dietsch GN, Matthews MA, et al. VTX-2337 is a novel TLR8 agonist
Treatment of Recurrent or Metastatic Squamous Cell Cancer of the that activates NK cells and augments ADCC. Clin Cancer Res. 2012;18:
Head and Neck (MK-3475-048/KEYNOTE-048). https://clinicaltrials. 499-509.
gov/ct2/show/NCT02358031. Accessed March 18, 2016. 21. NCT01836029. Chemotherapy Plus Cetuximab in Combination With
9. NCT02105636. Trial of Nivolumab vs Therapy of Investigators Choice in VTX-2337 in Patients With Recurrent or Metastatic Squamous Cell
Recurrent or Metastatic Head and Neck Carcinoma (CheckMate 141). Carcinoma of the Head and Neck. https://clinicaltrials.gov/ct2/show/
https://clinicaltrials.gov/ct2/show/NCT02105636. Accessed March 18, NCT01836029. Accessed March 18, 2016.
2016. 22. NCT02110082. Combination Study of Urelumab and Cetuximab in
10. NCT01693562. A Phase 1/2 Study to Evaluate MEDI4736. https:// Patients With Advanced/Metastatic Colorectal Cancer or Advanced/
clinicaltrials.gov/ct2/show/NCT01693562. Accessed March 18, 2016. Metastatic Head and Neck Cancer. https://clinicaltrials.gov/ct2/show/
11. Segal NH, Hamid O, Hwu W, et al. A phase I multi-arm dose-expansion NCT02110082. Accessed March 18, 2016.
study of the anti-programmed cell death-ligand-1 (PD-L1) antibody 23. NCT02253992. Study of the Safety and Tolerability of Urelumab Ad-
MEDI4736: preliminary data. Ann Oncol. 2014;25:iv365 (suppl 4; abstr ministered in Combination With Nivolumab in Solid Tumors and B-cell
1058PD). Non-Hodgkins Lymphoma. https://clinicaltrials.gov/ct2/show/NCT02253992.
12. Segal NH, Ou SHI, Balmanoukian AS, et al. Safety and efficacy of Accessed March 18, 2016.
MEDI4736, an anti-PD-L1 antibody, in patients from a squamous cell 24. Grandis JR, Zeng Q, Drenning SD, et al. Normalization of EGFR mRNA
carcinoma of the head and neck (SCCHN) expansion cohort. J Clin Oncol. levels following restoration of wild-type p53 in a head and neck
2015;33: (suppl; abstr 3011). squamous cell carcinoma cell line. Int J Oncol. 1998;13:375-378.
13. Larkin J, Hodi FS, Wolchok JD. Combined nivolumab and ipilimumab 25. Vermorken JB, Trigo J, Hitt R, et al. Open-label, uncontrolled, multi-
or monotherapy in untreated melanoma. N Engl J Med. 2015;373: center phase II study to evaluate the efficacy and toxicity of cetuximab
1270-1271. as a single agent in patients with recurrent and/or metastatic squamous

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e281


HASHEMI SADRAEI, SIKORA, AND BRIZEL

cell carcinoma of the head and neck who failed to respond to platinum- 41. Argiris A, Ghebremichael M, Gilbert J, et al. Phase III randomized,
based therapy. J Clin Oncol. 2007;25:2171-2177. placebo-controlled trial of docetaxel with or without gefitinib in re-
26. Bonner JA, Harari PM, Giralt J, et al. Radiotherapy plus cetuximab for current or metastatic head and neck cancer: an eastern cooperative
squamous-cell carcinoma of the head and neck. N Engl J Med. 2006;354: oncology group trial. J Clin Oncol. 2013;31:1405-1414.
567-578. 42. Fury MG, Sherman E, Lisa D, et al. A randomized phase II study of
27. Srivastava RM, Lee SC, Andrade Filho PA, et al. Cetuximab-activated cetuximab every 2 weeks at either 500 or 750 mg/m2 for patients with
natural killer and dendritic cells collaborate to trigger tumor antigen- recurrent or metastatic head and neck squamous cell cancer. J Natl
specific T-cell immunity in head and neck cancer patients. Clin Cancer Compr Canc Netw. 2012;10:1391-1398.
Res. 2013;19:1858-1872. 43. Stewart JS, Cohen EE, Licitra L, et al. Phase III study of gefitinib
28. Ferris RL. Immunology and immunotherapy of head and neck cancer. compared with intravenous methotrexate for recurrent squamous
J Clin Oncol. 2015;33:3293-3304. cell carcinoma of the head and neck [corrected]. J Clin Oncol. 2009;27:
29. NCT02124850. A Phase Ib Study of Neoadjuvant of Cetuximab Plus 1864-1871.
Motolimod and Cetuximab Plus Motolimod Plus Nivolumab. https:// 44. Hoos A, Wolchok JD, Humphrey RW, et al. CCR 20th Anniversary
clinicaltrials.gov/ct2/show/NCT02124850. Accessed March 18, 2016. commentary: immune-related response criteriacapturing clinical ac-
30. NCT01860430. A Phase Ib Trial of Concurrent Cetuximab (ERBITUX) and tivity in immuno-oncology. Clin Cancer Res. 2015;21:4989-4991.
Intensity Modulated Radiotherapy (IMRT) With Ipilimumab (YERVOY) in 45. Concha-Benavente F, Srivastava RM, Trivediet S, et al. Identification of
Locally Advanced Head and Neck Cancer. https://clinicaltrials.gov/ct2/ the cell-intrinsic and -extrinsic pathways downstream of EGFR and
show/NCT01860430. Accessed March 18, 2016. IFNgamma that induce PD-L1 expression in head and neck cancer.
31. Schiller JT, Castellsague X, Garland SM. A review of clinical trials of Cancer Res. 2016;76:1031-1043.
Human papillomavirus prophylactic vaccines. Vaccine. 2012;30(Suppl 46. Abiko K, Matsumura N, Hamanishi J, et al. IFN-gamma from lympho-
5):F123-F138. cytes induces PD-L1 expression and promotes progression of ovarian
32. Zandberg DP, Rollins S, Goloubeva O, et al. A phase I dose escalation trial cancer. Br J Cancer. 2015;112:1501-1509.
of MAGE-A3- and HPV16-specific peptide immunomodulatory vaccines in 47. Fang W, Zhang J, Honget S, et al. EBV-driven LMP1 and IFN-gamma up-
patients with recurrent/metastatic (RM) squamous cell carcinoma of the regulate PD-L1 in nasopharyngeal carcinoma: implications for onco-
head and neck (SCCHN). Cancer Immunol Immunother. 2015;64:367-379. targeted therapy. Oncotarget. 2014;5:12189-12202.
33. Sewell DA, Pan ZK, Paterson Y. Listeria-based HPV-16 E7 vaccines limit 48. Vassilakopoulou M, Avgeris M, Velcheti V, et al. Evaluation of PD-L1
autochthonous tumor growth in a transgenic mouse model for HPV-16 expression and associated tumor-infiltrating lymphocytes in laryngeal
transformed tumors. Vaccine. 2008;26:5315-5320. squamous cell carcinoma. Clin Cancer Res. 2016;22:704-713.
34. NCT02526316. Cisplatin-based Chemotherapy Combined With P16_37- 49. Oliveira-Costa JP, de Carvalho AF, da Silveira da GG, et al. Gene ex-
63 Peptide Vaccination in Patients With HPV-positive Cancers (VICORYX-2). pression patterns through oral squamous cell carcinoma development:
https://clinicaltrials.gov/ct2/show/NCT02526316. Accessed March 18, PD-L1 expression in primary tumor and circulating tumor cells. Onco-
2016. target. 2015;6:20902-20920.
35. NCT00019110. Vaccine Therapy in Treating Patients With Advanced 50. Droeser RA, Hirt C, Viehl CT, et al. Clinical impact of programmed cell
or Recurrent Cancer. https://clinicaltrials.gov/ct2/show/NCT00019110. death ligand 1 expression in colorectal cancer. Eur J Cancer. 2013;49:
Accessed March 18, 2016. 2233-2242.
36. Hodi FS, ODay SJ, McDermott DF, et al. Improved survival with ipili- 51. Lipson EJ, Vincent JG, Loyo M, et al. PD-L1 expression in the Merkel cell
mumab in patients with metastatic melanoma. N Engl J Med. 2010;363: carcinoma microenvironment: association with inflammation, Merkel
711-723. cell polyomavirus and overall survival. Cancer Immunol Res. 2013;1:
37. Weber JS, ODay S, Urba W, et al. Phase I/II study of ipilimumab 54-63.
for patients with metastatic melanoma. J Clin Oncol. 2008;26:5950-5956. 52. Taube JM, Anders RA, Young GD, et al. Colocalization of inflammatory
38. Seiwert TY, Haddad RI, Gupta S, et al. Antitumor activity and safety of response with B7-h1 expression in human melanocytic lesions supports
pembrolizumab in patients (pts) with advanced squamous cell carci- an adaptive resistance mechanism of immune escape. Sci Transl Med.
noma of the head and neck (SCCHN): Preliminary results from KEYNOTE- 2012;4:127ra37.
012 expansion cohort. J Clin Oncol. 2015;33 (suppl; abstr LBA6008). 53. Gajewski TF, Louahed J, Brichard VG. Gene signature in melanoma
39. Wolchok JD, Hoos A, ODay S, et al. Guidelines for the evaluation of associated with clinical activity: a potential clue to unlock cancer im-
immune therapy activity in solid tumors: immune-related response munotherapy. Cancer. 2010;16:399-403.
criteria. Clin Cancer Res. 2009;15:7412-7420. 54. Seiwert TY, Burtness B, Weiss J, et al. Inflamed-phenotype gene ex-
40. Seiwert T. Immunotherapy for head and neck cancer. Paper presented pression signatures to predict benefit from the anti-PD-1 antibody
at: 2016 Multidisciplinary Head and Neck Cancer Symposium; February pembrolizumab in PD-L1+ head and neck cancer patients. J Clin Oncol.
2016; Scottsdale, AZ. 2015;33:15s (suppl; abstr 6017).

e282 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


HEAD AND NECK CANCER

Multimodality Management of
Locoregionally Recurrent or
Second Primary Head and Neck
Cancer

CHAIR
Stuart J. Wong, MD
Medical College of Wisconsin
Milwaukee, WI

SPEAKERS
Dwight E. Heron, MD
University of Pittsburgh Medical Center
Pittsburgh, PA

Kerstin Stenson, MD
Rush University Medical Center
Chicago, IL
WONG ET AL

Locoregional Recurrent or Second Primary Head and Neck


Cancer: Management Strategies and Challenges
Stuart J. Wong, MD, Dwight E. Heron, MD, MBA, FACRO, FACR, Kerstin Stenson, MD, FACS, Diane C. Ling,
MD, and John A. Vargo, MD

OVERVIEW

Treatment of patients with locoregional recurrent or second primary head and neck squamous cell cancer (HNSCC) has been
guided by well-reasoned principles and informed by carefully tested chemotherapy and radiation regimens. However,
clinical decision making for this population is complicated by many factors. Although surgery is generally considered the
treatment of choice for patients with HNSCC with recurrent disease or new second primary disease in a previously irradiated
field, operability of cases is not always straightforward. Postoperative treatment is frequently warranted but carries
significant risk. In addition, the rapid rise in the incidence of HPV-associated HNSCC raises the question of whether
established treatment paradigms should be re-examined in this population of patients with a much better prognosis than the
non-HPV population. Furthermore, new radiation techniques and new systemic agents show early promising results in
recent clinical studies, suggesting potential for practice-changing effects in the future management of this disease. This
article examines each of the treatment modalities used in the care of patients with HNSCC with recurrent or new second
primary disease and provides a perspective to aid clinicians in the management of this disease.

T he majority of patients newly diagnosed with HNSCC


present with locally advanced cancer and undergo
combined modality therapy, including radiation therapy. For
appreciated. Examination of the treatment for patients
with HPV-positive OPC in Radiation Therapy Oncology
Group (RTOG) studies provides a clear picture as to the
this patient group who subsequently fail in a local or regional failure patterns of this patient population.2 This analysis
site, the therapeutic options are complicatedmost com- showed that following disease progression, 2-year overall
monly because of overlap of the previous radiation portal. In survival was nearly twice as high for patients with HPV-
addition, the management of locoregional recurrent or positive OPC compared with patients who were HPV-
second primary head and neck cancer has grown in com- negative (55% vs. 28%, respectively). The majority of
plexity as a result of the increase of the relative incidence these patients fail within 1 year after completing de-
of HPV-associated HNSCC compared with non-HPV HNSCC. finitive therapy. Although ostensibly it is logical to apply
Practicing oncologists have more treatment options at their the same oncologic principles to each of these two patient
disposal, which further complicates the treatment decision- groups, it is important to note that we do not have
making process. For instance, reirradiation was considered abundant clinical trial information with long-term follow-
a treatment option limited to high-volume centers with up data to guide these treatment decisions. The sequence
abundant experience with this therapy. It is now far more of salvage treatment options for these two categories of
acceptable to use reirradiation outside of the clinical trial patients given the differences in outcome remains the
setting. The wide use of this salvage treatment modality is subject of future investigation.
evident by numerous publications demonstrating safety and In the following discussion, we will review treatment
efficacy of concurrent chemotherapy and reirradiation. options for patients with recurrent and second primary
It is well recognized that diagnosis of HPV-associated HNSCC in the context of the evolving epidemiologic patterns
HNSCC carries a more favorable prognosis compared with of this disease. In particular, we will discuss surgical ap-
non-HPV HNSCC.1 However, the failure pattern of HPV- proaches to this disease, the use of new and conventional
associated oropharynx cancer (OPC) and its sensitivity to radiation therapy techniques, and systemic therapeutic
treatment in the setting of recurrent disease is less well options.

From the University of Pittsburgh Cancer Institute, Pittsburgh, PA; University of Pittsburgh School of Medicine, Pittsburgh, PA; Rush University Medical Center, Chicago, IL; Medical
College of Wisconsin, Milwaukee, WI.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Stuart J. Wong, MD, Medical College of Wisconsin, 9200 West Wisconsin Ave., Milwaukee, WI 53226; email: swong@mcw.edu.

2016 by American Society of Clinical Oncology.

e284 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


RECURRENT HEAD AND NECK CANCER

SURGICAL MANAGEMENT OF PREVIOUSLY radiation or CRT.5,6 For patients and physicians to gain
IRRADIATED HEAD AND NECK CANCER: SALVAGE perspective and evaluate risk/benefit ratio of surgery, pa-
AND TREATMENT MANAGEMENT STRATEGIES tients with advanced head and neck cancer who have
BEFORE AND AFTER REIRRADIATION completed radiation or CRT need a full metastatic workup
The goals of multimodality strategies for patients with ad- before surgery. This includes CT scan of the head, neck, and
vanced head and neck cancer encompass organ preservation chest and/or PET scan.
through less radical surgery while improving outcomes. As
survival improved and multimodality organ preservation Creating a safe wound. Experience and literature docu-
strategies became more successful, the role of surgery was ment the increased risk of fistula and other complications for
redefined and has become increasingly more prominent for patients undergoing salvage surgery who have received
patients with recurrent disease. Most surgeons hold a general prior radiation or CRT. Use of nonirradiated free tissue has
dogma that performing surgery on a patient who has pre- been shown to provide reliable reconstruction that allows
viously undergone radiation or chemoradiation (CRT) can be for maximal resection, unhampered by concerns of ade-
a challenging task. The salvage surgical approach may be not quate tissue availability.7-10 For patients who undergo
only technically challenging but also associated with a higher surgery after prior irradiation, free tissue transfer with
incidence of complications, such as wound-healing problems, microvascular anastomoses allows for successful wound
fistula, and infection. This discussion will address some of the rehabilitation.11 Paramount in caring for our patients who
challenges that patients with recurrent HNSCC confront when will undergo resection in an irradiated bed is the surgeons
they are faced with salvage surgery. Important preoperative understanding of the type and volume of tissue needing
considerations will be discussed, specific site considerations resection, the vulnerability of the surrounding structures,
will be presented, and the rationale for surgical palliation will and the amount of nonirradiated tissue needed to re-
be addressed. construct the defect. In essence, a safe wound must be
created to limit the potential toxicity from wound break-
down after salvage surgery or tissue necrosis from reirra-
Preoperative Considerations diation. A safe wound is one that will offer protection of
Patient factors. Adequate access to nutrition is critical not carotid and vertebral arteries (preventing major vessel
only for perioperative health and wound healing but also for rupture) and cover potential exposed bone in a hostile
efficient postoperative care of the patient. Extended post- wound environment (fistula or tissue necrosis). Surgeons
operative nil per os status should be anticipated for patients must carefully evaluate preoperative imaging to determine
with previous radiation or CRT who undergo large re- what anatomic areas will be vulnerable in these situations
sections. One should strongly consider placement of a (Figs. 1 and 2). Some patients are scheduled to undergo
gastric tube. Previous studies have shown the benefit of a (chemo-)reirradiation without planned prior surgery. The
gastric tube compared with a nasogastric tube in terms of team is responsible for determining if carotid arteries or
patient comfort, cosmesis, durability, and ease of tube other areas will become exposed with the added high
feeding.3,4 It is also essential that patients are euthyroid
before any planned surgery. Thyroid-stimulating hormone
levels are routinely evaluated not only before surgery but FIGURE 1. Patient With Recurrent Palate Carcinoma
also for the entire lifetime of the patient who has undergone After Chemoradiation
radiation or CRT. Hypothyroidism negatively affects wound
healing and occurs in at least 40% of patients treated with

KEY POINTS
HPV remains prognostic in recurrent HNSCC, challenging
existing paradigms.
Salvage surgery remains standard whenever feasible.
Modern reirradiation with intensity-modulated
radiation therapy or stereotactic body radiation therapy
widens the therapeutic ratio of reirradiation.
The addition of cetuximab to 5-fluorouracil and cisplatin
has redefined the standard for systemic therapy in
HNSCC not amendable to surgery or reirradiation, even
in HPV-positive HNSCC.
Novel systemic therapies, including immune checkpoint
inhibitors, show promising early results warranting
additional ongoing prospective study.
Arrow shows internal carotid artery and adjacent tumor.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e285


WONG ET AL

FIGURE 2. Same Patient After Resection and surgery. The influence of these latter factors has driven on-
Reconstruction With Radial Forearm Free Flap cologists and scientists to develop reirradiation treatment
strategies. Salvage surgery as a first step seems to positively
affect survival, as does concomitant chemo-reirradiation.22,23
Despite these improvements, acute and late treatment toxic-
ities remain high.

Specific Sites and Surgical Approaches


Neck management. Salvage neck dissections for isolated
neck recurrence or in combination with primary site re-
currence are a heterogeneous group. Dissections may be
straightforward radical neck dissection with coverage of
vessels with a simple pectoralis myocutaneous flap. Alter-
natively, salvage neck dissections may involve resection/
reconstruction of the carotid artery, cranial nerves, deep
neck muscles, and/or vertebral arteries. Jones et al noted a
31% 5-year survival in the 69 (of 589) patients undergoing a
second neck dissection. Younger and more fit patients and
those with low T and N stage at presentation had better
survival than older patients and those with high T and
N stage.24 Mourad et al presented data for 51 patients (nine
who had undergone prior radiation) who underwent carotid
resection and in-line carotid artery bypass grafting. There
was an 82% 2-year survival and low neuromorbidity (2 of 51
had perioperative stroke).25 These findings and others sup-
Vulnerable area of carotid is covered with healthy tissue. port aggressive neck resection and carotid replacement
particularly for younger patients.
radiation doses. If the patient is at risk for wound break-
down, if feasible, the surgeon should plan to cover vul- Primary sites. There have been great technical advances in
nerable vessels or bony areas with vascularized tissue prior surgical approaches for salvage of primary site recurrences.
to reirradiation. Often, the team must weigh the risk of These developments mirror those occurring in surgical ap-
major vessel rupture (if the wound is not safe) with the risk of proaches for the untreated patient. For example, in re-
delaying the start of reirradiation. current nasopharyngeal cancers, image-guided endosurgery
is precise and effective, offers improved recovery, and
Rationale for salvage surgery. The treatment of patients with is safer than open approaches.26 Salvage endoscopic
recurrent advanced disease remains a challenge and demands nasopharyngectomy has also been found to be superior to
the thoughtful input of multidisciplinary teams. Although his- intensity-modulated radiation therapy (IMRT) in terms of
torical series have shown overall poor prognosis for this group of survival and quality of life parameters.27 Transoral robotic
patients in general, surgical salvage is considered the standard surgery has been found to be an oncologically sound al-
treatment and offers the best opportunity for locoregional ternative to open surgical procedures and is superior in
control and perhaps cure when compared with chemotherapy terms of functional and recovery outcomes.28 In addition,
or reirradiation alone. In arguably one of the most notable transoral laser microsurgery has been shown to have
studies of salvage surgery, Goodwin et al found that 2-year comparable oncologic outcomes with superior function
survival was predicted by stage (73% for stage I and 22% for and less morbidity compared with open partial or total
stage IV; p = .0005) and site (laryngeal 76%, oral cavity 47%, and laryngectomy.29
pharyngeal sites 24%; p = .0645).12 Others have found that time
to recurrence and interval from previous radiation, use of Palliation
chemotherapy, comorbidity, performance status, pre-existing Survival statistics, surgical complication rates, and treatment
organ dysfunction, and N3 neck disease are also negatively toxicities, as significant as they are, do not provide a full
correlated with survival.13-22 Surgical salvage, even with neg- picture of the positive and negative effects of surgical
ative margins, is associated with high recurrence rates, with treatment on patients with recurrent cancer. More impor-
approximately 67% overall failure rate. Multiple factors in- tantly, surgery has a compelling role in palliation for patients
cluding tumor-acquired radiation and chemotherapy resistance, with recurrent head and neck cancers. All major U.S.
pathologic patterns of submucosal multiple microscopic nests, medical, ethical, and religious organizations recognize that it
and/or perineural, perivascular, or perilymphatic invasion are is imperative to treat the distressing symptoms of patients
thought to contribute to the recurrence rates after salvage who are dying.30 Palliative surgical treatment is nuanced and

e286 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


RECURRENT HEAD AND NECK CANCER

requires knowledge of a patients unique medical condition tissue toxicity. An early retrospective study by the Institut
at the time, as well as the patients personality and family Gustave-Roussy reported outcomes on 169 patients with
dynamics. A clear definition of palliative surgery has evolved unresectable disease treated with curative-intent full-dose
and remains consistent with those standardized principles of reirradiation with or without chemotherapy.37 At a median
palliative care in general: Surgical procedures used for the follow-up of 70 months, the median overall survival was
primary intention of improving quality of life or relieving 10 months with an overall survival rate of 21% at 2 years and
symptoms caused by advanced disease.30 9% at 5 years. This modest improvement in salvage outcome
The indications for palliative surgery are not well de- came at the expense of high toxicity rates, with 46% of
fined, though many would support palliative resection when patients experiencing grade 3 or greater mucositis. Others
confronted with potential suffocation, carotid rupture, have reported similar results using conventional radiation
prevention of quadriplegia (in the instance of tumor erosion therapy techniques.38-40
into cervical spine), or control of foul, ulcerative wounds. Two phase II RTOG trials using split-course hyper-
Patients typically do not live longer (though a small per- fractionated reirradiation and chemotherapy subsequently
centage are cured by the operation with or without post- followed. RTOG 9610 found that concurrent hydroxyurea
operative adjuvant treatment), but there is great potential and 5-fluorouracil (5-FU) resulted in a median survival of
for better living during the patients remaining life. The 8.5 months and overall survival rates of 15.2% and 3.8% at
pattern of disease progression can change, where instead of 2 and 5 years, respectively.41 RTOG 9911 used concurrent
dying with a painful, odiferous, ulcerative mass, the patient cisplatin and paclitaxel, achieving a slightly better median
is allowed to die of distant disease.31-34 Resource utilization survival of 12.1 months and a 2-year survival rate of 25.9%.42
associated with palliative-intent surgeries have been shown However, high toxicity was reported in both studies, with a
to be similar to that associated with curative intent sur- 63%78% rate of grade 3 or greater acute toxicity, 22%37%
geries.35 However, patients with palliative-intent surgery rate of grade 3 or greater late toxicity, and overall 8%9%
have differing resource needs. treatment-related death.
A quote by Gaisford perhaps best summarizes the goals in Given the consistently high rates of toxicity reported,
palliative surgery: Palliative surgical procedures can be of many have questioned whether treatment toxicity out-
greater magnitude than curative operations, and the head weighs the potential benefits of reirradiation, especially
and neck cancer surgeon must be ready, willing, and given that the median survival only marginally exceeds the
ablebut mostly willingto carry them out. There is in the 6 to 9 months of chemotherapy alone.43-46 Efforts have been
last analysis no absolute rulethe surgeon who accepts made to identify patients who might benefit the most from
patients with cancer for their initial care is obligated to care this highly aggressive approach. In RTOG 9610, an interval
for them throughout the course of their disease.36 of more than 1 year from prior radiation therapy was
The multidisciplinary teams ability to support our pa- identified as a significant predictor of improved survival
tients who undergo salvage surgery has improved over (median survival, 9.8 months vs. 5.8 months; p = .036).41 In
the years through more robust reconstructions and ad- a long-term study at The University of Chicago, the authors
vances in perioperative care. Nonetheless, salvage surgery is identified reirradiation dose, triple-agent chemotherapy
nearly always radical, morbid, and technically challenging. (cisplatin, paclitaxel, and gemcitabine), and surgery as in-
Awareness of a patients social support is as critical as dependent prognostic factors for overall survival, progression-
knowledge of their comorbidities and physical ability to free survival, and locoregional control.22 Nineteen (16.5%)
withstand an often-long operation with even longer re- patients died of treatment toxicity in this study, attesting to
covery. What must be clear is the patients willingness to the morbidity of this approach. More recently, HPV status
accept that survival, not function, is the overriding goal in has been shown to be strongly prognostic in the setting
salvage treatment. Some patients may want to maintain as of reirradiation and may help to select patients more likely
much quality of life (as defined by the individual patient) and to benefit from aggressive salvage treatments such as
choose palliative chemotherapy or hospice care. Knowing reirradiation.2,47
our patients well enough to help guide them in their choice
of treatment remains one of the great privileges of being a Intensity-Modulated Radiation Therapy
doctor. Recent technologic advances such as IMRT offer potentially
superior local control rates in the reirradiation setting
ROLE OF CONVENTIONAL AND ABLATIVE with fewer late effects compared with more conventional
RADIOTHERAPY IN THE MANAGEMENT OF techniques. For instance, Lee et al reported a 2-year
RECURRENT, SECOND PRIMARY, AND/OR locoregional control rate of 52% compared with 20% among
PREVIOUSLY IRRADIATED HEAD AND NECK patients treated with IMRT and those treated with con-
CANCER ventional radiation techniques, respectively.48 In addition,
Conventional Reirradiation patients with locoregional progression-free disease had
Historically, reirradiation to locoregional recurrences or better 2-year overall survival compared with those with
new primary cancers within a previously irradiated field has locoregional failure, further demonstrating the need to
been discouraged because of concern over excessive normal maintain locoregional control to improve survival. High

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e287


WONG ET AL

treatment morbidity occurred, although at a lower rate than had previously undergone radiation were randomly assigned to
reported in prior studies utilizing conventional techniques. reirradiation (60 Gy over 11 weeks in 2 Gy daily fractions) with
Acute and late-grade 3-4 toxicities were reported in 23% and concurrent hydroxyurea and 5-FU or observation following
15% of patients, respectively.48 Similar findings have been salvage surgery. Disease-free survival and locoregional control
published by other authors.49-51 were both significantly superior in the reirradiation group, but
there was no difference in overall survival. The reirradiation
arm experienced significantly higher rates of grade 3-4 toxic-
Stereotactic Body Radiation Therapy
ities compared with the observation arm (39% vs. 10%).64
Stereotactic body radiation therapy (SBRT) offers further op-
These results support a high threshold for postoperative
portunities to deliver highly conformal treatments with com-
reirradiation following salvage surgery. Primary considerations
parable local control and overall survival, shorter treatment
include high complication rates from combined therapy, large
times, and decreased toxicity compared with conventional
radiation volumes required to cover the postoperative bed,
techniques.52 The most common SBRT regimen consists of five
high competing risks of outside-field recurrence (such as dis-
fractions delivered on alternating days,52 which has been
tant metastases and second primary cancer), and a lack of a
shown to be safer than treatment delivered on consecutive
survival advantage. However, small-field postoperative reir-
days.53,54 There has been increasing interest in SBRT as an
radiation may be appropriate in select cases with high-risk
alternative technique for reirradiation, with several phase I and
features such as extracapsular extension or positive margins.
phase II trials showing promising safety and efficacy results.55-58
The use of SBRT in this setting was recently explored in a
A phase I dose-escalation trial from the University of Pittsburgh
small retrospective study of 28 patients with high-risk fea-
Cancer Institute showed that reirradiation up to 44 Gy using
tures (predominately extracapsular extension or close/positive
SBRT is well tolerated in the acute setting.58 A subsequent
margins). At a median follow-up of 14 months, the 1-year
dose-escalation study identified a clear dose-volume response
locoregional control and overall survival were 51% and 64%,
with local control that became more prominent for gross tumor
respectively, while rates of acute and late-grade 3 and higher
volumes greater than 25 cc with longer follow-up times.59
toxicities were low at 0% and 8%, respectively.23 The role of
The use of novel EGFR-targeted agents may help to further
postoperative SBRT is currently being explored in a randomized
improve disease outcome while minimizing additional toxicity.
phase II trial at the University of Pittsburgh Cancer Institute.
Because concurrent cetuximab with radiation therapy has been
shown to significantly improve local control and survival for
primary head and neck cancer without major increases in Considerations to Reduce Toxicity
mucosal toxicity,60 there has been interest in the application of Patient selection with regard to performance status, medical
this agent as a radiosensitizer in the reirradiation setting. This comorbidities, time interval since last radiation, prior dose
was explored in a single-institution, matched, case-control received, extent of existing late toxicity burden, and tumor
study, which found that cetuximab conferred a locoregional location and size should be carefully performed. With regard
and overall survival advantage without a major increase in to treatment parameters, it has been reported that 96% of
toxicity.61 In a subsequent phase II trial on SBRT with con- failures occur within the gross tumor volume 95% isodose
current cetuximab for previously irradiated HNSCC, the authors line,65 suggesting that target volume should be limited to
reported a median overall survival of 10 months, with a 1-year only high-risk areas to minimize toxicity. A clinical target
progression-free survival rate of 60%. Acute and late-grade 3 volume confined to the gross target volume plus a margin is
toxicity were each observed in 6% of patients, much lower than recommended.48,49 Induction or concurrent systemic ther-
reported in studies of other techniques.57 A similar French apy, including novel targeted therapy agents, may allow a
phase II study reported a median survival of 11.8 months, with reduction in radiation dose. An ongoing phase II trial is being
32% of patients experiencing grade 3 toxicities.55 However, conducted by the GORTEC to evaluate reirradiation using
although the risk of severe late toxicity increases significantly a hyperfractionated scheme with concurrent cetuximab
beyond 5 years,62 follow-up for most studies have been limited (NCT01237483). Finally, recent technologic advancements
to 1 to 2 years, pointing to the need for long-term studies. such as IMRT and SBRT offer potentially noninferior onco-
Local control and lower rates of toxicity with SBRT may logic outcomes with lower toxicity rates compared with
theoretically affect quality of life. In a prospective evaluation conventional methods, although supporting phase III data
of patient-reported quality of life outcomes following SBRT continues to evolve.
with or without cetuximab, overall quality of life and mul-
tiple domains commonly affected by reirradiation pro-
gressively improved following an initial 1-month decline,
SYSTEMIC THERAPEUTIC OPTIONS FOR
speaking to the positive palliative benefits of improved local
RECURRENT/SECOND PRIMARY HEAD AND NECK
control afforded by conformal reirradiation with SBRT.63
CANCERS
Combination Chemotherapy for Unresectable
Recurrent or Second Primary HNSCC
Postoperative Reirradiation Systemic therapy, in general, is considered to be the
Reirradiation in the postoperative setting has been studied in least effective of the three treatment modalities used in
GETTEC/GORTEC 9901, a phase III trial in which patients who the management of HNSCC. This is because, as a single

e288 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


RECURRENT HEAD AND NECK CANCER

treatment modality, chemotherapy alone does not have the extremely unlikely that a study with this design will ever
capability of eliciting curative treatment effectsunlike come to fruition based on previous experience with this
surgery or radiation therapy. In terms of achieving a cure for disease.
head and neck cancer, the primary role of systemic therapy RTOG 0421 (NCT00113399) was a phase III study launched
is as a sensitizer for radiation therapy. As described in in 2005 that compared reirradiation and concurrent che-
the preceding section, reirradiation has the possibility of motherapy with chemotherapy alone. This trial used the
achieving long-term tumor control and survival for patients cisplatin and paclitaxel reirradiation regimen of RTOG 9911
with unresectable, locoregional recurrent HNSCC. This raises (as described in the preceding section), which showed an
the difficult question that oncologists frequently struggle incremental improvement over the preceding study, RTOG
with in the clinicwhether systemic chemotherapy or 9610. The 2-year survival rate demonstrated in RTOG 9911
concurrent chemo-reirradiation should be used as the first- was 26%, which compared favorably with survival data from
line treatment for a patient with unresectable locoregional contemporary chemotherapy-alone studies.69 It was within
recurrent HNSCC. To address this question, it will be useful to this backdrop that RTOG 0421 was launched.
first review clinical trials that have examined systemic Simultaneously, the GORTEC group initiated a phase III
chemotherapy regimens. study in France, GORTEC 9803, which also compared chemo-
The EXTREME study was a phase III trial comparing cis- reirradiation with chemotherapy alone.70 Unfortunately,
platin, 5-FU, and cetuximab with cisplatin and 5-FU for neither of these studies were able to complete accrual,
patients with HNSCC with recurrent or metastatic disease.66 leaving this important clinical question unanswered. Even
A maximum of six cycles of chemotherapy was admin- though no conclusion could be drawn from these studies,
istered in both arms. In the experimental arm, weekly some useful observations were made. First, the inability to
maintenance cetuximab was prescribed following the complete these studies strongly suggested a bias on behalf
course of combination systemic therapy. The results of this of treating physicians favoring reirradiation that hampered
study demonstrated an overall survival advantage favoring accrual. Second, there seemed to be a shift in the wide-
the experimental arm, with a median overall survival of spread use and acceptance of reirradiation. Since the early
10.1 months and 7.4 months for the chemotherapy- days after reirradiation was first described,37,40 it had been
cetuximab arm compared with the chemotherapy-alone considered a novel and potential very dangerous therapy,
arm, respectively. A subsequent ad hoc analysis of this performed primarily at larger-volume academic centers with
study showed that the beneficial effects of adding cetuximab experience in this technique. However, by the time RTOG
to chemotherapy were independent of HPV status.67 The 0421 was activated, a shift toward general acceptance of
results of EXTREME study were surprising in the sense that reirradiation had occurred regarding the safety and a level of
numerous phase III studies in the preceding decades failed to comfort with the procedure. The confluence of these two
demonstrate superiority of newer, presumably more active, issues doomed these trials from achieving completion,
drug combinations compared with older, presumably less leaving the central question unanswered.
active, drug regimens. ECOG 1395 was a phase III trial that Both of RTOG 0421 and the GORTEC 9803 studies were
compared cisplatin and paclitaxel with cisplatin and 5-FU.46 designed in an era prior to the availability of the results of the
The experimental armcisplatin and paclitaxelfailed to EXTREME study. The positive results of the EXTREME trial
demonstrate an overall survival advantage; the median actually make the question of reirradiation or systemic
overall survival was 8.1 months and 8.7 months, re- chemotherapy for unresectable recurrent HNSCC even more
spectively, for the cisplatin-paclitaxel and the cisplatin/5-FU relevant and pressing. So in the absence of high-level evi-
arms. A preceding phase III study compared cisplatin plus dence comparing reirradiation with systemic therapy, how is
5-FU with either drug alone.68 Although the cisplatin/5-FU it possible to choose the most appropriate therapy for a
arm achieved a higher overall response rate compared with patient with unresectable recurrent HNSCC in a previously
the single-agent arms, this did not translate into an im- irradiated field?
provement in overall survival. Hence, in the context of this Although a simple solution is not possible, a number of
historical background it is clear why the results of the EX- considerations make it permissible to approach the decision
TREME trial were practice changing and why this regimen in a logical, evidenced-based manner. The first issue to be
has been solidified as the current standard of care. considered is toxicity. Considerable toxicity is associated
with the EXTREME regimen; approximately 80% of patients
experienced grade 3 or 4 adverse events in either arm of the
Systemic Therapy Compared With Reirradiation EXTREME trial. Likewise concurrent chemo-reirradiation is
The results of the EXTREME study raise a logical and highly associated with high toxicity, including grade 5 events that
clinically relevant question: will the EXTREME regimen have been reported in nearly all studies examining con-
provide a superior outcome compared with concurrent current chemo-reirradiationtypically in the range of
chemo-reirradiation for patients with unresectable locore- 3%.71,72 Most of the deaths associated with reirradiation are
gional recurrent HNSCC? To answer this question would related to bleeding events; carotid rupture accounts for the
require a head-to-head study comparing these two ap- majority of life-threatening bleeding events. Hence, it is
proaches; to date no such data exist. However, it is advisable to carefully consider patients with tumors that

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e289


WONG ET AL

have significant carotid involvement, such as circumferential Emerging in the literature is a growing body of evidence
tumor encasement of the carotid. Likewise, structural issues that suggests radiation therapy and immune checkpoint
preclude other patients from reirradiation, such as recurrent have complimentary effects.75,76 These studies indicate that
laryngeal cancer where retreatment can cause cartilaginous immune checkpoint inhibitors can overcome the negative
necrosis. Similarly, patients may not be eligible for reirra- effects of radiation on the tumor microenvironment. Con-
diation when it is not feasible to spare the spinal cord from a versely, these data also suggest that the antigenic response
lifetime cumulative conventionally fractionated (1.8-2.0 Gy to radiation therapy may induce a heightened effectiveness
per fraction) equivalent dose less than 54 Gy. A nomogram from immune checkpoint inhibition. It is possible to now
published by Tanvetyanon et al provides a tool by which speculate that by combining an immune checkpoint inhibitor
practitioners can identify poor prognostic factors that may with reirradiation, the best of both worlds may be achievable
help identify favorable populations and avoid exposure of in a single treatment plan. In other words, it is conceivable
dangerous toxicity to other patients.15 to concurrently potentiate the effects of reirradiation
(i.e., enhancing locoregional disease control) while simul-
Immune Checkpoint Inhibitors taneously augmenting the systemic immunologic effects
The emergence of data indicating activity of immune (i.e., enhancing durable treatment response and long-term
checkpoint inhibitors in HNSCC has potential major impli- survival). To test this hypothesis, a new study, the
cations for management of this disease, including treatment KEYSTROKE trial (RTOG Foundation Study 3507), will com-
for patients with HNSCC with unresectable locoregional pare reirradiation with SBRT plus pembrolizumab with
recurrent disease. An expansion cohort of patients with reirradiation with SBRT alone. It is conceivable that future
HNSCC were enrolled in the KEYNOTE-012 phase IB clinical studies examining immune checkpoint inhibitors with reir-
trial of pembrolizumab for recurrent and metastatic disease radiation may demonstrate favorable results that could
who had 1% or greater PD-L1positive staining by immu- reduce our reliance on dogmatic approaches to patients with
nohistochemistry.73 Of the 56 evaluable patients, the best unresectable recurrent or new second primary HNSCC.
overall response rate was 20%, with similar response rates
seen in patients with HPV-positive disease and those with
HPV-negative disease. An expansion cohort of patients with CONCLUSION
PD-L1unselected HNSCC enrolled in the KEYNOTE-012 Locoregional recurrent or second primary HNSCC in a pre-
demonstrated a best overall response rate of 25% for all viously irradiated field represents a challenging clinical
patients, 21% for patients with HPV-positive disease, and scenario for which surgical, reirradiation, and systemic
27% for patients with HPV-negative disease.74 Of note, both treatment options continue to evolve. As outlined herein,
of the reports included patients whose response was management in general would favor salvage surgical re-
ongoingsuggesting durable response to therapy. section for patients with resectable disease, followed by
In contrast to conventional chemotherapy regimens, such consideration for adjuvant reirradiation with chemotherapy.
as EXTREME, long-term durable response and survivorship For patients with unresectable disease, modern reirradiation
occurs in less than 5% of patients. Phase III studies of im- (IMRT or SBRT) with concurrent systemic therapy is favored.
mune checkpoint inhibitors for recurrent metastatic HNSCC Finally, in cases where surgery is not feasible and reirra-
are in progress and potentially could establish a new diation may pose too high of a risk of severe toxicity either
standard of care in years to come. If long-term survivorship is because of prior radiation dose to normal structures or
achievable with immune checkpoint inhibitor therapy, this extent/involvement of the recurrence, palliative systemic
treatment option would put systemic therapy in a playing therapy is favored. Patients should be encouraged to enroll
field with reirradiation. in clinical trials incorporating novel systemic agents.

References
1. Gillison ML, Harris J, Westra W, et al. Survival outcomes by tumor 4. Wang J, Liu M, Liu C, et al. Percutaneous endoscopic gastrostomy versus
human papillomavirus (HPV) status in stage III-IV oropharyngeal nasogastric tube feeding for patients with head and neck cancer:
cancer (OPC) in RTOG 0129. J Clin Oncol. 2009;27:15 (suppl; abstr a systematic review. J Radiat Res (Tokyo). 2014;55:559-567.
6003). 5. Rivelli TG, Mak MP, Martins RE, et al. Cisplatin based chemoradiation
2. Fakhry C, Zhang Q, Nguyen-Tan PF, et al. Human papillomavirus and late toxicities in head and neck squamous cell carcinoma patients.
overall survival after progression of oropharyngeal squamous cell Discov Med. 2015;20:57-66.
carcinoma. J Clin Oncol. 2014;32:3365-3373. 6. Cannon CR. Hypothyroidism in head and neck cancer patients: Ex-
3. Canadian Agency for Drugs and Technologies in Health. Nasogastric Feeding perimental and clinical observations. Laryngoscope. 1994;104:1-21.
Tubes versus Percutaneous Endoscopic Gastrostomy for Patients with Head 7. Kim AJ, Suh JD, Sercarz JA, et al. Salvage surgery with free flap re-
or Neck Cancer: A Review of Clinical Effectiveness and Guidelines. Cana- construction: factors affecting outcome after treatment of recurrent
dian Agency for Drugs and Technologies in Health Ottawa (ON). http:// head and neck squamous carcinoma. Laryngoscope. 2007;117:
www.ncbi.nlm.nih.gov/books/NBK253809/. Accessed February 19, 2016. 1019-1023.

e290 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


RECURRENT HEAD AND NECK CANCER

8. Dirven R, Swinson BD, Gao K, et al. The assessment of phar- of selected T1-T3 nasopharyngeal carcinoma - A case-matched com-
yngocutaneous fistula rate in patients treated primarily with definitive parison. Radiother Oncol. 2015;115:399-406.
radiotherapy followed by salvage surgery of the larynx and hypo- 28. White H, Ford S, Bush B, et al. Salvage surgery for recurrent cancers
pharynx. Laryngoscope. 2009;119:1691-1695. of the oropharynx: comparing TORS with standard open surgical
9. Kostrzewa JP, Lancaster WP, Iseli TA, et al. Outcomes of salvage surgery approaches. JAMA Otolaryngol Head Neck Surg. 2013;139:
with free flap reconstruction for recurrent oral and oropharyngeal 773-778.
cancer. Laryngoscope. 2010;120:267-272. 29. Canis M, Martin A, Ihler F, et al. Transoral laser microsurgery in
10. Suh JD, Kim BP, Abemayor E, et al. Reirradiation after salvage surgery treatment of pT2 and pT3 glottic laryngeal squamous cell carcinoma -
and microvascular free flap reconstruction for recurrent head and neck results of 391 patients. Head Neck. 2014;36:859-866.
carcinoma. Otolaryngol Head Neck Surg. 2008;139:781-786. 30. Palliative Surgery: Definition, Principles, Outcomes Assessment. In
11. Cohn AB, Lang PO, Agarwal JP, et al. Free-flap reconstruction in the Dunn GP, Martensen R, Weissman D (eds). Surgical Palliative Care: A
doubly irradiated patient population. Plast Reconstr Surg. 2008;122: Residents Guide. Chicago, IL: American College of Surgeons, 2009;131-
125-132. 139.
12. Goodwin WJ Jr. Salvage surgery for patients with recurrent squamous 31. Jang DW, Teng MS, Ojo B, et al. Palliative surgery for head and neck
cell carcinoma of the upper aerodigestive tract: when do the ends justify cancer with extensive skin involvement. Laryngoscope. 2013;123:
the means? Laryngoscope. 2000;110:1-18. 1173-1177.
13. Kim J, Kim S, Albergotti WG, et al. Selection of ideal candidates for 32. Miglani A, Patel VM, Stern CS, et al. Palliative reconstruction for the
surgical salvage of head and neck squamous cell carcinoma: effect of the management of incurable head and neck cancer. J Reconstr Microsurg.
Charlson-Age Comorbidity Index and oncologic characteristics on 1-year Epub 2015 Dec 4.
survival and hospital course. JAMA Otolaryngol Head Neck Surg. 2015; 33. Copeland EM III. The art of medicine at the end of life: a surgeons point
141:1059-1065. of view. Bull Am Coll Surg. 2008;93:17-18.
14. Ho AS, Kraus DH, Ganly I, et al. Decision making in the management of 34. Conley JJ. Palliative surgery in the head and neck. Ann Otol Rhinol
recurrent head and neck cancer. Head Neck. 2014;36:144-151. Laryngol. 1962;71:585-590.
15. Tanvetyanon T, Padhya T, McCaffrey J, et al. Prognostic factors for 35. Cullinane CA, Borneman T, Smith DD, et al. The surgical treatment of
survival after salvage reirradiation of head and neck cancer. J Clin Oncol. cancer: a comparison of resource utilization following procedures
2009;27:1983-1991. performed with a curative and palliative intent. Cancer. 2003;98:
16. Gleich LL, Ryzenman J, Gluckman JL, et al. Recurrent advanced (T3 or T4) 2266-2273.
head and neck squamous cell carcinoma: is salvage possible? Arch 36. Gaisford JC. Palliative surgery. JAMA. 1972;221:83-84.
Otolaryngol Head Neck Surg. 2004;130:35-38. 37. De Crevoisier R, Bourhis J, Domenge C, et al. Full-dose reirradiation for
17. Richey LM, Shores CG, George J, et al. The effectiveness of salvage unresectable head and neck carcinoma: experience at the Gustave-
surgery after the failure of primary concomitant chemoradiation in Roussy Institute in a series of 169 patients. J Clin Oncol. 1998;16:
head and neck cancer. Otolaryngol Head Neck Surg. 2007;136:98-103. 3556-3562.
18. Arnold DJ, Goodwin WJ, Weed DT, et al. Treatment of recurrent and 38. Brockstein B, Haraf DJ, Stenson K, et al. A phase I-II study of concomitant
advanced stage squamous cell carcinoma of the head and neck. Semin chemoradiotherapy with paclitaxel (one-hour infusion), 5-fluorouracil
Radiat Oncol. 2004;14:190-195. and hydroxyurea with granulocyte colony stimulating factor support for
19. Mandapathil M, Roessler M, Werner JA, et al. Salvage surgery for head patients with poor prognosis head and neck cancer. Ann Oncol. 2000;11:
and neck squamous cell carcinoma. Eur Arch Otorhinolaryngol. 2014; 721-728.
271:1845-1850. 39. Spencer SA, Wheeler RH, Peters GE, et al. Concomitant chemotherapy
20. Zafereo M. Surgical salvage of recurrent cancer of the head and neck. and reirradiation as management for recurrent cancer of the head and
Curr Oncol Rep. 2014;16:386-391. neck. Am J Clin Oncol. 1999;22:1-5.
21. Omura G, Saito Y, Ando M, et al. Salvage surgery for local residual or 40. Haraf DJ, Weichselbaum RR, Vokes EE. Reirradiation with concomitant
recurrent pharyngeal cancer after radiotherapy or chemoradiotherapy. chemotherapy of unresectable recurrent head and neck cancer: a po-
Laryngoscope. 2014;124:2075-2080. tentially curable disease. Ann Oncol. 1996;7:913-918.
22. Salama JK, Vokes EE, Chmura SJ, et al. Long-term outcome of concurrent 41. Spencer SA, Harris J, Wheeler RH, et al. Final report of RTOG 9610, a
chemotherapy and reirradiation for recurrent and second primary multi-institutional trial of reirradiation and chemotherapy for unre-
head-and-neck squamous cell carcinoma. Int J Radiat Oncol Biol Phys. sectable recurrent squamous cell carcinoma of the head and neck. Head
2006;64:382-391. Neck. 2008;30:281-288.
23. Vargo JA, Kubicek GJ, Ferris RL, et al. Adjuvant stereotactic body 42. Langer CJ, Harris J, Horwitz EM, et al. Phase II study of low-dose
radiotherapy6cetuximab following salvage surgery in previously paclitaxel and cisplatin in combination with split-course concomitant
irradiated head and neck cancer. Laryngoscope. 2014;124: twice-daily reirradiation in recurrent squamous cell carcinoma of the
1579-1584. head and neck: results of Radiation Therapy Oncology Group Protocol
24. Jones AS, Tandon S, Helliwell TR, et al. Survival of patients with neck 9911. J Clin Oncol. 2007;25:4800-4805.
recurrence following radical neck dissection: utility of a second neck 43. Argiris A, Ghebremichael M, Gilbert J, et al. Phase III randomized,
dissection? Head Neck. 2008;30:1514-1522. placebo-controlled trial of docetaxel with or without gefitinib in re-
25. Mourad M, Saman M, Stroman D, et al. Carotid artery sacrifice and current or metastatic head and neck cancer: an eastern cooperative
reconstruction in the setting of advanced head and neck cancer. oncology group trial. J Clin Oncol. 2013;31:1405-1414.
Otolaryngol Head Neck Surg. 2015;153:225-230. 44. Bourhis J, Rivera F, Mesia R, et al. Phase I/II study of cetuximab in
26. Naara S, Amit M, Billan S, et al. Outcome of patients undergoing salvage combination with cisplatin or carboplatin and fluorouracil in patients
surgery for recurrent nasopharyngeal carcinoma: a meta-analysis. Ann with recurrent or metastatic squamous cell carcinoma of the head and
Surg Oncol. 2014;21:3056-3062. neck. J Clin Oncol. 2006;24:2866-2872.
27. You R, Zou X, Hua Y. Salvage endoscopic nasopharyngectomy is 45. Burtness B, Goldwasser MA, Flood W, et al. Phase III randomized trial
superior to intensity-modulated radiation therapy for local recurrence of cisplatin plus placebo compared with cisplatin plus cetuximab in

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e291


WONG ET AL

metastatic/recurrent head and neck cancer: an Eastern Cooperative 62. Owen D, Iqbal F, Pollock BE, et al. Long-term follow-up of stereotactic
Oncology Group study. J Clin Oncol. 2005;23:8646-8654. radiosurgery for head and neck malignancies. Head Neck. 2015;37:
46. Gibson MK, Li Y, Murphy B, et al. Randomized phase III evaluation of 1557-1562.
cisplatin plus fluorouracil versus cisplatin plus paclitaxel in advanced 63. Vargo JA, Heron DE, Ferris RL, et al. Prospective evaluation of patient-
head and neck cancer (E1395): an intergroup trial of the Eastern Co- reported quality-of-life outcomes following SBRT 6 cetuximab for
operative Oncology Group. J Clin Oncol. 2005;23:3562-3567. locally-recurrent, previously-irradiated head and neck cancer. Radio-
47. Davis KS, Vargo JA, Ferris RL, et al. Stereotactic body radiotherapy for ther Oncol. 2012;104:91-95.
recurrent oropharyngeal cancer - influence of HPV status and smoking 64. Janot F, de Raucourt D, Benhamou E, et al. Randomized trial of post-
history. Oral Oncol. 2014;50:1104-1108. operative reirradiation combined with chemotherapy after salvage
48. Lee N, Chan K, Bekelman JE, et al. Salvage reirradiation for recurrent surgery compared with salvage surgery alone in head and neck car-
head and neck cancer. Int J Radiat Oncol Biol Phys. 2007;68:731-740. cinoma. J Clin Oncol. 2008;26:5518-5523.
49. Sulman EP, Schwartz DL, Le TT, et al. IMRT reirradiation of head and neck 65. Popovtzer A, Gluck I, Chepeha DB, et al. The pattern of failure after
cancer-disease control and morbidity outcomes. Int J Radiat Oncol Biol reirradiation of recurrent squamous cell head and neck cancer: im-
Phys. 2009;73:399-409. plications for defining the targets. Int J Radiat Oncol Biol Phys. 2009;74:
50. Biagioli MC, Harvey M, Roman E, et al. Intensity-modulated radio- 1342-1347.
therapy with concurrent chemotherapy for previously irradiated, re- 66. Vermorken JB, Mesia R, Rivera F, et al. Platinum-based chemotherapy
current head and neck cancer. Int J Radiat Oncol Biol Phys. 2007;69: plus cetuximab in head and neck cancer. N Engl J Med. 2008;359:
1067-1073. 1116-1127.
51. Duprez F, Madani I, Bonte K, et al. Intensity-modulated radiotherapy for 67. Vermorken JB, Psyrri A, Mesa R, et al. Impact of tumor HPV status on
recurrent and second primary head and neck cancer in previously ir- outcome in patients with recurrent and/or metastatic squamous cell
radiated territory. Radiother Oncol. 2009;93:563-569. carcinoma of the head and neck receiving chemotherapy with or
52. Rwigema JC, Vargo JA, Clump DA, et al. Stereotactic body radiotherapy without cetuximab: retrospective analysis of the phase III EXTREME
in the management of head and neck malignancies. Curr Cancer Ther trial. Ann Oncol. 2014;25:801-807.
Rev. 2014;10:314-322. 68. Jacobs C, Lyman G, Velez-Garca E, et al. A phase III randomized study
53. Kodani N, Yamazaki H, Tsubokura T, et al. Stereotactic body radiation comparing cisplatin and fluorouracil as single agents and in combination
therapy for head and neck tumor: disease control and morbidity for advanced squamous cell carcinoma of the head and neck. J Clin
outcomes. J Radiat Res (Tokyo). 2011;52:24-31. Oncol. 1992;10:257-263.
54. Yazici G, Sanl TY, Cengiz M, et al. A simple strategy to decrease fatal 69. Wong SJ, Machtay M, Li Y. Locally recurrent, previously irradiated head
carotid blowout syndrome after stereotactic body reirradiaton for and neck cancer: concurrent reirradiation and chemotherapy, or
recurrent head and neck cancers. Radiat Oncol. 2013;8:242. chemotherapy alone? J Clin Oncol. 2006;24:2653-2658.
55. Lartigau EF, Tresch E, Thariat J, et al. Multi institutional phase II study of 70. Tortochaux J, Tao Y, Tournay E, et al. Randomized phase III trial (GORTEC
concomitant stereotactic reirradiation and cetuximab for recurrent 98-03) comparing reirradiation plus chemotherapy versus metho-
head and neck cancer. Radiother Oncol. 2013;109:281-285. trexate in patients with recurrent or a second primary head and neck
56. Comet B, Kramar A, Faivre-Pierret M, et al. Salvage stereotactic reir- squamous cell carcinoma, treated with a palliative intent. Radiother
radiation with or without cetuximab for locally recurrent head-and- Oncol. 2011;100:70-75.
neck cancer: a feasibility study. Int J Radiat Oncol Biol Phys. 2012;84: 71. Wong SJ, Bourhis J, Langer CJ. Retreatment of recurrent head and neck
203-209. cancer in a previously irradiated field. Semin Radiat Oncol. 2012;22:
57. Vargo JA, Ferris RL, Ohr J, et al. A prospective phase II trial of re- 214-219.
irradiation with stereotactic body radiotherapy plus cetuximab in pa- 72. McDonald MW, Moore MG, Johnstone PA. Risk of carotid blowout after
tients with recurrent previously-irradiated squamous cell carcinoma of reirradiation of the head and neck: a systematic review. Int J Radiat
the head and neck. Int J Radiat Oncol Biol Phys. 2015;91:480-488. Oncol Biol Phys. 2012;82:1083-1089.
58. Heron DE, Ferris RL, Karamouzis M, et al. Stereotactic body radiotherapy 73. Seiwert TY, Burtness B, Weiss J, et al. A phase Ib study of MK-3475 in
for recurrent squamous cell carcinoma of the head and neck: results of a patients with human papillomavirus (HPV)-associated and non-HPV
phase I dose-escalation trial. Int J Radiat Oncol Biol Phys. 2009;75: associated head and neck (H/N) cancer. J Clin Oncol. 2014;32:5s (suppl;
1493-1500. abstr 6011).
59. Rwigema JC, Heron DE, Ferris RL, et al. The impact of tumor volume and 74. Seiwert T, Haddad R, Gupta S. Antitumor activity and safety of
radiotherapy dose on outcome in previously irradiated recurrent pembrolizumab in patients (pts) with advanced squamous cell carci-
squamous cell carcinoma of the head and neck treated with stereotactic noma of the head and neck (SCCHN) (2015) preliminary results from
body radiation therapy. Am J Clin Oncol. 2011;34:372-379. KEYNOTE-012 expansion cohort. J Clin Oncol. 2015;33s (suppl; abstr
60. Bonner JA, Harari PM, Giralt J, et al. Radiotherapy plus cetuximab for LBA6008).
squamous-cell carcinoma of the head and neck. N Engl J Med. 2006;354: 75. Deng L, Liang H, Burnette B, et al. Irradiation and anti-PD-L1 treatment
567-578. synergistically promote antitumor immunity in mice. J Clin Invest. 2014;
61. Heron DE, Rwigema JC, Gibson MK, et al. Concurrent cetuximab with 124:687-695.
stereotactic body radiotherapy for recurrent squamous cell carcinoma 76. Twyman-Saint Victor C, Rech AJ, Maity A, et al. Radiation and dual
of the head and neck: a single institution matched case-control study. checkpoint blockade activate non-redundant immune mechanisms in
Am J Clin Oncol. 2011;34:165-172. cancer. Nature. 2015;520:373-377.

e292 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


HEALTH SERVICES RESEARCH AND QUALITY
OF CARE

Defining and Measuring Quality

CHAIR
Patricia A. Ganz, MD, FASCO
University of California, Los Angeles
Los Angeles, CA

SPEAKERS
David C. Miller, MD, MPH
University of Michigan Health System
Ann Arbor, MI

Michael J. Hassett, MD, MPH


Dana-Farber Cancer Institute
Boston, MA
GANZ, HASSETT, AND MILLER

Challenges and Opportunities in Delivering High-Quality


Cancer Care: A 2016 Update
Patricia A. Ganz, MD, FASCO, Michael J. Hassett, MD, MPH, and David C. Miller, MD, MPH

OVERVIEW

Herein, both the rationale and scope of current initiatives aimed at improving the quality of cancer care delivery in the United
States are described. First, we discuss a recent report from the Institute of Medicine that issued a strong call for both the
development of quality measures in oncology and implementation of a learning health care system in which data and
experience from clinical practice can inform continuous improvements in cancer care. Second, we review the multiple,
diverse initiatives that are underway to identify, test, and validate quality measures for the entire spectrum of cancer care.
Finally, we discuss regional quality improvement collaboratives as one successful approach to creating a cycle of quality
measurement, identification of best practices, and implementation of changes in practice patterns that ultimately yield
improved care and outcomes for patients with cancer.

I n 2013, the Institute of Medicine (IOM) released a report


with comprehensive, specific recommendations for im-
proving the quality of cancer care in the United States, with a
hold great promise, are substantially more expensive and
require individualized treatments that will be challenging to
deliver to large populations of patients. To some extent
special focus on addressing the challenges associated with these novel therapies remain experimental, but there is
the expanding number of patients with cancer and cancer tremendous demand by the public and clinicians to adopt
survivors.1 A major factor that will strain the cancer care these therapies in the earliest stages of development. This
delivery system is the increasing numbers of persons older is the context for consideration of the challenges and
than age 65 who will be at highest risk for the development opportunities in the delivery of high-quality cancer care.
of cancer because of the known association of age with many
cancers (e.g., breast, colon, prostate, lung, bladder, kidney,
and lymphoma). In addition, the projected shortfall in on- DIRECT RESPONSES AND CHAMPIONS FOR
cology providers will exacerbate this situation.2 We also SPECIFIC INSTITUTE OF MEDICINE REPORT
anticipate tremendous growth in the number of survivors of RECOMMENDATIONS
cancer who may remain on treatments for extended periods The IOM report specifically cited the central position of the
of time and/or have long-term and late effects of treatments patient in the care delivery system and the importance of
that will require ongoing medical care and surveillance.3 effective communication of diagnosis, prognosis, and
The increasing cost of care complicates these demographic treatment options at the point of care, with an emphasis on
changes and expanding demand for services. Costs are not patient preferences and shared decision making.1 Team-
limited to pharmaceuticals, but also include widespread based and coordinated care also were proposed as impor-
adoption and use of many new and expensive technologies tant strategies to reduce the fragmentation of care and to
in the diagnosis and treatment of cancer (e.g., next- address the shortages of some clinicians, as well as to
generation sequencing) to refine understanding of tumor maximize opportunities for receipt of psychosocial and
characteristics for use of more specific and directed ther- palliative care along with cancer-directed therapies. In ad-
apies. Furthermore, the extensive subtyping of cancers will dition, there was a strong call for the development of quality
create many small subsets of patients with cancer, making metrics specific to oncology, elimination of wasteful and
some types of cancer even rarer than before. Immuno- low-value care, and for implementation of a learning health
modulatory treatments and cell therapies, although they care system in which information from clinical practice in the

From the Department of Health Policy and Management, UCLA Fielding School of Public Health, Center for Cancer Prevention and Control Research, Jonsson Comprehensive Cancer
Center, Los Angeles, CA; Department of Medicine, Harvard Medical School, Division of Population Sciences, Dana-Farber Cancer Institute, Boston, MA; Dow Division of Health Services
Research, Department of Urology, Institute for Healthcare Policy and Innovation, University of Michigan Medical School, Ann Arbor, MI.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Patricia A. Ganz, MD, Center for Cancer Prevention and Control Research, Jonsson Comprehensive Cancer Center, 650 Charles Young Dr. South, Room A2-125
CHS, Los Angeles, CA 90049; email: pganz@mednet.ucla.edu.

2016 by American Society of Clinical Oncology.

e294 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


IMPROVING THE QUALITY OF CANCER CARE

real world could inform improvements in the delivery of centered care in their episode of care payment model di-
cancer care. Although the IOM report addresses many other rectly from the IOM report (reflecting recommendation 10
issues, the areas mentioned above have been the focus of and other aspects of the report). The latter also encouraged
considerable activity in the last few years, and they will be participation from private health insurers. As of this writing,
the major focus of this update. the participating clinician groups and insurance plans have
Recommendations 1 and 2 from the IOM report focused not been announced, but this is expected to be implemented
on the central role of engaged patients in delivering high- in early 2016. Providing more financial resources to support
quality cancer care. The goals of these recommendations are the degree of patient engagement in treatment decision
to ensure that the cancer care team has provided patients making and care, as well as ancillary supportive services, will
and their families with understandable information on be critical if we are to enhance the delivery of quality cancer
cancer prognosis, treatment benefits and harms, palliative care.
care, psychosocial support, and estimates of total and out- Other efforts underway have begun to address the cost of
of-pocket costs of cancer care. These recommendations cancer care and the development of tools to assist clinicians
pertain to newly diagnosed patients at entry into the cancer at the point of care in discussing preferences and toxicities,
care setting, as well as patients with advanced and recurrent along with cost (multiple IOM report recommendations).
cancer, where the goals also include providing patients with Among these are the ASCO Value Framework5 and work
end-of-life care consistent with their needs, values, and underway by the National Comprehensive Care Network in
preferences. The IOM report identified specific actors to its framework for resource stratification. There is increasing
take the lead in accomplishing these goals, tasking a broad dialogue regarding these issues in the lay press and in
array of stakeholders in developing better ways to com- professional oncology journals.6 It is heartening to see so
municate this complex information to patients and their many stakeholders beginning to focus on the value of care
families, and calling out the need to develop a personalized that is being delivered, including elimination of low-value
cancer care plan that would reflect the patients needs and and wasteful care.7-9 Additional efforts in progress are
preferences. working to promote a learning health care system (IOM
This aspirational goal is very important, but it will take report recommendation 7), with multiple experiments in
some time to accomplish. However, we were heartened by progress and development of oncology-specific quality
proposals from several organizations to modify cancer care metrics. To provide further elaboration on several of these
reimbursement to account for the complexity of initial di- issues, the following sections describe ongoing research on
agnosis and treatment planning (American Society of Clinical quality metrics and work being done to address the need to
Oncology [ASCO] Plan), and an aggressive proposal for a improve the quality of cancer care.
payment modification scheme from the Centers for Medi-
care & Medicaid Services (CMS) called the Oncology Care
Model (OCM),4 which identified the elements of patient- METRICS FOR ASSESSING QUALITY OF CANCER
CARE
A fundamental requirement of any effort to improve the
quality of health care is the ability to measure practice
KEY POINTS performance reliably, validly, and efficiently. Without
quality measures, identifying gaps in performance and
The 2013 IOM report, Delivering High-Quality Cancer assessing the extent to which improvement efforts ame-
Care: Charting a New Course for a System in Crisis,
liorate these gaps are impossible. In fact, quality measures
identified the aging of the population, the exploding cost
of care, and fragmentation and complexity of cancer
could help address many of the challenges highlighted by the
treatment as major threats to the care delivery system. IOM and outlined above, including the need to reduce care
Recommendations from the report are being fragmentation, improve access to psychosocial and palliative
championed by various entities, and this article care, administer precision cancer treatments, provide end-
highlights a number of areas of progress. of-life care consistent with patients wishes, and eliminate
Measuring the quality of care is a major challenge, and wasteful low-value care. In this section, we will identify
new strategies are being implemented to report on and organizations that have taken the lead in developing cancer
evaluate quality in cancer care delivery. quality measures; provide an overview of the quality
Among the experiments in place are physician learning measures that have been developed; and identify important
collaboratives, such as the Michigan Urological Surgery topics for which quality measures are lacking. Finally, we will
Improvement Collaborative, to improve on the quality
review some of the biggest challenges to the development
of prostate cancer care delivery.
The development of new payment models that fund
and use of measures to assess and improve care quality.
episodes of care or provide reimbursement to groups of In the 2 decades since the IOM issued a call to action
physicians responsible for the entire continuum of through a series of landmark publications, including To Err Is
cancer care will likely hasten greater accountability and Human: Building a Safer Health System10 and Crossing the
coordination of care. Quality Chasm: A New Health System for the 21st Century,11
professional societies have taken the lead in developing

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e295


GANZ, HASSETT, AND MILLER

quality measures for cancer care. Substantial contributors to FIGURE 1. Schematic Example of Donabedian Model
this effort have included the American College of Surgeons/ Describing the Components of Quality Care
Commission on Cancer, American Medical Association,
ASCO, American Society of Hematology, American Society
for Radiation Oncology, College of American Pathologists,
National Comprehensive Cancer Network, and National
Hospice and Palliative Care Organization. In addition, a
number of organizations have endeavored to collect and
endorse quality measures to facilitate their use, including
the Agency for Healthcare Research and Qualitys National
Quality Measures Clearing House12 and the National Quality
Forums (NQFs) endorsement process.13-15
The framework proposed by Donabedian16 defines three
types of quality measures: structure, process, and outcomes
(Fig. 1). Although all measure types are relevant to cancer
care, most available measures focus on processes because (e.g., treatment plan generation, fertility counseling);
they are easier to develop, implement, and interpret. In fact, (3) the provision of interdisciplinary and multidisciplinary
process-based quality measures are currently used to sup- coordinated care; (4) comprehensive symptom assessment
port accountability, pay-for-performance, contractual, and and treatment; and (5) patient experience of care. Fur-
regulatory programs such as the Physician Quality Reporting thermore, it emphasized the importance of measures that
System, Meaningful Use, alternative quality contracts, and assess outcomes, address cross-cutting topics that pertain to
others.17-19 One notable exception is the Quality Oncology different cancer types and care settings (e.g., chemotherapy-
Practice Initiative (QOPI) from ASCO, which was designed to induced nausea), and focus on issues of interest to patients
foster practice-based quality improvement specifically to rather than the health care system.
facilitate an oncology practices effort to monitor and improve Past efforts to develop and implement quality mea-
its performance.20-22 QOPI assesses performance relative to sures highlight a number of challenges that will have to be
more than 100 cancer-focused measures twice yearly for addressed if we are to build the comprehensive measure-
hundreds of practices and thousands of patients.20-23 ment system advocated for by IOM and NQF. First, we must
Although many valid and reliable process-based cancer determine what aspects of care should be measured.
quality measures have been developed, existing measures tend Usually, decisions regarding what to measure are based on
to cover a limited spectrum of cancer care services. Specifically, consensus opinions about which performance gaps are
they focus on common malignancies (i.e., breast cancer, co- important and where improvement efforts could have the
lorectal cancer, lung cancer), assess the performance of phy- greatest impact rather than on the use of actual practice
sicians or hospitals, focus on widely accepted standards, and performance data to identify substantial gaps and define
address initial management strategies. This leaves many im- benchmarks.27 Second, we must gather the data needed to
portant aspects of cancer care underevaluated, including less assess performance efficiently and accurately. With limited
common cancers, nonphysician providers, long-term follow-up resources, we can only assess performance relative to a limited
care, and end-of-life issues. In a 2009 report, the NQF high- set of measures. And third, we must interpret the results of
lighted these hurdles and suggested that building a compre- measurement efforts appropriately, so that the findings can be
hensive measurement system would require new measures to used to create real and meaningful improvements in quality.
be developed to address gaps in episodes of care and focus on Figuring out what to measure seems simple, but it can
patients rather than individual providers or distinct care set- present a number of pitfalls. Historically, selecting topics for
tings.24 In 2012, the Measure Applications Partnership (MAP) measure development has followed one of two paths. In the
suggested that there was an immediate need to assess cross- first, interested parties, often professional societies, lead
cutting aspects of care, meaning those that are relevant to all rigorous measure development efforts on topics that are of
patients with cancer throughout the trajectory of their illness.25 particular interest to them. This tends to produce measures
Both organizations noted that new measures should focus on for which there is high-quality evidence and widespread
outcomes, care coordination, care transitions, quality of life, consensus. However, measures can take a long time to
patient safety, and experience of care.15,25 develop, are hard to update, and often focus on areas where
In December 2012, ASCO convened a Collaborative Cancer quality problems are less pronounced. In the second, hos-
Measure Summit to identify topics for which new quality pitals, clinics, and provider organizations develop measures
measures were needed.26 The summit, which was attended on their own to support local quality improvement efforts.
by 12 specialty societies and patient advocacy organizations, This provides more flexibility, but it can generate measures
highlighted issues similar to those raised by NQF and the based on lower-quality evidence or less consensus, and
MAP. Specifically, ASCO encouraged the development of testing for reliability and validity are less common. In both
measures to address (1) the use of palliative care/hospice paths, topic selection is usually driven by providers and
services; (2) planning and counseling prior to therapy payers, rather than patients.

e296 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


IMPROVING THE QUALITY OF CANCER CARE

Once specifications have been developed, the next chal- physicians and surgeons have established practice- or hospital-
lenge is determining how to gather the data needed to based regional quality improvement collaboratives.33,34 The
reliably and efficiently report on a measure. Three common general organizational principles of such collaboratives include
methods are used. Many measures rely on administrative centralized and standardized collection of high-quality clinical
data collected for other purposes, such as claims for health outcomes data; provision of performance feedback to indi-
care services rendered. Administrative data are readily vidual clinicians and practices; identification of clinical care
available and are often based on a common standard (e.g., processes (e.g., diagnostic tests, type of surgical procedure)
ICD-9, ICD-10, CPT), but they lack clinical granularity and that correlate with optimal patient outcomes; implementation
substantial data-quality issues can exist (i.e., missing data, of these best clinical practices; and dissemination of findings
consistency, accuracy). Some measures depend on manual from collaborative activities to benefit patients across a broad
data abstraction. This is often done by a cancer registrar (e.g., geographic region.
the National Cancer Databases Rapid Quality Reporting The Michigan Urological Surgery Improvement Collab-
System)28 or a quality improvement staff member (e.g., orative (MUSIC) is an example of such a collaborative.
ASCOs QOPI).22 Although these data can be quite granular, Established in 2011 with funding and support from Blue
they are costly and time consuming to collect. With the recent Cross Blue Shield of Michigan, MUSIC is a physician-led
widespread adoption of electronic health records (EHRs), quality improvement collaborative currently comprised of
there is growing interest in the use of EHR data for quality 42 urology practices and 235 urologists from across Mich-
measurement. EHR data offer great promise, but identifying igan (roughly 85% of the urologists in the state).35 The
important concepts from these data can be challenging be- collaborative also includes four patient advocates who play a
cause they contain a lot of unstructured free text. EHRs central role in all of our quality improvement activities. The
encourage users to enter information into structured data mission of MUSIC is to make Michigan the best place in the
fields to facilitate quality analyses, but this creates a burden world for prostate cancer care.
on EHR users who must spend time entering data that may The collaborative is managed by a coordinating center,
not be integral to the care they are providing. housed administratively in the University of Michigan De-
After all the data have been collected, the final challenge is to partment of Urology, and governed by an executive com-
interpret the results. In many circumstances, there are justi- mittee (comprised of MUSIC urologists from across the
fiable reasons for not providing recommended treatments, state) and a core set of operating principles. The partici-
making 100% concordance an unattainable goal. That said, pation of each individual practice has been reviewed and
determining what the actual target should be is not straight- deemed not regulated by a local institutional review board.
forward. Using high-performing organizations to define a A centerpiece of the collaborative is a secure, web-based
benchmark is one option, but even high-performing organi- clinical registry. In each practice, a trained data coordinator
zations can improve in some situations and adjusting for le- screens case encounters to identify patients who are eligible
gitimate differences between organizations can be difficult. for inclusion in the MUSIC registry. Eligible cases include
Models that account for health-risk and case-mix exist, but patients undergoing prostate biopsy, as well as patients with
they are imperfect, can be challenging to implement, and only newly diagnosed prostate cancer that have not received
pertain to some measures.29-32 Often, organizations conducting prior cancer-directed treatment. Data are collected on pa-
quality improvement projects select internal benchmarks and tient demographics, cancer severity (including pathologic
plan to demonstrate improvement relative to past perfor- details from needle biopsies and radical prostatectomies),
mance. But, how much improvement can actually be attained? utilization and outcomes for radiographic staging studies,
Lastly, if suboptimal performance is identified, who should be comorbidities, cancer-directed therapies, and perioperative
responsible for that deficiency? Attribution is a particular complications and patient-reported outcomes for men un-
challenge in oncology, in which care often requires a multi- dergoing radical prostatectomy. MUSIC uses multiple quality
disciplinary approach spanning several different care settings. assurance techniques to ensure the integrity of the registry
These limitations notwithstanding, some organizations have data.35 The overall quality of the registry is reflected in its
been able to apply quality measures in the real-world setting inclusion as a Qualified Clinical Data Registry for the CMS
and have used the results of these efforts to effect meaningful Physician Quality Reporting System.
improvements in cancer care quality. The third section of this
manuscript highlights a number of these successes. Translating Data Into Quality Improvement
Although essential, a registry (and its associated reports and
USING PRACTICE VARIABILITY TO IDENTIFY metrics) is only the first link in a chain that leads to improved
OPPORTUNITIES FOR INTERVENTIONS TO quality and better patient outcomes. Data from the registry
IMPROVE QUALITY must be analyzed and presented back to clinicians in an
The Role of Quality Improvement Collaboratives understandable and actionable format. Most importantly,
As described previously, patients, providers, health care there must be engagement among participating clinicians,
payers, and policymakers are increasingly interested in including a desire to understand and respond to the data in
expanding efforts to measure and improve the quality of a way that ultimately yields better patient outcomes. Ex-
cancer care. As one mechanism for achieving this goal, some amples of such engagement include efforts to identify,

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e297


GANZ, HASSETT, AND MILLER

communicate, and disseminate specific processes of care or FIGURE 2. Example of a Quality Improvement Cycle
techniques (e.g., postoperative pain control pathways) that
are used by higher performing surgeons or practices. Im-
portantly, the collaborative model relies on learning from all
participants because no single hospital, practice, or surgeon
excels in all clinical domains or has a monopoly on the
knowledge and experience that leads to superior outcomes.35
This learning most commonly occurs through direct in-
teractions and discussions at collaborative-wide meetings
held several times per year. The meetings are attended by
clinical champions from each participating hospital or prac-
tice. These individuals review data and engage in vigorous
discussion and learning with peers; they also maintain re-
sponsibility for sharing the data and lessons learned with
partners and clinical team members in their local practices
and hospitals. Ultimately, the quality improvement loop must
be closed by translating the knowledge gained in the col-
laborative into specific interventions that yield measurable
improvements in patient care. This cycle of data collection and
measurement, analysis, discussion, interventions and then re- lowest reported at approximately 0.7%.37 We also recently
measurement is repeated for topics identified and deemed disseminated a toolkit that discusses the components of a
important by collaborative members (Fig. 2).35 The net effect biopsy bundle that may help to sustain and/or further reduce
is creation of an engine for comparative effectiveness re- the rate of infectious hospitalizations. In addition to the use of
search that is linked to a statewide quality improvement tailored or augmented antibiotic prophylaxis, the bundle
consortium that serves as the effector arm for translating data includes the use of a needle disinfectant, a practice associated
into action and, ultimately, better patient outcomes. with lower infection risk based on our own data in the MUSIC
registry and reports in the published literature.38
Specific MUSIC Initiatives
In terms of specific quality initiative priorities, MUSIC is Optimizing radiographic staging for men with early-stage
focused on reducing morbidity with prostate biopsy, opti- prostate cancer. As one of its first activities, MUSIC
mizing radiographic staging for men with newly diagnosed addressed ASCOs participation in the Choosing Wisely
prostate cancer, improving patient outcomes after radical priority of avoiding the use of staging radionuclide bone
prostatectomy, and leveraging clinical evidence and shared scans and CT scans among men with early-stage prostate
decision making to enhance the appropriateness of local cancer at low risk for metastases.39 By providing urologists
therapy for men with early-stage prostate cancer. Several of individual and practice-level feedback on their own practice
these initiatives are described in more detail below. patterns relative to their peers, as well as levels of adherence
with guideline recommendations, the collaborative achieved a
Reducing hospitalizations following prostate biopsy. Over rapid statewide decrease in the use of these radiographic
the last 2 years, MUSIC has implemented process changes staging evaluations for men with low-risk tumors.40
for antibiotic prophylaxis that have achieved and sustained a However, based on further analysis of both the existing
large reduction in prostate biopsy-related infectious hos- literature and our own registry data, it became clear that
pitalizations. After identifying a baseline hospitalization rate these imaging studies could safely be avoided in a much
of approximately 1.5% (with wide variability across practices larger population of men with newly diagnosed prostate
in the state), and fluoroquinolone resistance as the root cancer. Therefore, we developed and disseminated new
cause for most episodes of postbiopsy sepsis, MUSIC de- imaging appropriateness criteria that both minimize un-
veloped two pathways to address the rising prevalence of necessary imaging and are extremely unlikely to omit a
these bacteria.36 These included tailored antibiotic pro- positive study. The published MUSIC imaging criteria
phylaxis based on results from a rectal swab culture and recommend a bone scan for men with a Gleason score of 8
augmented antibiotic prophylaxis (i.e., the addition of a or higher or prostate-specific antigen (PSA) level of 20 ng/mL
second agent to standard fluoroquinolone therapy) based or higher, and a CT scan for men with a Gleason score of 8 or
on each communitys local antibiotic resistance profiles. The higher or a PSA level of 20 or higher, or clinical stage greater
development of these pathways was based on published than or equal to T3.41,42 After baseline variation in imag-
literature in this area and the specific clinical experience of ing practice patterns was established in 2012 and 2013, a
MUSIC urologists with the lowest rates of hospitalization. multidimensional quality improvement intervention was
The net result of this intervention has been a nearly 50% implemented in 2014, which included video presentations,
reduction in the frequency of these significant clinical events. dissemination of written guidelines, and site visits to each
Our current statewide rate of hospitalization is among the practice. Each component of the intervention aimed to

e298 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


IMPROVING THE QUALITY OF CANCER CARE

educate providers on the use and rationale for the MUSIC In summary, the partnership with Blue Cross Blue Shield of
imaging criteria. Michigan and patients with prostate cancer, urologists in
Among more than 10,000 patients newly diagnosed with Michigan are collecting clinically credible data on prostate
prostate cancer that were entered into the MUSIC registry cancer care and outcomes, comparing and capitalizing on
from March 2012 through July 2015, 8,271 patients (80%) variation in performance among practices and individual
met the criteria to avoid imaging. Among this group of surgeons, sharing best practices, and implementing changes
patients, we observed a large decrease in the use of both in clinical behavior. The net result has been more efficient
bone scans and CT scans from before and after dissemi- utilization of health care resources, improved care delivery
nation of the improvement interventions. We estimate that in our own environments, and enhancements in the quality,
application of the MUSIC criteria for imaging saved patients value, and outcomes of treatment provided to men in
in Michigan from undergoing 196 and 260 potentially un- Michigan with prostate cancer.
necessary bone scans and CT scans, respectively, over an 18-
month period. By decreasing the use of both bone scans and
CT scans among men with prostate cancer at low risk for CONCLUSION
metastases, this statewide intervention enhanced the value As can be seen by the preceding discussion, there are many
of care for this patient population through targeted im- opportunities and challenges associated with improving the
provement interventions. quality of cancer care delivery. With so many different clinicians
involved in the care of patients with cancer (e.g., surgeons,
Patient-reported outcomes after radical prostatectomy. medical oncologists, radiation oncologists, diagnostic radiolo-
Radical prostatectomy is a common treatment for men with gists, pathologists, nurses, primary care providers, and internal
early-stage prostate cancer. Improving functional outcomes medicine specialists), it is challenging to identify which prac-
after this procedure remains a priority for the urological titioners are accountable for specific aspects of care. In
community. To address this quality improvement challenge, addition, as cancer is a disease process with many clinical
MUSIC is also creating a statewide electronic infrastructure, manifestations depending on the organ site and disease
known as MUSIC-PRO, which will allow measurement of presentation, there are many processes and outcomes of
urinary and sexual function outcomes at a population-level care that need our attention. In terms of measurement, one
across diverse academic and community practices. does not know where to start.
MUSIC-PRO began with five practices in April 2014, and In contrast, the example of the hands-on urology col-
expanded to 17 practices by December 2015. For men laboratives described in Michigan provides a window on
undergoing radical prostatectomy in participating practices, what can be accomplished when a group of clinicians set out
the coordinating center emails patients with a link to an to examine how they are delivering care in specific situa-
online questionnaire on urinary function (score range 0-21) tions. This is especially valuable when there is consensus on
and erectile function (score range 0-30) before and at 3, 6, what types of tests or procedures provide little value or
12, and 24 months after radical prostatectomy. Patients cause substantial morbidity. It is also important that ex-
without email are provided with paper surveys. The results amination of clinical management practices occurs in a safe
of questionnaires are delivered to providers through the and constructive setting where the primary goal is to un-
registry, including patient-specific reports that can be derstand how and why some unnecessary tests or pro-
reviewed during clinic visits. Surgeons also receive summary cedures are performed, and then to address the situation
data on their own patients in comparison with patients in based on this understanding. Learning how to implement
their practice and at all sites. these best practices can be shared and supported through
Among 989 men participating in MUSIC-PRO through late the safety of the collaborative environment.
2015, the overall response rate was 85%, with 70% of the The issue of accountability for correct or harmful processes
surveys completed electronically. Across the collaborative, or outcomes will likely be resolved with the emergence of
52% and 71% of patients recovered urinary function (de- physician groups being merged into organizational units that
fined as a score greater than 17) at 3 and 6 months, re- will deliver cancer care from diagnosis through end of life in a
spectively. However, recovery of urinary function varied bundled or episode-based reimbursement scheme, such as
substantially across five pilot practices. Among patients with accountable care organizations. Under this circumstance,
good baseline erectile function (defined as a score greater coordination and quality of care will be the preeminent goal,
than 22), 15% and 21% recovered erectile function at 3 and and elimination of wasteful practices will be a central dogma.
6 months. Improvements in health and well-being will be the primary
We are now capitalizing on this variation in patient-reported goal, with measurement of patient-reported outcome mea-
outcomes across practices and surgeons to identify top per- sures (e.g., psychosocial well-being, control of symptoms).
formers and areas needing improvement. Further, these data The prevention of hospitalizations or emergency department
can be used for preoperative counseling and managing post- visits that are costly and dreaded by patients, will become
operative expectations. Leveraging this infrastructure, MUSIC is even more important. Fortunately, there are many experi-
developing a menu of strategies designed to improve func- ments in process across the country that will help bring these
tional recovery for patients across the state. important outcomes into mainstream cancer care.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e299


GANZ, HASSETT, AND MILLER

References

1. Levit L, Balogh E, Nass S, et al. Delivering High-Quality Cancer Care: 24. National Quality Forum. Measurement Framework: Evaluating Effi-
Charting a New Course for a System in Crisis. Washington, DC: The ciency Across Patient-Focused Episodes of Care. Washington, DC: Na-
National Academies Press; 2013. tional Quality Forum; 2009.
2. Erikson C, Salsberg E, Forte G, et al. Future supply and demand for 25. Measure Applications Partnership, The National Quality Forum. Per-
oncologists: challenges to assuring access to oncology services. J Oncol formance Measurement Coordination Strategy for PPS-Exempt Cancer
Pract. 2007;3:79-86. Hospitals. Washington, DC: National Quality Forum; 2012.
3. de Moor JS, Mariotto AB, Parry C, et al. Cancer survivors in the United 26. Hassett MJ, McNiff KK, Dicker AP, et al. High-priority topics for cancer
States: prevalence across the survivorship trajectory and implications quality measure development: results of the 2012 American Society of
for care. Cancer Epidemiol Biomarkers Prev. 2013;22:561-570. Clinical Oncology Collaborative Cancer Measure Summit. J Oncol Pract.
4. Kline RM, Bazell C, Smith E, et al. Centers for Medicare and Medicaid 2014;10:e160-e166.
Services: using an episode-based payment model to improve oncology 27. Hassett MJ, Hughes ME, Niland JC, et al. Selecting high priority quality
care. J Oncol Pract. 2015;11:114-116. measures for breast cancer quality improvement. Med Care. 2008;46:
5. Schnipper LE, Davidson NE, Wollins DS, et al; American Society of 762-770.
Clinical Oncology. American Society of Clinical Oncology statement: 28. Winchester DP, Stewart AK, Phillips JL, et al. The National Cancer Data
a conceptual framework to assess the value of cancer treatment op- Base: past, present, and future. Ann Surg Oncol. 2010;17:4-7.
tions. J Clin Oncol. 2015;33:2563-2577. 29. Ellis RP, Fernandez JG. Risk selection, risk adjustment and choice:
6. Saltz LB. The value of considering cost, and the cost of not considering concepts and lessons from the Americas. Int J Environ Res Public Health.
value. J Clin Oncol. 2015; JCO647867. Epub 2015 Dec 14. 2013;10:5299-5332.
7. Schnipper LE, Lyman GH, Blayney DW, et al. American Society of Clinical 30. Pouw ME, Peelen LM, Lingsma HF, et al. Hospital standardized mortality
Oncology 2013 top five list in oncology. J Clin Oncol. 2013;31:4362-4370. ratio: consequences of adjusting hospital mortality with indirect
8. Schnipper LE. Quality oncology practice initiative: an approach to standardization. PLoS One. 2013;8:e59160.
solving the value equation. J Oncol Pract. 2014;10:219-220. 31. Wennberg DE, Sharp SM, Bevan G, et al. A population health ap-
9. Nekhlyudov L, Levit L, Hurria A, et al. Patient-centered, evidence-based, and proach to reducing observational intensity bias in health risk ad-
cost-conscious cancer care across the continuum: translating the Institute of justment: cross sectional analysis of insurance claims. BMJ. 2014;
Medicine report into clinical practice. CA Cancer J Clin. 2014;64:408-421. 348:g2392.
10. Kohn LT, Corrigan J, Donaldson MS. To Err Is Human: Building a Safer 32. Fiscella K, Burstin HR, Nerenz DR. Quality measures and sociodemo-
Health System. Washington, DC: National Academy Press; 2000. graphic risk factors: to adjust or not to adjust. JAMA. 2014;312:
11. Institute of Medicine. Committee on Quality of Health Care in America: 2615-2616.
Crossing the Quality Chasm: A New Health System for the 21st Century. 33. Miller DC, Murtagh DS, Suh RS, et al. Establishment of a urological
Washington, DC: National Academy Press; 2001. surgery quality collaborative. J Urol. 2010;184:2485-2490.
12. NQMC. National Quality Measures Clearinghouse (NQMC). https:// 34. Share DA, Campbell DA, Birkmeyer N, et al. How a regional collaborative
www.qualitymeasures.ahrq.gov/. Accessed February 27, 2016. of hospitals and physicians in Michigan cut costs and improved the
13. National Quality Forum. Measure Evaluation Criteria and Guidance for quality of care. Health Aff (Millwood). 2011;30:636-645.
Evaluating Measures for Endorsement. Washington, DC: National 35. Montie JE, Linsell SM, Miller DC. Quality of care in urology and the
Quality Forum; 2015. Michigan Urological Surgery Improvement Collaborative. Urol Pract.
14. The National Quality Forum. Quality of Cancer Care Measures. Project 2014;1:74-78.
Steering Committee Meeting; 2005. 36. Womble PR, Dixon MW, Linsell SM, et al; Michigan Urological Surgery
15. Hassett MJ, Bach PB. The Current State of Cancer Quality Measurement. Improvement Collaborative. Infection related hospitalizations after
https://www.qualityforum.org/Publications/2008/09/White_Paper, prostate biopsy in a statewide quality improvement collaborative.
_The_Current_State_of_Cancer_Quality_Measurement.aspx. Accessed J Urol. 2014;191:1787-1792.
February 27, 2016. 37. Womble PR, Linsell SM, Gao Y, et al; Michigan Urological Surgery
16. Donabedian A. The quality of care. How can it be assessed? JAMA. 1988; Improvement Collaborative. A statewide intervention to reduce hos-
260:1743-1748. pitalizations after prostate biopsy. J Urol. 2015;194:403-409.
17. The Tax Relief and Health Care Act, Pub. L No. 109-432, 2006. 38. Issa MM, Al-Qassab UA, Hall J, et al. Formalin disinfection of biopsy
18. Centers for Medicaid & Medicare Services. Physician Quality Reporting needle minimizes the risk of sepsis following prostate biopsy. J Urol.
System, 2013. 2013;190:1769-1775.
19. The Patient Protection and Affordable Care Act, Pub. L No. 111-148, 2010. 39. ABIM Foundation. Imaging Tests for Early Prostate Cancer. http://www.
20. Blayney DW. Defining quality: QOPI is a start. J Oncol Pract. 2006;2:203. choosingwisely.org/patient-resources/imaging-tests-for-early-pros-
21. Neuss M, Gilmore TR, Kadlubek P. Tools for measuring and improving tate-cancer/. Accessed January 20, 2016.
the quality of oncology care: the Quality Oncology Practice Initiative 40. Ross I, Womble P, Ye J, et al. MUSIC: patterns of care in the radiographic
(QOPI) and the QOPI certification program. Oncology (Williston Park). staging of men with newly diagnosed low risk prostate cancer. J Urol.
2011;25:880, 883, 886-887. 2015;193:1159-1162.
22. Jacobson JO, Neuss MN, McNiff KK, et al. Improvement in oncology 41. Merdan S, Womble PR, Miller DC, et al. Toward better use of bone scans
practice performance through voluntary participation in the Quality among men with early-stage prostate cancer. Urology. 2014;84:
Oncology Practice Initiative. J Clin Oncol. 2008;26:1893-1898. 793-798.
23. McNiff KK, Neuss MN, Jacobson JO, et al. Measuring supportive care in 42. Risko R, Merdan S, Womble PR, et al. Clinical predictors and recom-
medical oncology practice: lessons learned from the quality oncology mendations for staging computed tomography scan among men with
practice initiative. J Clin Oncol. 2008;26:3832-3837. prostate cancer. Urology. 2014;84:1329-1334.

e300 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


HEALTH SERVICES RESEARCH AND QUALITY
OF CARE

Removing Barriers to Clinical Trial


Participation

CHAIR
Joseph M. Unger, PhD, MS
Fred Hutchinson Cancer Center
Seattle, WA

SPEAKERS
Archie Bleyer, MD
St. Charles Medical Center
Bend, OR

Elise Cook, MD, MS


The University of Texas MD Anderson Cancer Center
Houston, TX
CLINICAL TRIAL PARTICIPATION

The Role of Clinical Trial Participation in Cancer Research:


Barriers, Evidence, and Strategies
Joseph M. Unger, PhD, Elise Cook, MD, Eric Tai, MD, and Archie Bleyer, MD

OVERVIEW

Fewer than one in 20 adult patients with cancer enroll in cancer clinical trials. Although barriers to trial participation have
been the subject of frequent study, the rate of trial participation has not changed substantially over time. Barriers to trial
participation are structural, clinical, and attitudinal, and they differ according to demographic and socioeconomic factors. In
this article, we characterize the nature of cancer clinical trial barriers, and we consider global and local strategies for reducing
barriers. We also consider the specific case of adolescents with cancer and show that the low rate of trial enrollment in this
age group strongly correlates with limited improvements in cancer population outcomes compared with other age groups.
Our analysis suggests that a clinical trial system that enrolls patients at a higher rate produces treatment advances at a faster
rate and corresponding improvements in cancer population outcomes. Viewed in this light, the issue of clinical trial en-
rollment is foundational, lying at the heart of the cancer clinical trial endeavor. Fewer barriers to trial participation would
enable trials to be completed more quickly and would improve the generalizability of trial results. Moreover, increased
accrual to trials is important for patients, because trials provide patients the opportunity to receive the newest treatments.
In an era of increasing emphasis on a treatment decision-making process that incorporates the patient perspective, the
opportunity for patients to choose trial participation for their care is vital.

T he path from initial development of a new cancer drug to


diffusion of the new therapy into the cancer treatment
community relies on clinical trials, which represent the final
To understand the effect of clinical trial participation on
cancer population mortality and survival, one might
imagine a counterfactual system in which the cancer clinical
step in evaluating the efficacy of new therapeutic ap- trial participation rate was much higher. Fortunately, such a
proaches. It has been consistently estimated that less than system already exists. Enrollment of children (younger than
5% of adult patients with cancer enroll in cancer clinical age 15) to clinical trials has historically been much higher
trials.1,2 Conversely, the vast majority of adult patients with than for adult cancers (greater than 50%).2,6,7 At the same
cancer (greater than 95%) do not participate in clinical trials, time, mortality rates have for children have been decreasing
even though 70% of Americans are estimated to be inclined since the 1970s, whereas for adults they have been de-
or very willing to participate in clinical trials.3 Thus, a large gap creasing only since the 1990s.8 The average reduction in the
exists between trial participation rates and the willingness of rate of mortality from 1975 to 1995 was 2.6% per year for
patients to participate, suggesting that barriers to trial par- patients younger than age 20.9 Interestingly, the reduction
ticipation are numerous and frequently insurmountable. was weakest among older children (age 15 to 19; 2.0% per
Barriers to trial participation have been the subject of fre- year), reflecting other studies which have found both lower
quent study, but the rate of trial participation has not changed trial enrollment for adolescents and young adults with
substantially over time. The infrastructure supporting the cancer and lower rates of mortality reduction.10,11
conduct of clinical trials has been designed to anticipate a These data are consistent with the idea that a clinical trial
low, albeit steady, trial participation rate. The National Cancer system that enrolls patients at a higher rate produces
Institutes (NCI) cooperative group clinical trial treatment treatment advances at a faster rate, and concurrent survival
program caps enrollment for its funded groups at 17,000 total increases and mortality reductions in the cancer population.
patients per year, representing 1% of the estimated 1.7 million In this context, the issue of clinical trial enrollment is viewed
new cancer diagnoses in the United States in 2015.4,5 as foundational, lying at the heart of the cancer clinical trial

From the Fred Hutchinson Cancer Research Center, Seattle, WA; The University of Texas MD Anderson Cancer Center, Houston, TX; Centers for Disease Control and Prevention, Atlanta,
GA; St. Charles Health System, Quality Department, Bend, OR.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Joseph M. Unger, PhD, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N, M3-C102, P.O. Box 19024, Seattle, WA 98109;
email: junger@fredhutch.org.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 185


UNGER ET AL

endeavor.12 Therefore, the identification of specific barriers FIGURE 1. Model Pathway of Trial Enrollment Process
to trial enrollment and efforts to remove such barriers rep-
resent critical research objectives for cancer investigators.
In this article, we attempt to characterize the specific
barriers to cancer clinical trial participation. In addition, we
consider the distinction between clinical trial enrollment
between children and adolescents with cancer. As suggested
above, these age-proximal patient groups provide a natural
observational contrast illuminating the association between
clinical trial enrollment rates and corresponding improve-
ments in outcomes in the cancer population. We present
original data to make this case. Finally, we examine global and
local strategies to improve cancer clinical trial participation.

UNDERSTANDING BARRIERS TO CLINICAL TRIAL


PARTICIPATION
A patients decision of which cancer treatment to receive is Abbreviation: SES, socioeconomic status.
complex and deeply personal; the prospect of incorporating
clinical trial treatment into a patients care adds another point the patient makes a decision about whether to par-
level of complexity. In this multifactorial decision-making ticipate. An important note is that under this model, patient
environment, patients may face several barriers to trial par- attitudes toward clinical trial participation only come into
ticipation. As a guide to understanding the trial decision- play at the end of an otherwise long process.
making process, we present a simplified flow diagram (Fig. 1) We have categorized barriers to trial participation as
illustrating a representative pathway through which a patient structural (especially the absence of an available clinical
may receive care. This model has been the basis for multiple trial), clinical (i.e., not meeting eligibility), attitudinal (with
studies examining barriers to clinical trial participation.13-15 respect to both patients and physicians), and demographic
The model indicates that after cancer diagnosis and a clinic and socioeconomic. It is recognized that there is greater
visit, an assessment of trial availability is made to identify fluidity between these categories than the model allows,
whether a trial exists at the institution for the patients but simplifications were made to facilitate discussion.
histology and stage. If a trial is available, an evaluation of trial
eligibility is made, and, if eligible, a trial is discussed with the
patient. The trial may then be offered to the patient, at which Structural Barriers
To participate in a clinical trial, patients must first have
access to a cancer clinic. Access to a clinic can be influenced
by many different structural factors such as transportation,
KEY POINTS travel costs, access to insurance, and availability of child
care.16 Uninsured patients, in particular, present with later
Although barriers to trial participation have been the stage of disease and have worse cancer outcomes.17,18 To
subject of frequent study, the rate of trial participation the extent that such patients present at their cancer di-
has not changed substantially over time.
agnosis with a greater comorbid burden, their likelihood of
Barriers to trial participation are structural, clinical, and
attitudinal, and they differ according to demographic
eventually participating in a clinical trial is lower.19
and socioeconomic factors. Once a patient has access to cancer care, a major structural
An analysis of the specific case of adolescents with barrier pertains to the availability of a clinical trial for the
cancer illustrates how a clinical trial system that enrolls patients histology and stage. Multiple prospective studies of
patients at a higher rate produces treatment advances the cancer care decision-making process have examined the
at a faster rate and corresponding improvements in extent to which trials are unavailable for patients. Lara et al
cancer population outcomes. prospectively tracked barriers to cancer clinical trials at the
Fewer barriers to trial participation would enable trials University of California (UC) Davis Cancer Center from 1997
to be completed more quickly and would improve the to 2000.20 Among patients considered for trial availability,
generalizability of trial results. Crucially, increased there was no trial available to 47% of patients (Table 1). Javid
accrual to trials is important for patients because trials
et al registered patients to a prospective survey study prior
provide patients the opportunity to receive the newest
treatments.
to their treatment decision regarding their cancer care at a
In an era of increasing emphasis on a treatment diverse set of eight institutions. No trial was available for
decision-making process that incorporates the patient nearly half of the patients (46%).13 Together, these and
perspective, the opportunity for patients to choose trial earlier studies consistently show that once a patient has
participation for their care is vital. access to cancer care, the absence of an available clinical trial
precludes participation for about half of all patients.14,21,22

186 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


CLINICAL TRIAL PARTICIPATION

TABLE 1. Clinical Trial Enrollment Patterns for Multiple Studies in the Literature
No. Patients Examined* Trial Unavailable Ineligible Trial Participation Rate Did Not Participate
Lara et al20 171 47% 8% 23% 22%
13
Javid et al 9,09 46% 14% 16% 24%
Klabunde et al14 2,339 60% 16% 7% 17%
Begg et al21 3,534 33% 33% 16% 16%
22
Hunter et al 44,156 60% 18% 8% 14%
Average** 49% 18% 14% 19%
*Assessed for trial availability.
**Unweighted.

Clinical Barriers Multiple earlier studies found physician decision or pref-


Even if a trial is available, patients may not be eligible. Studies erence was the primary reason for nonparticipation in half of
have found that a common reason for patient ineligibility the patients for whom a protocol was available and the
to available protocols is narrow eligibility criteria.3,14,21-24 patient was eligible.21,22
Trial eligibility attempt to satisfy two opposing criteria. A number of factors have been found to deter physician
On the one hand, eligibility must be sufficiently narrow to recommendation for trial participation. In their role of
produce a treatment effect that is approximately consistent guiding patient care, physicians may have a strong inclina-
across the cohort. On the other hand, eligibility should be tion toward a specific treatment for a given patient.31-33 The
sufficiently inclusive that the trial targets a meaningful pop- prospective study by Javid and colleagues found that the
ulation of patients for whom a new treatment would apply.25 nature of the study regimen was cited as a reason for not
Eligibility criteria may also exclude patients due to safety discussing a trial with eligible patients by 56% of physi-
concerns. However, trials are often criticized for having cians.13 Physicians are also frequently concerned that clinical
eligibility criteria that are too narrow, sacrificing general- trial participation can interfere with the physician-patient
izability.26 These exclusions also make trials less accessible relationship.31,34,35 Random assignment into a phase III trial
for patients. in particular subjects the treatment choice to uncertainty,
In the previously described prospective studies of trial and physicians may anticipate that the introduction of un-
barriers, ineligibility was identified as a reason for nonpartic- certainty will subvert patient confidence in the physicians
ipation by 18% of all patients on average (Table 1).13,14,20-22,27 expertise, even if, as indicated by the existence of a ran-
The dominant reason for ineligibility exclusions is likely ex- domized clinical trial, multiple treatments of potentially
clusions due to comorbid conditions. One recent study com- similar efficacy are available.
prehensively cataloged the trial eligibility criteria for a set Practical considerations may also influence physicians
of 21 trials in diverse cancer settings.28 The authors found willingness to participate in trials. Physicians often lack
that the average number of eligibility criteria per trial was appropriate incentives to participate in clinical research.36
16, 60% of which were related to comorbidity or perfor- The time spent attending to the details of clinical trial en-
mance status. Although prespecified trial eligibility criteria rollment and explaining clinical trials to patients can often
that protect patient safety are crucial, it is also possible that be prohibitive for physicians.33 In addition, in a busy clinic
certain kinds of exclusions are unnecessary. A recent report environment, physicians may be less likely to refer patients
indicated trial eligibility criteria have increased in recent years to trials if they believe the process will be too time
for both academic group and pharmaceutical-sponsored consuming.30,31,37 Oncologists who consider trial paper-
clinical trials.29 This trend not only renders trials less acces- work time consuming or who otherwise believe trial ef-
sible for many patients, it may also limit the generalizability fort would be extra work are less likely to refer a patient
of trial results. to a clinical trial.37 Finally, some physicians find the re-
quirement of obtaining informed consent to be problem-
atic, even though nearly all agree that informed consent is
Physician Attitudes necessary.30,31
As the agent linking patients to their cancer care, physicians
play an obvious and vital role in clinical trial participation.
A survey of oncologists in community cancer clinics found Patient Attitudes
that most agreed that clinical trials provide high-quality Efforts to reduce structural, clinical, and physician barriers to
care (87%) and benefit enrolled patients (83%).30 However, trial participation are critical. However, the ultimate decision
physicians face their own barriers to trial enrollment, so regarding trial participation rests with the patient. In-
even if quality cancer care is assumed, physicians may treat evitably, the decision about whether to participate in a trial
otherwise eligible patients off-protocol with one arm of will reflect a patients personal preferences, which may also
a trial, without actually entering the patient on the trial.31 be influenced by family and friends.38

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 187


UNGER ET AL

Some proportion of patients are influenced by altruistic likely to have more comorbid burdens, inducing clinical
motivations.15 However, the majority of patients are (ap- exclusions.57,58 To the extent that trials seek to reflect the
propriately) concerned primarily with finding the best population of patients for whom new trial-proven treat-
possible treatment of their disease.15,39,40 In the absence of ments will be administered, better representation of older
other barriers, a patient who believes that the best possible patients in trials is critical. Recognizing this, in 2000,
treatment option is to be found in a clinical trial is more likely Medicare was directed to cover the routine care costs of
to participate in that clinical trial.33 clinical trial participation for its patients.59 Unfortunately,
Patients have frequently reported being uneasy or fearful the proportion of older patients in trials remains well below
about the prospect of participating in a clinical trials.41 In the expected rate.28,51
some cases this could be due to a residual mistrust of Evidence as to the association of race with trial partici-
medical science due to past abuses, such as the infamous pation is mixed. A study by Murthy et al found that black
Tuskegee Syphilis Study or the history of human experi- patients were underrepresented in NCI-sponsored breast,
mentation with radiation following World War II.42,43 At- lung, colorectal, and prostate cancer clinical trials from 2000
tention in the last several decades to the process of rigorous to 2002.1,60 In contrast, in sequential studies within SWOG, a
consent may have reduced these fears, especially for national clinical trials consortium, black patients were en-
younger generations of patients. Attention must also be paid rolled to trials in a representative fashion over an extended
to providing consent forms which are easy to read, because period of examination.28,50,53 This was confirmed in a sample
more complicated consent forms can induce anxiety.44 of older patients with breast cancer.61 Evidence has also
More generally, a fear of experimentation may be been mixed for Hispanic patients.1,53 Even if enrollment of
expressed through a dislike of randomization.14,15,22,45-47 minorities is adequate in the treatment trial setting, en-
There is perhaps no stronger indication that a patient is rollment of minority healthy volunteers for prevention trials
about to participate in an experiment than the revelation has been decidedly more difficult and has generated well-
that the patient will be randomly allocated to one of two or designed outreach programs for large individual trials.62-65
more treatments. Fear of randomization has been identified Given the increasingly diverse nature of the U.S. population,
as the most commonly cited reason by patients for declining continued attention to this issue is required.
trial participation.15 Recognizing this, some physicians avoid Females may be somewhat under-represented in the
the word randomization, relying instead on analogy to nonsex-specific cancers, although the magnitude of the
describe the randomization process, though this may lead to disparity is likely small.28,50,53 In addition, some evidence
situations where patients sometimes do not understand that suggests that any sex disparity in clinical trial enrollment
their treatment has been randomly assigned.48,49 could be age-related, with older women less likely to enroll
Patients are sometimes uneasy as well about the potential in trials than their younger counterparts.1 Such a pattern
toxic effects of chemotherapy in trials, especially for the might indicate generational differences in attitudes toward
experimental therapies.38 Patients may already have a clinical research.
strong sense of the particular treatment they wish to receive The examination of socioeconomic factors as a barrier to
after discussion with their physicians.33 Because trials participation has historically been hindered by the lack of
sometimes require more frequent monitoring than nontrial collection of patient-level socioeconomic status (SES) data.
care, traveling to and from a cancer clinic has been indicated This is unfortunate given the frequency with which the direct
by many patients to be a reason for nonparticipation.15,20 and indirect costs of trial participation have been cited as
Concern about how to pay for trials has been cited as a meaningful barriers.41 Two recent articles overcame this
reason for nonparticipation among about a quarter of pa- limitation. The first used a web-based survey to engage
tients, despite the fact that the majority of states mandate patients in their decision-making process.15 Among the
that insurers cover the routine care costs of trials, as does numerous demographic and socioeconomic factors exam-
Medicare.15 In a review by Ford et al, cost concerns were ined, patients with lower income (less than $50,000 per
identified as the second most frequently indicated reason year) were 29% less likely to participate in trials than higher-
for nonparticipation in trials in the literature.41 income patients. Utilizing data from a prospective barriers
study, this observation was confirmed.13,66 Thus, the evi-
dence to date of income disparities in trial enrollment is
Demographic and Socioeconomic Disparities fairly consistent.61 Given that trial treatment costs are not
Demographic and socioeconomic disparities in trial enroll- substantially different than nontrial treatment costs,67 this
ment can occur anywhere along the pathway from initial suggests that marginal direct costs play a role. A prior study
clinic visit until the patient ultimately makes their treatment of the effect of the 2000 Medicare policy change on trial
decision. The most consistent and largest disparity pertains participation found that patients with Medicare plus private
to age.1,15,50-53 Hutchins et al found that patients in co- insurance participated at a higher rate following the policy
operative group trials were much less likely to be age 65 or change, whereas patients with Medicare alone participated
older than those in the U.S. cancer population.50 Some at rates similar to those prior to the policy change.53 This
evidence suggests that attitudinal barriers on the part of finding points to the prohibitive influence of copays and
physicians play a role.13,54-56 In addition, older patients are coinsurance for some patients.

188 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


CLINICAL TRIAL PARTICIPATION

EVIDENCE FOR THE BENEFIT OF CLINICAL TRIALS advent of the Affordable Care Act, which could be associated
ON CANCER POPULATION OUTCOMES with delays in diagnosis and compromises in optimum di-
OBSERVED THROUGH THE RELATIVE LACK OF agnostic and therapeutic interventions.72 At the same time,
PROGRESS IN ADOLESCENTS AND YOUNG AYAs have had the lowest participation in clinical trials in
ADULTS absolute terms than any other major age group.10 The
The potential barriers to trial enrollment that patients face central issue then is the extent to which lack of clinical trial
are numerous. But, just how important are clinical trials for activity in AYA patients with cancer accounts for the relative
progress against cancer? The answer to this question is lack of progress in improving cancer population outcomes.
crucial, because if trial participation is ultimately unrelated
to cancer population survival gains, the issue of barriers to All Cancers
trial participation has little importance. To examine this, we The Cancer Therapy Evaluation Program (CTEP) of NCIs
studied the relationship between adolescents and young Division of Cancer Therapy and Diagnosis has patient accrual
adults (AYAs) and cancer population outcomes over time. data from phase I, II, and III cancer treatment trials con-
This is an ideal group in which to examine the impact of ducted by the NCI cooperative groups and NCI-designated
clinical trials given that since 1980, AYA patients have had a cancer centers. For this analysis, 371,302 patient entries
slower rate of cancer population survival improvement than during 1997 to 2009 were examined. In addition, we used
younger and older age groups by 5% to 13% in absolute cancer population data derived from the Surveillance, Epi-
differences (Fig. 2). Concomitantly, cancer has become the demiology, and End Results registry, U.S. Census data, and
most frequent cause of death due to disease in AYAs.68 joinpoint statistical software analyses to examine trends in
During the past decade (2000 to 2009), deaths due to cancer U.S. cancer population estimates.73-78
declined in all age groups except in young adults age 20 to Figure 3 shows the relationship between the average
29; in 25- to 29-year-olds, deaths due to cancer actually percent change (APC) in the 5-year cancer-specific survival
increased.69 rate from 1985 to 1999 and the accrual rate to national
The survival disparity between AYAs and other patients cancer treatment trials during 2001 to 2006. Although this
may be due in part to early achievements in improving comparison is confounded by time, there was a nearly 1:1
survival for AYAs, after which resources were directed to- correlation over the entire age range that was strongly
ward research in other age groups. Also, cancer in AYAs have significant. Patients age 15 to 34 had the lowest APC in
potentially complex biologic signatures that neither pedi- 5-year survival. A similar pattern was found with respect to
atric oncologists nor adult-treating medical and hematolo- cancer mortality, which isolates patients age 0 to 40 (Fig. 4).
gist oncologists are accustomed to treating.70 AYAs are also Patients age 20 to 24 had a particularly poor reduction in
less likely to have health insurance,71 especially prior to the cancer mortality, as well as the lowest absolute number of
clinical trial accruals. In both cases, the correlation between
trial enrollment and, respectively, APC in 5-year survival and
FIGURE 2. Increase in Absolute Percentage of Annual mortality is clearly evident and highly significant (p , .001).
5-Year Cancer-Specific Survival Rates Since 1973 to
1975 by Calendar Year and Age
Acute Lymphoblastic Leukemia
The greatest effort during the last decade to increase ac-
cruals in AYAs was within the population with acute lym-
phoblastic leukemia (ALL), the most common pediatric
cancer. New clinical trials in ALL specifically designed for
AYAs were launched,79-81 the National Comprehensive
Cancer Network released practice guidelines for ALL,82 and
an increasing number of presentations and publications on
the topic occurred at national meetings and appeared in the
peer-reviewed medical literature.83-89 The effort was ef-
fective, with AYAs having the greatest accrual increase of all
cancers in ALL, up to twice that of the cancer with the next
greatest increase, acute myeloid leukemia.74,90 Perhaps in
part for this reason, the only increase in either absolute
accruals or accrual as a proportion of cases during the first
decade of the 21st century occurred among patients age
10 to 20 (Fig. 5).
Hence, the population with ALL was examined more
Baseline is 1973 to 1975 average. Kaposi sarcoma is excluded due to the HIV/ closely for a relationship between survival improvement and
AIDS epidemic during the 1980s and early 1990s; thyroid cancer is excluded
because of overdiagnosis and increasing survival inflation. Regressions are 4
clinical trial participation. As shown in Fig. 6, clinical trial
polynomials. Data source is SEER 9 regions.75 accruals as a proportion of ALL cases in the United States

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 189


UNGER ET AL

FIGURE 3. Comparison of Average Percent Change in referrals, participation, and conduct. Fortunately, NCI-
the 5-Year Cancer Survival Rate and Treatment Trial designated cancer centers are evaluating their own AYA
Accruals, by 5-Year Age Intervals referral patterns and clinical trial determinants,92,93 and
intergroup efforts are underway within the current orga-
nizational structure of the federal clinical trials enterprise,
including the NCIs National Clinical Trials Network (NCTN),
to create novel opportunities for collaborative AYA oncology
research among the pediatric and adult NCTN groups.94,95 As
is done in England, age-specific biology, pharmacology,
proteomics, genomics, and clinician and patient behavior
studies embedded within clinical trials are required to
further improve survival for AYAs.96

GLOBAL AND LOCAL STRATEGIES TO IMPROVE


CLINICAL TRIAL PARTICIPATION
We have illustrated the nature of clinical trial enrollment
barriers and established the potential link between trial
enrollment and improvements in cancer population survival.
The open columns represent trial accruals during 2001 to 2006 and the colored Efforts to improve trial enrollment of cancer patients are
bars the average percent change (APC) in 5-year relative survival rate of all
invasive cancer except Kaposi sarcoma during 1985 to 1999. The red bars indicate
clearly needed. In this context, we propose the following
the adolescent and young adult (AYA) age group. The inset compares the APC in global and local strategies to improve trial participation.
5-year survival rate with the treatment trial accruals. Accrual data from the
National Cancer Institute Cancer Therapy Evaluation Program (CTEP) were
provided by Steve Friedman, Michael Montello, Troy Budd, and Samantha
Finnegan via the Freedom of Information Act. Survival data were obtained from
Overall Strategies
SEER 9 Regions.75 Kaposi sarcoma is excluded from the survival statistic because the In 2010, the NCI and the American Society of Clinical On-
HIV/AIDS epidemic occurred during the 1980s and early 1990s, which substantively
altered the overall cancer survival rate in AYAs during those years.
cology sponsored a Cancer Trial Accrual Symposium to
provide recommendations for trial recruitment. Summary
recommendations centered on the patient, community,
during 2000 to 2009 drops precipitously for patients age physician/provider, and site.97 This symposium led to many
15 to 20. Figure 6 also shows the 5-year leukemia-specific recommendations at each level consistent with the over-
survival rate for patients with ALL as a function of single year arching view that one size does not fit all when it comes
of age. Joinpoint analysis identified two inflections, ages 17 and to recruitment to clinical trials. The organizers noted in
20, during which the 5-year survival rate decreased 23%. This
AYA ALL cliff constituted 30% of the overall decline from
95% at age 5 to less than 20% at age 70. The cliff patterns for FIGURE 4. Comparison of Average Percent Reduction
both accrual and survival are virtually superimposable, in the Annual National Cancer Mortality Rate and
which strongly suggests they are related, although other Treatment Trial Accruals, by 5-Year Age Intervals, Age
factors, such as a switch from pediatric to adult treatment Younger Than 40
regimens, could also contribute.91

Clinical Trials Effect Summary


These data enable three fundamental conclusions. First,
both survival prolongation and mortality reduction in pa-
tients with cancer are correlated with clinical trial activity.
Second, the dependency of survival prolongation on treat-
ment trial accrual has been apparent for all ages. Third, AYAs
have had the least trial participation and the least survival
prolongation and mortality reduction, particularly among
patients age 20 to 29.
It has been previously observed that the age-dependent
rate in the reduction of deaths attributed to cancer in the
United States is correlated with the age-dependent accrual The open columns represent trial accruals during 2000 to 2006 and the colored
of young adults to national cancer treatment trials during the bars the average percent reduction in national cancer mortality rate during 1990
same era.69 After suicide, cancer is the second leading cause to 1998. The red bars indicate the adolescent and young adult (AYA) age group.
The inset compares the mortality rate reduction with the treatment trial accruals.
of death due to disease among AYAs. More is needed to Accrual data from the National Cancer Institute Cancer Therapy Evaluation
Program (CTEP) were provided by Steve Friedman, Michael Montello, Troy Budd,
overcome the national AYA cancer-related death problem, and Samantha Finnegan via the Freedom of Information Act. Mortality data were
beginning with increased clinical trial availability, access, obtained from the National Center for Health Statistics via the SEER program.78

190 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


CLINICAL TRIAL PARTICIPATION

FIGURE 5. Absolute and Relative Trial Accrual Rates to 6. Decentralize laboratories in favor of developing re-
NCI Treatment Trials: Comparison of 2001 to 2003 gional expertise to provide carry-over benefits after
Versus 2007 to 2009 Estimates trial completion.
7. Streamline and advance bio-bank regulatory issues.
8. Invest in research and evidence-based cancer care
relevant to each region, including cancer registries
and clinical trial infrastructure.
In the authors view, the globalization of clinical research
is vital to speed up availability of life-saving medicines
throughout the world.99 In the setting of a domestic clinical
trial system that has been described as being in a state of
crisis, this view has added weight.12
The importance of differing cultural, scientific, ethical,
government, and logistic issues in each region must be
considered.100 One key ethical issue is whether the study
drug will be available at the end of the clinical trial. Avail-
ability may be affected by local religious customs concerning
Comparison of 2001 to 2003 versus 2007 to 2009 for annual accruals to
contraceptive studies and ethical guidelines limiting pe-
treatment trials sponsored by National Cancer Institute (NCI)-sponsored diatric clinical trials. An additional ethical issue in low re-
cooperative groups and NCI-designated cancer centers (red curves) and accrual
proportion of all patients in the United States with invasive cancer into the trials source countries involves whether to develop local resources
by 5-year age intervals (green curves), by single years of age. The heavy curves for testing or to use international vendors for that purpose.
represent 2007 to 2009 and the thin curves 2001 to 2003. Accrual data from the
NCI Cancer Therapy Evaluation Program (CTEP) were provided by Steve Friedman, Although developing local resources provides an often needed
Michael Montello, Troy Budd, and Samantha Finnegan via the Freedom of benefit, this could involve higher costs and may also be deterred
Information Act. Accrual proportion (%) was estimated from cancer incidence in
SEER 9 regions and population data from the U.S. Census Bureau.73,75 by local shipping laws and other logistic barriers.
An excellent example of global recruitment is the START
trial. 101 START is a multicenter, phase III, randomized,
particular that although clinical trial enrollment barriers double-blind, placebo-controlled trial of the cancer vaccine
have been extensively studied, Few rigorously conducted tecemotide in patients with non-small cell lung cancer with
studies have tested interventions to address challenges to unresectable stage III disease. The trial is sponsored by
clinical trials accrual.97 In this broader context, the NCIs Merck KGaA and EMD Serono, Inc., in 275 study centers in
AccrualNet website provides strategies, tools, and other 33 countries worldwide. A continuous series of strategies
resources to support clinical trials.98 was implemented for patient recruitment and retention
throughout the duration of the trial.102 These strategies
were referred to as the START global patient recruitment
Global Strategies and retention continuum, with the overarching purpose of
At the beginning of this article, we delineated many of the raising awareness of the START trial and keeping it in the
specific challenges to clinical trial enrollment. Given the forefront in physician communities and in the local START
need to accrue large numbers of patients in a shortened sites to increase patient enrollment and retention. The
timeline and the increased complexity of U.S.-based clin- strategies target physicians, research staff, local sites, pa-
ical trials, academic and industry sponsors are increasingly tients, and the local community. Physicians were provided
exploring regions outside of the United States to conduct START informational calls (sometimes physician-to-physician),
trials, including in less developed regions of the world. visits, and meetings, and START information at national on-
In the view of Barrios et al, the globalization of clinical trial cology meetings. Research staff received START education,
research is unavoidable.99 In a wide ranging review, they and recruitment tools including motivational videos. The
propose the following solutions to some of the challenges local sites were offered additional site funding for acceler-
of clinical trial globalization: ated recruitment and START educational teleconferencing.
1. Harmonize and share standards and goals for product Patients received holiday and thank you cards and patient
safety, quality, and efficacy. START educational materials. The local community was
2. Use finite resources more efficiently, share knowl- reached through local media outreach, public awareness
edge, and optimize inspection resources. advertisements, and engagement of local site liaisons. En-
3. Engage global partners to advance regulatory science rollment to the trial occurred from 2007 through 2011 and
and public health solutions. reached full accrual of 1,513 patients, indicating a highly
4. Implement risk-based monitoring and clinical inspection. successful recruitment effort.
5. Incorporate error reduction strategies and consider Domestic trials may also partner with international col-
regional variations in the standards of care and their laborators to augment trial enrollment. The SWOG SELECT
effect on trial results. trial for prostate cancer prevention used similar recruitment

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 191


UNGER ET AL

FIGURE 6. 5-Year Leukemia-Specific Survival Rates in Moreover, predetermined IRB-approved responses to


Patients with Acute Lymphoblastic Leukemia anticipated user-generated content should be available to
Diagnosed From 2000 to 2012, and Estimated ALL allow immediate responses when required, and links to
Treatment Trial Accrual Proportion From 2000 to the study website should be included to allow for integration
2009, by Single Years of Age of study platforms.
The U.S. Food and Drug Administration (FDA) has provided
no specific guidance on the use of social media in clinical
research.107 The Recruitment Information Sheet states that
in the case of direct advertising, the information and mode
of communication should be reviewed by the IRB for evi-
dence of coercion or implication of benefits to participation.
The Office of the Inspector General has released guidance
regarding the use of clinical trial websites, indicating that IRB
approval of a clinical trial listing, if limited to selected basic
information, is not required.108
InVentiv Health, a contract research organization, has
provided specific plans to assist researchers to plan digital
recruitment campaigns (Table 2).109 Their plans are pre-
mised on the idea that recruitment for global clinical trials is
an area ripe for re-engineering. More broadly, Facebook has
often been listed as the social media of choice; of the 73% of
online U.S. adults using social media, 71% use Facebook.
Each year of age was averaged from two consecutive years. Joinpoint analysis of Often researchers using Facebook attempt to recruit from
survival data identified ages 17 and 2074; linear regressions for survival data are for
age ranges 5 to 17, 17 to 20, and 20 to 70. Survival data were obtained from SEER 18 the initial audience prior to forming a relationship. A better
regions.77 Accrual data from the NCI Cancer Therapy Evaluation Program (CTEP) were approach may be to grow and engage your audience first,
provided by Steve Friedman, Michael Montello, Troy Budd, and Samantha Finnegan
via the Freedom of Information Act. Accrual proportion (%) was estimated from before patients are recruited.110
cancer incidence in SEER 9 regions and population data from the U.S. Census
Bureau.73,75

Strategies to Address Demographic and


strategies as the START trial. The SELECT trial enrolled over Socioeconomic Barriers
35,000 men in the United States, Canada, and Puerto Rico, Elderly recruitment. Patients with concomitant illnesses
and completed enrollment 2 years ahead of schedule.62,63 are often excluded from trials to ensure safety and to isolate
On a smaller accrual scale, the contributions of the In- the cancer as the primary source of morbidity in the patient.
ternational Breast Cancer Study Group, in collaboration with Unfortunately, this has the effect of excluding many patients
the cooperative groups of the NCI, provided necessary ac- from trials, especially older patients with a greater comorbid
crual to a trial evaluating ovarian failure in premenopausal burden.28,57,58 Further, trials typically exclude patients with
women with breast cancer.103 prior cancers, even as the population of cancer survivors in
the United States is growing and which currently numbers
around 15 million.111 In this context, one strategy to remove
Local Strategies
barriers to trials would be to remove unnecessary eligibility
Social media. Increasingly, social media platforms provide
criteria. Such an approach would improve access to trials,
an opportunity to communicate about clinical trials with
especially for older patients, andbecause histology and
potential trial researchers and participants.104 The Quorum
stage explain the vast majority of variation in cancer out-
Review Institutional Review Board (IRB) offered the fol-
comes, rather than comorbid conditionswould result in
lowing considerations for a plan to use social media in
only limited loss of power to test the efficacy of new
research105,106:
treatments.28 One study estimated that if protocol exclu-
1. Provide a rationale for the application of social media
sions related to organ system abnormalities and functional
to the target population. status were relaxed, participation of older patients in clinical
2. Address the privacy and confidentiality concerns of trials would approach 60%, in line with cancer population
the social media applications to be used. rates.51
3. Vet all communications for sensitivity and potential for Researchers should also consider increasing the number of
harm, even if the content does not require IRB approval. trials targeted to older patients, with due consideration to
4. Provide a summary statement regarding the intended potential safety issues.112 Several trials found no more
uses of the social media account. toxicity in elderly patients in chemotherapy-containing trials
5. Closely monitor user-generated content (if allowed), than in younger patients, when patients were appropriately
which is essential to a robust online community, for selected.113,114 However, when chemotherapy was given
patient protection and study integrity. to patients age 80 or older, high risk of hospitalization or

192 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


CLINICAL TRIAL PARTICIPATION

TABLE 2. Proposed Steps to Plan Digital Recruitment evidence that payment inducements do or do not affect
Campaigns109 assessment.123,124 A careful calibration of the size of any
monetary incentive would be necessary to avoid undue
Steps for Campaign influence.125
Steps for Strategy Development Implementation
Measures to address socioeconomic disparities in re-
Audit digital channels and social Place ads with key messages and cruitment may have a preferentially beneficial impact on
media sites to determine a call to action in the selected
where the targeted patients/ sites and social media platforms minority patients. For the SELECT trial, several strategies
caregivers can be found specifically addressed patients with low socioeconomic
Monitor existing online chats for Link banner or pop-up ads to status.61,62 SELECT provided funds to sites semiannually to
the area of research interest search engine results for offset travel expenses and meals, in addition to providing
to understand the issues and targeted health-related
concern to patients/caregivers searches
patient retention items. Larger supplemental site grants
and listen for patient speak so were awarded to 15 SELECT sites with potential to increase
it can be replicated in your minority recruitment through a competitive award mech-
messages
anism. These additional funds were most commonly used to
Map the recruitment messages Partner with advocacy groups and provide additional staff time for minority recruitment. Sites
to the targeted patients/ place trial ads on these groups
caregivers and appropriate online sites also provided reimbursement for food and/or trans-
channels portation costs expended to participate in the trial.62
Identify key bloggers and others Engage key opinion leaders as
who could eventually raise online ambassadors to raise
awareness and support trial awareness of the trial
participation
DISCUSSION
Both patients and physicians have been found to regard
Develop the creative concepts
for advertising and test the clinical trial participation as a positive approach to cancer
messaging and imagery for care.3 Despite this, the complexity of the enrollment process
effectiveness and the potential barriers faced by patients have combined
to make a successful clinical trial enrollment a rare event.
Clinical trials are the key step in advancing new treatments
treatment discontinuation as a result of toxicity (even with from the research setting to the cancer care clinic.
frequent dose modifications) were observed.115 The In- Therefore, a thorough understanding of the nature of trial
ternational Society of Geriatric Oncology recommends the enrollment patterns and barriers to enrollment is of para-
use of comprehensive geriatric assessment (CGA) in cancer mount importance. The literature indicates that structural
patients older than age 70.116,117 The CGA is time con- barriers preclude patient participation in trials for half of all
suming, often leading to physician abandonment. Fortu- cancer patients. Among patients for whom a trial is available,
nately, CGA time requirements have led to the development about half (or a quarter of all patients) are excluded due to
of prescreening tools used to determine whether full eligibility issues with trial exclusion criteria. The remaining
screening with CGA is required, though there are in- patients are sometimes not offered the chance to participate
consistent results regarding the validity of these tools.118-120 because of physician concerns, or decline due to patient
More generally, it is important to develop prediction models concerns. Structural, clinical, and attitudinal barriers to trials
capable of estimating risk of chemotherapy for octogenar- can differ according to some important factors, especially
ians and nonagenarians with regards to toxicity and hos- age. As a result, only a small portion of adult patients with
pitalization.115 A comprehensive approach to the evaluation cancer participate in trials, less than 5%, a rate that has
of the older patient with cancer considers the patients remained fairly constant over decades.
residence and fitness and includes an interdisciplinary team Increasing accrual to clinical trials is important for multiple
to provide individualized care.121 reasons. Faster accrual would enable trials to be conducted
more quickly. The predominant reason that trials fail to
Socioeconomic barriers. If marginal direct costs are pro- complete is poor accrual.126 These failures represent a lost
hibitive for some patients, then measures to cover these investment on the part of funding agencies. Moreover, when
costs would remove a critical barrier to enrollment. One clinical trials close because of failure to accrue, nonfinancial
approach would be to cover the excess costs of clinical trials costs are also incurred. Patients have exposures to study
for all patients, because even in an insured population, drugs with potential or realized adverse events. Patients and
copays and coinsurance have been shown to deter clinical research staff may experience psychologic effects such as
trial participation.53 Another potential approach would be to loss of trust and morale. Informed consent documents rarely
provide payments to patients. In the United States, the include the risk of closure because of lack of study partici-
practice of paying patients for trial participation is wide- pation, despite the fact that about one in four randomized,
spread, but also contentious, highly variable, and lacking in phase III trials have such an outcome.126
general guidance.122 One concern is that a payment in- The more rapid completion of trials would enable new
ducement might alter a subjects assessment of potential treatments to be developed more quickly. We have shown
risks or impair their judgment, although there is little data indicating a compelling relationship between the

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 193


UNGER ET AL

incidence of clinical trial enrollments and improvements in FIGURE 7. Estimated Treatment Trial Accrual
cancer population survival outcomes. Our focus was a natural Proportion of Patients Diagnosed With Cancer From
observational contrast between AYAs and other age groups 2008 to 2010 by Single Year of Age and the History of
with cancer. We found a slower rate of progress in AYAs SEER Representation of the United States Population
compared with younger and older patients, which underscores
the need to increase the number of clinical trials available to
AYAs with cancer and their participation in them. By extension,
this observation also points to the need to increase trial en-
rollment for patients of any age group, or any demographic
group, because this could have a beneficial impact on in-
creasing survival and reducing mortality from cancer.
Another important reason to increase clinical trial accrual is
to improve the generalizability of clinical trial results. Figure 7
illustrates how the vast majority of patients with cancer of all
agesbut, especially patients older than about age 15do
not participate in clinical trials.127 However, we have made
the case that there is a strong correlation between trial
participation and cancer population survival improvements.
Thus, trial results must generalize to nontrial patients, at
least to some degree. But, they may not do so in an efficient
manner, and cancer population survival gains may be lost in
Accrual data from the NCI Cancer Therapy Evaluation Program (CTEP) were
the process. Under this rubric, greater participation leads to provided by Steve Friedman, Michael Montello, Troy Budd, and Samantha
greater generalizability, which leads to better cancer pop- Finnegan via the Freedom of Information Act. Accrual proportion (%) was
estimated from cancer incidence in SEER 9, SEER 13, and SEER 18 regions and
ulation outcomes. Patients who participate in cancer trials population data from the U.S. Census Bureau.73,75-77
are usually younger, healthier, and perhaps wealthier than Modified from Bleyer A.127
the typical patient who is not a trial participant. To the extent
that trials are more inclusive with respect to comorbid or
other conditions, adequately represent the demographic access to potentially less expensive therapies (if the agent
makeup of the United States, and are easier to pay for, the is provided at no or reduced cost to the patient), to teams
generalizability of trials would likely improve. of professional dedicated to the patients care, and to care
Finally, increased accrual to trials is important for patients, that is strictly directed by a protocol. Moreover, in an era
because trials provide opportunity to receive the newest of increasing emphasis on a treatment decision-making
treatments. The principle of equipoise posits that a properly process that incorporates the patient perspective, the
designed treatment trial tests a new or modified form of opportunity for patients to choose trial participation for
therapy that is not known to have that benefit (otherwise their care should not be hindered by unnecessary barri-
the trial would not be justified). In contrast, in addition to ers. In the end, the potential benefits of trial participa-
access to the newest treatments, patients who participate tion will be shared by patients, researchers, and future
in clinical trials have other potential advantages, such as generations.

References
1. Murthy VH, Krumholz HM, Gross CP. Participation in cancer clinical trials: 7. Hunger SP, Lu X, Devidas M, et al. Improved survival for children and
race-, sex-, and age-based disparities. JAMA. 2004;291:2720-2726. adolescents with acute lymphoblastic leukemia between 1990 and
2. Tejeda HA, Green SB, Trimble EL, et al. Representation of African- 2005: a report from the Childrens Oncology Group. J Clin Oncol. 2012;
Americans, Hispanics, and whites in National Cancer Institute cancer 30:1663-1669.
treatment trials. J Natl Cancer Inst. 1996;88:812-816. 8. Kohler BA, Sherman RL, Howlader N, et al. Annual Report to the Nation
3. Comis RL, Miller JD, Aldige CR, et al. Public attitudes toward partic- on the status of cancer, 1975-2011, featuring incidence of breast
ipation in cancer clinical trials. J Clin Oncol. 2003;21:830-835. cancer subtypes by race/ethnicity, poverty, and state. J Natl Cancer
4. National Cancer Institute. The NCTN Budget. http://www.cancer.gov/ Inst. 2015;107:djv048.
research/areas/clinical-trials/nctn/budget#1. Accessed January 13, 9. Ries LAG, Smith MA, Gurney JG, et al (eds). Cancer incidence and
2016. survival among children and adolescents: United States SEER Program
5. Howlader N, Noone AM, Krapcho M, et al (eds). SEER Cancer Statistics 1975-1995. National Cancer Institute, SEER Program. NIH Pub. No. 99-
Review, 1975-2012, National Cancer Institute. April 2015. http://seer. 4649. Bethesda, MD: National Institutes of Health; 1999.
cancer.gov/csr/1975_2012/. Accessed January 13, 2016. 10. Bleyer A, Budd T, Montello M. Adolescents and young adults with
6. Bond MC, Pritchard S. Understanding clinical trials in childhood cancer: the scope of the problem and criticality of clinical trials.
cancer. Paediatr Child Health. 2006;11:148-150. Cancer. 2006;107:1645-1655.

194 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


CLINICAL TRIAL PARTICIPATION

11. Ferrari A, Bleyer A. Participation of adolescents with cancer in clinical 32. Melisko ME, Hassin F, Metzroth L, et al. Patient and physician attitudes
trials. Cancer Treat Rev. 2007;33:603-608. toward breast cancer clinical trials: developing interventions based on
12. Institute of Medicine. A National Cancer Clinical Trials System for the understanding barriers. Clin Breast Cancer. 2005;6:45-54.
21st Century: Reinvigorating the NCI Cooperative Group Program. 33. Prescott RJ, Counsell CE, Gillespie WJ, et al. Factors that limit the
Washington, DC: The National Academies Press; 2010. quality, number and progress of randomised controlled trials. Health
13. Javid SH, Unger JM, Gralow JR, et al. A prospective analysis of the Technol Assess. 1999;3:1-143.
influence of older age on physician and patient decision-making when 34. Ross S, Grant A, Counsell C, et al. Barriers to participation in rand-
considering enrollment in breast cancer clinical trials (SWOG S0316). omised controlled trials: a systematic review. J Clin Epidemiol. 1999;
Oncologist. 2012;17:1180-1190. 52:1143-1156.
14. Klabunde CN, Springer BC, Butler B, et al. Factors influencing en- 35. Taylor KM, Margolese RG, Soskolne CL. Physicians reasons for not
rollment in clinical trials for cancer treatment. South Med J. 1999;92: entering eligible patients in a randomized clinical trial of surgery for
1189-1193. breast cancer. N Engl J Med. 1984;310:1363-1367.
15. Unger JM, Hershman DL, Albain KS, et al. Patient income level and 36. Institute of Medicine. Transforming Clinical Research in the United
cancer clinical trial participation. J Clin Oncol. 2013;31:536-542. States: Challenges and Opportunities: Workshop Summary. Wash-
16. Rivers D, August EM, Sehovic I, et al. A systematic review of the factors ington, DC: The National Academies Press; 2010.
influencing African Americans participation in cancer clinical trials. 37. Siminoff LA, Zhang A, Colabianchi N, et al. Factors that predict the
Contemp Clin Trials. 2013;35:13-32. referral of breast cancer patients onto clinical trials by their surgeons
17. Roetzheim RG, Pal N, Tennant C, et al. Effects of health insurance and and medical oncologists. J Clin Oncol. 2000;18:1203-1211.
race on early detection of cancer. J Natl Cancer Inst. 1999;91: 38. Ellis PM, Butow PN, Tattersall MH, et al. Randomized clinical trials in
1409-1415. oncology: understanding and attitudes predict willingness to par-
18. Ward E, Halpern M, Schrag N, et al. Association of insurance with ticipate. J Clin Oncol. 2001;19:3554-3561.
cancer care utilization and outcomes. CA Cancer J Clin. 2008;58:9-31. 39. Cassileth BR, Lusk EJ, Miller DS, et al. Attitudes toward clinical trials
19. Robbins AS, Pavluck AL, Fedewa SA, et al. Insurance status, comor- among patients and the public. JAMA. 1982;248:968-970.
bidity level, and survival among colorectal cancer patients age 18 to 64 40. Daugherty C, Ratain MJ, Grochowski E, et al. Perceptions of cancer
years in the National Cancer Data Base from 2003 to 2005. J Clin Oncol. patients and their physicians involved in phase I trials. J Clin Oncol.
2009;27:3627-3633. 1995;13:1062-1072.
20. Lara PN Jr, Higdon R, Lim N, et al. Prospective evaluation of cancer 41. Ford JG, Howerton MW, Lai GY, et al. Barriers to recruiting un-
clinical trial accrual patterns: identifying potential barriers to en- derrepresented populations to cancer clinical trials: a systematic
rollment. J Clin Oncol. 2001;19:1728-1733. review. Cancer. 2008;112:228-242.
21. Begg CB, Zelen M, Carbone PP, et al. Cooperative groups and com- 42. Jones JH. Bad Blood: The Tuskegee Syphilis Experiment. New York: Free
munity hospitals. Measurement of impact in the community hospitals. Press; 1993.
Cancer. 1983;52:1760-1767. 43. Lederer S. Subjected to Science: Human Experimentation in America
22. Hunter CP, Frelick RW, Feldman AR, et al. Selection factors in clinical after the Second World War. Baltimore, MD: Johns Hopkins University
trials: results from the Community Clinical Oncology Program Phy- Press; 1995.
sicians Patient Log. Cancer Treat Rep. 1987;71:559-565. 44. Coyne CA, Xu R, Raich P, et al; Eastern Cooperative Oncology Group.
23. McCusker J, Wax A, Bennett JM. Cancer patient accessions into clinical Randomized, controlled trial of an easy-to-read informed consent
trials: a pilot investigation into some patient and physician de- statement for clinical trial participation: a study of the Eastern Co-
terminants of entry. Am J Clin Oncol. 1982;5:227-236. operative Oncology Group. J Clin Oncol. 2003;21:836-842.
24. Simon MS, Brown DR, Du W, et al. Accrual to breast cancer clinical 45. Jenkins V, Fallowfield L. Reasons for accepting or declining to par-
trials at a university-affiliated hospital in metropolitan Detroit. Am J ticipate in randomized clinical trials for cancer therapy. Br J Cancer.
Clin Oncol. 1999;22:42-46. 2000;82:1783-1788.
25. Green S, Benedetti J, Crowley J. Clinical Trials in Oncology, 2nd Ed. 46. Llewellyn-Thomas HA, McGreal MJ, Thiel EC, et al. Patients willing-
Boca Raton, FL: CRC Press; 2003. ness to enter clinical trials: measuring the association with perceived
26. Newhouse JP, McClellan M. Econometrics in outcomes research: the benefit and preference for decision participation. Soc Sci Med. 1991;
use of instrumental variables. Annu Rev Public Health. 1998;19:17-34. 32:35-42.
27. Langford AT, Resnicow K, Dimond EP, et al. Racial/ethnic differences in 47. McQuellon RP, Muss HB, Hoffman SL, et al. Patient preferences for
clinical trial enrollment, refusal rates, ineligibility, and reasons for treatment of metastatic breast cancer: a study of women with early-
decline among patients at sites in the National Cancer Institutes stage breast cancer. J Clin Oncol. 1995;13:858-868.
Community Cancer Centers Program. Cancer. 2014;120:877-884. 48. Jenkins VA, Fallowfield LJ, Souhami A, et al. How do doctors explain
28. Unger JM, Barlow WE, Martin DP, et al. Comparison of survival randomised clinical trials to their patients? Eur J Cancer. 1999;35:
outcomes among cancer patients treated in and out of clinical trials. 1187-1193.
J Natl Cancer Inst. 2014;106:dju002. 49. Hietanen P, Aro AR, Holli K, et al. Information and communication in
29. Clisant S, Clermont A, Adenis A, et al. Inflation in the number of the context of a clinical trial. Eur J Cancer. 2000;36:2096-2104.
eligibility criteria for industry-sponsored phase II cancer clinical 50. Hutchins LF, Unger JM, Crowley JJ, et al. Underrepresentation of
trial: illustration over a 20-year period. Contemp Clin Trials. 2012; patients 65 years of age or older in cancer-treatment trials. N Engl J
33:459. Med. 1999;341:2061-2067.
30. Somkin CP, Altschuler A, Ackerson L, et al. Organizational barriers to 51. Lewis JH, Kilgore ML, Goldman DP, et al. Participation of patients 65
physician participation in cancer clinical trials. Am J Manag Care. 2005; years of age or older in cancer clinical trials. J Clin Oncol. 2003;21:
11:413-421. 1383-1389.
31. Benson AB III, Pregler JP, Bean JA, et al. Oncologists reluctance to 52. Talarico L, Chen G, Pazdur R. Enrollment of elderly patients in clinical
accrue patients onto clinical trials: an Illinois Cancer Center study. trials for cancer drug registration: a 7-year experience by the US Food
J Clin Oncol. 1991;9:2067-2075. and Drug Administration. J Clin Oncol. 2004;22:4626-4631.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 195


UNGER ET AL

53. Unger JM, Coltman CA Jr, Crowley JJ, et al. Impact of the year 2000 73. U.S. Census Bureau. National Intercensal Estimates. (2000-2010).
Medicare policy change on older patient enrollment to cancer clinical http://www.census.gov/popest/data/intercensal/national/nat2010.
trials. J Clin Oncol. 2006;24:141-144. html. Accessed January 28, 2016.
54. Kemeny MM, Peterson BL, Kornblith AB, et al. Barriers to clinical trial 74. Joinpoint Regression Program. Version 4.1.1. August 2014; Statistical
participation by older women with breast cancer. J Clin Oncol. 2003; Research and Applications Branch, National Cancer Institute. http://
21:2268-2275. surveillance.cancer.gov/joinpoint. Accessed January 15, 2016.
55. Kornblith AB, Kemeny M, Peterson BL, et al; Cancer and Leukemia 75. Surveillance Epidemiology, and End Results (SEER) Program (www.
Group B. Survey of oncologists perceptions of barriers to accrual of seer.cancer.gov) SEER*Stat Database: Incidence - SEER 9 Regs Re-
older patients with breast carcinoma to clinical trials. Cancer. 2002;95: search Data, Nov 2014 Sub (1973-2012) ,Katrina/Rita Population
989-996. Adjustment. - Linked To County Attributes - Total U.S., 1969-2013
56. Townsley CA, Selby R, Siu LL. Systematic review of barriers to the Counties, National Cancer Institute, DCCPS, Surveillance Research
recruitment of older patients with cancer onto clinical trials. J Clin Program, Surveillance Systems Branch, released April 2015, based on
Oncol. 2005;23:3112-3124. the November 2014 submission.
57. Havlik RJ, Yancik R, Long S, et al. The National Institute on Aging and 76. Surveillance Epidemiology, and End Results (SEER) Program (www.
the National Cancer Institute SEER collaborative study on comorbidity seer.cancer.gov) SEER*Stat Database: Incidence - SEER 13 Regs Re-
and early diagnosis of cancer in the elderly. Cancer. 1994;74: search Data, Nov 2014 Sub (1992-2012) ,Katrina/Rita Population
2101-2106. Adjustment. - Linked To County Attributes - Total U.S., 1969-2013
58. Yancik R. Cancer burden in the aged: an epidemiologic and de- Counties, National Cancer Institute, DCCPS, Surveillance Research
mographic overview. Cancer. 1997;80:1273-1283. Program, Surveillance Systems Branch, released April 2015, based on
59. Centers for Medicare & Medicaid Services. Medicare Coverage. Clinical the November 2014 submission.
Trials: Final National Coverage Decision. https://www.cms.gov/Medicare/ 77. Surveillance Epidemiology, and End Results (SEER) Program (www.
Coverage/ClinicalTrialPolicies/downloads/finalnationalcoverage. seer.cancer.gov) SEER*Stat Database: Incidence - SEER 18 Regs Re-
pdf. Accessed January 13, 2016. search Data + Hurricane Katrina Impacted Louisiana Cases, Nov 2014
60. Stewart JH, Bertoni AG, Staten JL, et al. Participation in surgical on- Sub (1973-2012 varying) - Linked To County Attributes - Total U.S.,
cology clinical trials: gender-, race/ethnicity-, and age-based dispar- 1969-2013 Counties, National Cancer Institute, DCCPS, Surveillance
ities. Ann Surg Oncol. 2007;14:3328-3334. Research Program, Surveillance Systems Branch, released April 2015,
61. Gross CP, Filardo G, Mayne ST, et al. The impact of socioeconomic based on the November 2014 submission.
status and race on trial participation for older women with breast 78. Surveillance Epidemiology, and End Results (SEER) Program (www.
cancer. Cancer. 2005;103:483-491. seer.cancer.gov) SEER*Stat Database: Mortality - All COD, Aggregated
62. Cook ED, Moody-Thomas S, Anderson KB, et al. Minority recruitment With State, Total U.S. (1969-2012) ,Katrina/Rita Population
to the Selenium and Vitamin E Cancer Prevention Trial (SELECT). Clin Adjustment., National Cancer Institute, DCCPS, Surveillance Re-
Trials. 2005;2:436-442. search Program, Surveillance Systems Branch, released April 2015.
63. Cook ED, Arnold KB, Hermos JA, et al. Impact of supplemental site Underlying mortality data provided by NCHS (www.cdc.gov/nchs).
grants to increase African American accrual for the Selenium and 79. Breitenbach K, Stock W. Intergroup Trial C10403: a pediatric treat-
Vitamin E Cancer Prevention Trial. Clin Trials. 2010;7:90-99. ment approach to improve outcomes in adolescents and young adults
64. Fouad MN, Corbie-Smith G, Curb D, et al. Special populations re- with acute lymphoblastic leukemia. J Adolesc Young Adult Oncol.
cruitment for the Womens Health Initiative: successes and limita- 2011;1:107-108.
tions. Control Clin Trials. 2004;25:335-352. 80. NCT02043587. Chemotherapy With Liposomal Cytarabine CNS Pro-
65. Moinpour CM, Atkinson JO, Thomas SM, et al. Minority recruitment in phylaxis for Adult Acute Lymphoblastic Leukemia & Lymphoblas-
the prostate cancer prevention trial. Ann Epidemiol. 2000;10:S85-S91. tic Lymphoma. https://clinicaltrials.gov/ct2/show/NCT02043587?
66. Unger JM, Gralow JR, Albain KS, et al. Patient income level and cancer term=acute+lymphoblastic+leukemia+wieduwilt&rank=1. Accessed
clinical trial participation: a prospective survey study. JAMA Oncol. January 28, 2016.
2016;2:137-139. 81. DeAngelo DJ, Stevenson KE, Dahlberg SE, et al. Long-term outcome
67. Goldman DP, Berry SH, McCabe MS, et al. Incremental treatment costs of a pediatric-inspired regimen used for adults aged 18-50 years with
in national cancer institute-sponsored clinical trials. JAMA. 2003;289: newly diagnosed acute lymphoblastic leukemia. Leukemia. 2015;29:
2970-2977. 526-534.
68. Centers for Disease Control and Prevention National Center for Health 82. Bleyer A. How NCCN guidelines can help young adults and older
Statistics. http://www.cdc.gov/nchs. Accessed September 13, 2010. adolescents with cancer and the professionals who care for them.
69. Bleyer WA. Potential favorable impact of the affordable care act of J Natl Compr Canc Netw. 2012;10:1065-1071.
2010 on cancer in young adults in the United States. Cancer J. 2010;16: 83. Dombret H, Cluzeau T, Huguet F, et al. Pediatric-like therapy for adults
563-571. with ALL. Curr Hematol Malig Rep. 2014;9:158-164.
70. Bleyer A, Barr R, Hayes-Lattin B, et al; Biology and Clinical Trials 84. Hallboo k H, Gustafsson G, Smedmyr B, et al; Swedish Adult Acute
Subgroups of the US National Cancer Institute Progress Review Group Lymphocytic Leukemia Group; Swedish Childhood Leukemia Group.
in Adolescent and Young Adult Oncology. The distinctive biology of Treatment outcome in young adults and children .10 years of age
cancer in adolescents and young adults. Nat Rev Cancer. 2008;8: with acute lymphoblastic leukemia in Sweden: a comparison
288-298. between a pediatric protocol and an adult protocol. Cancer. 2006;107:
71. Bleyer WA, Barr R, Keohan ML. Adolescents and young adults with 1551-1561.
cancer: what will it take to improve outcome. Am Soc Clin Oncol Educ 85. Hayakawa F, Sakura T, Yujiri T, et al; Japan Adult Leukemia Study Group
Book. 2001;21:125-140. (JALSG). Markedly improved outcomes and acceptable toxicity in
72. Martin S, Ulrich C, Munsell M, et al. Delays in cancer diagnosis in adolescents and young adults with acute lymphoblastic leukemia
underinsured young adults and older adolescents. Oncologist. 2007; following treatment with a pediatric protocol: a phase II study by the
12:816-824. Japan Adult Leukemia Study Group. Blood Cancer J. 2014;4:e252.

196 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


CLINICAL TRIAL PARTICIPATION

86. Hocking J, Schwarer AP, Gasiorowski R, et al. Excellent outcomes for presented at: National Cancer Institute American Society of Clinical
adolescents and adults with acute lymphoblastic leukemia and Oncology Cancer Trial Accrual Symposium: Science and Solutions;
lymphoma without allogeneic stem cell transplant: the FRALLE-93 April 2010; Bethesda, MD.
pediatric protocol. Leuk Lymphoma. 2014;55:2801-2807. 103. Moore HC, Unger JM, Phillips KA, et al; POEMS/S0230 Investigators.
87. Ram R, Wolach O, Vidal L, et al. Adolescents and young adults with Goserelin for ovarian protection during breast-cancer adjuvant che-
acute lymphoblastic leukemia have a better outcome when treated motherapy. N Engl J Med. 2015;372:923-932.
with pediatric-inspired regimens: systematic review and meta- 104. Frandsen M, Walters J, Ferguson SG. Exploring the viability of using
analysis. Am J Hematol. 2012;87:472-478. online social media advertising as a recruitment method for smoking
88. Ribera JM, Oriol A, Sanz MA, et al. Comparison of the results of the cessation clinical trials. Nicotine Tob Res. 2014;16:247-251.
treatment of adolescents and young adults with standard-risk acute 105. Lamberti MJ, Kush R, Kubick W, et al. An examination of eClinical
lymphoblastic leukemia with the Programa Espan ~ol de Tratamiento en Technology usage and CDISC standard adoption. Ther Innov Regul Sci.
Hematologa pediatric-based protocol ALL-96. J Clin Oncol. 2008;26: 2015:2168479015586003.
1843-1849. 106. Top considerations for using social media in researchit all starts
89. Usvasalo A, Raty R, Knuutila S, et al. Acute lymphoblastic leukemia in with a plan. Seattle, WA; Quorum Review IRB. https://www.ucdmc.
adolescents and young adults in Finland. Haematologica. 2008;93: ucdavis.edu/biorepositories/pdfs/misc-biobank/Top-considerations-
1161-1168. for-using-social-media-in-research.pdf. Accessed February 15, 2016.
90. Stock W, La M, Sanford B, et al; Childrens Cancer Group; Cancer and 107. U.S. Food and Drug Administration. Recruiting Study Subjects -
Leukemia Group B studies. What determines the outcomes for ad- Information Sheet. www.fda.gov/regulatoryinformation/guidances/
olescents and young adults with acute lymphoblastic leukemia ucm126428.htm. Accessed February 15, 2016.
treated on cooperative group protocols? A comparison of Childrens 108. Office of Inspector General. Clinical Trial Web Sites. A promising tool
Cancer Group and Cancer and Leukemia Group B studies. Blood. 2008; to foster informed consent. http://oig.hhs.gov/oei/reports/oei-01-
112:1646-1654. 97-00198.pdf. Accessed February 15, 2016.
91. Douer D. Is asparaginase a critical component in the treatment of 109. InVentiv Health Clinical Division. E-Recruiting: using digital plat-
acute lymphoblastic leukemia? Best Pract Res Clin Haematol. 2008; forms, social, media, and mobile technologies to improve clinical trial
214:647-658. enrollment. http://www.inventivhealth.com/docs/e-Recruiting_
92. Collins CL, Malvar J, Hamilton AS, et al. Case-linked analysis of clinical Using_Digital_Platforms_Social_Media_and_Mobile_Technologies_
trial enrollment among adolescents and young adults at a National to_Improve_Clinical_Trial_Enrollment.pdf. Accessed February 15,
Cancer Institute-Designated comprehensive cancer center. Cancer. 2016.
2015;121:4398-4406. 110. Gossen R. Great Facebook pages for research sites. Engage and recruit
93. Sender L, Zabokrtsky KB. Adolescent and young adult patients with clinical research participants. Paper presented at: 23rd Society of
cancer: a milieu of unique features. Nat Rev Clin Oncol. 2015;12: Clinical Research Associates Annual Meeting; September 2014;
465-480. Orlando, FL.
94. Freyer DR, Felgenhauer J, Perentesis J; COG Adolescent and Young 111. American Cancer Society. Cancer Treatment and Survivorship Facts &
Adult Oncology Discipline Committee. Childrens Oncology Groups Figures 2014-2015. Atlanta: American Cancer Society; 2014.
2013 blueprint for research: adolescent and young adult oncology. 112. Denson AC, Mahipal A. Participation of the elderly population in
Pediatr Blood Cancer. 2013;60:1055-1058. clinical trials: barriers and solutions. Cancer Contr. 2014;21:209-214.
95. Weiss AR, Nichols CR, Freyer DR. Enhancing adolescent and young 113. Chen H, Cantor A, Meyer J, et al. Can older cancer patients tolerate
adult oncology research within the National Clinical Trials Network: chemotherapy? A prospective pilot study. Cancer. 2003;97:
Rationale, progress, and emerging strategies. Semin Oncol. 2015;42: 1107-1114.
740-747. 114. Mariano C, Francl M, Pope J, et al. Comparison of toxicity experienced
96. Stark D, Bowen D, Dunwoodie E, et al. Survival patterns in teenagers by older versus younger patients enrolled in breast cancer clinical
and young adults with cancer in the United Kingdom: Comparisons trials. Clin Breast Cancer. 2015;15:73-79.
with younger and older age groups. Eur J Cancer. 2015;51:2643-2654. 115. Sud S, Lai P, Zhang T, et al. Chemotherapy in the oldest old: the
97. Denicoff AM, McCaskill-Stevens W, Grubbs SS, et al. The National feasibility of delivering cytotoxic therapy to patients 80 years old and
Cancer Institute-American Society of Clinical Oncology Cancer Trial older. J Geriatr Oncol. 2015;6:395-400.
Accrual Symposium: summary and recommendations. J Oncol Pract. 116. Extermann M, Aapro M, Bernabei R, et al; Task Force on CGA of the
2013;9:267-276. International Society of Geriatric Oncology. Use of comprehensive
98. AccrualNet. Strategies, tools, and resources to support accrual to geriatric assessment in older cancer patients: recommendations from
clinical trials. https://accrualnet.cancer.gov/. Accessed January 28, the task force on CGA of the International Society of Geriatric On-
2016. cology (SIOG). Crit Rev Oncol Hematol. 2005;55:241-252.
99. Barrios CH, Werutsky G, Martinez-Mesa J. The global conduct of 117. Extermann M, Hurria A. Comprehensive geriatric assessment for older
cancer clinical trials: challenges and opportunities. Am Soc Clin Oncol patients with cancer. J Clin Oncol. 2007;25:1824-1831.
Educ Book. 2015:e132-e139. 118. Hamaker ME, Jonker JM, de Rooij SE, et al. Frailty screening methods
100. Tierney G, Sieher S. Key strategies for effective globalization of clin- for predicting outcome of a comprehensive geriatric assessment in
ical trials. Pharmaceutical Outsourcing. http://www.pharmoutsourcing. elderly patients with cancer: a systematic review. Lancet Oncol. 2012;
com/Featured-Articles/150621-Key-Strategies-for-Effective-Globalization- 13:e437-e444.
of-Clinical-Trials/. Accessed September 25, 2013. 119. Kellen E, Bulens P, Deckx L, et al. Identifying an accurate pre-screening
101. Butts CA, Socinski MA, Mitchell P, et al. START: a phase III study of tool in geriatric oncology. Crit Rev Oncol Hematol. 2010;75:243-248.
L-BLP25 cancer immunotherapy for unresectable stage III non-small 120. Luciani A, Ascione G, Bertuzzi C, et al. Detecting disabilities in older
cell lung cancer. J Clin Oncol. 2013;31:15s (suppl; abstr 7500). patients with cancer: comparison between comprehensive geriatric
102. Gaumond B, Ferrande L, Houde J, et al. Global patient recruitment assessment and vulnerable elders survey-13. J Clin Oncol. 2010;28:
continuum for a phase III non-small cell lung cancer clinical trial. Paper 2046-2050.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 197


UNGER ET AL

121. Balducci L, Colloca G, Cesari M, et al. Assessment and treatment of 125. U.S. Department of Health and Human Services. Federal Policy for the
elderly patients with cancer. Surg Oncol. 2010;19:117-123. Protection of Human Subjects (Common Rule). http://www.hhs.gov/
122. Grady C. Payment of clinical research subjects. J Clin Invest. 2005;115: ohrp/humansubjects/commonrule/. Accessed February 15, 2016.
1681-1687. 126. Unger JM, Barlow WE, Ramsey SD, et al. The scientific impact of
123. Bernstein M. Payment of research subjects involved in clinical trials is positive and negative phase 3 cancer clinical trials. JAMA Oncol. Epub
unethical. J Neurooncol. 2003;63:223-224. 2016 Mar 10.
124. Grady C, Dickert N, Jawetz T, et al. An analysis of U.S. practices of 127. Bleyer A. In and out, good and bad news, of generalizability of SWOG
paying research participants. Contemp Clin Trials. 2005;26:365-375. treatment trial results. J Natl Cancer Inst. 2014;106:dju027.

198 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


HEALTH SERVICES RESEARCH AND QUALITY
OF CARE

Using Social Media, Wearables,


and Electronic Medical Records to
Improve Quality of Cancer Care

CHAIR
Michael J. Fisch, MD, MPH
The University of Texas MD Anderson Cancer Center
Houston, TX

SPEAKERS
Arlene E. Chung, MD, MHA, MMCi
University of North Carolina at Chapel Hill School of Medicine
Durham, NC

Melissa K. Accordino, MD
New York-Presbyterian Hospital
New York, NY
FISCH, CHUNG, AND ACCORDINO

Using Technology to Improve Cancer Care: Social Media,


Wearables, and Electronic Health Records
Michael J. Fisch, MD, MPH, Arlene E. Chung, MD, MHA, MMCi, and Melissa K. Accordino, MD

OVERVIEW

Digital engagement has become pervasive in the delivery of cancer care. Internet- and cellular phonebased tools and
systems are allowing large groups of people to engage with each other and share information. Health systems and individual
health professionals are adapting to this revolution in consumer and patient behavior by developing ways to incorporate the
benefits of technology for the purpose of improving the quality of medical care. One example is the use of social media
platforms by oncologists to foster interaction with each other and to participate with the lay public in dialogue about science,
medicine, and cancer care. In addition, consumer devices and sensors (wearables) have provided a new, growing dimension
of digital engagement and another layer of patient-generated health data to foster better care and research. Finally,
electronic health records have become the new standard for oncology care delivery, bringing new opportunities to measure
quality in real time and follow practice patterns, as well as new challenges as providers and patients seek ways to integrate
this technology along with other forms of digital engagement to produce more satisfaction in the process of care along with
measurably better outcomes.

H igh-quality medical care is expected to be efficacious,


safe, timely, patient centered, efficient, and equitable.1
None of these attributes of high-quality medical care are
published literature and prioritize what to read continues
to evolve. Traditional techniques involved subscribing to
key journals, browsing other journals in shared collections
possible in a vacuum. Physicians must be engaged with each (libraries, offices), gathering in journal clubs, discussing
other to share ideas, collaborate, and establish best practice specific articles at morning reports or interdisciplinary
norms and creative new solutions. Social media has rapidly rounds, and attending local and national meetings where
become one of the normative approaches to foster these new data are presented and reference literature is cited.
critical connections. Compared with traditional media, social We occasionally visit online textbooks or information
media is updated more frequently and it is more interactive, resources and we also encounter lay press stories citing
while also being archivable and searchable.2 Three major medical articles. In todays world, as noted by Dr. Bryan
needs for connection that social media helps oncologists Vartebedian (@Doctor_V), weve reached a point where
meet are (1) finding and prioritizing medical literature and social media is now part of the professional workflow.3
medical meetings information; (2) finding the current lo- Oncology clinicians are finding both efficiency and en-
cation, interests, and qualifications of other physicians; and joyment from using social media platforms such as Twitter
(3) crowdsourcing or surveying each other about best to keep up with the medical literature and share in-
practices or approaches to clinical scenarios. formation from meetings. On Twitter, one can follow
many key journals (and can even follow collections of
FINDING AND PRIORITIZING MEDICAL journals on a list) and quickly scan through articles as they
LITERATURE come out and are tweeted by the journals. More im-
The medical literature is central to ongoing medical ed- portantly, when oncologists follow each other on Twitter
ucation. Early findings of new research as well as expert (and collections of colleagues on lists), there is a sort of
summaries of existing literature are shared at medical Bayesian quality to social media whereby the prior
meetings, and this work is more formally disseminated in probability of seeing an article or hearing about a new
medical journal articles. How oncologists and other health finding at a meeting4 is enhanced by retweets that amplify
professionals identify new information from meetings and the most important articles.

From AIM Specialty Health, Chicago, IL; Outcomes Research Program, Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC;
Department of Medicine, Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Michael J. Fisch, MD, MPH, AIM Specialty Health, 8600 West Bryn Mawr Ave., Suite 800, Chicago, IL 60631; email: fischm@aimspecialtyhealth.com.

2016 by American Society of Clinical Oncology.

200 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


USE OF SOCIAL MEDIA IN ONCOLOGY PRACTICE

FINDING AND CONNECTING TO OTHER directly through Doximity and commented on and/or
PHYSICIANS liked. The latter allows others to see what articles are
In addition to curating the medical literature, Twitter and most endorsed. The most powerful feature of Doximity is
other social media platforms (such as Facebook) also pro- that it allows clinicians to find each other by searching the
vide a venue for connection. For example, if a physician posts Doximity search engine (even when the physician you are
or tweets the link to an article, other colleagues can reply searching for has not engaged with Doximity). Try it: search
with their own comments. Patient and layperson perspec- for yourself on this site. You will find that Doximity identifies
tives come forth frequently because others besides oncol- not only your location but also your medical school, resi-
ogists can see these tweets and engage in dialogue. This dency and fellowship (along with listing your colleagues who
element helps health professionals maintain a broader and trained with you), certifications, licenses, and insurance that
more patient-centered perspective on the literature. An ex- your practice accepts. Individuals can also list their publi-
ample of an exchange between oncologists and a cancer cations and other aspects about themselves by expanding
survivor is shown in Fig. 1. the profile. Physicians could, for example, search for col-
Twitter does not lend itself to in-depth analysis or detailed leagues in their residency and find out where they now
commentary, as the comments are limited to 140 characters. practice and their current specialty. In addition, Medstro has
Oncology professionals also use Twitter to connect to blogs emerged as another popular social network for physicians.
with extended commentary on the literature and other key Medstro features discussion groups with a tight connection
issues in oncology care. These blogs can be individually to the New England Journal of Medicine, as well as contests
managed (such as 33charts by @Doctor_V) or they can be and interactive challenges featuring engagement with ac-
guest blogs distributed by a physicians institution or pro- ademic subspecialty experts. Job searching is also integral to
fessional organization (such as the American Society of both Doximity and Medstro.
Clinical Oncologys ASCO Connection) or through another
physicians popular blog (such as KevinMD). Another
professional-oriented site is ResearchGate. This site, which is CROWDSOURCING AND SURVEYING OTHER
geared toward academic researchoriented professionals, PHYSICIANS
allows oncology professionals to connect directly with in- Social media provides a convenient mechanism for checking
vestigators to ask questions about their articles, to follow in with other physicians, even when those physicians are
their most recent publications, and to even request reprints outside of the physicians local peer group or organization.
from individuals. For example, if a clinician wonders Is there a valid in-
Doximity, a unique platform restricted to health pro- strument that can be used to measure fear of cancer re-
fessionals with a National Provider Identifier number, is a currence?, the question could be crowdsourced on Twitter
more restricted setting for conversation and, this is more by simply typing the question and pressing send. This
comfortable for some clinicians. Articles can be found would be distributed to the clinicians followers. One could
also distribute the question outside of ones followers to
other groups of oncology professionals using hashtags. For
example, adding #pallonc at the end of the question would
KEY POINTS distribute the tweet not only to an individuals followers but
also to anybody else worldwide who is tuned in to this
Social media is increasingly used by oncology clinicians topic by searching or streaming tweets with this hashtag.
to find and prioritize medical literature, connect with
Dr. Matthew Katz (@subatomicdoc), a radiation oncologist,
other providers, and interact with the public about
cancer care.
has led the oncology community by developing Cancer Tag
Consumer devices and sensors (called wearables) are Ontology and Oncology Tag Ontology (via symplur.com) to
growing in popularity and allow for patient monitoring codify this way of indexing tweets.5
in their own environments outside of clinical Another sort of question might be a clinical case scenario,
encounters. such as, Is there a role for post-treatment PET/CT following
Patient-generated health data from wearables provide chemoRT for stage III NSCLC? This type of question could be
an opportunity to enhance comparative effectiveness directed to Twitter also, and those replying are likely to give a
research in oncology. short answer and include a link to a relevant article or web
Electronic health records are widespread in oncology resource. However, there is a forum for oncologists available
and are being used to measure adherence to quality for addressing clinical scenarios via theMednet.org. This new
metrics and often to connect with patients through
forum is facilitated by Dr. Nadine Housri (@nadinehousri),
secure portals.
Electronic health records have the potential to improve
who is also a radiation oncologist. It is free and, like Doximity,
the quality of cancer care by enhancing education and restricted to health professionals. It is not a place for dis-
awareness of evidence-based guidelines and by serving cussion of specific patients, but it is useful for general case
as a resource for quality assessment and as a tool for scenarios, and the forum is well facilitated and allows for
clinical and population-based research social interactions between various disciplines and be-
tween community and academic practitioners. Another

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 201


FISCH, CHUNG, AND ACCORDINO

FIGURE 1. Example of an Oncology-Related Exchange on Twitter

This example of an exchange on Twitter begins with a tweet by an oncology fellow in Boston sharing an ASCO Post article describing a peer-reviewed manuscript about the
association between symptoms and employment outcomes in cancer survivors. Hashtags and specific Twitter handles are used to help disseminate the tweet. A picture of the data is
also included by adding a tiny URL link to the picture file. A cancer survivor replies to the tweet, raising the issue of fear of recurrence. Another oncologist replies to the survivor,
referencing a specific article about key associations related to fear of recurrence.

kind of question that lends itself to social interaction organizations and by individual oncologists as an edu-
between physicians is How many oncologists are us- cational tool and a practical tool for clinical research
ing the XYZ regimen in first-line treatment of multiple planning.
myeloma? Dr. Simrit Parmar (@spa718), an academic
hematologist/oncologist, has developed a platform called CONNECTING WITH PATIENTS
MDRing that works via the web or smartphone (as an Physicians do not need social media to connect with their
iPhone application) to bring cancer professionals to- own patients. Connecting with our patients occurs through
gether to answer survey-type questions like this. MDRing direct interactions and also through secure interactions
has been used by clinical cooperative groups and other available through electronic health record (EHR) systems or

202 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


USE OF SOCIAL MEDIA IN ONCOLOGY PRACTICE

other platforms approved by our clinical organizations. It is WEARABLE TECHNOLOGY


well recognized that use of social media can bring risks to In addition to connecting through words and media, another
individual health professionals and their organizations. dimension of digital engagement comes from the growing
Physicians must be aware of unintentional online disclosure availability and affordability of various consumer devices
of patient information, because even subtle oversteps of and sensors that can be worn or placed on the body. Known
boundaries raise concerns for professional reputation and as wearables, these devices provide an unparalleled op-
responsibility.6 Nevertheless, there is nothing that prevents portunity to monitor a persons health and experience in his
our patients from engaging us through publicly available or her real-world environment. The current model of health
social media sites such as Facebook and Twitter. Fear of this care for treatment or prevention is still mainly focused on
sort of contact is one reason that some clinicians are re- face-to-face encounters with providers in the clinical setting;
luctant to use social media. Dr. Don Dizon and a working however, as we move away from this traditional model of
group of the ASCO Integrated Media and Technology care, wearables could provide additional insights about the
Committee noted that oncologists must not confuse the role patient experience and enhance the quality of cancer care.
of provider of information and provider of care, must un- Wearable technology refers collectively to electronics that
derstand regulations related to state licensure and medical can be worn on or close to the body. Wearables encompass a
records, must recognize the importance of transparency and growing number of different types of items such as wrist-
disclosure of potential conflicts of interest, and must avoid worn sensors that detect and measure physical activity and
disclosures of preliminary results or nonpublic information.7 intensity (such as a Fitbit device or Apple Watch), rings and
Guidance is also commonly included in the social media glasses with sensors, earbuds, and textiles or fabrics with
policies of health care organizations and clinical research embedded sensors to monitor heart rate and temperature,
organizations. to more invasive versions such as an implantable microchip
With boundaries understood, it is appropriate for cli- or smart tattoo. It is estimated that there will be over 111
nicians to interact with patients and the lay public in the million worldwide shipments of wearables in 2016, with
broader sense. That is, individual cancer survivors and most being in the form of watches and wristbands (92%).9
their family members can be followed on social media, Users are almost equally split between males and females
and they are a key component of the healthy exchange of and are mostly younger than age 55,9 although adoption
ideas and perspectives about illness, science, and health among older adults is expected to be the sector of largest
care. Oncologists can use this opportunity to help educate growth. It is also anticipated that the wearables market will
the public, to advocate for science and for clinical trial grow over 64% to $25 billion in 2019, with over 245 million
participation, and to promote good health behaviors and devices sold.10
principles of sound medical care. Social media can also be
used by oncologists to demonstrate their humanity,
compassion, and deep dedication to the service of others. HEALTH MEASUREMENT OPPORTUNITIES
Some oncologists use Facebook for this purpose. For THROUGH GROWING ADOPTION OF
example, Dr. Anas Younes (@DrAnasYounes), an academic WEARABLES
lymphoma specialist, has shared information about Patient-generated health data (PGHD) from wearables, such
lymphoma, cancer, and other health care topics using this as biometric data like heart rate and blood pressure readings
platform and built a following represented by greater than or physical activity data, are some of the more commonly
1,500 likes on his Facebook page. Moreover, organi- captured data from wearables. PGHD are defined as health-
zations such as ASCO and specific health care organiza- related dataincluding health history, symptoms, biometric
tions widely use public Facebook pages. The Mayo Clinic is data, etc.created, recorded, gathered, or inferred by or
one example of a health care organization that has fully from patients/caregivers to help address a health con-
embraced social media, adopting a patient-centered ap- cern.11 PGHD from wearables could provide a more holistic
proach of reaching out to innovate in this evolving space view of the health and status of a patient.
where patients are seeking information. The Mayo Clinic As consumer adoption of wearables increases and there
Center for Social Media (#mccsm) has greater than 1.25 are advances in the underlying technologies, the opportu-
million followers on Twitter and greater than 750,000 nity to collect data on a population level is more feasible and
likes on its public Facebook page. Of note, Facebook use could provide important advances in oncology comparative
is most effective when the user is able to provide regular effectiveness research. Recently, there have also been
input, whereas Twitter is easier to use sporadically. Other several key funding initiatives that aim to incorporate
social media platforms such as Instagram, Pinterest, and wearable device data. For example, the National Institutes of
even Snapchat are used less often for sharing information Health Precision Medicine Initiative Cohort Program will
about cancer, but there is certainly opportunity to har- capture physiologic and environmental exposures. More-
ness these popular sites for the same purposes. As noted by over, several of the Patient-Centered Outcomes Research
Dr. Michael Thompson (@mtmdphd), social media platforms Institutefunded Patient-Powered Research Networks col-
will always evolve, but it is the content, curation, and lect PGHD from wearables in an effort to collect data
connectivity that matter.8 in a nationally representative sample at lower costs than

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 203


FISCH, CHUNG, AND ACCORDINO

traditional methods of data collection. Dietary data and for wearables in cancer care are likely to emerge where
physical activity data are examples of data that are tradi- the wearable provides utility and value.
tionally difficult to capture on a continuous basis in research
studies or for clinical care. Capturing data such as these CHALLENGES IN THE USE OF WEARABLES
could be used to better understand the influence of these As is the case with other emerging technologies, the op-
factors on cancer risk and progression among populations of portunity to monitor patients more continuously with
patients. wearables comes with its own unique challenges. Because
Physical activity, in particular, is an important factor in the use of wearables in health care is still nascent, it is not
cancer survivorship, both for prevention and quality of life clear what types of data from wearables will be the most
and symptom management, and may also improve a effective to improve symptoms, the patient experience, and
patients ability to tolerate cancer treatment by improving short- and long-term health outcomes. Best practices for
fatigue, quality of life, and physical functioning.12-14 integrating this PGHD into clinical workflows to ensure
Pedometry using commercially available fitness trackers appropriate review and action are not yet available, but as
such as Fitbits has shown that large decreases in daily practices and health care systems begin to have more ex-
steps may correlate with more severe symptoms, im- perience with PGHD, there will be better guidance on how to
paired physical health, and restricted daily activities.15 engage patients in a learning health ecosystem and what will
Thus, tracking physical activity using a wearable device be acceptable within provider workflows. In addition, in the
could potentially be used for monitoring symptoms and next 2 years, the Office of the National Coordinator for
severity in those who may not be able to self-report Health Information Technology will be developing a policy
symptoms because of various barriers such as literacy framework for identifying best practices, gaps, and oppor-
or severity of illness. The use of a wearable physical ac- tunities for the use of PGHD, which will provide further
tivity tracker could also potentially monitor levels of guidance.
activity and provide a target for behavioral interventions Among the largest vendors of EHRs, most allow patients to
during treatment and survivorship using just-in-time submit wearables data in various formats for provider re-
adaptive methodologies that would not have been pos- view. An example of a wearables workflow within clinical
sible without the real-time continuous activity tracking care might include the following: (1) a patient receives a
afforded by wearables. One might imagine that a patient message from his or her provider in the patients portal
with breast cancer undergoing treatment could wear a account, which provides instructions on how to connect the
physical activity tracker and that the PGHD captured wearable device account to allow for transmission of these
showed lower than usual levels of activity for this patient. data to the health care system via the patient portal; (2) the
Decreased physical activity could trigger a cascade of patient uses the wearable device for a certain interval to
activities such as offering a patient-reported outcome capture data; and (3) the PGHD are transmitted either in real
questionnaire to determine whether the patient has time or asynchronously and made available for providers to
worsening symptoms. If there was no detection of review within EHR workflows. Currently, data visualization
worsening of symptoms, the patient could be encouraged and presentation is generally constrained by the EHR ven-
to be more active, which could be personalized based on dors specifications and is presented mostly in tabular for-
prior activity levels and other factors such as whether the mat. These data are usually presented at the patient level
patient has anemia. The patient could receive a person- and not at a population level. Wearables could generate
alized and tailored text message to her wrist-worn large volumes of asynchronous or continuous streams of
wearable about the benefits of activity during treat- data, which would be very challenging to manage and review
ment and could be given specific goals for increasing without intelligent filtering and summarization for both
activity safely. This PGHD could also be shared with her patients and providers.16 Therefore, informatics solutions to
oncologist and primary care physicians, such that the care assist with transforming these data into actionable in-
team was aware of the patients health status and ex- formation will be imperative.
periences, which could inform treatment decisions or Current adoption of wearables is low and rarely sustained
changes in management. Wearables could also be in- beyond several months. However, this is felt to be attrib-
tegrated with other smart home monitors and other utable to cost, the lack of integration with other devices, and
sensing systems, such as smart bottles that could detect the absence of interactive and dynamic feedback. Security
when a user has not opened a pill bottle and send alerts to and privacy concerns are also a barrier to adoption. Overall,
patients or caregivers, or these devices may be able to the tangible benefits of wearables currently lag behind rapid
detect a fall or impaired gait such that assistance could growth of the device market, so it is expected that adoption
be provided to prevent complications. Another scenario will ramp up as software platforms provide better person-
in which wearables also have promise is for remote alization and insights, which could also better sustain patient
monitoring for cardiovascular complications such as engagement. In part, the lack of accurate, validated bio-
arrhythmias during treatment with cardiotoxic chemo- metric data from the majority of consumer wearables is
therapeutics and in survivorship care. As digital patient also a barrier to adoption within health care. In the present
engagement advances and matures, further applications state, it is difficult to decipher which wearable devices and

204 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


USE OF SOCIAL MEDIA IN ONCOLOGY PRACTICE

what types of PGHD will provide the most value within also have the ability to share secured information with
cancer care. Current studies in progress piloting the use of patients directly through portals, which may improve pa-
PGHD will be important in determining what may be the tient satisfaction through increased data sharing.25,26 Lim-
most effective types of wearables or PGHD that could im- itations of EHRs include negative effects in provider-patient
prove the patient experience during cancer treatment and in communication, such as reduced eye contact, higher op-
survivorship. erating costs, unreliability of electronic systems, and work-
flow limitations.27,28
There are notable racial and age-related disparities in
FUTURE DIRECTIONS WITH WEARABLES
the delivery of cancer care.29-31 An evaluation of ad-
As the wearables market matures, we will likely see a
ministrative data showed a 66% increase in adoption of
movement away from primarily wrist-worn devices to body-
basic EHRs in federally qualified health centers from 2010
conforming sensors embedded in flexible patches or stickers
to 2012 (from 29.8% to 49.6%). However, disparities
and toward medical-grade sensors that provide accurate and
existed; centers with higher proportions of low-income
valid data. In addition, we should see improvements in
patients had lower EHR adoption rates compared with
physical comfort, devices being more unobtrusive, battery
centers with smaller proportions of low-income patients
life, and interoperability with other devices and software
(45.1% vs. 55.5%, p , .05).32 These results were similar
applications. We will also see more of a connected eco-
to another population-based study that showed EHR
system of appliances, home-based sensors, and wearables
adoption to be lowest in areas with high concentrations of
such that richer information about an individuals activities
minority and low-income populations.33 In regard to EHR
of daily living alongside biometric data could be captured.
portal access, disparities also exist. A review of four
These data could provide insights about various facets of a
university nephrology practices showed that older age
patients health and environment, which, if collected on a
(odds ratio [OR] 0.29), black race (OR 0.50), and having
population level, could provide important insights about the
Medicaid insurance compared with private insurance
influence of behavioral, lifestyle, and environmental factors
(OR 0.53) were associated with decreased portal enroll-
on cancer risk and progression.
ment.34 Another cross-sectional evaluation of portal uti-
lization revealed that older and nonwhite patients were
ELECTRONIC HEALTH RECORDS IN CANCER CARE less likely to enroll; after enrolling, younger patients and
DELIVERY men were less likely to solicit advice or medication refills
Social media and wearables reflect interactions that occur compared with older patients and women.35 Potential
largely outside of the health care environment. In the very barriers to portal enrollment include lack of resources,
fabric of health care delivery is the need to document health patient comfort and proficiency, lack of provider enthu-
status and elements of care. In recent years, the practice of siasm and communication, health literacy, and distrust in
medical oncology has become increasingly complicated. As the health system.32,34,36-43 Interventions to reduce dis-
reimbursement models transition from fee-for-service to parities in enrollment must address both access and at-
newer shared-risk models, a higher emphasis is placed on titudinal barriers.
the delivery of high-quality and high-value care. EHR use
reflects the new digital landscape that extends far beyond THE EFFECT OF ELECTRONIC HEALTH RECORDS
mere documentation, as the EHR brings features that have ON QUALITY OF CARE
been shown to improve quality and efficiency of care.17 As EHRs have the ability to improve the quality of cancer care
such, practices have been incentivized to use an EHR be- in a multitude of ways, including educational alerts to im-
cause it is seen as a generalizable intervention to expand the prove adherence to evidence-based guidelines, improved
use of technology to improve the quality of our care.18 patient communication through patient portals, as a re-
Within oncology, EHR use can improve quality in real time source for individual and practice-based quality self-
and can be used as a tool to measure adherence to quality assessment, and as a tool for cancer care delivery research.
metrics within individual practices and help identify care Educational electronic alerts have been successful
disparities. at improving quality measures in a number of clinical
An EHR, as defined by the Institute of Medicine, is an settings.44-48 Kucher et al found that an EHR alert that
electronic system with the ability to collect and store patient identified patients at risk for deep-vein thrombosis (DVT)
data, supply information, permit providers to enter orders increased use of appropriate DVT prophylaxis and led to
(known as computerized provider-order entry), and provide reduced rates of venous thromboembolism.46 Another study
decision support. In addition, EHRs may be able to provide alerted providers to a patients risk of contrast-induced
electronic communication, administrative support, patient acute kidney injury (AKI) when imaging studies with con-
support, and population health management reporting.19 trast were ordered. After the alert was implemented, rates
Benefits of EHRs include improved quality of care by in- of prophylaxis increased and incidence of contrast-induced
creased provider adherence to evidence guidelines, re- AKI decreased.47 Selvan et al investigated an electronic alert
duction in medical errors by improvement in accuracy and triggered by erythropoietin-stimulating agent (ESA) order
clarity of data, and the potential for cost reduction.20-24 EHRs entry if a patients hemoglobin was 12 g/dL or above

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 205


FISCH, CHUNG, AND ACCORDINO

(considered inappropriate use) at a single institution. antineoplastic and supportive medications, and be as-
Postalert rates of inappropriate ordering of ESAs decreased sociated with better quality of care.
by 80%.48 An EHR alert triggered by a patient-reported To provide high-quality care, it is important to system-
pain score of 4 or greater led to increased documentation atically assess our own practice to identify areas for im-
of pain care plans for patients with cancer with moderate provement as well as disparities in care. The 1999 Institute of
to severe pain by 20%.49 Another EHR intervention, which Medicine report Ensuring Quality of Cancer Care called for a
has the ability to provide real-time clinical decision sup- system to measure and monitor quality of care and to reduce
port through a combination of alerts and links to clinical disparities in cancer care.73 In response to this report, ASCO
guidelines, is ASCO CancerLinQ, a rapid learning system.50-52 developed the Quality Oncology Practice Initiative (QOPI) to
A rapid learning system refers to a technology platform measure quality at the individual and practice levels by
that allows data to be collected from different EHR systems medical record abstraction and facilitate continuous im-
to be collected, aggregated, analyzed, and translated into provement.74 To globally address quality improvement
information aimed to improve treatment and drive scientific throughout the physician workforce, the Accreditation
learning and discovery from every patient encounter.50,53,54 Council for Graduate Medical Education has revised its
As health care delivery continues to shift toward system- training requirements and implemented the Clinical
based practice, knowledge of detailed evidence-based Learning Environmental Review (CLER) program designed to
guidelines does not solely fall on individual providers but improve education in patient safety and health care qual-
rather the system as a whole, and EHRs have the capability of ity.75 As part of the CLER program, residents and fellows are
providing this type of decisional support to improve quality expected to receive specialty-specific data on their adher-
of care. ence to quality metrics and benchmarks within their patient
Another important quality measure in oncology is ensuring populations, to identify and reduce health care disparities
that patients receive appropriate treatment of their indi- within their practice, and to foster habits of continuous
vidual cancer.50 Because more cancer therapies are now practice improvement.75 Through ASCOs CancerLinQ proj-
delivered orally, a focus on medication adherence is be- ect, data from the EHR can be pulled to measure QOPI
coming increasingly important. Medication nonadherence is compliance in real time.35 An EHR is a robust tool that can
common among patients with chronic conditions, in whom pull individual and practice-specific data in real time to allow
adherence rates are estimated to be around 50%.55,56 In providers the opportunity to measure their own perfor-
women with early-stage hormone receptorpositive breast mances and compare themselves with their peers, and thus
cancer, noninitiation, early discontinuation, and non- foster practice improvements on the individual, practice,
adherence to hormonal therapy are common and are as- and, potentially, global levels.
sociated with poorer survival.57-63 Approximately one-third In addition to continuous improvement, EHRs can affect
of patients with chronic myelogenous leukemia are non- cancer care delivery on a larger scale as a tool for cancer care
adherent to their tyrosine kinase inhibitors during treat- delivery research. An upcoming pragmatic trial to be con-
ment, and this is associated with poorer outcomes, including ducted through SWOG will use the EHR to assess the efficacy
treatment resistance.64,65 Barriers to nonadherence are of prophylactic colony-stimulating factors during the first
multifactorial but include poor provider-patient communi- cycle of intermediate febrile neutropenia risk chemotherapy
cation, side effects, costs of and access to medications, and (10%20%). Sites will be randomized to a standing-order
patient behaviors and education.56,66-68 Through patient entry system for prophylactic colony-stimulating factors via
portals, EHRs have the ability to help foster secure com- manipulation of the individual site EHR or usual care. In
munication and thus bridge the gap between patients and addition to evaluating rates and resource utilization related
providers and allow improved communication between all to febrile neutropenia, this study will evaluate whether a
members of the health care team, including staff who can standing-order entry system helps improve adherence to
answer questions, provide symptom management, and clinical practice guidelines.76 Other areas for future cancer
assist with prior authorizations to help offset high out-of- care delivery research include improvement of content,
pocket costs. A recent randomized trial demonstrated that a dissemination, and implementation of cancer survivorship
web-based platform of symptom self-reporting between care plans; EHR triggers aimed at improving adherence to
patients with cancer who are receiving chemotherapy and medication at both the patient and physician levels; and EHR
providers was associated with improved quality of life, fewer educational alerts to help providers adhere to best practice
hospitalizations, and a longer duration of palliative che- guidelines.77
motherapy.69 In addition, single-institution studies have Many opportunities exist to improve quality of care
shown that access to medical records and the ability to among patients with cancer, especially in improving treat-
request medication refills (via patient portals) were both ment adherence and reducing disparities. The EHR is a
associated with increased adherence to medications and valuable tool to aid in this regard, both in daily clinical
medical advice.70-72 Furthermore, patient portals may be a practice and in cancer care delivery research. However,
valuable resource to provide patient education and even much like social media and wearables, interventions to re-
individualized medication plans. Improved communica- duce disparities of adoption and patient participation are
tion through the EHR may improve adherence to both needed.

206 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


USE OF SOCIAL MEDIA IN ONCOLOGY PRACTICE

References

1. U.S. Institute of Medicine Committee on Quality of Health Care in 20. Chaudhry B, Wang J, Wu S, et al. Systematic review: impact of health
America. Crossing the Quality Chasm: A New Health System for the 21st information technology on quality, efficiency, and costs of medical care.
Century. Washington, DC: National Academy Press; 2001. Ann Intern Med. 2006;144:742-752.
2. Thompson MA, Majhail NS, Wood WA, et al. Social media and the 21. Amarasingham R, Plantinga L, Diener-West M, et al. Clinical information
practicing hematologist: Twitter 101 for the busy healthcare provider. technologies and inpatient outcomes: a multiple hospital study. Arch
Curr Hematol Malig Rep. 2015;10:405-412. Intern Med. 2009;169:108-114.
3. Vartebedian B. Social Media Has Been Introduced to Physicians. 22. Porterfield A, Engelbert K, Coustasse A. Electronic prescribing: im-
33charts, January 25, 2015. http://33charts.com/2015/01/social- proving the efficiency and accuracy of prescribing in the ambulatory
media-introduced-to-physicians.html. care setting. Perspect Health Inf Manag. 2014;11:1g.
4. Fisch M. Putting Twitter to Use Among Oncologists: Shared Note 23. Bubalo J, Warden BA, Wiegel JJ, et al. Does applying technology
Taking at National Meetings and Other Stuff. ASCO Connection, throughout the medication use process improve patient safety with
December 5, 2012. https://connection.asco.org/commentary/putting- antineoplastics? J Oncol Pharm Pract. 2014;20:445-460.
twitter-use-among-oncologists-shared-note-taking-national-meetings- 24. Meisenberg BR, Wright RR, Brady-Copertino CJ. Reduction in chemo-
and-other. therapy order errors with computerized physician order entry. J Oncol
5. Katz MS, Utengen A, Anderson PF, et al. Disease-specific hashtags for Pract. 2014;10:e5-e9.
online communication about cancer care. J Clin Oncol. 2015;33 (suppl; 25. McNamara M, Arnold C, Sarma K, et al. Patient portal preferences:
abstr 6520). perspectives on imaging information. J Assoc Inf Sci Technol. 2015;66:
6. Mostaghimi A, Crotty BH. Professionalism in the digital age. Ann Intern 1606-1615.
Med. 2011;154:560-562. 26. Beckjord EB, Rechis R, Nutt S, et al. What do people affected by cancer
7. Dizon DS, Graham D, Thompson MA, et al. Practical guidance: the use of think about electronic health information exchange? Results from the
social media in oncology practice. J Oncol Pract. 2012;8:e114-e124. 2010 LIVESTRONG Electronic Health Information Exchange Survey and
8. Thompson MA. Using social media to learn and communicate: it is not the 2008 Health Information National Trends Survey. J Oncol Pract.
about the tweet. Am Soc Clin Oncol Educ Book. 2015;35:206-211. 2011;7:237-241.
9. Plank W, Wyatt T. The Future of the Wearables Market. Wall Street 27. Kazmi Z. Effects of exam room EHR use on doctor-patient communi-
Journal, January 13, 2016. http://www.wsj.com/articles/the-future-of- cation: a systematic literature review. Inform Prim Care. 2013;21:30-39.
the-wearables-market-1452736738. Accessed January 18, 2016. 28. Blumenthal D, Glaser JP. Information technology comes to medicine.
10. CCS Insight. Market Forecast: Wearables. Cupertino, CA: CCS Insight; N Engl J Med. 2007;356:2527-2534.
2014. 29. Hershman DL, Wang X, McBride R, et al. Delay of adjuvant chemo-
11. Shapiro M, Johnston D, Wald J, et al. Patient-Generated Health Data: therapy initiation following breast cancer surgery among elderly
White Paper Prepared for the Office of the National Coordinator for women. Breast Cancer Res Treat. 2006;99:313-321.
Health Information Technology, 2012. http://www.rti.org/pubs/ 30. Katz SJ, Lantz PM, Paredes Y, et al. Breast cancer treatment experiences
patientgeneratedhealthdata.pdf. Accessed December 1, 2014. of Latinas in Los Angeles County. Am J Public Health. 2005;95:
12. Hooke MC, Gilchrist L, Tanner L, et al. Use of a fitness tracker to promote 2225-2230.
physical activity in children with acute lymphoblastic leukemia. Pediatr 31. Maggard MA, OConnell JB, Lane KE, et al. Do young breast cancer
Blood Cancer. Epub 2016 Jan 12. patients have worse outcomes? J Surg Res. 2003;113:109-113.
13. Galv~ao DA, Taaffe DR, Spry N, et al. Combined resistance and aerobic 32. Jones EB, Furukawa MF. Adoption and use of electronic health records
exercise program reverses muscle loss in men undergoing androgen among federally qualified health centers grew substantially during
suppression therapy for prostate cancer without bone metastases: 2010-12. Health Aff (Millwood). 2014;33:1254-1261.
a randomized controlled trial. J Clin Oncol. 2010;28:340-347. 33. King J, Furukawa MF, Buntin MB. Geographic variation in ambulatory
14. Schmitz KH, Courneya KS, Matthews C, et al; American College of Sports electronic health record adoption: implications for underserved com-
Medicine. American College of Sports Medicine roundtable on exercise munities. Health Serv Res. 2013;48:2037-2059.
guidelines for cancer survivors. Med Sci Sports Exerc. 2010;42: 34. Jhamb M, Cavanaugh KL, Bian A, et al. Disparities in electronic health
1409-1426. record patient portal use in nephrology clinics. Clin J Am Soc Nephrol.
15. Bennett AV, Reeve BB, Basch EM, et al. Evaluation of pedometry as a 2015;10:2013-2022.
patient-centered outcome in patients undergoing hematopoietic 35. Goel MS, Brown TL, Williams A, et al. Disparities in enrollment and use
cell transplant (HCT): a comparison of pedometry and patient re- of an electronic patient portal. J Gen Intern Med. 2011;26:1112-1116.
ports of symptoms, health, and quality of life. Qual Life Res. Epub 36. Goel MS, Brown TL, Williams A, et al. Patient reported barriers to
2015 Nov 17. enrolling in a patient portal. J Am Med Inform Assoc. 2011;18:i8-i12
16. Chung AE, Basch EM. Potential and challenges of patient-generated (suppl 1).
health data for high-quality cancer care. J Oncol Pract. 2015;11:195-197. 37. Ferreira A, Correia A, Silva A, et al. Why facilitate patient access to
17. Cheriff AD, Kapur AG, Qiu M, et al. Physician productivity and the medical records. Stud Health Technol Inform. 2007;127:77-90.
ambulatory EHR in a large academic multi-specialty physician group. Int 38. Wright Nunes JA, Osborn CY, Ikizler TA, et al. Health numeracy: per-
J Med Inform. 2010;79:492-500. spectives about using numbers in health management from African
18. Centers for Medicare & Medicaid Services (CMS), HHS. Medicare and American patients receiving dialysis. Hemodial Int. 2015;19:287-295.
Medicaid Programs; electronic health record incentive programstage 3 39. Yamin CK, Emani S, Williams DH, et al. The digital divide in adoption and
and modifications to meaningful use in 2015 through 2017. Final rules use of a personal health record. Arch Intern Med. 2011;171:568-574.
with comment period. Fed Regist. 2015;80:62761-62955. 40. Sarkar U, Karter AJ, Liu JY, et al. Social disparities in internet patient
19. Institute of Medicine. Key Capabilities of an Electronic Health Record portal use in diabetes: evidence that the digital divide extends beyond
System. Washington, DC: Institute of Medicine; 2003. access. J Am Med Inform Assoc. 2011;18:318-321.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 207


FISCH, CHUNG, AND ACCORDINO

41. Butler JM, Carter M, Hayden C, et al. Understanding adoption of a 60. Partridge AH, Wang PS, Winer EP, et al. Nonadherence to adjuvant
personal health record in rural health care clinics: revealing barriers and tamoxifen therapy in women with primary breast cancer. J Clin Oncol.
facilitators of adoption including attributions about potential patient 2003;21:602-606.
portal users and self-reported characteristics of early adopting users. 61. Hershman DL, Kushi LH, Shao T, et al. Early discontinuation and non-
AMIA Annu Symp Proc. 2013;2013:152-161. adherence to adjuvant hormonal therapy in a cohort of 8,769 early-
42. Wade-Vuturo AE, Mayberry LS, Osborn CY. Secure messaging and di- stage breast cancer patients. J Clin Oncol. 2010;28:4120-4128.
abetes management: experiences and perspectives of patient portal 62. Hershman DL, Shao T, Kushi LH, et al. Early discontinuation and non-
users. J Am Med Inform Assoc. 2013;20:519-525. adherence to adjuvant hormonal therapy are associated with increased
43. Ronda MC, Dijkhorst-Oei LT, Rutten GE. Reasons and barriers for using a mortality in women with breast cancer. Breast Cancer Res Treat. 2011;
patient portal: survey among patients with diabetes mellitus. J Med 126:529-537.
Internet Res. 2014;16:e263. 63. Yood MU, Owusu C, Buist DS, et al. Mortality impact of less-than-
44. Klatt TE, Hopp E. Effect of a best-practice alert on the rate of in- standard therapy in older breast cancer patients. J Am Coll Surg. 2008;
fluenza vaccination of pregnant women. Obstet Gynecol. 2012;119: 206:66-75.
301-305. 64. Noens L, van Lierde MA, De Bock R, et al. Prevalence, determinants, and
45. Meyfroidt G, Wouters P, De Becker W, et al. Impact of a computer- outcomes of nonadherence to imatinib therapy in patients with chronic
generated alert system on the quality of tight glycemic control. In- myeloid leukemia: the ADAGIO study. Blood. 2009;113:5401-5411.
tensive Care Med. 2011;37:1151-1157. 65. Ganesan P, Sagar TG, Dubashi B, et al. Nonadherence to imatinib ad-
46. Kucher N, Koo S, Quiroz R, et al. Electronic alerts to prevent venous versely affects event free survival in chronic phase chronic myeloid
thromboembolism among hospitalized patients. N Engl J Med. 2005; leukemia. Am J Hematol. 2011;86:471-474.
352:969-977. 66. Sonpavde G, Periman PO, Bernold D, et al. Sunitinib malate for met-
47. Cho A, Lee JE, Yoon JY, et al. Effect of an electronic alert on risk of astatic castration-resistant prostate cancer following docetaxel-based
contrast-induced acute kidney injury in hospitalized patients un- chemotherapy. Ann Oncol. 2010;21:319-324.
dergoing computed tomography. Am J Kidney Dis. 2012;60:74-81. 67. Neugut AI, Subar M, Wilde ET, et al. Association between prescription
48. Selvan MS, Sittig DF, Thomas EJ, et al. Improving erythropoietin- co-payment amount and compliance with adjuvant hormonal therapy
stimulating agent administration in a multihospital system through in women with early-stage breast cancer. J Clin Oncol. 2011;29:
quality improvement initiatives: a pre-post comparison study. J Patient 2534-2542.
Saf. 2011;7:127-132. 68. Accordino MK, Hershman DL. Disparities and challenges in adherence to
49. Ranpura V, Agrawal S, Chokshi P, et al. Improving documentation of pain oral antineoplastic agents. Am Soc Clin Oncol Educ Book. 2013;33:
management at MedStar Washington Cancer Institute. J Oncol Pract. 271-276.
2015;11:155-157. 69. Basch E, Deal AM, Kris MG, et al. Symptom monitoring with patient-
50. Jacobson JO, Neuss MN, Hauser R. Measuring and improving value of reported outcomes during routine cancer treatment: a randomized
care in oncology practices: ASCO programs from Quality Oncology controlled trial. J Clin Oncol. 2016;34:557-565.
Practice Initiative to the Rapid Learning System. Am Soc Clin Oncol Educ 70. Wright E, Darer J, Tang X, et al. Sharing physician notes through an
Book. 2012;32:e70-e76. electronic portal is associated with improved medication adherence:
51. American Society of Clinical Oncology. ASCO CancerLinQ. http:// quasi-experimental study. J Med Internet Res. 2015;17:e226.
cancerlinq.org/. Accessed January 4, 2016. 71. Lyles CR, Sarkar U, Schillinger D, et al. Refilling medications through an
52. Sledge GW Jr, Miller RS, Hauser R. CancerLinQ and the future of cancer online patient portal: consistent improvements in adherence across
care. Am Soc Clin Oncol Educ Book. 2013;33:430-434. racial/ethnic groups. J Am Med Inform Assoc. Epub 2015 Sep 2.
53. Abernethy AP, Etheredge LM, Ganz PA, et al. Rapid-learning system for 72. Ross SE, Moore LA, Earnest MA, et al. Providing a web-based online
cancer care. J Clin Oncol. 2010;28:4268-4274. medical record with electronic communication capabilities to patients
54. Etheredge LM. A rapid-learning health system. Health Aff (Millwood). with congestive heart failure: randomized trial. J Med Internet Res.
2007;26:w107-w118. 2004;6:e12.
55. Osterberg L, Blaschke T. Adherence to medication. N Engl J Med. 2005; 73. Hewitt ME, Simone JV (eds); National Cancer Policy Board, Institute of
353:487-497. Medicine and National Research Council. Ensuring Quality Cancer Care.
56. McDonald HP, Garg AX, Haynes RB. Interventions to enhance patient Washington, DC: National Academy Press; 1999.
adherence to medication prescriptions: scientific review. JAMA. 2002; 74. Neuss MN, Desch CE, McNiff KK, et al. A process for measuring the
288:2868-2879. quality of cancer care: the Quality Oncology Practice Initiative. J Clin
57. Livaudais JC, Hershman DL, Habel L, et al. Racial/ethnic differences in Oncol. 2005;23:6233-6239.
initiation of adjuvant hormonal therapy among women with hormone 75. Accreditation Council for Graduate Medical Education. CLER Pathways
receptor-positive breast cancer. Breast Cancer Res Treat. 2012;131: to Excellence. https://www.acgme.org/acgmeweb/Portals/0/PDFs/
607-617. CLER/CLER_Brochure.pdf. Accessed December 28, 2015.
58. Kimmick G, Anderson R, Camacho F, et al. Adjuvant hormonal therapy 76. Ramsey SD. A Pragmatic Trial to Improve Colony Stimulating Factor
use among insured, low-income women with breast cancer. J Clin Oncol. Use in Cancer. http://www.pcori.org/research-results/2015/pragmatic-
2009;27:3445-3451. trial-improve-colony-stimulating-factor-use-cancer. Accessed December
59. Neugut AI, Hillyer GC, Kushi LH, et al. Non-initiation of adjuvant hor- 28, 2015.
monal therapy in women with hormone receptor-positive breast 77. Mayer DK, Birken SA, Check DK, et al. Summing it up: an integrative
cancer: the Breast Cancer Quality of Care Study (BQUAL). Breast Cancer review of studies of cancer survivorship care plans (2006-2013). Cancer.
Res Treat. 2012;134:419-428. 2015;121:978-996.

208 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


HEMATOLOGIC MALIGNANCIESLEUKEMIA,
MYELODYSPLASTIC SYNDROMES, AND
ALLOTRANSPLANT

Evolving Therapies in Acute


Myeloid Leukemia: Progress
at Last?

CHAIR
Daniel J. DeAngelo, MD, PhD
Dana-Farber Cancer Institute
Boston, MA

SPEAKERS
Eytan M. Stein, MD
Memorial Sloan Kettering Cancer Center
New York, NY

Farhad Ravandi, MD
The University of Texas MD Anderson Cancer Center
Houston, TX
DEANGELO ET AL

Evolving Therapies in Acute Myeloid Leukemia: Progress at


Last?
Daniel J. DeAngelo, MD, PhD, Eytan M. Stein, MD, and Farhad Ravandi, MD

OVERVIEW

Acute myeloid leukemia (AML) is an acquired disease characterized by chromosomal translocations and somatic mutations
that lead to leukemogenesis. Systemic combination chemotherapy with an anthracycline and cytarabine remains the
standard induction regimen for fit adults. Patients who achieve complete remission generally receive postinduction
therapy with cytarabine-based chemotherapy or an allogeneic bone marrow transplant. Those unfit for induction che-
motherapy are treated with hypomethylating agents (HMAs), low-dose cytarabine, or they are offered supportive care alone
with transfusions and prophylactic antimicrobials. The revolution in understanding the genetics of AML, facilitated by next-
generation sequencing, has led to many new drugs against driver mutations. Better methods of identification of leukemic
blasts have provided us with better means to detect the disease left behind after cytotoxic chemotherapy regimens. This
measurable residual disease has been correlated with poorer relapse-free survival, demonstrating the need for novel
strategies to eradicate it to improve the outcome of patients with acute leukemias. In this article, we discuss adapting and
improving AML therapy by age and comorbidities, emerging targeted therapies in AML, and minimal residual disease (MRD)
assessment in AML.

A cute myeloid leukemia is most commonly diagnosed in


older adults, with a median age of diagnosis of 66 years.1
Given the aging population in the United States, the in-
these factors do not sufficiently address the heterogeneity
present in this older population. New studies suggest
that geriatric evaluation including psychological, cognitive,
cidence of AML will grow dramatically in the coming physical, and comorbidities can identify deficits affecting
decades.2 Older patients with AML define a distinct subgroup morbidity and mortality among patients with cancer and
with a worse prognosis and different biological character- should be incorporated into the initial evaluation for older
istics as compared to their younger counterparts.3 In ad- adults with AML.8-11 In addition, molecular profiling can help
dition, many older patients have significant comorbidities to further identify a biologically resistant group of patients
and functional impairment that further complicates thera- who may benefit from targeted therapeutic approaches.
peutic options.3,4 As a result of the biologic complexity, high Regardless of whether standard induction chemotherapy,
rates of chemotherapy drug resistance, and poor host fac- less-intensive therapy, or targeted approaches are used, the
tors, the 5-year disease-specific survival of patients older definition of complete remission is being redefined by our
than age 65 remains less than 5%. ability to detect small residual subclones or MRD.
In spite of the many advances made in other hematologic
diseases, induction chemotherapy with cytarabine and an ADAPTING AND IMPROVING THERAPY
anthracycline remains the standard initial approach for older ACCORDING TO AGE AND COMORBIDITIES
patients with AML. Although associated with high rates of Acute myeloid leukemia is a clonal hematopoietic stem cell
death from toxicity, it still offers the best chance of longer- disorder, for which the treatment, and in fact cure, hinges
term survival.5 Less-intensive treatment and palliative ap- upon the successful eradication of the leukemic clone. To
proaches are currently being explored for patients who this end, induction chemotherapy is given to reduce the level
cannot tolerate standard anthracycline induction therapy.6 of the leukemic burden below the level of detection and to
Previous studies have identified age, performance status, restore normal hematopoiesis. Postremission consolidation,
comorbidities, and cytogenetic risk groups as important alone or followed by an autologous or allogeneic stem cell
factors in stratifying older patients with AML.7 However, transplant, is then used to eradicate the remaining leukemic

From the Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA; Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, NY;
The University of Texas MD Anderson Cancer Center, Houston, TX.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Daniel J. DeAngelo, MD, PhD, Dana-Farber Cancer Institute, 450 Brookline Ave., Boston, MA 02215; email: daniel_deangelo@dfci.harvard.edu.

2016 by American Society of Clinical Oncology.

e302 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


EVOLVING THERAPIES IN ACUTE MYELOID LEUKEMIA

burden in hopes of cure. The backbone of remission in- spent a mean of 28.3% of their life from diagnosis in the
duction therapy for younger patients (age , 60) and a sub- hospital and 13.8% of their life attending outpatient ap-
set of older patients deemed fit enough to undergo such pointments, and only 23% used hospice services. Within
treatment has consisted of high-dose cytotoxic chemo- 30 days before death, 84.5% of patients were hospitalized,
therapy often consisting of 7 days of continuous infusion and 61.0% died in the hospital. Although intensive induction
cytarabine combined with 3 days of an anthracycline, offers a minority of patients a potentially curative therapy,
otherwise known as the 7 plus 3 regimen. Remission rates the patients who died spent more than 50% of their life from
with induction therapy vary and depend on many factors diagnosis in the hospital or clinic. However, with the in-
including patient age, cytogenetics, molecular profiling, troduction of HMAs, as well as improvement in supportive
antecedent hematologic disorder, prior chemotherapy, and care measures, it is unclear whether the findings of this study
type of leukemia. Over the past several decades, induction would be replicable in the modern era.
therapy has been intensified with the aim of improving
remission rates, extending disease-free survival and, ulti-
mately, overall survival. For elderly patients (age . 60) Pretreatment Evaluation
induction strategies have concentrated on less-toxic in- Induction chemotherapy is a complicated and rigorous pro-
duction strategies using HMAs, monoclonal antibodies, cess that is required to obtain clinical remission.14 Before
nucleoside analogs, and other novel compounds. the initiation of induction chemotherapy, it is important to
establish the presence of comorbid conditions that could
potentially complicate the treatment of the patient. It is
Health Care Utilization clear that all elements of the patients history and physi-
Older patients with AML spend a large portion of their life cal examination are important. However, particular atten-
after diagnosis in the hospital or clinic. Given the short life tion should be paid to a prior history of a hematologic
expectancy of the vast majority of older patients with AML disorderfor example, a myelodysplastic or myeloprolif-
and the low likelihood of cure, it is important to consider the erative disorder, prior therapy for malignancies including
impact of cancer therapy on their quality of life, which in- chemotherapy or radiation therapy, and a family history of
cludes the time spent in the hospital and receiving medical disease. Patients with secondary or treatment-related AML
care throughout their illness. One randomized study com- have a significantly worse prognosis, which is predominantly
pared intensive induction versus supportive care of older influenced by the higher incidence of unfavorable cytoge-
patients with AML, with hydroxyurea or low-dose cytarabine netic abnormalities. A history of cardiac disease such as
used as cytoreductive agent when needed.12 In this study, congestive heart failure would mandate careful monitoring
patients treated with intensive chemotherapy had a and potential alteration of induction chemotherapy because
10-week survival advantage compared with those treated anthracycline-based regimens are standard. Patients typi-
with supportive care. However, there were no major dif- cally receive large amounts of intravenous fluids that ac-
ferences in patients time spent in the hospital. Another company initial chemotherapy as well as antibiotics and
study looked retrospectively at 330 consecutive older pa- blood and platelet transfusions, and therefore patients with
tients diagnosed with AML to examine their health care cardiac abnormalities at the time of diagnosis need rigorous
utilization.13 The median number of hospitalizations for the and careful monitoring.
entire cohort was 4.2 (range, 1 to 18). Patients who died A study of 101 patients with AML age 65 or older at the
Dana-Farber Cancer Institute underwent geriatric assess-
ment.15 With a median age of 72, only 38% of patients had a
KEY POINTS hematopoietic stem cell transplant comorbidity index of
greater than 1, and only three patients required help with
The treatment of AML, especially in elderly patients, activities of daily living. The majority of patients (78%) re-
remains a difficult endeavor with high therapy-related ceived treatment and, of those, 45% underwent induction
toxicity and poor long-term survival. chemotherapy and 44% received either decitabine or aza-
Efficacy of therapy in AML has been assessed by citidine. The 1-year disease-specific survival was 39%. On
morphologic assessment, with complete remission multivariate analysis, age at diagnosis, cytogenetic risk
correlating with improved outcomes. group, secondary AML versus de novo, comorbidity score
Several retrospective studies have clearly demonstrated (hematopoietic stem cell transplant comorbidity index) of
the adverse prognostic value of persistent MRD after at least 1, difficulty with strenuous activity, and pain were
induction and consolidation therapy.
independent prognostic factors for survival. The most
Standardization and universal availability of these
assays are key to their potential future use to direct
amazing fact is that even when the analysis was limited to
risk-adapted therapy. patients with ECOG performance status of 1 or lower, diffi-
New target-specific agents with novel mechanisms of culty with strenuous activities, pain, and comorbidity
action are likely to be more effective in eradicating MRD, remained independent predictors. Thus, geriatric assessment
thereby improving outcomes. variables are independent prognostic factors for survival,
even among patients with the best performance status.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e303


DEANGELO ET AL

Acute Myeloid Leukemia Ontogeny improved median overall survival among patients treated
Acute myeloid leukemia is a biologically heterogeneous with acacitadine compared with conventional care ap-
disease that is typically classified into three clinically proaches, which included supportive care, LDAC, and 7 plus 3.25
distinct categories. Secondary AML (s-AML) represents Although most studies using HMA therapy in AML pa-
transformation from a prior diagnosis of myelodysplastic tients demonstrate lower complete remission rates
syndrome or myeloproliferative neoplasm (MPN); therapy- (10%20%) compared with conventional intensive chemo-
related AML develops as a complication of exposure to therapy, approximately 30% of patients have evidence of
leukemogenic therapies; and de novo AML arises in the disease response or stabilization and, importantly, have
absence of an identified exposure or prior stem cell dis- overall survival rates equivalent or superior to other con-
order. A retrospective, targeted, mutational analysis of ventional treatments. In addition, HMA therapy is well
194 patients with s-AML or therapy-related AML was tolerated over long periods of time, with fewer hospital days
compared with 100 unselected older patients with AML.16 and transfusions.
The presence of a mutation in SRSF2, SF3B1, U2AF1, ZRSR2, At The University of Texas MD Anderson Cancer Center,
ASXL1, EZH2, BCOR, or STAG2 was greater than 95% specific the outcomes of 671 older patients with AML treated
for the diagnosis of s-AML. The presence of one of these with up-front intensive chemotherapy or HMA therapy
secondary-type mutations defines a subtype of elderly were retrospectively analyzed.26 Although the complete
de novo AML that share clinical and pathologic simi- remission rates were significantly lower with HMA therapy
larities with confirmed cases of s-AML. Patients with versus intensive chemotherapy (28% vs. 42%), there was no
secondary-type mutations have a higher likelihood of in- difference in 8-week early death, 2-year relapse-free sur-
trinsic therapy resistance, as reflected by a lower rate vival, or median overall survival (6.7 vs. 6.5 months). The
of complete remission with standard intensive induction only group of patients who benefited from intensive in-
chemotherapy.3,17,18 In contrast, older patients with de duction were those with NPM-1mutated AML, coming back
novo/pan-AML mutations achieved a higher rate of com- to the importance of AML ontogeny. The group from The
plete remission and rarely required reinduction. Thus, ge- Ohio State University demonstrated a very high overall
netic ontogeny may highlight a subset of older patients with response rate of 75% using a 10-day decitabine regimen,
secondary-type mutations who, on the basis of age, genetic even in those patients with a complex karyotype.27 How-
characteristics, and induction outcomes, may have had an ever, the 10-day decitabine regimen is associated with
unrecognized period of antecedent myelodysplasia before treatment mortality rates similar to those of intensive
AML diagnosis. In elderly AML, genetic ontogeny may ac- chemotherapy in elderly individuals. Thus, the vast majority
count for the differences in chemosensitivity and clinical of older individuals desiring largely outpatient therapy
outcomes. should be considered for subcutaneous azacitidine, 5-day
decitabine, or LDAC therapy.
The novel nucleoside analog clofarabine (Genzyme,
Less Intensive Strategies Cambridge, MA) has been studied extensively in older pa-
Low-dose cytarabine (LDAC) has been used for the treat- tients with de novo AML. Clofarabine inhibits ribonucleotide
ment of elderly, unfit AML patients for several decades. reductase and DNA polymerase. It is resistant to cleavage by
Many studies have highlighted the fact that even very small purine nucleoside phosphorylase and to deamination by
doses of cytarabine (20 mg/m2 administered once or twice adenosine deaminase.28-30 A complete remission rate of
daily for 1014 days per month) can induce complete re- 32% was seen in a phase II trial of patients with relapsed or
mission in 8% to 18% of patients with AML and can prolong refractory hematologic malignancies.31,32 Clofarabine has
survival compared with best supportive care.19 However, been combined in an effort to modulate cytarabine tri-
patients with adverse cytogenetics seldom achieve re- phosphate accumulation in subsequent phase I and II trials.33,34
mission with LDAC. Despite this, LDAC may still be a Clofarabine as a single agent was studied in 112 patients
therapeutic option for geriatric patients without poor-risk older than age 60 who were deemed to be unlikely can-
karyotype AML who prefer to self-administer drugs at home didates to benefit from standard conventional chemo-
without the need for daily clinic visits or hospitalization. therapy. A complete remission rate was 38%, and the
Because of the tolerability of LDAC, there are many clinical overall response rate was 46% with a median disease-free
trials exploring the combination of LDAC with novel in- survival of approximately 33 weeks.35 Clofarabine has now
vestigational agents in an attempt to improve outcomes. been compared in multiple randomized phase III studies,
Inhibitors of the DNA methyltransferase (DNMT) azaciti- including the MRC study of older adults who are not fit for
dine (Vidaza, Celgene) and decitabine (Dacogen, Bloo- chemotherapy, comparing low dose ara-c versus clofar-
mington, MN) are approved for the treatment of patients abine. Although the clofarabine arm had a higher remission
with myelodysplastic syndrome.20-22 The current World rate, there was no difference in overall survival.36 In ad-
Health Organization definition of AML now includes patients dition, the recent ECOG 2906 study was presented at the
with a blast count of between 20% and 29% because these American Society of Hematology (ASH) Annual Meeting
patients were previously considered as having myelodys- comparing 3 plus 7 versus clofarabine, and the standard
plasia.23,24 The study by Fenaux et al demonstrated an arm was superior.37

e304 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


EVOLVING THERAPIES IN ACUTE MYELOID LEUKEMIA

Conclusion The most targeted mutated protein is FLT3 because of


The treatment of AML, especially for elderly patients, its role as the most common recurrent genetic alteration
remains a difficult endeavor with high therapy-related among patients with de novo AML. FLT3 internal tandem
toxicity and poor long-term survival. The impact on health duplications (FLT3-ITD) occurs in 30% of AML patients and
care utilization should not be underestimated, even in those portend a poor prognosis, particularly among patients with a
patients choosing less-intensive strategies. It is hoped that high allelic ratio.39,40
the development of targeted therapies either as single
agents of in combination with current modalities will result Midostaurin. Midostaurin (PKC-412) is a moderately potent
in improved outcomes. inhibitor of FLT3-ITD and FLT3 tyrosine kinase domain (TKD)
mutations and inhibits other kinases such as c-KIT, PDGFR-b,
VEGFR-2, and protein kinase C. A phase II study of mid-
EMERGING TARGETED THERAPIES IN ACUTE ostaurin monotherapy for patients with relapsed and re-
MYELOID LEUKEMIA fractory FLT3-positive AML showed minimal clinical activity;
Acute myeloid leukemia is an acquired disease character-
only 1 patient achieved a partial remission (PR).41 A follow-
ized by chromosomal translocations and somatic mutations
up phase IB study combined midostaurin with induction
that lead to leukemogenesis. Systemic combination che-
chemotherapy (7 plus 3) without regard to FLT3 status and
motherapy with an anthracycline and cytarabine remains
demonstrated that the combination was safe and well tol-
the standard induction regimen for fit adults. Patients
erated; complete remissions were seen in 92% of patients
who achieve a complete remission generally receive post-
with FLT3-ITD compared with 74% of patients with FLT3 wild-
induction therapy with cytarabine-based chemotherapy or
type.42 On the basis of these data, the RATIFY study, an
an allogeneic bone marrow transplant.34 Those unfit for
international randomized phase III study of midostaurin or
induction chemotherapy are treated with hypomethylating
placebo in combination with induction and consolidation
agents or low-dose cytarabine, or they are offered sup-
chemotherapy, was designed. The outcomes, reported at
portive care alone with transfusions and prophylactic
the 2015 American Society of Hematology (ASH) Annual
antimicrobials.35,38
Meeting, showed improved 5-year overall survival in the
The revolution in understanding the genetics of AML,
midostaurin arm (51.4% vs. 44.2%), regardless of whether
facilitated by next-generation sequencing, has led to many
patients were censored at the time of stem cell transplant,
new drugs against driver mutations. These novel thera-
despite no difference in the rates of complete remission at
pies have emerged in parallel with antibodies against cell
60 days.43 The superiority of midostaurin/chemotherapy
surface proteins expressed on leukemic myeloblasts. The
over placebo/chemotherapy was consistent regardless of
past 10 years have seen the development of small-molecule
allelic burden (high vs. low), FLT3-ITD, or FLT3-TKD. Pa-
inhibitors of commonly mutated proteins and antibody-based
tients receiving midostaurin had an increased frequency of
therapies with early clinical efficacy unseen in prior studies.
grade 3-4 desquamating rash. The overall survival benefit
in combination with the favorable toxicity profile makes
Small-Molecule Inhibitors midostaurin in combination with induction and consolida-
A variety of mutated and overexpressed proteins contrib- tion chemotherapy the new standard of care for patients
ute alone or in combination to leukemogenesis. This ever- with FLT3-mutated AML.
enlarging number of molecular targets has led to small
molecules that inhibit mutant or overexpressed proteins. Quizartinib. More potent inhibitors of FLT3-ITD and FLT3-TKD
The list of agents in development is long, but a few inhibitors are in development. Quizartinib is a selective inhibitor of
stand out as showing exceptional promise in recent clinical FLT3-ITD but lacks activity against FLT3-TKD. The composite
studies (Table 1). complete remission rate in phase II studies of quizartinib

TABLE 1. Response Rate in Clinical Trials of Targeted Agents for the Treatment of Acute Myeloid Leukemia*

Agent Mechanism of Action Suggested Patient Population Notes


Quizartinib FLT3 inhibitor FLT3 + AML Single-agent activity against FLT3-ITD
Resistance emerges in most patients
ASP-2215 FLT3 inhibitor with activity FLT3-ITD or FLT3-TKD Single-agent activity against FLT3-ITD and FLT3-TKD
against TKD resistance mutation with CRc rate of 46% in relapsed/refractory AML
AG-221 IDH2 inhibitor IDH2 mutated Single-agent activity (37% ORR) in R/R AML
AG-120 IDH1 inhibitor IDH1 mutated Single-agent activity (35% ORR) in R/R AML
ABT-199 BCL-2 inhibitor Ongoing investigation CR/CRi rate of 71% when combined with HMAs
in untreated AML in older patients
Abbreviations: AML, acute myeloid leukemia; CRc, cytogenic complete remission; ORR, overall response rate; R/R, relapsed/refractory; CRi, incomplete peripheral blood count
recovery; HMA, hypomethalyting agent.
*Response criteria between trials do not necessarily use the international working group criteria and, therefore, between-trial comparisons are difficult to make.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e305


DEANGELO ET AL

as a single agent in relapsed and refractory FLT3-mutated TABLE 2. Antibody-Based Therapies for Acute Myeloid
AML ranges between 44% and 54%.44-47 The median du- Leukemia (Excluding Immunotherapy) in Clinical
ration of response is between 11.3 and 12.7 weeks. Many Development
patients who receive quizartinib acquire mutations in
the TKD of the FLT3 gene (D835 and F691) and resistance Mechanism of
Agent Action Notes
to ongoing treatment. Because of this, quizartinib may be
best used as monotherapy bridge to a potentially cura- Gemtuzumab Anti-CD33 antibody-drug Withdrawn from U.S.
Ozagomicin conjugate market; efforts to
tive allogeneic bone marrow transplant. A randomized reintroduce on the
open-label phase III study of quizartinib versus chemo- basis of ongoing
therapy (NCT02039726) with a primary endpoint of overall clinical studies
survival is currently accruing patients, and a randomized Vadastuximab Anti-CD33 antibody-drug CR/CRi of 65% when
Tariline conjugate given in combination
double-blind study of quizartinib or placebo in combination (SGN-CD33A) with HMAs in older
with induction and consolidation chemotherapy is in devel- patients with
opment (NCT02668653). untreated AML in
ongoing study
Gilteritinib. Gilteritinib (ASP-2215), also a potent inhibitor IMGN779 Anti-CD33 antibody-drug Payload of alkylating
conjugate agent (DGN-462);
of FLT3, differs from quizartinib in its ability to inhibit both studies beginning
FLT3-ITD and FLT3-TKD mutations. Results of a phase I/II in early 2016
study among 165 patients receiving 80 mg or greater of Lintuzumab- Anti-CD33 antibody- Ongoing phase I/II study
gilteritinib were reported at the 2015 ASH Annual Meeting.48 AC225 radiopharmaceutical in combination with
Among patients with FLT3-ITD, the composite complete low-dose cytarabine
remission rate was 46%, and it was 9% for those with a JNJ-56022473 Anti-CD123 Antibody against CD123;
trials beginning
FLT3-TKD. Responses were similar regardless of whether
patients had received prior FLT3directed therapy. The KHK2823 Anti-CD123 Antibody against CD123;
trials beginning
median duration of response was 15.9 weeks, similar
ADCT-301 Anti-CD25 Antibody-drug conjugate
to what is seen in clinical studies with quizartinib. A phase against CD25; trials
I study of ASP2215 in combination with induction and beginning
consolidation chemotherapy is ongoing (NCT02236013), AGS67E Anti-CD37 Antibody-drug conjugate
and a randomized phase III study of ASP2215 versus salvage (Anti-CD37) against CD37; trials
chemotherapy is accruing patients (NCT02421939). beginning
Abbreviations: CR, complete remission; CRi, complete response with incomplete blood
count recovery; HMA, hypomethylating agent; AML, acute myeloid leukemia.
IDH1 and IDH2
IDH, the enzyme that converts isocitrate to alpha-ketoglutarate
in the mitochondria (IDH2) or the cytoplasm (IDH1) as part BCL-2 Inhibitors
of the citric acid cycle, is mutated in 15% and 10% of pa- BCL-2 overexpression has been implicated in the mainte-
tients with de novo AML, respectively.49 The prevalence of nance and survival of AML cells in vitro and is associated with
IDH mutations increases with age.50,51 Mutant IDH enzymes resistance to chemotherapy and poor survival among pa-
acquire neomorphic activity and catalyze the conversion of tients with AML. The BCL-2 inhibitor venetoclax (ABT-199)
alpha-ketoglutarate into beta-hydroxygutarate (2-HG). In- showed a complete remission/complete response with
creased intracellular 2-HG causes inhibition of TET enzymes incomplete blood count recovery in five of 32 patients
and subsequent arrest in myeloblast maturation.52-54 In- in a phase II clinical study.57 Preclinical models suggest that
hibitors of mutant IDH1 and mutant IDH2 are currently the combination of venetoclax and HMAs are synergistic. On
in phase I clinical trials (NCT02381886, NCT01915498, and the basis of preclinical data, a phase I study of the combi-
NCT02074839). nation of venetoclax and decitabine or 5-azacitidine was
Interim results of a phase I/II study of the IDH2 inhibitor initiated. As of September 2015, the overall response rate
AG-221 (Agios/Celgene), presented at the 2015 ASH Annual (34 patients) is 76%, with 71% of patients having a complete
Meeting, demonstrated an overall response rate of 37% remission or complete response with incomplete blood
among 159 patients with relapsed/refractory AML with count recovery, without differences in response between
a composite complete remission of 27%. Duration of patients who received decitabine or patients who received
response was 6.9 months.55 Similarly, a phase I study of 5-azacitidine.58 The study is proceeding to an expansion
the IDH1 inhibitor AG-120 demonstrated an overall re- stage (NCT02203773).
sponse rate of 35%, with a composite complete remission
rate of 33%.56 Accrual has started on a phase I study
exploring the safety of combining AG-120 and AG-221 Cell Surface Targets: Antibody-Based Therapy
with both induction and consolidation chemotherapy Antibody-based therapies against cell surface proteins, ei-
and with the HMA, 5-azacitidine (NCT02632708 and ther alone or conjugated to chemotherapeutic or radio-
NCT0267792). pharmaceutical agents, continue to be developed. To date,

e306 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


EVOLVING THERAPIES IN ACUTE MYELOID LEUKEMIA

only one antibody-drug conjugate against CD33, gemtuzu- Lack of achieving a response to the initial therapy, per se,
mab ozagomicin, was granted accelerated approval by the identifies the individual as having a less favorable disease,
U.S. Food and Drug Administration but was subsequently thereby diminishing their likelihood of survival.65 Tradi-
withdrawn from the market because of the inability to tionally, response has been determined using morphologic
confirm clinical benefit in a phase III study. Despite this, the assessment declaring a patient as being in complete re-
effort to target CD33 and other cell surface proteins mission when microscopic examination of the bone mar-
continues (Table 2). row and peripheral blood fails to detect abnormal immature
Perhaps the antibody-drug conjugate with the most en- cells, and there is evidence of resumption of normal bone
couraging clinical data is SGN-CD33A, a novel antibody-drug marrow function with normalization of peripheral blood
conjugate with highly stable dipeptide linkers that enable counts. This assessment is highly insensitive, typically relying
uniform drug loading of a pyrrolobenzodiazapene dimer, on the examination of only 500 cells in the marrow specimen
which cross-links DNA, leading to cell death. An interim and is very much dependent on the skills of the examiner and
analysis of a phase I dose-escalation study of SGN-CD33A the sampling technique. It has also been clear that such
among patients with relapsed CD33-positive AML or patients morphologic remission, although associated with a higher
who declined intensive therapy, the composite complete likelihood of achieving a cure, is not equivalent with com-
remission rate was 27%.59 More impressive is an ongoing plete eradication of the leukemia cells and, without further
study of SGN-CD33A in combination with HMAs (23 pa- postremission therapy, will likely be associated with relapse
tients), where the complete remission/complete response in most, if not all, patients.66 This is a result of the persistence
with incomplete blood count recovery rate is 65%.60 The of morphologically undetectable leukemia, commonly re-
median overall survival has not yet been reached, and at a ferred to a MRD. Detection of this residual leukemia is highly
median follow-up of 7.7 months, 72% of patients remained dependent on the sensitivity and specificity of the technique
alive and in the study. This impressive early clinical activity used, leading to its description by some as measurable
has led to the development of the CASCADE study, a ran- residual disease.67
domized phase III study of HMAs in combination with Therefore, the traditional definition of complete remis-
SGN-CD33A versus HMAs alone for patients with newly sion is an insensitive and imperfect assessment of the
diagnosed AML who are not candidates for intensive in- susceptibility of leukemic cells to the treatment admin-
duction chemotherapy. istered. More sensitive assays such as multiparameter flow
cytometry (MFC), polymerase chain reaction (PCR) analysis
of aberrant and leukemia-specific genes, and, more recently,
MINIMAL RESIDUAL DISEASE ASSESSMENT IN next-generation sequencing have been used to try to detect
ACUTE MYELOID LEUKEMIA: DOES IT MATTER? submicroscopic leukemia cells persisting in complete re-
Why Is Detection of Minimal Residual Disease mission, thereby providing an improved assessment of the
Relevant? effectiveness of therapy.68 Several published reports among
Predicting outcome of therapy has been and continues to be patients with AML have examined the utility of such residual
an integral component of the treatment of patients with disease assessment and have demonstrated the prognostic
AML.61 This is based largely on the realization that certain value of detecting MRD at the time of morphologic remission
subtypes of AML are less sensitive to the effects of cyto- in AML. This section reviews the available literature and
toxic chemotherapy and, as such, less likely to be cured discusses the potential ways that this can influence future
with the established and currently available combina- therapeutic decision making for patients with AML.
tion chemotherapy regimens. This prediction, be it highly
imperfect, has been largely used to determine the
best postremission strategy (including allogeneic stem cell Available Assays for Detecting Minimal Residual
transplantation or continued chemotherapy) and has been Disease
based mainly on pretreatment patient- and disease-related Currently available techniques for detecting residual dis-
variables such as age, performance status, cytogenetic and ease in complete remission include MFC, which relies on
molecular aberrations, and presence or absence of ante- detection of leukemia-associated immunophenotypes, PCR,
cedent hematologic disorders or prior DNA-damaging and, more recently, next-generation sequencingbased
therapy for other disorders or malignancies.61-64 As such, assays to detect aberrant genetic events in the leukemic
this determination is based largely on collective historical cells.69-71 Although karyotypic analysis to detect persistent
information and does not rely on the response of the in- cytogenetically abnormal clones at the time of remission is
dividual patient to the specific therapeutic intervention he an old and less sensitive strategy, its value in predicting
or she received for induction of their initial remission. outcome has been reported by several investigators.72,73
Clearly, the sensitivity of an individual patients leukemic Leukemia-associated immunophenotypes are aberrant
cells to the initial therapy is likely to be important in de- patterns of antigen expression that can be identified in the
termining the degree of reduction of disease burden and the leukemic blasts of most patients at diagnosis.74,75 They in-
probability that it would be associated with relapse-free clude aberrant expression of lymphoid antigens, aberrant
survival and cure. expression levels of normal antigens, and co-expression or

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e307


DEANGELO ET AL

asynchronous expression of early and late antigens, leading independent prognostic factor for relapse on multivariate
to temporal aberrancy. The advantages of this assay include analysis, and the prediction was not improved by PCR or
its wide availability, its applicability to more than 95% cases morphologic data.83 Other investigators have reported that
of AML, and its relative rapidity of assessment, which offers MRD by flow and type of response (complete remission,
its potential utility for rapid therapeutic decision making. complete remission with incomplete platelet recovery, or
The disadvantages often include challenging interpretations complete remission with incomplete blood count recovery)
requiring expert analysis (multiple leukemia-associated are important prognostic factors in AML and should be used
immunophenotypes in different blast subsets for the same independently to predict outcome.84
patients, potential for phenotypic shifts between diagnosis,
and relapse blasts) and lower sensitivity depending on the
antibody panel used. Reported Studies of Minimal Residual Disease
Molecular markers such as gene fusion transcripts, gene Monitoring in Acute Myeloid Leukemia
mutations, and aberrantly overexpressed genes have also The potential value of detection of aberrant mutant genes in
been used effectively as markers for detecting residual predicting outcome in AML is best apparent among patients
leukemia burden.67,71 Quantitative reverse-transcription with normal karyotype, because this is the group in which
PCR assays are now well established for detecting re- there is most uncertainty regarding the role of allogeneic
current fusion transcripts such as PML-RARA, CBFB-MYH11, stem cell transplant in first remission. Mutations in several
and RUNX1-RUNX1T1 and have the advantage of being genes including FLT3, NPM1, CEBPA, DNMT3A, TET2, and
highly sensitive and specific in these subsets of AML that IDH1/2 have been described and have been correlated with
account for approximately 20% of cases overall.76 Their outcome in large cohorts of patients with AML.85-87 Several
value in predicting outcome is now universally accepted. of these have been studied as potential markers for MRD
This is particularly true for patients with acute promyelocytic monitoring. The potential utility of mutant FLT3 as a marker
leukemia in whom achievement of a complete molecular of MRD has been controversial, with some investigators
remission with a sensitive assay is now considered as a suggesting that it can be reliably used in complete remission
requisite for long-term leukemia-free survival.77 to predict relapse, and others arguing that it may not be ideal
To increase the applicability of molecular testing to a larger because of the probability of its gain or loss at the time of
population of AML patients (particularly those with a normal relapse.78,88 In contrast, the utility of NPM1 mutations as re-
karyotype), a number of recurrent aberrant gene muta- liable markers of persistent disease is becoming more estab-
tions such as FLT3, NPM1, and DNMT3A have been evaluated lished, with several studies demonstrating that, using real-time
as markers for MRD assessment.78-80 However, despite quantitative PCR for NPM1, achieving an MRD-negative status
this, a large proportion of patients with AML lack a leukemia- after induction and after consolidation is associated with a
specific aberrant molecular target for MRD monitoring. This considerable reduction in the risk of relapse and improvement
has led some investigators to examine normal genes such as in survival.79,89,90 Most recently, a large prospective trial
WT1 that are overexpressed in leukemia cells as potential conducted by the United Kingdom National Cancer Research
markers for MRD assessment, establishing thresholds that Institute AML Working Group (UK NCRI) further demonstrated
predict higher likelihood of disease relapse.81 Novel and that MRD monitoring for mutated NPM1 was the only major
sensitive next-generation sequencing assays that can detect prognostic factor for relapse or death, and, on sequential
all coexisting mutations in leukemic clones at the time of monitoring, relapse was reliably predicted by a rising level of
diagnosis and remission have also begun to be used for MRD NPM1-mutated transcripts.91
monitoring, although there are few retrospective studies The value of MRD monitoring for acute promyelocytic
reported to date.82 The advantage of molecularly-based leukemia and for core-binding factor (CBF) leukemias has
MRD monitoring is its high sensitivity, particularly using been more established. In a study by the U.K. group of
the more modern assays. patients with acute promyelocytic leukemia treated with all-
Comparative studies evaluating morphology, flow, and trans retinoic acid and chemotherapy, persistent MRD after
PCR assays in the same patients have been limited. In a study consolidation and MRD recurrence were the most important
from St. Jude Childrens Hospital, follow-up bone marrow predictors of relapse in multivariate analysis.77 Similarly,
samples from 203 children and adolescents with newly di- several studies have reported the value of establishing
agnosed AML were evaluated by morphologic assessment, thresholds for RUNX1-RUNX1T1 and CBFB-MYH11 after in-
flow cytometry, and PCR for fusion transcripts.83 Among duction and consolidation in predicting the likelihood of
samples with more than 5% blasts by morphology, 8.2% relapse and survival.92-94 Furthermore, recent reports have
were MRD-positive ($ 0.1%) by flow cytometry, whereas suggested a value for MRD-directed risk stratification to
57.5% of samples with more than 5% blasts by morphology improve outcome for patients with CBF AML.95
were negative by flow. Virtually all (99%) of samples negative Although MFC has been extensively used to detect MRD
by PCR were also negative by flow, but only 9.6% of sam- in pediatric studies (and particularly for patients with acute
ples positive by PCR were also flow-positive, with analy- lymphoblastic leukemia), data in the adult population are
sis of RUNX1-RUNX1T1 and CBFB-MYH11 accounting for more limited.96 Early reports using more limited assays
most discrepancies. Flow MRD after induction was an demonstrated the feasibility of this strategy in predicting

e308 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


EVOLVING THERAPIES IN ACUTE MYELOID LEUKEMIA

relapse and survival.97,98 Several reports in pediatric AML has a decreased likelihood of success, and a change in
further established the prognostic value of MRD assessed treatment, if available, is necessary. Clearly, this is de-
by flow and demonstrated its potential use in risk-adapted pendent on the availability of agents with alternative
therapy.99,100 More recently, investigators from the HOVAN/ mechanisms of action that are able to eradicate MRD. Al-
SAKK AML study group and the UK NCRI have reported the though allogeneic stem cell transplant has been traditionally
prognostic value of flow-based MRD assessment in pre- considered to be the alternate strategy in this setting, the
dicting the outcome in younger and older AML cohorts, available data referred to before suggest that it may not be
respectively.101,102 Furthermore, data from the Fred the most effective strategy to eradicate MRD.103,104 Novel
Hutchison Cancer Center have clearly demonstrated that agents such as molecularly targeted drugs (FLT3 or IDH
patients transplanted in CR1 or CR2 have a substantially inhibitors) or monoclonal antibodybased agents including
lower likelihood of relapse post-transplant and improved antibody-drug conjugates and bispecific antibodies, and,
survival if they are determined to be MRD-negative using a potentially, checkpoint inhibitors and chimeric antigen re-
flow cytometrybased assay.103-105 ceptor T cells, may provide us with such alternate strategies
to deal with persistent MRD remaining after cytotoxic
Future Prospects for Minimal Residual Disease regimens. Whether these concepts for maintenance therapy
Eradication Using Target-Specific Agents can lead to improved outcomes with or without subsequent
Perhaps the most important value of persistent MRD is that transplantation will require complex design of future clinical
it indicates that continuation of current treatment strategy trials.

References
1. Howlader N, Krapcho M, Neyman N, et al (eds). SEER Cancer Statistics 13. El-Jawahri AR, Abel GA, Steensma DP, et al. Health care utilization and
Review, 1975-2012, National Cancer Institute. http://seer.cancer.gov/ end-of-life care for older patients with acute myeloid leukemia.
csr/1975_2012/. Accessed April 6, 2016. Cancer. 2015;121:2840-2848.
2. U.S. Census Bureau. U.S. Interim Projections by Age, Sex, Race 14. Lowenberg B, Downing JR, Burnett A. Acute myeloid leukemia. N Engl J
and Hispanic Origin. https://www.census.gov/population/projections/ Med. 1999;341:1051-1062.
data/national/usinterimproj.html. Accessed April 7, 2016. 15. Sherman AE, Motyckova G, Fega KR, et al. Geriatric assessment in
3. Appelbaum FR, Gundacker H, Head DR, et al. Age and acute myeloid older patients with acute myeloid leukemia: a retrospective study of
leukemia. Blood. 2006;107:3481-3485. associated treatment and outcomes. Leuk Res. 2013;37:998-1003.
4. Wedding U, Rohrig B, Klippstein A, et al. Impairment in functional 16. Lindsley RC, Mar BG, Mazzola E, et al. Acute myeloid leukemia
status and survival in patients with acute myeloid leukaemia. J Cancer ontogeny is defined by distinct somatic mutations. Blood. 2015;125:
Res Clin Oncol. 2006;132:665-671. 1367-1376.
5. Behringer B, Pitako JA, Kunzmann R, et al. Prognosis of older patients 17. Anderson JE, Kopecky KJ, Willman CL, et al. Outcome after induction
with acute myeloid leukemia receiving either induction or noncurative chemotherapy for older patients with acute myeloid leukemia is not
treatment: a single-center retrospective study. Ann Hematol. 2003; improved with mitoxantrone and etoposide compared to cytarabine
827:381-389. and daunorubicin: a Southwest Oncology Group study. Blood. 2002;
6. ODonnell MR, Abboud CN, Altman J, et al. Acute myeloid leukemia. 100:3869-3876.
J Natl Compr Canc Net. 2011;9:280-317. 18. Goldstone AH, Burnett AK, Wheatley K, et al; Medical Research
7. Sekeres MA, et al. Decision-making and quality of life in older adults Council Adult Leukemia Working Party. Attempts to improve treat-
with acute myeloid leukemia or advanced myelodysplastic syndrome. ment outcomes in acute myeloid leukemia (AML) in older patients: the
Leukemia. 2004;18:809-816. results of the United Kingdom Medical Research Council AML11 trial.
8. Hurria A, Browner IS, Cohen HJ, et al. Senior adult oncology. J Natl Blood. 2001;98:1302-1311.
Compr Canc Netw. 2012;10:162-209. 19. Burnett AK, Milligan D, Prentice AG, et al. A comparison of low-dose
9. Hurria A, Gupta S, Zauderer M, et al. Developing a cancer-specific cytarabine and hydroxyurea with or without all-trans retinoic acid for
geriatric assessment: a feasibility study. Cancer. 2005;104: acute myeloid leukemia and high-risk myelodysplastic syndrome in
1998-2005. patients not considered fit for intensive treatment. Cancer. 2007;109:
10. Klepin HD, Geiger AM, Tooze JA, et al. The feasibility of inpatient 1114-1124.
geriatric assessment for older adults receiving induction chemo- 20. Kantarjian H, Issa JP, Rosenfeld CS, et al. Decitabine improves patient
therapy for acute myelogenous leukemia. J Am Geriatr Soc. 2011;59: outcomes in myelodysplastic syndromes: results of a phase III ran-
1837-1846. domized study. Cancer. 2006;106:1794-1803.
11. Sorror ML, Maris MB, Storb R, et al. Hematopoietic cell trans- 21. Silverman LR, Demakos EP, Peterson BL, et al. Randomized controlled
plantation (HCT)-specific comorbidity index: a new tool for risk as- trial of azacitidine in patients with the myelodysplastic syndrome:
sessment before allogeneic HCT. Blood. 2005;106:2912-2919. a study of the cancer and leukemia group B. J Clin Oncol. 2002;20:
12. Lowenberg B, Zittoun R, Kerkhofs H, et al. On the value of intensive 2429-2440.
remission-induction chemotherapy in elderly patients of 65+ years 22. Kantarjian H, Oki Y, Garcia-Manero G, et al. Results of a randomized
with acute myeloid leukemia: a randomized phase III study of the study of 3 schedules of low-dose decitabine in higher-risk myelo-
European Organization for Research and Treatment of Cancer Leu- dysplastic syndrome and chronic myelomonocytic leukemia. Blood.
kemia Group. J Clin Oncol. 1989;7:1268-1274. 2007;109:52-57.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e309


DEANGELO ET AL

23. Harris NL, Jaffe ES, Diebold J, et al. The World Health Organization 39. Kottaridis PD, Gale RE, Frew ME, et al. The presence of a FLT3 internal
classification of neoplastic diseases of the hematopoietic and lym- tandem duplication in patients with acute myeloid leukemia (AML)
phoid tissues. Report of the Clinical Advisory Committee meet- adds important prognostic information to cytogenetic risk group and
ing, Airlie House, Virginia, November, 1997. Ann Oncol. 1999;10: response to the first cycle of chemotherapy: analysis of 854 patients
1419-1432. from the United Kingdom Medical Research Council AML 10 and 12
24. Vardiman JW, Harris NL, Brunning RD. The World Health Organization trials. Blood. 2001;98:1752-1759.
(WHO) classification of the myeloid neoplasms. Blood. 2002;100: 40. Frohling S, Schlenk RF, Breitruck J, et al; AML Study Group Ulm. Acute
2292-2302. myeloid leukemia. Prognostic significance of activating FLT3 muta-
25. Fenaux P, Mufti GJ, Hellstrom-Lindberg E, et al; International Vidaza tions in younger adults (16 to 60 years) with acute myeloid leukemia
High-Risk MDS Survival Study Group. Efficacy of azacitidine compared and normal cytogenetics: a study of the AML Study Group Ulm. Blood.
with that of conventional care regimens in the treatment of higher- 2002;100:4372-4380.
risk myelodysplastic syndromes: a randomised, open-label, phase III 41. Stone RM, DeAngelo DJ, Klimek V, et al. Patients with acute myeloid
study. Lancet Oncol. 2009;10:223-232. leukemia and an activating mutation in FLT3 respond to a small-
26. Quintas-Cardama A, Ravandi F, Liu-Dumlao T, et al. Epigenetic therapy molecule FLT3 tyrosine kinase inhibitor, PKC412. Blood. 2005;105:
is associated with similar survival compared with intensive chemo- 54-60.
therapy in older patients with newly diagnosed acute myeloid leu- 42. Stone RM, Fischer T, Paquette R, et al. Phase IB study of the FLT3
kemia. Blood. 2012;120:4840-4845. kinase inhibitor midostaurin with chemotherapy in younger newly
27. Blum W, Garzon R, Klisovic RB, et al. Clinical response and miR-29b diagnosed adult patients with acute myeloid leukemia. Leukemia.
predictive significance in older AML patients treated with a 10-day 2012;26:2061-2068.
schedule of decitabine. Proc Natl Acad Sci USA. 2010;107:7473-7478. 43. Stone RM, Mandrekar S, Sanford BL. The multi-kinase inhibitor
28. Jeha S, Razzouk B, Rytting M, et al. Phase II study of clofarabine in midostaurin (M) prolongs survival compared with placebo (P) in
pediatric patients with refractory or relapsed acute myeloid leukemia. combination with daunorubicin (D)/cytarabine (C) induction (ind),
J Clin Oncol. 2009;27:4392-4397. high-dose C consolidation (consol), and as maintenance (maint)
29. Jeha S, Gaynon PS, Razzouk BI, et al. Phase II study of clofarabine in therapy in newly diagnosed acute myeloid leukemia (AML) pa-
pediatric patients with refractory or relapsed acute lymphoblastic tients (pts) age 18-60 with FLT3 mutations (muts): an international
leukemia. J Clin Oncol. 2006;24:1917-1923. prospective randomized (rand) P-controlled double-blind trial (CALGB
30. Jeha S, Gandhi V, Chan KW, et al. Clofarabine, a novel nucleoside 10603/RATIFY [Alliance]). Blood. 2015;126:6.
analog, is active in pediatric patients with advanced leukemia. Blood. 44. Levis MJ, Perl AE, Dombret H, et al. Final results of a phase 2 open-
2004;103:784-789. label, monotherapy efficacy and safety study of quizartinib (AC220) in
31. Kantarjian H, Gandhi V, Cortes J, et al. Phase 2 clinical and phar- patients with FLT3-ITD positive or negative relapsed/refractory acute
macologic study of clofarabine in patients with refractory or relapsed myeloid leukemia after second-line chemotherapy or hematopoietic
acute leukemia. Blood. 2003;102:2379-2386. stem cell transplantation. Blood. 2012;120:673.
32. Kantarjian HM, Gandhi V, Kozuch P, et al. Phase I clinical and phar- 45. Cortes JE, Kantarjian H, Foran JM, et al. Phase I study of quizartinib
macology study of clofarabine in patients with solid and hematologic administered daily to patients with relapsed or refractory acute
cancers. J Clin Oncol. 2003;21:1167-1173. myeloid leukemia irrespective of FMS-like tyrosine kinase 3-internal
33. Kantarjian HM, Erba HP, Claxton D, et al. Phase II study of clofarabine tandem duplication status. J Clin Oncol. 2013;31:3681-3687.
monotherapy in previously untreated older adults with acute myeloid 46. Cortes JE, Perl AE, Dombret H, et al. Final results of a phase 2 open-
leukemia and unfavorable prognostic factors. J Clin Oncol. 2010;28: label, monotherapy efficacy and safety study of quizartinib (AC220) in
549-555. patients .= 60 years of age with FLT3 ITD positive or negative
34. Bloomfield CD, Lawrence D, Byrd JC, et al. Frequency of prolonged relapsed/refractory acute myeloid leukemia. Blood. 2012;120:48.
remission duration after high-dose cytarabine intensification in acute 47. Schiller GJ, TM, Goldberg SL, et. al. Final results of a randomized phase
myeloid leukemia varies by cytogenetic subtype. Cancer Res. 1998;58: 2 study showing the clinical benefit of quizartinib (AC220) in patients
4173-4179. with FLT3-ITD positive relapsed or refractory acute myeloid leukemia.
35. Kantarjian HM, Thomas XG, Dmoszynska A, et al. Multicenter, ran- J Clin Oncol. 2014;32:5s (suppl; abstr 7100).
domized, open-label, phase III trial of decitabine versus patient choice, 48. Altman JK, Perl AE, Cortes JE. Antileukemic activity and tolerability of
with physician advice, of either supportive care or low-dose cytar- ASP2215 80mg and greater in FLT3 mutation-positive subjects with
abine for the treatment of older patients with newly diagnosed acute relapsed or refractory acute myeloid leukemia: results from a phase
myeloid leukemia. J Clin Oncol. 2012;30:2670-2677. 1/2, open-label, dose-escalation/dose-response study. Blood. 2015;
36. Burnett AK, Russell NH, Hunter AE, et al. Clofarabine doubles the 126:321.
response rate in older patients with acute myeloid leukemia but does 49. Green CL, Evans CM, Zhao L, et al. The prognostic significance of IDH2
not improve survival. Blood. 2013;122:1384-1394. mutations in AML depends on the location of the mutation. Blood.
37. Foran JM, Zhuoxin S, Claxton DF, et al. North American Leukemia, 2011;118:409-412.
Intergroup Phase III Randomized Trial of Single Agent Clofarabine As 50. Paschka P, Schlenk RF, Gaidzik VI, et al. IDH1 and IDH2 mutations are
Induction and Post-Remission Therapy, and Decitabine As Mainte- frequent genetic alterations in acute myeloid leukemia and confer
nance Therapy in Newly-Diagnosed Acute Myeloid Leukemia in Older adverse prognosis in cytogenetically normal acute myeloid leukemia
Adults (Age $60 Years): A Trial of the ECOG-ACRIN Cancer Research with NPM1 mutation without FLT3 internal tandem duplication. J Clin
Group (E2906). Presented at: 57th ASH Annual Meeting & Exposition; Oncol. 2010;28:3636-3643.
December 2015; Orlando, FL. 51. Marcucci G, Maharry K, Wu YZ, et al. IDH1 and IDH2 gene mutations
38. Dombret H, Seymour JF, Butrym A, et al. International phase 3 study of identify novel molecular subsets within de novo cytogenetically
azacitidine vs conventional care regimens in older patients with newly normal acute myeloid leukemia: a Cancer and Leukemia Group B
diagnosed AML with .30% blasts. Blood. 2015;126:291-299. study. J Clin Oncol. 2010;28:2348-2355.

e310 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


EVOLVING THERAPIES IN ACUTE MYELOID LEUKEMIA

52. Ward PS, Patel J, Wise DR, et al. The common feature of leukemia- 70. Kern W, Voskova D, Schoch C, et al. Determination of relapse risk
associated IDH1 and IDH2 mutations is a neomorphic enzyme activity based on assessment of minimal residual disease during complete
converting alpha-ketoglutarate to 2-hydroxyglutarate. Cancer Cell. remission by multiparameter flow cytometry in unselected patients
2010;17:225-234. with acute myeloid leukemia. Blood. 2004;104:3078-3085.
53. Figueroa ME, Abdel-Wahab O, Lu C, et al. Leukemic IDH1 and IDH2 71. Grimwade D, Freeman SD. Defining minimal residual disease in acute
mutations result in a hypermethylation phenotype, disrupt TET2 myeloid leukemia: which platforms are ready for prime time? Blood.
function, and impair hematopoietic differentiation. Cancer Cell. 2010; 2014;124:3345-3455.
18:553-567. 72. Chen Y, Cortes J, Estrov Z, et al. Persistence of cytogenetic abnor-
54. Lu C, Ward PS, Kapoor GS, et al. IDH mutation impairs histone de- malities at complete remission after induction in patients with acute
methylation and results in a block to cell differentiation. Nature. 2012; myeloid leukemia: prognostic significance and the potential role of
483:474-478. allogeneic stem-cell transplantation. J Clin Oncol. 2011;29:2507-2513.
55. Stein EM, DiNardo C, Altman JK. Safety and efficacy of AG-221, a 73. Marcucci G, Mrozek
K, Ruppert AS, et al. Abnormal cytogenetics at
potent inhibitor of mutant IDH2 that promotes differentiation of date of morphologic complete remission predicts short overall and
myeloid cells in patients with advanced hematologic malignancies: disease-free survival, and higher relapse rate in adult acute myeloid
results of a phase 1/2 trial. Blood. 2015;126:323. leukemia: results from cancer and leukemia group B study 8461. J Clin
56. DiNardo C, de Botton S, Pollyea DA. Molecular profiling and re- Oncol. 2004;22:2410-2418.
lationship with clinical response in patients with IDH1 mutation- 74. San-Miguel JF, Vidriales MB, Orf~ao A. Immunological evaluation of
positive hematologic malignancies receiving AG-120, a first-in-class minimal residual disease (MRD) in acute myeloid leukaemia (AML).
potent inhibitor of mutant IDH1, in addition to data from the com- Best Pract Res Clin Haematol. 2002;15:105-118.
pleted dose escalation portion of the phase 1 study. Blood. 2015;126: 75. Kern W, Haferlach C, Haferlach T, et al. Monitoring of minimal residual
1306. disease in acute myeloid leukemia. Cancer. 2008;112:4-16.
57. Konopleva M, Pollyea DA, Potluri J, et al. A phase 2 study of ABT-199 76. Schnittger S, Weisser M, Schoch C, et al. New score predicting for
(GDC-0199) in patients with acute myelogenous leukemia. Blood. prognosis in PML-RARA+, AML1-ETO+, or CBFBMYH11+ acute myeloid
2014;124:118. leukemia based on quantification of fusion transcripts. Blood. 2003;
58. DiNardo C, Pollyea D, Pratz K. Phase 1b study of venetoclax (ABT-
102:2746-2755.
199/GDC-0199) in combination with decitabine or azacitidine in
77. Grimwade D, Jovanovic JV, Hills RK, et al. Prospective minimal residual
treatment-naive patients with acute myelogenous leukemia who
disease monitoring to predict relapse of acute promyelocytic leu-
are $ to 65 years and not eligible for standard induction therapy.
kemia and to direct pre-emptive arsenic trioxide therapy. J Clin Oncol.
Blood. 2015;126:327.
2009;27:3650-3658.
59. Stein AS, Walter RB, Erba HP, et al. A phase 1 trial of SGN-CD33A as
78. Schnittger S, Schoch C, Dugas M, et al. Analysis of FLT3 length mu-
monotherapy in patients with CD33-positive acute myeloid leukemia
tations in 1003 patients with acute myeloid leukemia: correlation to
(AML). Blood. 2015;126:324.
cytogenetics, FAB subtype, and prognosis in the AMLCG study and
60. Fathi AT, Erba HP, Lancet JE, et al. A novel well-tolerated regimen with
usefulness as a marker for the detection of minimal residual disease.
high remission rate in frontline unfit AML. Blood. 2015;126:454.
Blood. 2002;100:59-66.
61. Dohner H, Weisdorf DJ, Bloomfield CD. Acute myeloid leukemia.
79. Kronke J, Schlenk RF, Jensen KO, et al. Monitoring of minimal residual
N Engl J Med. 2015;373:1136-1152.
disease in NPM1-mutated acute myeloid leukemia: a study from the
62. Dohner H, Estey EH, Amadori S, et al; European LeukemiaNet. Di-
German-Austrian acute myeloid leukemia study group. J Clin Oncol.
agnosis and management of acute myeloid leukemia in adults: rec-
2011;29:2709-2716.
ommendations from an international expert panel, on behalf of the
80. Debarri H, Lebon D, Roumier C, et al. IDH1/2 but not DNMT3A mu-
European LeukemiaNet. Blood. 2010;115:453-474.
63. Grimwade D, Hills RK, Moorman AV, et al; National Cancer Research tations are suitable targets for minimal residual disease monitoring in
Institute Adult Leukaemia Working Group. Refinement of cytogenetic acute myeloid leukemia patients: a study by the Acute Leukemia
classification in acute myeloid leukemia: determination of prognostic French Association. Oncotarget. 2015;6:42345-42353.
significance of rare recurring chromosomal abnormalities among 5876 81. Cilloni D, Renneville A, Hermitte F, et al. Real-time quantitative po-
younger adult patients treated in the United Kingdom Medical Re- lymerase chain reaction detection of minimal residual disease by
search Council trials. Blood. 2010;116:354-365. standardized WT1 assay to enhance risk stratification in acute myeloid
64. Grimwade D, Ivey A, Huntly BJ. Molecular landscape of acute myeloid leukemia: a European LeukemiaNet study. J Clin Oncol. 2009;27:
leukemia in younger adults and its clinical relevance. Blood. 2016;127: 5195-5201.
29-41. 82. Pastore F, Levine RL. Next-generation sequencing and detection of
65. Ravandi F. Primary refractory acute myeloid leukaemia - in search of minimal residual disease in acute myeloid leukemia: ready for clinical
better definitions and therapies. Br J Haematol. 2011;155:413-419. practice? JAMA. 2015;314:778-780.
66. Stone RM. Is it time to revisit standard post-remission therapy? Best 83. Inaba H, Coustan-Smith E, Cao X, et al. Comparative analysis of dif-
Pract Res Clin Haematol. 2012;25:437-441. ferent approaches to measure treatment response in acute myeloid
67. Hourigan CS, Karp JE. Minimal residual disease in acute myeloid leukemia. J Clin Oncol. 2012;30:3625-3632.
leukaemia. Nat Rev Clin Oncol. 2013;10:460-471. 84. Chen X, Xie H, Wood BL, et al. Relation of clinical response and minimal
68. Ravandi F, Jorgensen JL. Monitoring minimal residual disease in acute residual disease and their prognostic impact on outcome in acute
myeloid leukemia: ready for prime time? J Natl Compr Canc Netw. myeloid leukemia. J Clin Oncol. 2015;33:1258-1264.
2012;10:1029-1036. 85. Mrozek
K, Marcucci G, Paschka P, et al. Clinical relevance of mutations
69. Klco JM, Miller CA, Griffith M, et al. Association between mutation and gene-expression changes in adult acute myeloid leukemia with
clearance after induction therapy and outcomes in acute myeloid normal cytogenetics: are we ready for a prognostically prioritized
leukemia. JAMA. 2015;314:811-822. molecular classification? Blood. 2007;109:431-448.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e311


DEANGELO ET AL

86. Patel JP, Gonen M, Figueroa ME, et al. Prognostic relevance of in- 96. Paietta E. Minimal residual disease in acute myeloid leukemia: coming
tegrated genetic profiling in acute myeloid leukemia. N Engl J Med. of age. Hematology Am Soc Hematol Educ Program. 2012;2012:35-42.
2012;366:1079-1089. 97. San Miguel JF, Martnez A, Macedo A, et al. Immunophenotyping in-
87. Schlenk RF, Dohner K, Krauter J, et al; German-Austrian Acute Myeloid vestigation of minimal residual disease is a useful approach for pre-
Leukemia Study Group. Mutations and treatment outcome in cyto- dicting relapse in acute myeloid leukemia patients. Blood. 1997;90:
genetically normal acute myeloid leukemia. N Engl J Med. 2008;358: 2465-2470.
1909-1918. 98. Venditti A, Buccisano F, Del Poeta G, et al. Level of minimal residual
88. Nazha A, Cortes J, Faderl S, et al. Activating internal tandem dupli- disease after consolidation therapy predicts outcome in acute myeloid
cation mutations of the fms-like tyrosine kinase-3 (FLT3-ITD) at leukemia. Blood. 2000;96:3948-3952.
complete response and relapse in patients with acute myeloid leu- 99. Rubnitz JE, Inaba H, Dahl G, et al. Minimal residual disease-directed
kemia. Haematologica. 2012;97:1242-1245. therapy for childhood acute myeloid leukaemia: results of the AML02
89. Schnittger S, Kern W, Tschulik C, et al. Minimal residual disease levels multicentre trial. Lancet Oncol. 2010;11:543-552.
assessed by NPM1 mutation-specific RQ-PCR provide important 100. Langebrake C, Creutzig U, Dworzak M, et al; MRD-AML-BFM Study
Group. Residual disease monitoring in childhood acute myeloid
prognostic information in AML. Blood. 2009;114:2220-2231.
leukemia by multiparameter flow cytometry: the MRD-AML-BFM
90. Shayegi N, Kramer M, Bornhauser M, et al; Study Alliance Leukemia
Study Group. J Clin Oncol. 2006;24:3686-3692.
(SAL). The level of residual disease based on mutant NPM1 is an
101. Terwijn M, van Putten WL, Kelder A, et al. High prognostic impact of
independent prognostic factor for relapse and survival in AML. Blood.
flow cytometric minimal residual disease detection in acute myeloid
2013;122:83-92.
leukemia: data from the HOVON/SAKK AML 42A study. J Clin Oncol.
91. Ivey A, Hills RK, Simpson MA, et al; UK National Cancer Research
2013;31:3889-3897.
Institute AML Working Group. Assessment of minimal residual disease
102. Freeman SD, Virgo P, Couzens S, et al. Prognostic relevance of
in standard-risk AML. N Engl J Med. 2016;374:422-433.
treatment response measured by flow cytometric residual disease
92. Corbacioglu A, Scholl C, Schlenk RF, et al. Prognostic impact of minimal
detection in older patients with acute myeloid leukemia. J Clin Oncol.
residual disease in CBFB-MYH11-positive acute myeloid leukemia.
2013;31:4123-4131.
J Clin Oncol. 2010;28:3724-3729. 103. Walter RB, Gooley TA, Wood BL, et al. Impact of pretransplantation
93. Krauter J, Gorlich K, Ottmann O, et al. Prognostic value of minimal minimal residual disease, as detected by multiparametric flow
residual disease quantification by real-time reverse transcriptase cytometry, on outcome of myeloablative hematopoietic cell trans-
polymerase chain reaction in patients with core binding factor leu- plantation for acute myeloid leukemia. J Clin Oncol. 2011;29:
kemias. J Clin Oncol. 2003;21:4413-4422. 1190-1197.
94. Yin JA, OBrien MA, Hills RK, et al. Minimal residual disease monitoring 104. Walter RB, Buckley SA, Pagel JM, et al. Significance of minimal residual
by quantitative RT-PCR in core binding factor AML allows risk strat- disease before myeloablative allogeneic hematopoietic cell trans-
ification and predicts relapse: results of the United Kingdom MRC plantation for AML in first and second complete remission. Blood.
AML-15 trial. Blood. 2012;120:2826-2835. 2013;122:1813-1821.
95. Zhu HH, Zhang XH, Qin YZ, et al. MRD-directed risk stratification 105. Araki D, Wood BL, Othus M, et al. Allogeneic hematopoietic cell
treatment may improve outcomes of t(8;21) AML in the first complete transplantation for acute myeloid leukemia: time to move toward a
remission: results from the AML05 multicenter trial. Blood. 2013;121: minimal residual disease-based definition of complete remission?
4056-4062. J Clin Oncol. 2016;34:329-336.

e312 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


HEMATOLOGIC MALIGNANCIESLEUKEMIA,
MYELODYSPLASTIC SYNDROMES, AND
ALLOTRANSPLANT

Molecularly and Phenotypically


Defined Subtypes of Acute
Lymphoblastic Leukemia:
Implications for Management

CHAIR
Charles G. Mullighan, MD, MBBS, MSc
St. Jude Childrens Research Hospital
Memphis, TN

SPEAKERS
Sabina Chiaretti, MD, PhD
Sapienza University of Rome
Rome, Italy

Susan M. OBrien, MD
University of California, Irvine
Irvine, CA
CHIARETTI ET AL

Advances in the Genetics and Therapy of Acute Lymphoblastic


Leukemia
Sabina Chiaretti, MD, PhD, Valentina Gianfelici, MD, PhD, Susan M. OBrien, MD, and
Charles G. Mullighan, MBBS, MD

OVERVIEW

Acute lymphoblastic leukemia (ALL) remains an important cause of morbidity in children and adults. In this article, we
highlight advances in the genetics and therapy of three key subtypes of ALL: T-cell ALL, BCR-ABL1 (Philadelphia [Ph]
chromosonepositive), and Ph-like ALL. T-ALL is an aggressive disease that accounts for about 15% and 25% of ALL among
pediatric and adult cohorts, respectively, and exhibits a multistep nature of cancer initiation and progression. The integration
of cytogenetics, molecular biology, and immunophenotype analyses has led to the identification of defined T-ALL subgroups,
such as early T-cell precursor ALL and novel lesions with a prognostic role, for which specific inhibitors are being developed.
Phpositive ALL was historically regarded as a subtype of ALL with a poor prognosis, and allogeneic stem cell transplant was
recommended for all patients who could undergo this procedure. The deep complete responses seen with combination
tyrosine kinase inhibitors (TKIs) and chemotherapy in Ph-positive ALL, and the reports of long-term survival among some
patients not undergoing allogeneic stem cell transplant, has raised the question of whether there is a subset of patients who
could be cured without this intervention. Ph-like ALL is a subtype of B-progenitor ALL common among older children and
adults and associated with a diverse range of genetic alterations that activate kinase signaling. Ph-like ALL is also associated
with poor outcome, for which precision medicine trials identifying kinase alterations and testing TKI therapy are being
developed.

T -cell ALL (T-ALL) is a genetically heterogeneous disease


that is caused by the accumulation of lesions acting in a
multistep pathogenic process involving cell growth, pro-
chromosomal translocations, duplications and deletions of
DNA, deregulated gene expression, and mutations.
The most frequent translocations involve the 14q11 (T-cell
liferation, survival, and differentiation during thymocyte receptor alpha and delta, TRA and TRD) and 7q34 (TRB) re-
development.1 It accounts for about 15% and 25% of ALL gions, juxtaposing the T-cell receptor (TCR) genes to pivotal
among pediatric and adult cohorts, respectively, and is transcription factor genes, such as TAL1, TAL2, LYL1, OLIG2
slightly more frequent in males than females, particularly (BHLHB1), LMO1, LMO2, TLX1, TLX3, NKX2-1, NKX2-2, NKX2-5,
in older children and adolescents.2 As for other ALL sub- HOXA genes, MYC, and MYB. Further improvement of cyto-
types, prognosis is far superior for children than adults; genetic analysis and the advent of NGS will likely result in
however, it must be emphasized that the outcome of identification of additional partners of rearrangement to TCR
these patients is now similar to that of patients with genes. Chromosomal rearrangements can also lead to fusion
B-cell ALL (B-ALL) because of the use of more intensive genes, among which the most important from a clinical
treatments. standpoint are represented by those involving ABL1, mostly
Until recently, there was a limited understanding of the represented by NUP214-ABL1, EML1-ABL1, and ETV6-ABL1
genetic basis of T-ALL. The improvement of cytogenetic rearrangement, for which the addition of TKIs may be of
assays and integration with techniques including gene ex- benefit, as well as those involving KMT2A (MLL; MLL-
pression profiling, single nucleotide polymorphism micro- rearranged). Finally, DNA copy number alterations are iden-
array analysis, and next-generation sequencing (NGS) have tified including deletion of CDKN2A/B and duplication of MYB.3
enabled detailed characterization of the genomic complexity Gene expression profiling studies have refined the char-
of T-ALL. The most frequent lesions can be categorized as acterization of molecular groups of T-ALL, which are reflective

From the Department of Cellular Biotechnologies and Hematology, Sapienza University of Rome, Rome, Italy; Chao Family Comprehensive Cancer Center, School of Medicine, University
of California, Irvine, CA; Department of Pathology, St. Jude Childrens Research Hospital, Memphis, TN.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Charles G. Mullighan, MBBS, MD, St. Jude Childrens Research Hospital, 252 Danny Thomas Place, MS 342, Memphis, TN 38105; email: charles.mullighan@
stjude.org.

2016 by American Society of Clinical Oncology.

e314 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


GENETICS AND THERAPY OF ACUTE LYMPHOLASTIC LEUKEMIA

of a maturational arrest and are characterized by the over- been described in rare cases. Moreover, NRAS/KRAS mu-
expression of known transcription factors (TLX1, TLX3, TAL1, tations are found in around 10% of adults and 4% to 10% of
LMO1, HOXA genes, NKX genes, LYL1, and MEF2C) and, more children.10-14 Finally, interleukin (IL)-4 and IL-7 may upre-
importantly, to identify novel subgroups (early T-cell pre- gulate PI3K/AKT/mTOR signaling in T-ALL.15 The prognostic
cursor ALL, see below).4-7 role of PTEN and NRAS/KRAS alterations is still under in-
The most frequent mutations are those in NOTCH1 and vestigation and may differ between children and adults.
FBXW7, in genes involved in the JAK/STAT and Ras/PI3K/AKT Among children, the presence of inactivating PTEN lesions,
pathways, in epigenetic regulators (PHF6, SUZ12, EZH2, as well as NRAS/KRAS mutations, do not affect outcome,11,14
TET2, H3F3A, and KDM6A), in transcription factors or reg- whereas among adults their presence is associated with
ulators of transcription (i.e., LEF1, WT1, BCL11B) and genes poorer prognosis.10,12
involved in mRNA maturation and ribosome activity The JAK/STAT pathway is also often aberrantly activated in
(i.e., CNOT3, RPL5, and RPL10). Furthermore, at relapse, T-ALL. Activating mutations are found mainly in IL7R, JAK1,
mutations of NT5C2 known to confer resistance to che- JAK3, and STAT5B; moreover, inactivating mutations and
motherapy with 6-mercaptopurine and 6-thioguanine are deletions have been described in PTPRC and PTPN2, which
common.8,9 encode phosphatases that regulate the activation of JAK1.16,17
NOTCH1 signaling is critical for T-cell differentiation, and The prognostic role of these lesions is still debated; JAK1
mutations in this gene are present in up to 60% of T-ALL mutations have been associated with chemotherapy refrac-
cases. In the same pathway, mutations in FBXW7, which toriness but with contrasting results. Similarly, the prognos-
increase NOTCH1 stability, can be detected in about 20% of tic role of JAK3 mutations needs to be more extensively
cases. Although a favorable outcome has been overall re- assessed.18-21 Finally, STAT5B mutations have been correlated
ported for NOTCH1 mutations, the scenario is more complex with an increased risk of recurrence in pediatric cases.22,23
if concomitant lesions are identified. Indeed, a comprehen- Given this complex genomic scenario, several targeted
sive prognostic model has been recently proposed by the approaches have been developed and might be used in the
GRAAL group10 that defined low-risk patients as those har- context of a personalized medicine.
boring NOTCH1 and FBXW7 mutations and high-risk patients NOTCH1 inhibitors represent an attractive therapeutic
as those without these mutations or harboring lesions in- target, in light of the high incidence of mutations. g-secretase
volving Ras/PTEN. inhibitors (GSI), in use for patients with Alzheimer dis-
Similarly, the PI3K/PTEN/AKT/mTOR pathway is critical for ease, were the first compounds developed for T-ALL.
cell metabolism, proliferation, and survival. Several mech- However, their side effects, particularly at the gastro-
anisms can lead to its deregulation: mutations or deletions in intestinal level, made their use not feasible24; these effects
PTEN have been reported in around 12% of adults and 13% might be overcome by the concomitant administration of
to 22% of children, whereas PI3K and AKT mutations have dexamethasone.25
Similarly, PI3K/PTEN/AKT/mTOR inhibitors are appealing
agents. Among them, rapamycin has been extensively in-
vestigated. Although its use as a single agent can be limited
KEY POINTS by the prolonged inhibition of TORC1 and activation of al-
ternative pathways, a synergistic effect between rapamycin
T-cell acute lymphoblastic leukemia is an aggressive (or its derivatives) and various chemotherapeutic agents
disease that accounts for about 15% and 25% of ALL (i.e., idarubicin, doxorubicin, and dexamethasone) have
among pediatric and adult cohorts, respectively.
been described.26 Therefore, several clinical trials are cur-
Recent scientific advances have allowed identification of
T-ALL subgroups such as early T-cell precursor ALL and
rently evaluating the effectiveness of the combination of
novel lesions with a prognostic role. rapamycin derivatives with chemotherapy among pediatric
Ph-positive ALL was historically regarded as a subtype of patients with relapsed ALL. In particular, in the phase I
ALL with a poor prognosis, and allogeneic stem cell RAD001 study, everolimus is combined with reinduction
transplant was recommended for all patients who could chemotherapy for pediatric patients with relapsed ALL
undergo this procedure. (NCT01523977). Furthermore, a second phase I study
The deep complete responses seen with combination designed to assess the effectiveness of CCI-779 (temsirolimus)
tyrosine kinase inhibitors and chemotherapy in Ph- among children whose disease has relapsed has recently
positive ALL, and the reports of long-term survival in concluded, with results not published (NCT01403415).
some patients not undergoing allogeneic stem cell The novel dual PI3K/mTOR inhibitor NVP-BEZ235 is an
transplant, has raised the question of whether there is a
orally bioavailable imidazoquinoline derivative, which ex-
subset of patients who could be cured without this
intervention.
erts antiproliferative and proapoptotic effects in several
Ph-like ALL, common among older children and adults, is T-ALL cell lines.27 Similarly, NVP-BKM120 is a proapoptotic
associated with poor outcome and is characterized by pan-PI3K inhibitor. Currently, two ongoing clinical trials are
a range of genetic alterations activating kinase signaling evaluating the effectiveness of BEZ235 and NVP-BKM120
that are sensitive to TKI therapy. among patients with relapsed or refractory acute leukemia
(NCT01756118 and NCT01396499, respectively).

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e315


CHIARETTI ET AL

A variety of JAK inhibitors have been developed. The early T-cell precursorlike immunophenotype. Strikingly, the
JAK1/JAK2-selective inhibitor ruxolitinib is already approved presence of an early T-cell precursorlike immunophenotype
by the U.S. Food and Drug Administration (FDA) for the conferred marked differences in outcome within the HOXA-
treatment of patients with myelofibrosis, and the JAK3- positive group, which were mirrored by corresponding in-
selective inhibitor tofacitinib received FDA approval for creases in cumulative incidence of relapse. In contrast, these
the treatment of patients with rheumatoid arthritis. These survival differences were not seen among patients with
data demonstrate that JAK kinase inhibitors can be ad- HOXA-negative disease, where early T-cell precursor and non-
ministered safely and open new possibilities for the treat- early T-cell precursor cases had similar 5-year event-free
ment of T-ALL with IL7R, JAK1, or JAK3 mutations. In vitro survival (EFS) and cumulative incidence of relapse.31 Simi-
studies have shown the efficacy of ruxolitinib in reducing the larly, Matlawska-Wasowska analyzed a cohort of pediatric
proliferation of cells that express mutations in JAK1, IL7R, and young adults and found overlap between MLL-rearranged
and PTPRC.16 Furthermore, Degryse et al showed that and the early T-cell precursor ALL cases, and confirmed that
treatment with the JAK3-selective inhibitor tofacitinib re- both are risk features for induction failure.32
duced the white blood cell count and caused leukemic cell Overall, these findings lead to some important consider-
apoptosis in leukemic mice that expressed JAK3 mutant ations. The prompt recognition of early T-cell precursor
variants.28 cases is improving outcome because patients are rapidly
Finally, BCL-2 antagonists, and in particular ABT-199, directed toward intensive treatments, including allogeneic
which have proven effective in chronic lymphocytic leuke- transplant during the first complete response (CR). In con-
mia, might also represent a therapeutic option in T-ALL; trast, refined molecular characterization is permitting fur-
a specific effect has been demonstrated in TLX3- or HOXA- ther stratification of these cases and recognition of very
positive primary T-ALL and in more immature T-ALL,29 as well high-risk cases. Finally, the presence of acute myeloid
as among patients who harbor STAT5B mutations.22 leukemiarelated features prompts investigation of the
Lastly, the concomitance of different alterations suggests use of myeloid-directed therapies.
that the combination of specific inhibitors that target dif- In conclusion, extensive research has refined the genomic
ferent pathways might prove useful for these patients and profile of T-ALL. In fact, improvement in cytogenetic and mo-
minimize the risk of resistance to treatment with single lecular analysis and the integration with immunophenotype
agents. have led to the identification of different subgroups and
novel lesions that result in deregulated pathways. This effort
paves the way toward a personalized approach that can
EARLY T-CELL PRECURSOR ACUTE include targeted therapies, depending on the underlying
LYMPHOBLASTIC LEUKEMIA molecular lesion(s), and possibly should include a combined
A distinct subgroup of T-ALL is represented by early T-cell approach with conventional therapy and personalized tar-
precursor ALL.6 This subset, initially identified by gene ex- geted approaches.
pression profiling, can be easily recognized by flow
cytometry because it is characterized by distinct cell surface
features: absence of CD1a and CD8, weak CD5 expression, PHILADELPHIA CHROMOSOMEPOSITIVE ALL:
and the expression of one or more myeloid or stem cell TRANSPLANT OR NO TRANSPLANT
associated markers. Several genomic lesions have been Philadelphia chromosomepositive ALL was historically
identified, including a low frequency of NOTCH1 mutations, considered one of the most high risk forms of ALL. Although
and mutations in DNMT3A, FLT3, IDH1, IDH2, and ETV6 have CR rates with induction chemotherapy were reasonable,
been reported. Interestingly, FLT3 mutations can be de- relapse was almost inevitable, and a key strategy was to get
tected in up to 35% of cases, thus implying the possibility of patients to undergo allogeneic stem cell transplant (SCT),
novel therapeutic strategies.30 Furthermore, mutations which was considered the only potentially curative option.
occurring in genes that regulate cytokine receptor and Ras Given the age of patients with ALL, in particular Ph-positive
signaling (67%), inactivating lesions that disrupt hemato- ALL (which increases with each decade of life), many patients
poietic development (58%), and histone-modifying genes were unable to be undergo this potentially curative therapy.33
(48%) have been reported, suggesting that early T-cell Approximately 30% of patients were able to receive a SCT in
precursor ALL shares a similar genomic background with the past; in addition to older age, other contraindications to
acute myeloid leukemia. SCT included lack of remission, early relapse, or poor per-
From a clinical standpoint, early T-cell precursor ALL was formance status from complications due to chemotherapy.34
initially associated with a dismal outcome because of its poor Thus, long-term survival rates were quite poor.
response to chemotherapy and high rate of resistance or The advent of TKI therapy has dramatically improved
early relapse.6 Notably, recent studies have reported poor outcomes for patients with Ph-positive ALL. Complete re-
outcomes in cases with genomic rearrangements associated sponses are substantially deeper than those achieved with
with HOXA deregulation. Bond et al analyzed patients en- chemotherapy alone, as reflected in the high rate of major
rolled in the GRAAL-2003 and -2005 studies and showed that molecular response (MMR) or complete molecular response
patients with HOXA-positive disease had higher rates of an (CMR) after a combination of TKI and chemotherapy.35 The

e316 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


GENETICS AND THERAPY OF ACUTE LYMPHOLASTIC LEUKEMIA

fact that CMR can occur has raised the question of whether 33% for patients without a SCT (nine patients), and 80% after
some patients can be cured without undergoing SCT. Small autologous SCT (10 patients). The favorable outcomes with
cohorts of patients who were not candidates for SCT and autologous SCT may also have been related to the re-
were treated with chemotherapy and TKIs have provided quirement that patients have a marrow MRD ratio of less
some information on long-term outcomes without SCT. than 10e-4. Seven patients had a CMR. The three patients
No randomized trials have addressed this issue, and who did not achieve a CMR experienced disease relapse
all of the published clinical trials regarding outcomes in after SCT. Imatinib was given to four patients after SCT; three
Ph-positive ALL include variable transplant approaches were alive in first CR at 37, 41, and 46 months. Treatment-
(myeloablative versus reduced-intensity conditioning) and related mortality was particularly high for patients who
choice, dose, and duration of TKI. Post-transplant mainte- underwent a matched unrelated donor SCT, and the authors
nance is an additional variable. Most data involve a com- noted that whether autologous SCT with a low or negative
bination of imatinib and chemotherapy because imatinib MRD level should be performed instead of allogeneic SCT
was the first available TKI. There are also some data that using an HLA-matched unrelated donor in patients with
autologous SCT may be beneficial in this setting. Autologous Ph-positive ALL is a critical issue that should further be
SCT has not been shown to be an improvement over che- addressed through a prospective controlled study.
motherapy in the overall treatment of ALL, but one of the Another trial that utilized MRD status to guide choice of
presumed reasons is the lack of true minimal residual dis- therapy was recently published. The GRAALL group ran-
ease (MRD) among patients prior to undergoing autologous domly assigned 268 patients with Ph-positive ALL to 800 mg
SCT. With a TKI and chemotherapy combination, this low of imatinib therapy with either reduced-intensity chemo-
level or absence of disease is possible. therapy (arm A) or hyperfractionated cyclophosphamide plus
Bassan describes the northern Italy experience with 94 pa- vincristine, doxorubicin, and dexamethasone (hyperCVAD)
tients with Ph-positive ALL who received 600 mg of imatinib therapy (arm B).40 Patients then underwent allogeneic SCT
daily with chemotherapy; 31 patients did not receive ima- (myeloablative, later amended to allow reduced-intensity
tinib and constituted a control cohort.36 Patients were to conditioning) if they had a related or 9/10 or 10/10 HLA-
undergo myeloablative allogeneic SCT (if related or unre- compatible unrelated donor. Other patients who experi-
lated donor was available) or autologous SCT followed by enced MMR after two cycles (MMR2) were to undergo
imatinib maintenance. Nine patients, including four who had autologous SCT. Thirty-nine patients were eligible for au-
not received imatinib, underwent autologous SCT and fur- tologous SCT and 28 of those patients underwent SCT during
ther imatinib therapy, and their outcomes were similar to first remission. Among patients who achieved an MMR2, the
patients undergoing allogeneic SCT. A multivariate model outcome was similar for autologous and allogeneic SCT.
suggested that imatinib and SCT favorably affected overall Allogeneic SCT was as effective for patients who did not
survival (OS), but SCT included patients undergoing autol- experience early MMR as for those who did. Of note, pa-
ogous SCT. A confounding element to the data is that tients who underwent autologous SCT received mainte-
imatinib and/or dasatinib was administered to patients with nance with a TKI, which was not planned for the allogeneic
persistent or increasing partial CR after SCT. SCT cohort. This again speaks to the question of the benefit
The PETHEMA and GETH groups37 reported results from of autologous SCT compared with just continued TKI ther-
30 patients who received 400 mg of imatinib daily and che- apy. The authors concluded that the data strongly sug-
motherapy. Sixteen patients underwent allogeneic (primarily gests that favorable patients with low white blood cell
myeloablative) or autologous SCT. Interestingly, of the five count and/or those with good early MRD response could be
patients who underwent autologous SCT, four were MRD treated with nonallogeneic postremission therapies.
negative. The only patient who was MRD positive experienced The UKALLXII/ECOG 299 is the largest prospective trial that
relapse; none of the other four patients relapsed. Of the assessed the role of imatinib in the treatment of Ph-positive
patients who received imatinib maintenance, two were alive ALL.41 The analysis compared outcomes of a large group of
at 40 and 60 months into CR. In a Saudi Arabian analysis of Ph- patients treated between 1993 and 2003 (pre-imatinib) with
positive pediatric ALL, SCT was limited to only children with patients who received early or late imatinib (480 to 600 mg).
related donors.38 Twelve patients underwent allogeneic SCT Both the CR and OS rates were better in the imatinib cohort.
and 10 were treated with chemotherapy and imatinib. After a The improvement in OS was derived partially from the use of
median follow-up of 42.2 months, there was no statistical imatinib that enabled a higher percentage of patients to
difference in EFS or OS. Interestingly, two children who re- undergo allogeneic SCT. There was also a modest benefit to
lapsed after SCT responded to TKI and were still in CR. imatinib in terms of EFS. Again, a small number of five
Long-term follow-up of the GRAAPH-2003 trial was re- patients underwent autologous SCT. There was one early
cently published and involved 45 patients with Ph-positive death and one relapse. The other three patients were alive in
ALL.39 Notably, postremission therapy was partly decided by CR at 3.5, 8, and 8 years. The conclusion was that the data did
MRD results. Imatinib doses of 600 to 800 mg per day were not support the omission of allogeneic SCT from the
given with consolidation chemotherapy and improved OS to treatment of Ph-positive ALL, but that limited data on the
52% compared with 20% prior to imatinib. Overall survival use of reduced-intensity conditioning suggested that it could
was 50% after myeloablative allogeneic SCT (24 patients), be beneficial. No attempt was made to allocate patients

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e317


CHIARETTI ET AL

depending on their MRD status, and there was no analysis of therapy. The general thinking is that long-term survival is not
the relationship of MRD status to outcome. possible without achieving MRD negativity. Thus, most
The issue of the need for allogeneic SCT is also complicated physicians would still recommend SCT for patients who did
by the fact that most trials did not use TKI maintenance after not achieve this level of MRD. However, the question of
SCT. Recent data suggest that TKI maintenance after allo- whether MRD status can be used to select patients for less-
genic SCT can markedly improve outcomes. Pfeifer et al intensive therapy has not yet been answered in a ran-
performed a randomized trial of prophylactic or MRD- domized clinical trial. Of note, the upcoming Intergroup trial
triggered imatinib after allogeneic SCT.42 Five-year sur- of patients with Ph-positive ALL will randomly assign pa-
vival was high in both groups and substantially higher than in tients to SCT or no SCT (for patients who are MRD-negative).
their previous study in which the interval between SCT and This trial will provide important information about selecting
imatinib was 5 months compared with less than 2.5 months. patients who may experience long-term survival without
Early occurrence of MRD positivity or positivity at higher undergoing SCT.
levels identified a subset of patients who did not benefit
from imatinib; whether second-generation TKIs would
produce a better outcome in this group is not known. PH-LIKE ACUTE LYMPHOBLASTIC LEUKEMIA:
In addition to using MRD as a potential decision-making GENETICS, DIAGNOSIS, AND MANAGEMENT
tool for the use of SCT, another question is: are there Ph-like ALL is a recently described subtype of B-ALL
prognostic factors beyond the presence of Ph positivity that characterized by a gene expression profile similar to that
further define subgroups? Mullighan et al published data of BCR-ABL1 positive (Ph-positive) ALL, a diverse range of
that showed a high percentage of children with Ph-positive genetic alterations activating tyrosine kinase signaling,
ALL had a deletion in the Ikaros (IKZF1) gene.43 Kim et al mutation of lymphoid transcription factor genes such as
investigated its relevance among 118 adult patients with IKZF1 (Ikaros), and poor outcome. The high frequency of
Ph-positive ALL who received imatinib-based chemotherapy kinase activating alterations in this form of ALL and the
and underwent allogeneic SCT (myeloablative or reduced- potential for targeting these lesions with currently avail-
intensity conditioning depending on age).44 IKZF1 deletions able TKIs have resulted in intense interest in implementing
were detected among 78.8% of patients and such patients diagnosis and testing of Ph-like ALL and underlying genetic
had a trend for a higher rate of relapse. However, in mul- alterations in prospective clinical trials.
tivariate analysis, the MRD kinetics were most closely re- In 2009, two groups identified childhood ALL cases in which
lated to outcomes, whereas neither IKZF1 deletions nor their the gene expression profile of leukemic cells lacking a known
functional subtypes were independently important. chromosomal rearrangement was similar to that of Ph-positive
Dose intensity of imatinib and the effect on outcomes has ALL.46,47 The Childrens Oncology Group identified a group of
not been well-explored in the setting of Ph-positive ALL. Lim high-risk BCR-ABL1negative B-ALL cases with a high fre-
et al intended to administer 600 mg per day of imatinib to all quency of IKZF1 alteration (which is also a hallmark of
patients starting from day 8 of induction chemotherapy Ph-positive ALL)43 and poor outcome that exhibited simi-
through remission induction and consolidation chemo- larity of the transcriptomic signature of these cases to that
therapy.45 For patients not proceeding to SCT, a further of Ph-positive ALL using gene set enrichment analysis. The
2 years of imatinib was planned. Lim et al showed that dose Dutch used hierarchical clustering of gene expression
intensity during the initial 7 to 8 weeks strongly correlated profiling data to show clustering of Ph-positive with
with median CR duration as well as OS. The most common Ph-negative cases.47 As there is no single founding genomic
reason for less dose intensity was gastrointestinal toxicity. alteration (such as BCR-ABL1) common to all cases, and as
This suggests the choice of chemotherapy in conjunction identification rests on demonstrating similarity with the
with imatinib might also be important. expression profile of Ph-like ALL, the frequency of Ph-like ALL
In summary, the advent of TKIs has dramatically improved varies between studies and the gene expression classifier
the prognosis of patients with Ph-positive ALL. The goal of used.48 However, multiple studies have shown that Ph-like
treatment is still to deliver allogeneic SCT when possible. ALL is common, particularly with increasing age, and it
However, long-term data from cohorts of patients who could is associated with poor outcome and commonly harbors
not undergo SCT suggest that a cure faction is possible even kinase-activating lesions.
without this intervention. The key question is, how can we A study of over 1,700 childhood, adolescent (age 16 to 21),
identify patients who may enjoy long-term survival without and young adult (age 22 to 39) ALL cases performed by the
the risks of SCT? Examining data across trials is complicated Childrens Oncology Group, the St. JudeWashington Uni-
by the fact that although most trials have used imatinib, the versity Pediatric Cancer Genome Project, and institutions
doses have varied substantially and very little data using studying adults with ALL defined the prevalence and genetic
second-generation TKIs exist. In addition, autologous basis of ALL.49 Using gene expression microarrays to define
transplant has been used in a minority of patients, some of the signature of Ph-positive ALL and statistical prediction,
whom have had long-term survival but most of whom also Ph-like ALL was shown to be present in approximately 10% of
received imatinib maintenance, raising the question of the standard-risk childhood ALL cases, with prevalance in-
contribution of the autologous SCT versus continued TKI creasing by age to over 25% of cases among young adults.49

e318 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


GENETICS AND THERAPY OF ACUTE LYMPHOLASTIC LEUKEMIA

There are fewer data describing the prevalence of Ph-like ALL rearrangements, mutations or deletions activating JAK-STAT
in older adults. Data from the German Multicenter Study or FLT3 signaling, Ras mutations, uncommon kinase alter-
Group for Adult ALL (GMALL) suggested that the frequency ations (e.g., NTRK3, FGFR1), and, in a proportion of cases, no
of Ph-like cases peaked among adolescents with a sub- known kinase alteration. ABL1-class rearrangements include
sequent decline.50 Recent analysis of almost 700 adult ALL those involving ABL1, ABL2 (also known as ABL-related
cases from the United States, United Kingdom, and Europe gene, or ARG), CSF1R (the macrophage CSF receptor), and
identified a frequency of 26% among young adults and 20% PDGFRB.66 In each case, the kinase is the 39 (C-terminal) fusion
among adult patients over the age of 40.51 A similar prev- partner and rearranged to multiple different 59 (N-terminal)
alence among adults was reported in 127 patients enrolled in fusion partners, and the fusion preserves the tyrosine kinase
Dutch-Belgian HOVON protocols.52 The outcome of Ph-like domain, resulting in activation of signaling. This group of
ALL is poor in the majority of studies,46,47,49,52-57 particularly rearrangements is termed ABL1-class as the transforming
for adolescents and adults for whom EFS and OS rates are effects of each are abrogated by ABL1 inhibitors such as
equal or are inferior to other high-risk B-ALL subtypes such as imatinib and dasatinib. Cases with EBF1-PDGFRB rearrange-
Ph-positive and MLL-rearranged ALL.52 An exception is the ment exhibit a particularly high rate of suboptimal treatment
St. Jude Total Therapy Study XV, in which children with response, induction failure, and relapse.67 Similarly, JAK2
Ph-like ALL did not have an inferior outcome, although many rearrangements involve a highly diverse range of fusion
were reclassified as high risk due to suboptimal treatment partners (at least 20), preserve the JAK2 kinase domain,
response (as measured by levels of MRD) and subjected to remove part of or the entire inhibitory pseudokinase do-
more intensive therapy including allogeneic bone marrow main, and are particularly common with increasing age.
transplantation.58 Importantly, several children undergo- Rearrangements of EPOR (encoding the erythropoietin re-
ing bone marrow transplantation commonly had kinase- ceptor) involve at least three different immunoglobulin and
activating lesions targetable with ABL1 inhibitors such as immunoglobulin-like genes that result in deregulation of
imatinib and dasatinib. receptor expression and truncation of the cytoplasmic tail of
Multiple chromosomal rearrangements, sequence muta- the receptor, which results in stabilization of receptor ex-
tions, and structural genetic alterations activate cytokine pression and heightened JAK-STAT signaling following stim-
receptor and tyrosine kinase signaling pathways in Ph-like ulation with EPO.68
ALL. Approximately half of cases have rearrangement of Several observations indicate that these alterations acti-
CRLF2 (encoding cytokine receptorlike factor 2), either as a vate kinase signaling and are amenable to TKI therapy.
rearrangement with the immunoglobulin heavy chain locus ABL1-class and JAK fusions expressed in cell lines result in
(IGH-CRLF2) or a focal deletion upstream of CRLF2 resulting proliferation and activation of downstream signaling path-
in expression of a P2RY8-CRLF2 fusion, both of which result ways, both of which are inhibited by TKIs. Human leukemic
in deregulated expression of intact CRLF2, which may be cells also exhibit signaling activation sensitive to TKIs; TKIs
detected by flow cytometry.59-61 Less commonly, a point synergize with chemotherapeutic agents to inhibit pro-
mutation of CRLF2 is present (p.Phe232Cys) that also liferation of human Ph-like ALL cells in vivo in xenograft
results in receptor activation.62 Approximately half of CRLF2- models. Ph-like cells also exhibit activation of additional
rearranged cases have activating mutations of Janus kinases, signaling pathways such as PI3K/mTOR signaling,69 and
particularly in the pseudokinase domain of JAK2 but also in combinatorial treatment with PI3K inhibitors and BCL-2
JAK1 and JAK3. CRLF2, which forms a heterodimeric receptor inhibitors with a TKI is efficacious.70-72 Anecdotal reports
with IL-7 receptor alpha (IL7RA) for the cytokine thymic also indicate that patients with Ph-like ALL are responsive to
stromal lymphopoietin (TSLP), is a scaffold for mutant JAK2, the addition of TKI therapy.49,73-75 Multiple patients with
and the two alterations are cotransforming. CRLF2 cases poorly responsive or refractory B-ALL have been found to
lacking JAK2 mutations commonly have other mutations have Ph-like ALL by gene expression profiling and/or testing
deregulating JAK-STAT signaling, including mutations of for driving kinase rearrangements, and they have frequently
IL7R15,49 and deletions of SH2B3 (also known as LNK), which shown dramatic, complete, and durable responses with the
is a negative regulator of JAK-STAT signaling.63,64 addition of a TKI, particularly for ABL1-class fusions. Im-
Initial sequencing studies of tyrosine kinase genes failed to portantly, there are few data regarding the use of JAK in-
identify sequence mutations in Ph-like ALL apart from those hibitors in Ph-like cases with JAK-STAT activating lesions.
in Janus kinases.54 Subsequent transcriptome sequencing Existing preclinical data suggest that Ph-like cases with JAK2
(RNA-seq) of a small number of Ph-like ALL cases identified a fusions, EPOR rearrangements, and IL7R/SH2B3 alterations
diverse range of chimeric fusions involving tyrosine cases in are sensitive to type I JAK inhibitors such as ruxolitinib,
cases lacking CRLF2 rearrangement.65 Transcriptome se- particularly in combination with chemotherapy,49,65,76 but
quencing of over 150 cases, with whole-genome sequencing CRLF2-rearranged cells are less responsive yet sensitive to
of a subset, provided a detailed understanding of the genetic type II JAK inhibitors that inhibit JAK2 in its inactive
alterations driving kinase signaling in Ph-like ALL.58 Cases conformation.77
lacking CRLF2 rearrangements have alterations deregulating There is consequently great interest in identifying patients
multiple signaling pathways, including ABL1-class chro- with Ph-like ALL and defining the underlying genetic lesions
mosomal rearrangements, JAK2 rearrangements, EPOR to implement logical TKI therapy. In view of the diverse range

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e319


CHIARETTI ET AL

of kinase-activating alterations, which continues to grow, and NGS are available (e.g., Archer, Foundation Medicine79),
identification of all such lesions requires NGS-based tech- and it is likely that prospective trials will use these or agnostic
nology. The most appropriate diagnostic approach used is genome sequencing at diagnosis.
dependent on the clinical goal. For example, the majority of In view of the success of ABL1 inhibitors in the manage-
cases with ABL1-class rearrangements can be readily de- ment of Ph-positive ALL,80 the use of these agents in ABL1-
tected with fluorescence in situ hybridization for the kinase class Ph-like ALL is logical but requires testing in prospective
rearrangements, and CRLF2 overexpression arising from trials. There is limited experience regarding the safety of
rearrangement can be identified by flow cytometry. However, combining JAK inhibition with chemotherapy in ALL, and this
identification of all Ph-like cases requires gene expression will require evaluation prior to widespread clinical use. Fi-
profiling using microarray, RNA-seq, or Taqman low-density nally, there are less common kinase alterations (TYK2,
array (TLDA) approaches,78 which are informative for research NTRK3) for which drugs have not yet been tested in ALL, and
studies but less suited for real-time diagnostic testing. Several these are an active area of research and drug discovery in
commercial solutions for nucleic acid capture of kinase loci Ph-like ALL.

References
1. Van Vlierberghe P, Ferrando A. The molecular basis of T cell acute BFM-treated children with precursor T-cell acute lymphoblastic leu-
lymphoblastic leukemia. J Clin Invest. 2012;122:3398-3406. kemia. Haematologica. 2013;98:928-936.
2. Chiaretti S, Vitale A, Cazzaniga G, et al. Clinico-biological features of 15. Zenatti PP, Ribeiro D, Li W, et al. Oncogenic IL7R gain-of-function mu-
5202 patients with acute lymphoblastic leukemia enrolled in the tations in childhood T-cell acute lymphoblastic leukemia. Nat Genet.
Italian AIEOP and GIMEMA protocols and stratified in age cohorts. 2011;43:932-939.
Haematologica. 2013;98:1702-1710. 16. Porcu M, Kleppe M, Gianfelici V, et al. Mutation of the receptor tyrosine
3. Durinck K, Goossens S, Peirs S, et al. Novel biological insights in T-cell phosphatase PTPRC (CD45) in T-cell acute lymphoblastic leukemia.
acute lymphoblastic leukemia. Exp Hematol. 2015;43:625-639. Blood. 2012;119:4476-4479.
4. Ferrando AA, Neuberg DS, Staunton J, et al. Gene expression signatures 17. Kleppe M, Lahortiga I, El Chaar T, et al. Deletion of the protein tyrosine
define novel oncogenic pathways in T cell acute lymphoblastic leukemia. phosphatase gene PTPN2 in T-cell acute lymphoblastic leukemia. Nat
Cancer Cell. 2002;1:75-87. Genet. 2010;42:530-535.
5. Homminga I, Pieters R, Langerak AW, et al. Integrated transcript and 18. Flex E, Petrangeli V, Stella L, et al. Somatically acquired JAK1 mutations
genome analyses reveal NKX2-1 and MEF2C as potential oncogenes in in adult acute lymphoblastic leukemia. J Exp Med. 2008;205:751-758.
T cell acute lymphoblastic leukemia. Cancer Cell. 2011;19:484-497. 19. Jeong EG, Kim MS, Nam HK, et al. Somatic mutations of JAK1 and JAK3 in
6. Coustan-Smith E, Mullighan CG, Onciu M, et al. Early T-cell precursor acute leukemias and solid cancers. Clin Cancer Res. 2008;14:3716-3721.
leukaemia: a subtype of very high-risk acute lymphoblastic leukaemia. 20. Asnafi V, Le Noir S, Lhermitte L, et al. JAK1 mutations are not frequent
Lancet Oncol. 2009;10:147-156. events in adult T-ALL: a GRAALL study. Br J Haematol. 2010;148:
7. Chiaretti S, Messina M, Tavolaro S, et al. Gene expression profiling iden- 178-179.
tifies a subset of adult T-cell acute lymphoblastic leukemia with myeloid- 21. Vicente C, Schwab C, Broux M, et al. Targeted sequencing identifies
like gene features and over-expression of miR-223. Haematologica. 2010; associations between IL7R-JAK mutations and epigenetic modulators
95:1114-1121. in T-cell acute lymphoblastic leukemia. Haematologica. 2015;100:
8. Tzoneva G, Perez-Garcia A, Carpenter Z, et al. Activating mutations in 1301-1310.
the NT5C2 nucleotidase gene drive chemotherapy resistance in re- 22. Kontro M, Kuusanmaki H, Eldfors S, et al. Novel activating STAT5B
lapsed ALL. Nat Med. 2013;19:368-371. mutations as putative drivers of T-cell acute lymphoblastic leukemia.
9. Vicente C, Cools J. The origin of relapse in pediatric T-cell acute lym- Leukemia. 2014;28:1738-1742.
phoblastic leukemia. Haematologica. 2015;100:1373-1375. 23. Bandapalli OR, Schuessele S, Kunz JB, et al. The activating STAT5B N642H
10. Trinquand A, Tanguy-Schmidt A, Ben Abdelali R, et al. Toward a mutation is a common abnormality in pediatric T-cell acute lymphoblastic
NOTCH1/FBXW7/RAS/PTEN-based oncogenetic risk classification of leukemia and confers a higher risk of relapse. Haematologica. 2014;99:
adult T-cell acute lymphoblastic leukemia: a Group for Research in Adult e188-e192.
Acute Lymphoblastic Leukemia study. J Clin Oncol. 2013;31:4333-4342. 24. DeAngelo DJ, Stone RM, Silverman LB, et al. A phase I clinical trial of the
11. Jenkinson S, Koo K, Mansour MR, et al. Impact of NOTCH1/FBXW7 notch inhibitor MK-0752 in patients with T-cell acute lymphoblastic
mutations on outcome in pediatric T-cell acute lymphoblastic leukemia leukemia/lymphoma (T-ALL) and other leukemias. J Clin Oncol. 2006;24
patients treated on the MRC UKALL 2003 trial. Leukemia. 2013;27: (suppl; abstr6585).
41-47. 25. Real PJ, Tosello V, Palomero T, et al. Gamma-secretase inhibitors re-
12. Beldjord K, Chevret S, Asnafi V, et al; Group for Research on Adult Acute verse glucocorticoid resistance in T cell acute lymphoblastic leukemia.
Lymphoblastic Leukemia (GRAALL). Oncogenetics and minimal residual Nat Med. 2009;15:50-58.
disease are independent outcome predictors in adult patients with 26. Avellino R, Romano S, Parasole R, et al. Rapamycin stimulates apoptosis
acute lymphoblastic leukemia. Blood. 2014;123:3739-3749. of childhood acute lymphoblastic leukemia cells. Blood. 2005;106:
13. Gutierrez A, Sanda T, Grebliunaite R, et al. High frequency of PTEN, PI3K, 1400-1406.
and AKT abnormalities in T-cell acute lymphoblastic leukemia. Blood. 27. Chiarini F, Grimaldi C, Ricci F, et al. Activity of the novel dual phos-
2009;114:647-650. phatidylinositol 3-kinase/mammalian target of rapamycin inhibitor
14. Bandapalli OR, Zimmermann M, Kox C, et al. NOTCH1 activation NVP-BEZ235 against T-cell acute lymphoblastic leukemia. Cancer Res.
clinically antagonizes the unfavorable effect of PTEN inactivation in 2010;70:8097-8107.

e320 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


GENETICS AND THERAPY OF ACUTE LYMPHOLASTIC LEUKEMIA

28. Degryse S, de Bock CE, Cox L, et al. JAK3 mutants transform hemato- Philadelphia chromosome-positive ALL. Bone Marrow Transplant.
poietic cells through JAK1 activation, causing T-cell acute lymphoblastic 2015;50:354-362.
leukemia in a mouse model. Blood. 2014;124:3092-3100. 45. Lim SN, Joo YD, Lee KH, et al. Long-term follow-up of imatinib plus
29. Peirs S, Matthijssens F, Goossens S, et al. ABT-199 mediated inhibition combination chemotherapy in patients with newly diagnosed Philadelphia
of BCL-2 as a novel therapeutic strategy in T-cell acute lymphoblastic chromosome-positive acute lymphoblastic leukemia. Am J Hematol. 2015;
leukemia. Blood. 2014;124:3738-3747. 90:1013-1020.
30. Zhang J, Ding L, Holmfeldt L, et al. The genetic basis of early T-cell 46. Mullighan CG, Su X, Zhang J, et al; Childrens Oncology Group. Deletion
precursor acute lymphoblastic leukaemia. Nature. 2012;481:157-163. of IKZF1 and prognosis in acute lymphoblastic leukemia. N Engl J Med.
31. Bond J, Marchand T, Touzart A, et al. An early thymic precursor phe- 2009;360:470-480.
notype predicts outcome exclusively in HOXA-overexpressing adult 47. Den Boer ML, van Slegtenhorst M, De Menezes RX, et al. A subtype of
T-ALL: a GRAALL study. Blood. 2015;126:808. childhood acute lymphoblastic leukaemia with poor treatment outcome:
32. Matlawska-Wasowska K, Kang H, Devidas M, et al. Mixed lineage a genome-wide classification study. Lancet Oncol. 2009;10:125-134.
leukemia rearrangements (MLL-R) are determinants of high risk disease 48. Boer JM, Marchante JR, Evans WE, et al. BCR-ABL1-like cases in pediatric
in homeobox A (HOXA)-deregulated T-lineage acute lymphoblastic acute lymphoblastic leukemia: a comparison between DCOG/Erasmus
leukemia: a Childrens Oncology Group study. Blood. 2015;126:694. MC and COG/St. Jude signatures. Haematologica. 2015;100:e354-e357.
33. Chiaretti S, Fo`a R. Management of adult Ph-positive acute lympho- 49. Roberts KG, Li Y, Payne-Turner D, et al. Targetable kinase-activating
blastic leukemia. Hematology Am Soc Hematol Educ Program. 2015; lesions in Ph-like acute lymphoblastic leukemia. N Engl J Med. 2014;371:
2015:406-413. 1005-1015.
34. Fielding AK. Treatment of Philadelphia chromosome-positive acute 50. Herold T, Baldus CD, Gokbuget N. Ph-like acute lymphoblastic leukemia
lymphoblastic leukemia in adults: a broader range of options, improved in older adults. N Engl J Med. 2014;371:2235.
outcomes, and more therapeutic dilemmas. Am Soc Clin Oncol Educ 51. Roberts KG, Payne-Turner D, McCastlain K, et al. High frequency and
Book. 2015;35:e352-e359. poor outcome of Ph-like acute lymphoblastic leukemia in adults. Blood.
35. Daver N, Thomas D, Ravandi F, et al. Final report of a phase II study of 2015;126:2618.
imatinib mesylate with hyper-CVAD for the front-line treatment of adult 52. Boer JM, Koenders JE, van der Holt B, et al. Expression profiling of adult
patients with Philadelphia chromosome-positive acute lymphoblastic acute lymphoblastic leukemia identifies a BCR-ABL1-like subgroup
leukemia. Haematologica. 2015;100:653-661. characterized by high non-response and relapse rates. Haematologica.
36. Bassan R, Rossi G, Pogliani EM, et al. Chemotherapy-phased imatinib 2015;100:e261-e264.
pulses improve long-term outcome of adult patients with Philadelphia 53. van der Veer A, Waanders E, Pieters R, et al. Independent prognostic
chromosome-positive acute lymphoblastic leukemia: Northern Italy value of BCR-ABL1-like signature and IKZF1 deletion, but not high CRLF2
Leukemia Group protocol 09/00. J Clin Oncol. 2010;28:3644-3652. expression, in children with B-cell precursor ALL. Blood. 2013;122:
37. Ribera JM, Oriol A, Gonzalez M, et al; Programa Espan~ol de Tratamiento 2622-2629.
en Hematologa; Grupo Espan ~ol de Trasplante Hemopoyetico Groups. 54. Loh ML, Zhang J, Harvey RC, et al. Tyrosine kinome sequencing of
Concurrent intensive chemotherapy and imatinib before and after stem pediatric acute lymphoblastic leukemia: a report from the Childrens
cell transplantation in newly diagnosed Philadelphia chromosome- Oncology Group TARGET Project. Blood. 2013;121:485-488.
positive acute lymphoblastic leukemia. Final results of the CSTIBES02 55. Kiyokawa N, Iijima K, Yoshihara H, et al. An analysis of Ph-like ALL in
trial. Haematologica. 2010;95:87-95. Japanese patients. Blood. 2013;122:352 (suppl; abstr 352).
38. Salami K, Alkayed K, Halalsheh H, et al. Hematopoietic stem cell 56. Te Kronnie G, Silvestri D, Vendramini E, et al. Philadelphia-like signature
transplant versus chemotherapy plus tyrosine kinase inhibitor in the in childhood acute lymphoblastic leukemia: the AEIOP experience.
treatment of pediatric Philadelphia chromosome-positive acute lym- Blood. 2013;122:353 (suppl; abstr 353).
phoblastic leukemia (ALL). Hematol Oncol Stem Cell Ther. 2013;6:34-41. 57. Stock W, Luger SM, Advani AS, et al. Favorable outcomes for older
39. Tanguy-Schmidt A, Rousselot P, Chalandon Y, et al. Long-term follow-up adolescents and young adults (AYA) with acute lymphoblastic leukemia
of the imatinib GRAAPH-2003 study in newly diagnosed patients with de (ALL): early results of U.S. Intergroup Trial C10403. Blood. 2014;124:
novo Philadelphia chromosome-positive acute lymphoblastic leuke- 796.
mia: a GRAALL study. Biol Blood Marrow Transplant. 2013;19:150-155. 58. Roberts KG, Pei D, Campana D, et al. Outcomes of children with BCR-
40. Chalandon Y, Thomas X, Hayette S, et al; Group for Research on ABL1like acute lymphoblastic leukemia treated with risk-directed
Adult Acute Lymphoblastic Leukemia (GRAALL). Randomized study of therapy based on the levels of minimal residual disease. J Clin Oncol.
reduced-intensity chemotherapy combined with imatinib in adults with 2014;32:3012-3020.
Ph-positive acute lymphoblastic leukemia. Blood. 2015;125:3711-3719. 59. Russell LJ, Capasso M, Vater I, et al. Deregulated expression of cytokine
41. Fielding AK, Rowe JM, Buck G, et al. UKALLXII/ECOG2993: addition of receptor gene, CRLF2, is involved in lymphoid transformation in B-cell
imatinib to a standard treatment regimen enhances long-term out- precursor acute lymphoblastic leukemia. Blood. 2009;114:2688-2698.
comes in Philadelphia positive acute lymphoblastic leukemia. Blood. 60. Mullighan CG, Collins-Underwood JR, Phillips LA, et al. Rearrangement
2014;123:843-850. of CRLF2 in B-progenitor- and Down syndrome-associated acute lym-
42. Pfeifer H, Wassmann B, Bethge W, et al; GMALL Study Group. phoblastic leukemia. Nat Genet. 2009;41:1243-1246.
Randomized comparison of prophylactic and minimal residual 61. Harvey RC, Mullighan CG, Chen IM, et al. Rearrangement of CRLF2 is
disease-triggered imatinib after allogeneic stem cell transplantation associated with mutation of JAK kinases, alteration of IKZF1, Hispanic/
for BCR-ABL1-positive acute lymphoblastic leukemia. Leukemia. Latino ethnicity, and a poor outcome in pediatric B-progenitor acute
2013;27:1254-1262. lymphoblastic leukemia. Blood. 2010;115:5312-5321.
43. Mullighan CG, Miller CB, Radtke I, et al. BCR-ABL1 lymphoblastic leu- 62. Yoda A, Yoda Y, Chiaretti S, et al. Functional screening identifies CRLF2 in
kaemia is characterized by the deletion of Ikaros. Nature. 2008;453: precursor B-cell acute lymphoblastic leukemia. Proc Natl Acad Sci USA.
110-114. 2010;107:252-257.
44. Kim M, Park J, Kim DW, et al. Impact of IKZF1 deletions on long-term 63. Bersenev A, Wu C, Balcerek J, et al. Lnk constrains myeloproliferative
outcomes of allo-SCT following imatinib-based chemotherapy in adult diseases in mice. J Clin Invest. 2010;120:2058-2069.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e321


CHIARETTI ET AL

64. Bersenev A, Wu C, Balcerek J, et al. Lnk controls mouse hematopoietic 73. Weston BW, Hayden MA, Roberts KG, et al. Tyrosine kinase inhibitor
stem cell self-renewal and quiescence through direct interactions with therapy induces remission in a patient with refractory EBF1-PDGFRB-
JAK2. J Clin Invest. 2008;118:2832-2844. positive acute lymphoblastic leukemia. J Clin Oncol. 2013;31:e413-e416.
65. Roberts KG, Morin RD, Zhang J, et al. Genetic alterations activating 74. Lengline E, Beldjord K, Dombret H, et al. Successful tyrosine kinase
kinase and cytokine receptor signaling in high-risk acute lymphoblastic inhibitor therapy in a refractory B-cell precursor acute lymphoblastic
leukemia. Cancer Cell. 2012;22:153-166. leukemia with EBF1-PDGFRB fusion. Haematologica. 2013;98:
66. Kobayashi K, Mitsui K, Ichikawa H, et al. ATF7IP as a novel PDGFRB fusion e146-e148.
partner in acute lymphoblastic leukaemia in children. Br J Haematol. 75. Kobayashi K, Miyagawa N, Mitsui K, et al. TKI dasatinib monotherapy
2014;165:836-841. for a patient with Ph-like ALL bearing ATF7IP/PDGFRB translocation.
67. Schwab C, Ryan SL, Chilton L, et al. EBF1-PDGFRB fusion in paediatric Pediatr Blood Cancer. 2015;62:1058-1060.
B-cell precursor acute lymphoblastic leukaemia (BCP-ALL): genetic 76. Maude S, Tasian S, Vincent T, et al. Targeting Jak2 and Mtor in xenograft
profile and clinical implications. Blood. Epub 2016 Feb 12. models of Crlf2-overexpressing acute lymphoblastic leukemia (All).
68. Iacobucci I, Li Y, Roberts KG, et al. Truncating erythropoietin receptor Pediatr Blood Cancer. 2012;58:1014.
rearrangements in acute lymphoblastic leukemia. Cancer Cell. 2016;29: 77. Wu SC, Li LS, Kopp N, et al. Activity of the type II JAK2 inhibitor CHZ868 in
186-200. b cell acute lymphoblastic leukemia. Cancer Cell. 2015;28:29-41.
69. Tasian SK, Doral MY, Borowitz MJ, et al. Aberrant STAT5 and PI3K/mTOR 78. Harvey RC, Kang HN, Roberts KG, et al. Development and validation of a
pathway signaling occurs in human CRLF2-rearranged B-precursor highly sensitive and specific gene expression classifier to prospectively
acute lymphoblastic leukemia. Blood. 2012;120:833-842. screen and identify B-precursor acute lymphoblastic leukemia (ALL) pa-
70. Shi C, Han L, Zhang Q, et al. Combined targeting of JAK2 with a type II tients with a Philadelphia chromosome-like (Ph-like or BCR-ABL1-like)
JAK2 inhibitor and mTOR with a TOR kinase inhibitor constitutes signature for therapeutic targeting and clinical intervention. Blood. 2013;
synthetic activity in JAK2-driven Ph-like acute lymphoblastic leukemia. 122:826.
Blood. 2015;126:2529. 79. He J, Abdel-Wahab O, Nahas MK, et al. Integrated genomic DNA/RNA
71. Waibel M, Solomon VS, Knight DA, et al. Combined targeting of JAK2 and profiling of hematologic malignancies in the clinical setting. Blood.
Bcl-2/Bcl-xL to cure mutant JAK2-driven malignancies and overcome 2016. In press.
acquired resistance to JAK2 inhibitors. Cell Reports. 2013;5:1047-1059. 80. Schultz KR, Carroll A, Heerema NA, et al; Childrens Oncology Group.
72. Maude SL, Tasian SK, Vincent T, et al. Targeting JAK1/2 and mTOR in Long-term follow-up of imatinib in pediatric Philadelphia chromosome-
murine xenograft models of Ph-like acute lymphoblastic leukemia. positive acute lymphoblastic leukemia: Childrens Oncology Group
Blood. 2012;120:3510-3518. study AALL0031. Leukemia. 2014;28:1467-1471.

e322 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


HEMATOLOGIC MALIGNANCIESLEUKEMIA,
MYELODYSPLASTIC SYNDROMES, AND
ALLOTRANSPLANT

Progress in Myeloproliferative
Neoplasms: Are We Ready?

CHAIR
Ruben A. Mesa, MD, FACP
Mayo Clinic
Scottsdale, AZ

SPEAKERS
Francesco Passamonti, MD
Fondazione IRCCS Policlinico San Matteo
Pavia, Italy

Jeffrey Tyner, PhD


Oregon Health & Science University
Portland, OR
MESA AND PASSAMONTI

Individualizing Care for Patients With Myeloproliferative


Neoplasms: Integrating Genetics, Evolving Therapies, and
Patient-Specific Disease Burden
Ruben A. Mesa, MD, and Francesco Passamonti, MD

OVERVIEW

Individualized medicine is important for patients with myeloproliferative neoplasms (MPNs), including essential throm-
bocythemia, polycythemia vera, and myelofibrosis, which are heterogeneous in terms of genetic mutation profile, prognosis,
disease burden, and symptoms. Status of MPN driver mutations in JAK2, CALR, and MPL (or lack of one of these mutations)
and other myeloid mutations (ASXL1, SRSF2, CBL, and IDH1/2, among others) affects diagnosis and prognosis. Management
begins with estimating the prognosis, disease burden including MPN symptoms, and prevention of vascular events. Al-
logeneic stem cell transplantation is the definitive therapy in a subset of patients with myelofibrosis, the majority of whom
receive JAK inhibition with ruxolitinib to relieve splenomegaly and symptoms and to prolong survival. Ruxolitinib is now a
second-line therapy in polycythemia vera, with pegylated interferon being evaluated as a potential front-line therapy
compared with hydroxyurea. The therapeutic landscape is evolving to include new JAK inhibitors, which may affect
cytopenias (pacritinib and momelotinib), combination therapies including ruxolitinib, and novel targets such as pentraxin
and telomerase. Assessing the therapeutic efficacy (including symptom impact) and toxicity of these new approaches is
necessary to determine longitudinal management of MPNs in clinical practice and is a key component of individualizing
care for patients with MPNs.

M yeloproliferative neoplasms account for three main


entities named polycythemia vera, essential throm-
bocythemia, and primary myelofibrosis.1,2 Polycythemia
Classification of MPNs is performed according to the 2008
World Health Organization (WHO) criteria, which are based
on clinical, histopathologic, and molecular findings.12 An
vera is characterized by erythrocytosis with some degree of updated WHO classification is expected in 2016, which will
leukocytosis and thrombocytosis and an overall increase in likely include MPN driver mutations (JAK2, CALR, and MPL),
bone marrow cellularity with well-defined symptoms.3 The different cell count cutoffs, and an extended indication for
clinical picture of patients with essential thrombocythemia is bone marrow biopsy.1
dominated by isolated thrombocytosis.4,5 Polycythemia vera
and essential thrombocythemia share several clinical fea- CLINICAL BURDEN OF MPNs: THE EFFECTS OF
tures, most prominently a high risk of thrombosis and a MPN SYMPTOMS ON HEALTH-RELATED QUALITY
predisposition to evolve into acute myeloid leukemia or OF LIFE
secondary myelofibrosis.6-8 Among MPNs, primary myelo- Given their variable nature, MPNs have various clinical ef-
fibrosis has the most heterogeneous clinical presentation, fects such as variable cytopenias, variable degrees of
including anemia, splenomegaly, leukocytosis or leukopenia, splenomegaly, and a constellation of MPN-related symp-
thrombocytosis or thrombocytopenia, a variably high toms.13 A patients clinical course is also affected by
symptom burden, and a variability of clinical responses to comorbidities, therapy-related toxicity, and morbidity
therapy.9,10 A study among 1,581 patients with MPNs was owing to prior vascular events. Health-related quality of
recently conducted at the Mayo Clinic in the United States life for patients with MPNs is a broader construct that
and at the University of Florence and Papa Giovanni XXIII includes the above-listed features and also considers the
Hospital in Italy; median survival was 19.8 years, 13.5 years, burden of medical care, disease-related financial burden
and 5.9 years for individuals with essential thrombocythemia, (e.g., decreased ability to work or medical disability),
polycythemia vera, and primary myelofibrosis, respectively.11 and stress or effects on mood as a result of uncertainty

From the Mayo Clinic Cancer Center, Phoenix, AZ; Division of Hematology, Department of Clinical and Experimental Medicine, University of Insubria, Varese, Italy.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Ruben A. Mesa, MD, Mayo Clinic Cancer Center, 5777 East Mayo Blvd., Phoenix, AZ 85259; email: mesa.ruben@mayo.edu.

2016 by American Society of Clinical Oncology.

e324 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


INDIVIDUALIZED THERAPY FOR MYELOPROLIFERATIVE NEOPLASMS

about the future (Sidebar). As one quantifies MPN- suicide) to overwhelming fatigue (which can lead to
related symptom burden and assesses the effects of medical disability and exacerbate mood disturbances).15
therapy and disease course, it is important to consider The advancing symptom burden seen with secondary
that MPN symptom burden and health-related quality myelofibrosis is not only a problematic feature of the
of life are interrelated but distinct concepts. For ex- disease but also likely directly contributes to the mortality
ample, one could theoretically improve a specific MPN- of the disorder. Analysis of causes of death in secondary
associated symptom such as pruritus; however, if the myelofibrosis shows that approximately one-third of
agent used to achieve that response led to problematic patients will succumb to acute myeloid leukemia and
toxicity (e.g., gastrointestinal), then the net effect on likely mortal complications of cytopenias; however, the
health-related quality of life could be adverse despite remainder of patients succumb to a variety of causes
symptom-specific improvement. exacerbated by the debilitation and hypercatabolic state
caused by the disease (e.g., thrombosis, infection, or
exacerbation of comorbidities such as congestive heart
MPN SYMPTOM BURDEN PLAYS AN IMPORTANT failure). 14
ROLE IN MORBIDITY AND MORTALITY The identification of symptoms prevalent among patients
Individualized management of MPNs recognizes that MPNs
with MPNs is not new; symptom burden has long been
can have a long disease course. Although MPNs are clonal
recognized as a key clinical feature of MPNs. However, the
hematopoietic malignancies, their course can frequently
degree of symptom burden was first quantified in 2007 in
exceed a decade, even among some patients with secondary
a web-based survey of more than 1,000 patients, conducted
myelofibrosis. As a result, the cumulative effects of symptom
by the CMPD Education Foundation and Mayo Clinic.13
burden, decreased health-related quality of life, comor-
Identified symptoms included fatigue, night sweats, pruri-
bidities, and medication-related toxicity all affect morbidity
tus, and bone pain, among others.13
and mortality.14 Vascular events early in the course of MPNs
There is likely a pathogenetic link between the clonal
can affect disease morbidity, ranging from very serious
myeloid malignancy of an MPN and the patients direct
events such as intra-abdominal thrombosis (which can lead
symptom burden. Part of this link may potentially be related
to portal hypertension, esophageal varices, and risks of
to inflammatory cytokines. Previous studies demonstrated
hemorrhage) to the obvious implications of stroke and
that interleukin (IL)-1, IL-2, IL-6, tumor necrosis factor-alpha,
myocardial infarction or the more subtle effect of MPNs on
and interferons may have a direct link between these two
cognition, chronic headaches, and erythromelalgia. The
areas (particularly in constitutional symptoms such as fevers,
burden of specific symptoms can be debilitating, ranging
night sweats, and weight loss), as well as between symptoms
from unrelenting pruritus (e.g., lack of relief has led to
and prognostic significance.14 Indeed, increases in in-
flammatory cytokines were observed in early studies with
ruxolitinib and improvement in these cytokines was seen
KEY POINTS with response to therapy.16

Individualizing care for patients with MPNs begins with


an accurate assessment of prognosis with current ASSESSMENT OF THE MUTATION PROFILE IN
clinical scoring systems and an estimation of disease MPNs
burden. The same driver mutations of JAK2, MPL, and CALR genes are
Genetic mutation testing (driver mutations such as JAK2 present in polycythemia vera, essential thrombocythemia,
V617F, CALR, and MPL) in MPNs is helpful in making the
initial diagnosis, and more extensive mutation testing
(ASXL1, IDH1/2, etc.) is helpful in refining the prognosis SIDEBAR. Factors Impacting Health-Related
in select clinical scenarios, such as equivocal candidates Quality of Life for Patients With
for stem cell transplantation. Myeloproliferative Neoplasms
MPNs have a range of disease-associated symptoms
that arise from the clonal disease, are associated with MPN-derived constitutional symptoms (i.e., pruritus, night
pathologic cytokines and inflammation, and directly sweats)
contribute to disease morbidity and mortality. MPN-derived spleen-oriented symptoms (i.e., discomfort
Alleviation of MPNs symptoms, now measurable or pain)
through standardized instruments, is an integral part of MPN vascular symptoms arising from elevated blood
assessing therapeutic effects in both clinical trials and counts
clinical practice. Toxicity from MPN-directed therapies
JAK inhibition is the cornerstone of medical therapy for Complications from earlier MPN-related events such as
problematic myelofibrosis and polycythemia vera. thrombosis
Expanding investigations with interferons, new JAK Burden of medical care related to MPN
inhibitors, JAK inhibitor combinations, and new targets Financial burden related to illness
are on the horizon. Stress related to uncertainty in health

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e325


MESA AND PASSAMONTI

and primary myelofibrosis, both in sporadic17-20 and in fa- mutation status was associated with different survival
milial cases.21,22 These patients (10%15%, overall) are re- (CALR+ASXL12 and CALRASXL1+ for best and worse sur-
ferred to as having triple-negative status. Among patients vival, respectively).33 A recent analysis showed that the
with triple-negative status, new somatic JAK2 and MPL favorable prognostic effect of CALR mutations on primary
variants have been discovered.23 myelofibrosis might be restricted to only those patients with
In polycythemia vera, JAK2 mutations cover the whole type 1 CALR mutations.34 In a recent study of 1,118 patients
mutation profile, with V617F present among 95%97% with primary myelofibrosis, driver mutations were classified
of patients4 and exon 12 mutations present among the prognostically in two distinct categories: favorable (type
remainder.24 Very few patients present with CBL or LNK 1/type 1like CALR) and unfavorable (all other mutation
mutations. MPL mutations have been identified in ap- categories).35
proximately 5% of patients with essential thrombocythemia On the basis of this information, molecular profiling of
and in 5%10% of patients with secondary myelofibrosis.25 MPN at diagnosis must include analysis of JAK2 (looking for
Mutations in the CALR gene have been mainly identified among the V617F point mutation first),7 screening of exon 12
patients with essential thrombocythemia and/or primary mutations of JAK2 (only in V617F-negative PV),24 and
myelofibrosis without JAK2 or MPL mutations,18,26 covering screening of CALR and MPL mutations (in V617F-negative ET
70% of the mutation profile for patients without JAK2/MPL and primary myelofibrosis).19
mutations.
Detection of JAK2 mutations is critical in the diagnostic
process of erythrocytosis and is a major criterion for diagnosis IS IT MANDATORY TO STUDY ADDITIONAL
of polycythemia vera.6 Regarding JAK2 allele burden, higher MUTATIONS IN MPNs AT DIAGNOSIS?
burdens correlate unequivocally with enhanced myelopoiesis Additional mutations of CBL, TET2, SF3B1, SRSF2, DNMT3A,
of the bone marrow, leukocytosis, and increasing spleen size, IDH1/2, EZH2, and ASXL1 genes are variably reported in
whereas they inversely correlate with platelet count.7,27 Al- MPNs, although mainly among patients with secondary
though allele burden quantification is of interest, it is not myelofibrosis.36
mandatory to diagnose polycythemia vera; a qualitative test A total of 879 patients with secondary myelofibrosis were
on whole blood leukocytes is a correct approach outside of studied to determine the individual and combined prog-
the research setting. nostic relevance of somatic mutations in ASXL1, SRSF2, EZH2,
Concerning CALR mutations, all of the described mutations TET2, DNMT3A, CBL, IDH1, IDH2, MPL, and JAK2.37 Of these,
are found mainly in a heterozygous state and are insertions ASXL1, SRSF2, and EZH2 mutations interindependently
or deletions (type 1, 52-bp deletion, p.L367fs*46; and type 2, predicted shortened survival. However, only ASXL1 muta-
5-bp TTGTC insertion, p.K385fs*47) in the last exon encoding tions remained significant in the context of the International
the C-terminal amino acids of the CALR protein.18,26 Prognostic Scoring System (IPSS) prognostic model.14
Information on CALR mutation in essential thrombocy- Leukemia-free survival was negatively affected by IDH1/2,
themia is summarized as follows.19,28-30 First, 40%50% and SRSF2, and ASXL1 mutations, whereas it was affected by
30%40% of patients display type 1 and type 2 variants, IDH1 and SRSF2 mutations in a Mayo Clinic cohort.37
respectively. Compared with mutant JAK2, both variants Next-generation sequencing may simplify the identifica-
were associated with higher platelet and lower hemoglobin tion of new additional mutations. By applying this approach,
and leukocyte counts. Patients with a CALR mutation have a mutations other than JAK2, CALR, or MPL occur among more
lower risk of thrombosis than patients with a JAK2 mutation. than 80% of patients with primary myelofibrosis, including
Evolution from essential thrombocythemia to polycythemia those with triple-negative status. A prior study revealed that
vera seems unusual (even impossible). Patients with a CALR the absence of such mutations is independently favorable
mutation were classified into the lower-risk groups with for survival, whereas the prognostic effect of their presence
regard to both the standard risk stratification for thrombosis is influenced by ASXL1, CBL, RUNX1, and SRSF2 mutations.38
(older than age 60 and prior thrombosis) and the In- These mutations are not routinely assessed at the time of
ternational Prognostic System in Essential Thrombocythe- diagnosis in daily clinical practice, although they may be
mia (IPSET) score31 for survival. useful in some clinical situations as discussed above.
In the largest collaborative trial including 617 patients with
primary myelofibrosis, individuals with a CALR mutation WHICH PROGNOSTIC SCORE SHOULD BE USED
had a lower risk of developing anemia, thrombocytopenia, TO PREDICT SURVIVAL IN MYELOFIBROSIS?
and marked leukocytosis compared with other subtypes.32 IPSS and DIPSS Models Based on Clinical Parameters
Patients with a CALR mutation also had a lower risk of The IPSS was defined in 2009 on the basis of 1,054 patients
thrombosis compared with patients with a JAK2 mutation. In with primary myelofibrosis, excluding post-polycythemia
other studies, CALR mutations had a favorable effect on vera and post-essential thrombocythemia secondary mye-
survival that was independent of both risk (as determined by lofibrosis and prefibrotic primary myelofibrosis.14 Median
the Dynamic International Prognostic Scoring System survival was 69 months. Parameters affecting the survival
[DIPSS] Plus model) and ASXL1 mutation status.19 A sub- and scoring system are reported in Table 1. For the low,
sequent analysis identified that combined CALR and ASXL1 intermediate-1, intermediate-2, and high-risk categories,

e326 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


INDIVIDUALIZED THERAPY FOR MYELOPROLIFERATIVE NEOPLASMS

median survival was 135 months, 95 months, 48 months, and Molecular-Based Prognostic Models
27 months, respectively. On the basis of the IPSS and DIPSS prognostic models and the
The progressive nature of primary myelofibrosis gener- available data on mutation platforms, it seemed obvious to
ated interest in defining new so-called dynamic models, such combine different clinical/molecular parameters with the
as the DIPSS and the DIPSS Plus model. In a nontime- aim to distinguish patients at higher risk from those at lower
dependent analysis (models at diagnosis), patients are risk. To reinforce the role of mutations in disease prediction,
assigned to a risk group on the basis of the assessment of the additional prognostic value of the number of mutated
risk factors at diagnosis and they are followed in the same genes was taken into account.41 The presence of two or
category irrespective of the acquisition of other risk factors more mutations predicted the worst outcomes. Median
during the disease course. According to a dynamic model, survival was 2.6 years, 7.0 years, and 12.3 years for patients
patients contribute to the estimate of survival in a category with two, one, or no mutations, respectively. The presence
only as long as they do not acquire further risk factors, then of two or more mutations was also associated with short-
they shift to a higher category according to their new ened leukemia-free survival.
score. By combining CALR, JAK2, MPL, triple-negative status, and
The DIPSS was developed based on a study of 525 patients each single variable included in the IPSS, a molecular-based
with primary myelofibrosis who were regularly followed, risk model was developed.32 Five risk categories with sig-
using the same IPSS risk factors but a different scoring nificantly different survival were identified. Although this
system (Table 1).39 Median survival referring to patients model was based on the three driver mutations and some
maintaining the same risk category over time was not IPSS clinical parameters, the Mutation-Enhanced In-
reached in the low-risk category, whereas median survival ternational Prognostic Scoring System (MIPSS)42 also com-
was 14.2 years, 4 years, and 1.5 years in the intermediate-1, bined additional mutations (ASXL1, SRSF2, EZH2, and
intermediate-2, and high-risk categories, respectively. IDH1/2) previously identified by next-generation sequenc-
The DIPSS Plus model was produced as a result of an ing.37 Factors included in the MIPSS model are reported in
analysis of 793 patients with primary myelofibrosis; 428 Table 2. Four risk groups were defined.
patients were referred within their first year of diagnosis, The Genetics-Based Prognostic Scoring System prognostic
whereas 365 were referred after their first year of di- model was developed to further integrate cytogenetic and
agnosis.40 Unfavorable cytogenetics, red blood cell molecular profiles (Table 2). The four risk categories were
transfusion need, and low platelet counts were added to associated with different survival, ranging from 2.2 years to
the DIPSS categories. For the low-, intermediate-1, more than 17 years.43 Figure 1 reports how to use this
intermediate-2, and high-risk categories, median sur- clinicalmutational-based prognostic model in the clinical
vival was 185 months, 78 months, 35 months, and practice.
16 months, respectively.

TABLE 2. Genetically Driven Prognostic Model in


Primary Myelofibrosis
TABLE 1. Clinically Driven Prognostic Model in Primary
Myelofibrosis MIPSS GIPSS

IPSS DIPSS DIPSS Plus Age > 6 1.5 2


Constitutional Symptoms 0.5 No
Age > 65 1 1 To assess DIPSS status**
Hemoglobin < 10 g/dL 0.5 No
Hemoglobin < 10 g/dL 1 2 To assess DIPSS status**
Platelets < 200 3 10 /L 9
1.0 No
Leukocyte Count 1 1 To assess DIPSS status**
> 25 3 109/L Triple Negative 1.5 2
Blast Cells 1% 1 1 To assess DIPSS status** JAK2 or MPL Mutation 0.5 2
Constitutional Symptoms 1 1 To assess DIPSS status** ASXL1 Mutation 0.5 1
Unfavorable Cytogenetics* No No 1 SRSF2 Mutation 0.5 1
Red Blood Cell Need No No 1 CALR Type 2, Type 2Like No 2
Platelets No No 1 Unfavorable Cytogenetics* No 3 for very high risk;
< 100 3 109/L 2 for high risk
Data are presented as points values unless otherwise indicated. IPSS categories are as Data are presented as points values unless otherwise indicated. MIPSS categories: low
follows: low (score 0), intermediate-1 (score 1), intermediate-2 (score 2), and high (score 0 to 0.5), intermediate-1 (score 1 to 1.5), intermediate-2 (score 2 to 3.5), and
(score $ 3). DIPSS categories are as follows: low (score 0), intermediate-1 (score 1 to 2), high (score $ 4).
intermediate-2 (score 3 to 4), and high (score 5 to 6). DIPSS Plus categories are as *Very high risk indicates a monosomal karyotype, inv(3), i(17q), -7/7q-, 11q, or 12p
follows: low (score 0), intermediate-1 (score 1), intermediate-2 (score 2 to 3), and high abnormalities; high risk indicates complex nonmonosomal karyotype, 2 abnormalities
(score 4 to 6). not included in the very high-risk category, 5q-,18, other autosomal trisomies
*Complex karyotype or a single abnormality or two abnormalities, including 18, -7/7q-, except 19, and other sole abnormalities not included in other risk categories.
i(17q), -5/5q-, 12p-, inv(3), or 11q23 rearrangement. Intermediate risk indicates sole abnormalities of 20q-, 1q1, or any other sole
**DIPSS status of intermediate-1 (1 point); intermediate-2 (2 points); and high translocation, and Y or other sex chromosome abnormality. Low risk indicates normal
(3 points). cytogenetics or sole abnormalities of 13q- or 19.
Abbreviations: IPSS, International Prognostic Scoring System; DIPSS, Dynamic Abbreviations: MIPSS, Mutation-Enhanced International Prognostic Scoring System;
International Prognostic Scoring System. GPSS, Genetics-Based Prognostic Scoring System.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e327


MESA AND PASSAMONTI

PROGNOSTICATION IN POLYCYTHEMIA VERA model included the following parameters: older than age 60 (2
AND ESSENTIAL THROMBOCYTHEMIA points), leukocyte count greater than 11 3 109/L (1 point), and
There is a global consensus that being age 60 or older at prior history of thrombosis (1 point). Three risk categories with
diagnosis and presence of vascular events in the patients significantly different survival were identified: low (0 points,
history are the two most relevant prognostic factors in median survival not reached), intermediate (1 to 2 points,
predicting thrombosis in polycythemia vera and essential median survival 24.5 years), and high (3 to 4 points, median
thrombocythemia.44,45 Therefore, patients with at least one survival 13.8 years).
of these risk factors at diagnosis are considered at high risk, Concerning thrombosis prediction in WHO-diagnosed
whereas those with none are considered at low risk.45 This essential thrombocythemia, the IPSET thrombosis
risk classification affects the therapeutic approach for pa- model50 was generated for the same IPSET cohort but
tients with polycythemia vera and essential thrombocy- considers risk factors including being older than 60 (1 point),
themia. However, leukocyte counts, cardiovascular risk, JAK2/ history of thrombosis (2 points), presence of cardiovascular
CALR mutation status, and bone marrow features are reported risk factors (1 point), and JAK2 (V617F) mutation (2 points).
to affect thrombosis, progression, and survival.28,29,46,47 On the basis of these factors, the IPSET thrombosis model
In polycythemia vera, the ECLAP study48 showed that specifies the following risk categories: low (less than 2
patients younger than age 65 without prior thrombosis have points; thrombosis risk of 1.03 per 100 person-years), in-
an incidence of thrombosis of 2.5 per 100 persons per year, termediate (2 points; thrombosis risk of 2.35 per 100
those older than age 65 or with prior thrombosis have an person-years), and high (more than 2 points; thrombosis
incidence of 5.0 per 100 persons per year, and patients older risk of 3.56 per 100 person-years). Although CALR mutation
than age 65 with prior thrombosis have an incidence of 10.9 status was introduced into the statistical analysis, the IPSET
per 100 persons per year. model maintained its prognostic power.51
In essential thrombocythemia, physicians can predict survival In another study, survival was assessed in 1,545 patients
according to the IPSET model,49 which was tested among 867 with WHO-defined polycythemia vera.52 Parameters in-
patients with WHO-defined essential thrombocythemia. The cluded in this model were older age, leukocytosis, and

FIGURE 1. Stepwise Approach to Estimating Prognosis in Myeloproliferative Neoplasms Incorporating Clinical


Prognostic Scoring System and Then Refining Prognosis With Mutation Status

Abbreviations: DIPSS, Dynamic International Prognostic Scoring System; ET, essential thrombocythemia; f-up, follow-up; HR, high risk; Int-1, intermediate-1 risk; Int-2,
intermediate-2 risk; IPSET, International Prognostic Score Essential Thrombocythemia; IPSS, International Prognostic Scoring System; LR, low risk; Med OS, median overall
survival; PMF, primary myelofibrosis; PV, polycythemia vera; WHO, World Health Organization.

e328 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


INDIVIDUALIZED THERAPY FOR MYELOPROLIFERATIVE NEOPLASMS

TABLE 3. Myeloproliferative Neoplasm Symptom Assessment Tools and Associated Instruments

Questionnaire Goals Number of Items Reference


Myelofibrosis Symptom Assessment Form Myelofibrosis-specific symptom assessment 20 Mesa et al53
(MF-SAF)
Includes the Brief Fatigue Inventory
Myelofibrosis Symptom Assessment Form Myelofibrosis symptom serial assessment on 7 Mesa et al54
2.0 (MF-SAF 2.0) clinical trials
Validated in the COMFORT-1 trial
Myeloproliferative Neoplasm Symptom As- Expanded to measure MPN symptoms across 27 Scherber et al15
sessment Form (MPN-SAF) ET, PV, and MF
Myeloproliferative Neoplasm Symptom As- Derived as a subset of the most representative 10 Emanuel et al55
sessment Form Total Symptom Score symptoms from MPN-SAF
(MPN 10 or MPN-SAF TSS)
Abbreviations: MPN, myeloproliferative neoplasm; ET, essential thrombocythemia; PV, polycythemia vera; MF, myelofibrosis.

venous thrombosis, which generated three risk categories of clinical changes, whereas the MPN 10 is helpful for routine
with different survival ranging from 10.9 to 27.8 years. clinical monitoring visits or when used daily in a diary with
Figure 1 shows how to use prognostication in MPNs. clinical trials. The tools are meant to be a springboard for
discussion between the patient and his or her provider,
and improvement in symptoms is always weighed against
CREATION OF VALIDATION OF MPN SYMPTOM medication-related toxicities.
ASSESSMENT TOOLS
The spectrum of symptom-related difficulties encountered WHAT LESSONS HAVE WE LEARNED FROM MPN
by patients with MPNs is varied and includes several areas of SYMPTOMS AND DISEASE FEATURES?
symptom burden such as those that are constitutional in The MPN International Quality of Life Study Group has analyzed
origin and those that are those related to vascular flow or the aggregate data gathered on symptom burden among pa-
enlargement of the spleen. Given that there were no vali- tients with MPN to make several observations regarding these
dated instruments that incorporated this spectrum of dif- diseases. Observations made across the spectrum of MPNs in-
ficulties, MPN-specific tools were necessary and were thus clude that cytokine increases are likely linked to symptom bur-
developed utilizing the above-described 2007 web survey den.16 It has been observed that certain symptom profile clusters
as a foundation (Table 3).13 The Myelofibrosis Symptom can exist among patients with MPNs, and these symptom clusters
Assessment Form (MF-SAF) was the first instrument de- may potentially correlate with disease phenotypes with clinical
veloped. The MF-SAF is a 20-item survey validated against features (and possibly biologic features as well).56 Clinically rel-
other outcome tools reported for patients with cancer and is evant observations for problematic polycythemia vera (specifi-
effective in capturing the presence and intensity of mye- cally that hydroxyurea treatment will fail for individual subgroups
lofibrosis disease symptoms.53 Subsequently, this in- or these individuals will have either palpable splenomegaly or a
strument was further refined to a seven-item instrument persisting need for phlebotomies) show that patients with these
(MF-SAF 2.0) for use specifically in myelofibrosis clinical features all have worse symptom burden compared with patients
trials.54 In an effort to have a broader instrument that could without these features. Furthermore, individuals who have more
represent the difficulties seen in essential thrombocythemia than one of these problematic clinical features also have
and polycythemia vera, the MF-SAF was expanded to 27 worsening symptom burden.57 These latter observations may
items and was named the MPN Symptom Assessment have relevance regarding current choice of medical therapy.
Form.15 In addition to the initial MF-SAF questions related to
abdominal discomfort, fatigue, coughing, night sweats, bone
pain, weight loss, inactivity, abdominal pain, pruritus, fever, DETERMINING A TREATMENT PLAN FOR
and quality of life, there was an additional focus on mi- PATIENTS WITH MPNs
crovascular symptoms including insomnia, sexual dysfunc- When determining the treatment plan for an individual
tion, vertigo, lightheadedness, dizziness, numbness, tingling, patient, it is necessary to consider the nuanced assessment
headaches, and concentration. Each individual symptom of prognosis, the importance of avoidance of vascular
had a potential score ranging from 0 (absent) to 10 (worst events, the effect of symptom burden, the contribution of
imaginable/as bad as it could be). Further refinement of this splenomegaly to morbidity, and the likelihood of progres-
instrument for frequent serial use resulted in a 10-item total sion to acute myeloid leukemia.
symptom score (the MPN-SAF-TSS or MPN 10).55 These 10 core
items included worst fatigue, early satiety, abdominal dis- Which Patients Are Candidates for Allogeneic Stem
comfort, concentration problems, inactivity, night sweats, Cell Transplantation and When?
itching, bone pain, fever, and weight loss. We recommend Allogeneic stem cell transplantation can be curative for
using the comprehensive MPN-SAF at diagnosis and at the time patients with MPN; however, because of the associated

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e329


MESA AND PASSAMONTI

TABLE 4. JAK Inhibitors and New Agents and Their Use, or Possible Use, in Myeloproliferative Neoplasm
Treatment

Agent Disease Trial Comments Reference


Ruxolitinib (Approved) MF COMFORT-1 and Improved splenomegaly Verstovsek et al63 and Harrison et al64
COMFORT-2
Improved MF-associated symptoms
Improved survival
PV RESPONSE Improved complete response rate Vannucchi et al78
(second
Improved splenomegaly
line)
Decreased PV-associated symptoms
Pacritinib MF PERSIST-1 and Improved splenomegaly Vannucchi et al70
PERSIST-2
Improved MF-associated symptoms
(ongoing)
Able to be given in full dose
regardless of thrombocytopenia
Improvement in anemia
Momelotinib MF SIMPLIFY-1 and Ongoing phase III trials NCT0196838 and NCT02101268
SIMPLIFY-2
SIMPLIFY-2
Combination Trials All Including a
Ruxolitinib Base
Plus Azacitidine (Hypomethylating) MF Phase II Ongoing trial NCT01787487, Daver et al73
Plus Danazol (Androgen) MF Phase II Ongoing trial NCT01732445, Gowin et al72
Plus Pegylated Interferon Alfa-2a MF Phase II Ongoing trial Mikkelsen et al76
(Interferon)
Plus Panobinostat (HDAC Inhibitor) MF Phase II Ongoing trial NCT01433445, Harrison et al75
Plus Pomalidomide (IMID) MF Phase II Ongoing trial NCT01644110, Stegelmann et al74
Plus BKM-120 (PI3K Inhibitor) MF Phase II Ongoing study Durrant et al83
Plus LDE-225 (Hedgehog Inhibitor) MF Phase II Ongoing trial NCT01787552, Gupta et al84
New Agents for MPNs Testing as
Single Agents
P1101/AOP2014 PV Phase III Ongoing trial NCT01949805
Proline Pegylated Interferon Alfa-2b
PRM-151 (Antifibrosis) MF Phase II Ongoing trial NCT01981850
Imetelstat (Telomerase Inhibitor) MF Phase II Ongoing trial NCT02426086
Abbreviations: MF, myelofibrosis; PV, polycythemia vera; HDAC, histone deacetylase; ET, essential thrombocythemia; IMID, immunomodulatory drug; PI3K, phosphoinositide
3-kinase.

expense and risk of transplant-related morbidity and mor- basis. Patients with intermediate-2 or high-risk disease lacking
tality, HSCT is currently limited to a subset of patients with an HLA-matched sibling or unrelated donor should be enrolled
MF. The European Group for Blood and Marrow Trans- in a protocol using HLA nonidentical donors; the optimal in-
plantation and the European LeukemiaNet international tensity of the conditioning regimen must be defined.
working group recently provided indications for and man-
agement of allogeneic stem cell transplantation.58 Although
readers are referred to the guidelines by the European What Options Are Available to Control Splenomegaly,
Group for Blood and Marrow Transplantation and the Eu- Cytoses, and Symptoms?
The European LeukemiaNet guidelines recommend control
ropean LeukemiaNet for further information, some points
of hematocrit and use of low-dose aspirin (unless contra-
are summarized as follows. Patients with intermediate-2
indicated) for the prevention of thrombosis, as well as the
or high-risk disease and who are younger than age 70
selective use hydroxyurea as the drug of choice when an
should be considered candidates for allogeneic stem cell antimyeloproliferative effect is needed for patients with
transplantation.59 Patients with intermediate-1 risk of disease MPNs. 45 However, data available on hydroxyurea are
and who are younger than age 65 should be considered scant, with the most complete study on hydroxyurea in
candidates if they present with adverse cytogenetics, a per- myelofibrosis retrospectively evaluating 40 patients.60
centage of blasts in peripheral blood greater than 2%, or re- In the last few years, several medicines with anti-JAK
fractory, transfusion-dependent anemia. Decisions regarding a properties (named JAK inhibitors) have been studied
pretransplant splenectomy should be made on a case-by-case (Table 4).61 Among these, ruxolitinib is the only approved

e330 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


INDIVIDUALIZED THERAPY FOR MYELOPROLIFERATIVE NEOPLASMS

JAK inhibitor for MPNs and available for clinical practice. Five-year results of the COMFORT-2 trial were recently
Other compounds entered phase III investigation (fedrati- presented.68 At a median follow-up of 4.3 years, 27% of
nib,62 momelotinib, and pacritinib), and others are being patients in the ruxolitinib arm and 24% who crossed over
tested in phase I to phase II studies. from BAT completed 5 years of on-study treatment; primary
reasons for premature discontinuation were adverse events
Ruxolitinib (24%) and disease progression (22%). Overall, 54% of pa-
Two prospective randomized trials with ruxolitinib have tients in the ruxolitinib arm achieved a 35% or more re-
been published: COMFORT-1 (155 patients treated with duction in spleen volume from baseline at any time during
ruxolitinib vs. 151 treated with placebo)63 and COMFORT-2 treatment. The probability of maintaining this reduction was
(146 patients treated with ruxolitinib vs. 73 treated with best 0.51 at 3 years and 0.48 at 5 years. One-third of evaluable
available therapy [BAT]).64 In the COMFORT-1 trial, the patients with a JAK2 V617F mutation had a greater than 20%
primary endpoint (reduction of spleen volume of 35% or reduction in allele burden and 16% improved fibrosis.
more assessed by MRI) at week 24 was reached in 42% of Results of the RESPONSE trial were published in 2015. This
patients in the ruxolitinib arm and in 1% of the placebo arm. phase III open-label study evaluated the efficacy and safety
At week 24, 46% of patients receiving ruxolitinib and 5% of of ruxolitinib versus BAT in 110 and 112 patients with
those receiving placebo experienced symptom alleviation by splenomegaly, respectively, who had either hydroxyurea-
at least 50%, as measured by the modified MF-SAF.53 Pa- resistant or hydroxyurea-intolerant polycythemia vera.
tients treated with ruxolitinib experienced relief of ab- RESPONSE-2 is an ongoing study with a similar design but
dominal discomfort, early satiety, night sweats, itching, and includes patients without splenomegaly.69 The primary
musculoskeletal pain. In the COMFORT-2 trial, the primary endpoint is the proportion of patients who achieve both
endpoint (the same as the COMFORT-1 study but evaluated hematocrit control through week 32 and a 35% or greater
at week 48) was reached in 28% of patients treated with reduction in spleen volume from baseline shown on imaging
ruxolitinib and in 0% of those receiving BAT. Mean im- at week 32.
provements in Functional Assessment of Cancer Therapy The primary endpoint was achieved in 21% of patients
Lymphoma System scores from baseline were greater in the treated with ruxolitinib versus 1% of patients who received
ruxolitinib arm. BAT. Of patients who received ruxolitinib or BAT, 60% and
Improved survival was observed for ruxolitinib versus 20% achieved hematocrit control and 38% and 1%
placebo (27 vs. 41 deaths), with a hazard ratio of 0.58.65 The achieved a 35% or greater spleen volume reduction, re-
3-year analysis of the COMFORT-2 trial confirmed that dose- spectively. Complete hematologic remission was achieved in
dependent anemia and thrombocytopenia were the most 24% and 9% of ruxolitinib-treated and BAT-treated patients,
common adverse events in the ruxolitinib-treated group, but respectively; 49% and 5% had a 50% or greater improvement
these events rarely led to discontinuation.66 Other adverse in MPN-SAF-TSS score at week 32. Herpes zoster (all grade 1
events of interest included leukopenia, bleeding, infections, or 2) was reported in 6.4% and 0% of patients treated with
thromboembolic events, elevated transaminase levels, in- ruxolitinib and BAT, respectively. The rate of thromboem-
creased systolic blood pressure, and weight gain. The rate of bolic events was lower in the ruxolitinib arm versus the BAT-
these events generally decreased with longer exposure to treated arm (one and six events, respectively).
ruxolitinib treatment, with the highest rates occurring within
the first 6 months of treatment. Among these events, in- Pacritinib and Momelotinib: JAK Inhibition and
fections occurred in 50% of patients between weeks 0 and 24 Improving Cytopenias
and included bronchitis, gastroenteritis, nasopharyngitis, Pacritinib is a JAK2 and FLT3 inhibitor that distinguished itself in
and urinary tract infections. The rate of infections was 25% early testing by alleviating myelofibrosis-associated spleno-
for weeks 144 to 168. Finally, patients randomly assigned to megaly and symptoms without drug-emergent worsening of
ruxolitinib showed longer overall survival than those ran- anemia or thrombocytopenia (Table 4). The PERSIST-1 trial
domly assigned to BAT (hazard ratio 0.48). demonstrated that pacritinib was superior to the best alter-
Because all patients crossed over to ruxolitinib in both native therapy, even in those with severe thrombocytopenia
studies, it is difficult to compare the effect on long-term (i.e., platelets less than 50 3 109/L), for control of spleno-
survival. In a recent study of COMFORT-2 participants,67 megaly. Subset analysis presented at the 2015 American So-
survival from diagnosis was compared for 100 patients with ciety of Hematology Annual Meeting showed that all subgroups
intermediate-2 and high-risk primary myelofibrosis who (in terms of disease features, risks, demographics, and baseline
received ruxolitinib (the IPSS cohort) versus a comparable blood counts) of patients with myelofibrosis seemed to benefit
group of 250 patients with primary myelofibrosis who re- from pacritinib therapy.70 On the basis of these results, the
ceived conventional treatment (namely, the DIPSS cohort) manufacturers of pacritinib are seeking U.S. Food and Drug
when at the same risk. Patients treated with ruxolitinib at Administration approval for the drug for use for patients with
some point during their disease history had better survival myelofibrosis with severe thrombocytopenia, which is cur-
compared with those who continued standard treatment for rently an unmet MPN need.
the full duration of follow-up, ultimately suggesting that Investigators of a randomized phase III trial (a JAK1 and
ruxolitinib affects the natural history of primary myelofibrosis. JAK2 inhibitor) are currently enrolling participants to study

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e331


MESA AND PASSAMONTI

TABLE 5. Serial Use of Myeloproliferative Neoplasm Symptom Instruments in Randomized Clinical Trials

Disease Instrument Trial/Agent Reference


MF MF-SAF 2.0 COMFORT-1 (ruxolitinib) Verstovsek et al63
MF EORTC QLQc30 COMFORT-2 (ruxolitinib) Harrison et al64
MF mMF-SAF JAKARTA (fedratinib) Pardanani et al62
MF MPN-SAF TSS PERSIST-1 (pacritinib) Mesa et al77
MF MPN-SAF TSS PERSIST-2 (pacritinib); ongoing NCT02055781
PV MPN-SAF TSS SIMPLIFY (momelotinib); ongoing NCT0196838
PV MPN-SAF RESPONSE (ruxolitinib) Vannucchi et al78
PV MPN-SAF RELIEF (ruxolitinib) Mesa et al79
PV and ET MPN-SAF MPD-RC 112 (phase III) pegylated NCT01259856
interferon
Abbreviations: MF, myelofibrosis; MF-SAF, Myelofibrosis Symptom Assessment Form; MPN-SAF TSS, Myeloproliferative Neoplasm Symptom Assessment Form Total Symptom Score;
mMF-SAF, Myeloproliferative Neoplasm Symptom Assessment Form; PV, polycythemia vera; ET, essential thrombocythemia.

momelotinib as a first-line treatment for myelofibrosis affected both individual and aggregate symptoms for pa-
compared with ruxolitinib. Building on the experience of tients with myelofibrosis.63 Participants ability to track MPN
phase II trials, the SIMPLIFY 1 trial of momelotinib seeks to symptoms with a daily diary also gave insight into issues
demonstrate superiority of this agent, compared with rux- regarding symptomatic rebound with withdrawal of the
olitinib, for myelofibrosis-associated anemia and non- medication as well as durability of response. In a phase III
inferiority in regard to reduction in splenomegaly and study of the JAK inhibitor fedratinib, investigators also used
symptoms. patient-reported outcomes with the MF-SAF and similarly
showed improvement in individual and aggregate symp-
Combination JAK Inhibition: Ongoing Investigations toms.62 The PERSIST-1 study reported the ability to de-
Multiple combination approaches, with a base therapy of termine symptomatic improvement across all subgroups
ruxolitinib, have been tested during the past 2 years. The treated with pacritinib, and investigators subsequently an-
goals of combination therapy in the setting of myelofibrosis alyzed those individuals with severe thrombocytopenia with
are to augment single-agent ruxolitinib benefits by any of the the greatest unmet needs and the resolution of symptoms
following: improvement of cytopenias, reduction in marrow compared with the best alternative therapy.77 Use of JAK
fibrosis, or deeper or broader molecular responses (either in inhibitors as second-line therapy for polycythemia vera
driver mutations such as JAK2-V617F or CALR or associated demonstrated improvement in symptom burden alone or in
mutations such as ASXL1 or IDH1/2). Updates presented at aggregate for patients who received ruxolitinib compared
the 2015 American Society of Hematology Annual Meeting with individuals who received best alternative therapy (in
were most promising with combinations (Table 1) including whom hydroxyurea previously failed).78 In addition, these
danazol (with activity for cytopenias),72 azacitidine (de- similar methodologies were used in the RELIEF study com-
crease in blasts),73 pomalidomide (improved anemia),74 and paring patients who were treated with ruxolitinib versus hy-
panobinostat (deeper responses perhaps in fibrosis and droxyurea and those with inadequate symptomatic control.79
molecular markers).75 The addition of ruxolitinib to pegy- In the majority of ongoing clinical trials not only for myelofi-
lated interferon alfa-2a (IFN-a-2a) was also reported to help brosis but also essential thrombocythemia and polycythemia
overcome acquired resistance to single-agent interferon for vera (e.g., the randomized phase III trial of hydroxyurea vs.
patients with a mixture of MPNs.76 Although all of these pegylated IFN-a-2a; NCT01259856), researchers analyzed
trials showed benefits, given the significant activity of single- symptomatic response as an important component of disease
agent ruxolitinib, randomized trials that compare these activity, drug efficacy, and clinical trial endpoints.
agents with single-agent ruxolitinib will likely be required
to change practice patterns to include combination
SYMPTOMS AND MPNs: MAIN COURSE OR SIDE
approaches.
DISH?
Treatment for an MPN includes many potential areas of
EVOLVING OPTIONS IN MPN MANAGEMENT: clinical benefit, such as improved splenomegaly, blood
EXPERIENCE INCORPORATING SYMPTOM count, progression-free survival, and symptom resolution.
ASSESSMENT All of these areas contribute to morbidity, are related to
The current standard across evaluating patients with MPNs activity against the disease-associated clone, and may affect
participating in clinical trials includes an important baseline patient suffering and even survival. Symptom response
assessment of symptom burden and potential improvement should be considered as an integral part of assessment
with therapy (Table 5). In the inaugural phase III trials of of MPN therapy response that is as important as other
ruxolitinib in myelofibrosis, it was identified that ruxolitinib clinical features. Current response criteria for myelofibrosis,

e332 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


INDIVIDUALIZED THERAPY FOR MYELOPROLIFERATIVE NEOPLASMS

polycythemia vera, and essential thrombocythemia have all INDIVIDUALIZED CARE FOR MPNs: FUTURE
adopted symptomatic improvement as an important com- LANDSCAPE
ponent of disease response.9,80 Symptomatic response for The landscape of MPN therapy continues to evolve and does
therapy clearly must be weighed against the aggregate so around a better understanding of the effect of hetero-
impact of therapeutic efficacy. Clearly, the benefit of geneous mutation profiles, widely variable disease burden
symptomatic improvement can be diminished if significant and prognosis, and an evolving range of options that cur-
toxicity is experienced from a treatment. Individualizing care rently have a clearly important role in JAK inhibition with
for patients with MPNs requires an accurate assessment of ruxolitinib. The efficacy of pacritinib, momelotinib, combi-
prognosis, a well-informed patient, good understanding of nation therapy with ruxolitinib, or new targets such as
the morbidity and mortality a patient may expect, a com- pentraxin (PRM 151)81 or telomerase (imetelstat)82 will be
prehensive therapeutic plan, and a nuanced assessment of determined based on their effect on disease course, disease
drug efficacy and toxicity. burden, and individualized patient features.

References
1. Tefferi A, Thiele J, Vannucchi AM, et al. An overview on CALR and CSF3R 15. Scherber R, Dueck AC, Johansson P, et al. The Myeloproliferative Neoplasm
mutations and a proposal for revision of WHO diagnostic criteria for Symptom Assessment Form (MPN-SAF): international prospective valida-
myeloproliferative neoplasms. Leukemia. 2014;28:1407-1413. tion and reliability trial in 402 patients. Blood. 2011;118:401-408.
2. Tefferi A, Thiele J, Orazi A, et al. Proposals and rationale for revision of 16. Verstovsek S, Kantarjian H, Mesa RA, et al. Safety and efficacy of
the World Health Organization diagnostic criteria for polycythemia INCB018424, a JAK1 and JAK2 inhibitor, in myelofibrosis. N Engl J Med.
vera, essential thrombocythemia, and primary myelofibrosis: recom- 2010;363:1117-1127.
mendations from an ad hoc international expert panel. Blood. 2007; 17. Passamonti F, Maffioli M, Caramazza D, et al. Myeloproliferative
110:1092-1097. neoplasms: from JAK2 mutations discovery to JAK2 inhibitor therapies.
3. Geyer H, Scherber R, Kosiorek H, et al. Symptomatic profiles of patients Oncotarget. 2011;2:485-490.
with polycythemia vera: implications of inadequately controlled dis- 18. Nangalia J, Massie CE, Baxter EJ, et al. Somatic CALR mutations in
ease. J Clin Oncol. 2016;34:151-159. myeloproliferative neoplasms with nonmutated JAK2. N Engl J Med.
4. Passamonti F, Rumi E, Pietra D, et al. A prospective study of 338 patients 2013;369:2391-2405.
with polycythemia vera: the impact of JAK2 (V617F) allele burden and 19. Tefferi A, Lasho TL, Finke CM, et al. CALR vs JAK2 vs MPL-mutated or
leukocytosis on fibrotic or leukemic disease transformation and vas- triple-negative myelofibrosis: clinical, cytogenetic and molecular
cular complications. Leukemia. 2010;24:1574-1579. comparisons. Leukemia. 2014;28:1472-1477.
5. Passamonti F, Rumi E, Arcaini L, et al. Prognostic factors for thrombosis, 20. Passamonti F, Caramazza D, Maffioli M. JAK inhibitor in CALR-mutant
myelofibrosis, and leukemia in essential thrombocythemia: a study of myelofibrosis. N Engl J Med. 2014;370:1168-1169.
605 patients. Haematologica. 2008;93:1645-1651. 21. Maffioli M, Genoni A, Caramazza D, et al. Looking for CALR mutations in
6. Passamonti F. How I treat polycythemia vera. Blood. 2012;120:275-284. familial myeloproliferative neoplasms. Leukemia. 2014;28:1357-1360.
7. Passamonti F. How to manage polycythemia vera. Leukemia. 2012;26: 22. Rumi E, Passamonti F, Della Porta MG, et al. Familial chronic myelo-
870-874. proliferative disorders: clinical phenotype and evidence of disease
8. Mesa RA, Tibes R. MPN blast phase: clinical challenge and assessing anticipation. J Clin Oncol. 2007;25:5630-5635.
response. Leuk Res. 2012;36:1496-1497. 23. Milosevic Feenstra JD, Nivarthi H, Gisslinger H, et al. Whole-exome
9. Tefferi A, Cervantes F, Mesa R, et al. Revised response criteria for sequencing identifies novel MPL and JAK2 mutations in triple-negative
myelofibrosis: International Working Group-Myeloproliferative Neo- myeloproliferative neoplasms. Blood. 2016;127:325-332.
plasms Research and Treatment (IWG-MRT) and European Leuke- 24. Passamonti F, Elena C, Schnittger S, et al. Molecular and clinical features
miaNet (ELN) consensus report. Blood. 2013;122:1395-1398. of the myeloproliferative neoplasm associated with JAK2 exon 12
10. Cervantes F, Dupriez B, Passamonti F, et al. Improving survival trends in primary mutations. Blood. 2011;117:2813-2816.
myelofibrosis: an international study. J Clin Oncol. 2012;30:2981-2987. 25. Rumi E, Pietra D, Guglielmelli P, et al; Associazione Italiana per la Ricerca
11. Tefferi A, Guglielmelli P, Larson DR, et al. Long-term survival and blast sul Cancro Gruppo Italiano Malattie Mieloproliferative. Acquired copy-
transformation in molecularly annotated essential thrombocythemia, neutral loss of heterozygosity of chromosome 1p as a molecular event
polycythemia vera, and myelofibrosis. Blood. 2014;124:2507-2513; quiz associated with marrow fibrosis in MPL-mutated myeloproliferative
2615. neoplasms. Blood. 2013;121:4388-4395.
12. Tefferi A, Vardiman JW. Classification and diagnosis of myeloprolifer- 26. Klampfl T, Gisslinger H, Harutyunyan AS, et al. Somatic mutations of
ative neoplasms: the 2008 World Health Organization criteria and point- calreticulin in myeloproliferative neoplasms. N Engl J Med. 2013;369:
of-care diagnostic algorithms. Leukemia. 2008;22:14-22. 2379-2390.
13. Mesa RA, Niblack J, Wadleigh M, et al. The burden of fatigue and quality 27. Passamonti F, Rumi E. Clinical relevance of JAK2 (V617F) mutant allele
of life in myeloproliferative disorders (MPDs): an international Internet- burden. Haematologica. 2009;94:7-10.
based survey of 1179 MPD patients. Cancer. 2007;109:68-76. 28. Rumi E, Pietra D, Ferretti V, et al; Associazione Italiana per la Ricerca sul
14. Cervantes F, Dupriez B, Pereira A, et al. New prognostic scoring system Cancro Gruppo Italiano Malattie Mieloproliferative Investigators. JAK2 or
for primary myelofibrosis based on a study of the International Working CALR mutation status defines subtypes of essential thrombocythemia with
Group for Myelofibrosis Research and Treatment. Blood. 2009;113: substantially different clinical course and outcomes. Blood. 2014;123:
2895-2901. 1544-1551.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e333


MESA AND PASSAMONTI

29. Tefferi A, Wassie EA, Guglielmelli P, et al. Type 1 versus type 2 calre- morphologic diagnosis: an international study. J Clin Oncol. 2011;29:
ticulin mutations in essential thrombocythemia: a collaborative study of 3179-3184.
1027 patients. Am J Hematol. 2014;89:E121-E124. 47. Carobbio A, Antonioli E, Guglielmelli P, et al. Leukocytosis and risk
30. Rotunno G, Mannarelli C, Guglielmelli P, et al; Associazione Italiana per stratification assessment in essential thrombocythemia. J Clin Oncol.
la Ricerca sul Cancro Gruppo Italiano Malattie Mieloproliferative In- 2008;26:2732-2736.
vestigators. Impact of calreticulin mutations on clinical and hemato- 48. Landolfi R, Marchioli R, Kutti J, et al; European Collaboration on Low-
logical phenotype and outcome in essential thrombocythemia. Blood. Dose Aspirin in Polycythemia Vera Investigators. Efficacy and safety of
2014;123:1552-1555. low-dose aspirin in polycythemia vera. N Engl J Med. 2004;350:114-124.
31. Passamonti F, Thiele J, Girodon F, et al. A prognostic model to predict 49. Passamonti F, Theile J, Barbui T, et al. A prognostic model to predict
survival in 867 World Health Organization-defined essential throm- survival in WHO-defined essential thrombocythemia: a study by the
bocythemia at diagnosis: a study by the International Working Group on IWG-MRT (International Working Group for Myeloproliferative Neo-
Myelofibrosis Research and Treatment. Blood. 2012;120:1197-1201. plasms Research and Treatment). Blood. 2011;118 (suppl; abstr 1746).
32. Rumi E, Pietra D, Pascutto C, et al; Associazione Italiana per la Ricerca sul 50. Barbui T, Finazzi G, Carobbio A, et al. Development and validation of an
Cancro Gruppo Italiano Malattie Mieloproliferative Investigators. International Prognostic Score of thrombosis in WHO-Essential
Clinical effect of driver mutations of JAK2, CALR, or MPL in primary Thrombocythemia (IPSET-thrombosis). Blood. 2012;120:5128-5133;
myelofibrosis. Blood. 2014;124:1062-1069. quiz 5252.
33. Tefferi A, Guglielmelli P, Lasho TL, et al. CALR and ASXL1 mutations- 51. Finazzi G, Carobbio A, Guglielmelli P, et al. Calreticulin mutation does
based molecular prognostication in primary myelofibrosis: an in- not modify the IPSET score for predicting the risk of thrombosis among
ternational study of 570 patients. Leukemia. 2014;28:1494-1500. 1150 patients with essential thrombocythemia. Blood. 2014;124:
34. Tefferi A, Lasho TL, Finke C, et al. Type 1 vs type 2 calreticulin mutations 2611-2612.
in primary myelofibrosis: differences in phenotype and prognostic 52. Tefferi A, Rumi E, Finazzi G, et al. Survival and prognosis among 1545
impact. Leukemia. 2014;28:1568-1570. patients with contemporary polycythemia vera: an international study.
35. Tefferi A, Guglielmelli P, Lasho TL, et al. Driver mutations and prognosis Leukemia. 2013;27:1874-1881.
in 1118 patients with primary myelofibrosis. Blood. 2015;126 (suppl; 53. Mesa RA, Schwager S, Radia D, et al. The Myelofibrosis Symptom
abstr 2801). Assessment Form (MFSAF): an evidence-based brief inventory to
36. Passamonti F, Maffioli M, Caramazza D. New generation small-molecule measure quality of life and symptomatic response to treatment in
inhibitors in myeloproliferative neoplasms. Curr Opin Hematol. 2012; myelofibrosis. Leuk Res. 2009;33:1199-1203.
19:117-123. 54. Mesa RA, Gotlib J, Gupta V, et al. Effect of ruxolitinib therapy on
37. Vannucchi AM, Lasho TL, Guglielmelli P, et al. Mutations and prognosis myelofibrosis-related symptoms and other patient-reported outcomes
in primary myelofibrosis. Leukemia. 2013;27:1861-1869. in COMFORT-I: a randomized, double-blind, placebo-controlled trial.
38. Tefferi A, Lasho TL, Finke C, et al. A 27-gene NGS panel in primary J Clin Oncol. 2013;31:1285-1292.
myelofibrosis identifies ASXL1, CBL, RUNX1 and SRSF2 mutations as 55. Emanuel RM, Dueck AC, Geyer HL, et al. Myeloproliferative neoplasm
being unfavorable and absence of any non-driver mutation as being (MPN) symptom assessment form total symptom score: prospective
favorable to survival. Blood. 2015;126 (suppl; abstr 350). international assessment of an abbreviated symptom burden scoring
39. Passamonti F, Cervantes F, Vannucchi AM, et al. A dynamic prognostic system among patients with MPNs. J Clin Oncol. 2012;30:4098-4103.
model to predict survival in primary myelofibrosis: a study by the IWG- 56. Geyer HL, Scherber RM, Dueck AC, et al. Distinct clustering of symp-
MRT (International Working Group for Myeloproliferative Neoplasms tomatic burden among myeloproliferative neoplasm patients: retro-
Research and Treatment). Blood. 2010;115:1703-1708. spective assessment in 1470 patients. Blood. 2014;123:3803-3810.
40. Gangat N, Caramazza D, Vaidya R, et al. DIPSS Plus: a refined Dynamic 57. Geyer HL, Scherber RM, Dueck AC, et al. Symptom severity and clinical
International Prognostic Scoring System for primary myelofibrosis that variables of polycythemia vera patients with splenomegaly, phlebot-
incorporates prognostic information from karyotype, platelet count, omy requirements and/or hydroxyurea use: a retrospective evaluation
and transfusion status. J Clin Oncol. 2011;29:392-397. of 1334 patients. Blood. 2014;124 (suppl; abstr 1848).
41. Guglielmelli P, Lasho TL, Rotunno G, et al. The number of prognostically 58. Kroger NM, Deeg JH, Olavarria E, et al. Indication and management of
detrimental mutations and prognosis in primary myelofibrosis: an in- allogeneic stem cell transplantation in primary myelofibrosis: a con-
ternational study of 797 patients. Leukemia. 2014;28:1804-1810. sensus process by an EBMT/ELN international working group. Leukemia.
42. Vannucchi AM, Guglielmelli P, Rotunno G, et al. Mutation-Enhanced 2015;29:2126-2133.
International Prognostic Scoring System (MIPSS) for primary myelofi- 59. Kroger N, Giorgino T, Scott BL, et al. Impact of allogeneic stem cell
brosis: an AGIMM & IWG-MRT project. Blood. 2014;124 (suppl; abstr transplantation on survival of patients less than 65 years of age with
405). primary myelofibrosis. Blood. 2015;125:3347-3350, quiz 3364.
43. Tefferi A, Guglielmelli P, Finke C, et al. Integration of mutations and 60. Martnez-Trillos A, Gaya A, Maffioli M, et al. Efficacy and tolerability of
karyotype towards a Genetics-Based Prognostic Scoring System (GPSS) hydroxyurea in the treatment of the hyperproliferative manifestations
for primary myelofibrosis. Blood. 2014;124 (suppl; abstr 406). of myelofibrosis: results in 40 patients. Ann Hematol. 2010;89:
44. Cervantes F, Passamonti F, Barosi G. Life expectancy and prognostic 1233-1237.
factors in the classic BCR/ABL-negative myeloproliferative disorders. 61. Passamonti F. Balancing efficacy and safety of JAK inhibitors in mye-
Leukemia. 2008;22:905-914. lofibrosis. Leuk Res. 2014;38:290-291.
45. Barbui T, Barosi G, Birgegard G, et al; European LeukemiaNet. 62. Pardanani A, Harrison C, Cortes JE, et al. Safety and efficacy of fedratinib
Philadelphia-negative classical myeloproliferative neoplasms: critical in patients with primary or secondary myelofibrosis: a randomized
concepts and management recommendations from European Leuke- clinical trial. JAMA Oncol. 2015;1:643-651.
miaNet. J Clin Oncol. 2011;29:761-770. 63. Verstovsek S, Mesa RA, Gotlib J, et al. A double-blind, placebo-
46. Barbui T, Thiele J, Passamonti F, et al. Survival and disease progression in controlled trial of ruxolitinib for myelofibrosis. N Engl J Med. 2012;
essential thrombocythemia are significantly influenced by accurate 366:799-807.

e334 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


INDIVIDUALIZED THERAPY FOR MYELOPROLIFERATIVE NEOPLASMS

64. Harrison C, Kiladjian JJ, Al-Ali HK, et al. JAK inhibition with ruxolitinib versus 75. Harrison CN, Kiladjian JJ, Heidel FH, et al. Efficacy, safety, and confir-
best available therapy for myelofibrosis. N Engl J Med. 2012;366:787-798. mation of the recommended phase 2 starting dose of the combination
65. Verstovsek S, Mesa RA, Gotlib J, et al. Efficacy, safety and survival with of ruxolitinib (RUX) and panobinostat (PAN) in patients (pts) with
ruxolitinib in patients with myelofibrosis: results of a median 2-year myelofibrosis (MF). Blood. 2015;126 (suppl; abstr 4060).
follow-up of COMFORT-I. Haematologica. 2013;98:1865-1871. 76. Mikkelsen SU, Kjr L, Skov V, et al. Safety and efficacy of combination
66. Cervantes F, Vannucchi AM, Kiladjian JJ, et al. Three-year efficacy, therapy of interferon-alpha2 + JAK1-2 inhibitor in the Philadelphia-
safety, and survival findings from COMFORT-II, a phase 3 study com- negative chronic myeloproliferative neoplasms. preliminary results
paring ruxolitinib with best available therapy for myelofibrosis. Blood. from the Danish Combi-Trial - an open label, single arm, non-
2013;122:4047-4053. randomized multicenter phase II study. Blood. 2015;126 (suppl; abstr
67. Passamonti F, Maffioli M, Cervantes F, et al. Impact of ruxolitinib on the 824).
natural history of primary myelofibrosis: a comparison of the DIPSS and 77. Mesa RA, Egyed M, Szoke A, et al. Results of the PERSIST-1 phase III
the COMFORT-2 cohorts. Blood. 2014;123:1833-1835. study of pacritinib (PAC) versus best available therapy (BAT) in primary
68. Harrison CN, Vannucchi AM, Kiladjian JJ, et al. Long-term efficacy and myelofibrosis (PMF), post-polycythemia vera myelofibrosis (PPV-MF),
safety in COMFORT-II, a phase 3 study comparing ruxolitinib with best or post-essential thrombocythemia-myelofibrosis (PET-MF). J Clin
available therapy for the treatment of myelofibrosis: 5-year final study Oncol. 2015;33 (suppl; abstr LBA7006).
results. Blood. 2015;126 (suppl; abstr 59). 78. Vannucchi AM, Kiladjian JJ, Griesshammer M, et al. Ruxolitinib versus
69. Passamonti F, Griesshammer M, Cavo M, et al. Demographics, baseline standard therapy for the treatment of polycythemia vera. N Engl J Med.
characteristics, and disease symptom burden in RESPONSE-2: a ran- 2015;372:426-435.
domized, phase 3 study of ruxolitinib in polycythemia vera patients (pts) 79. Mesa R, Vannucchi AM, Yacoub A, et al. The efficacy and safety of
who are resistant to or intolerant of hydroxyurea (HU). Blood. 2015;126 continued hydroxyurea therapy versus switching to ruxolitinib in pa-
(suppl; abstr 2807). tients with polycythemia vera: a randomized, double-blind, double-
70. Vannucchi AM, Mesa RA, Cervantes F, et al. Analysis of outcomes by dummy, symptom study (RELIEF). Blood. 2014;124 (suppl; abstr 3168).
patient subgroups in patients with myelofibrosis treated with pacritinib 80. Barosi G, Mesa R, Finazzi G, et al. Revised response criteria for poly-
vs best available therapy (BAT) in the phase III Persist-1 Trial. Blood. cythemia vera and essential thrombocythemia: an ELN and IWG-MRT
2015;126 (suppl; abstr 58). consensus project. Blood. 2013;121:4778-4781.
71. Gupta V, Mesa RA, Deininger MW, et al. Circulating cytokines and 81. Verstovsek S, Mesa RA, Foltz LM, et al. PRM-151 in myelofibrosis:
markers of iron metabolism in myelofibrosis patients treated with durable efficacy and safety at 72 weeks. Blood. 2015;126 (suppl; abstr
momelotininb: correlatives from the Ym-387-II Study. Blood. 2015;126 56).
(suppl; abstr 1600). 82. Tefferi A, Lasho TL, Begna KH, et al. A pilot study of the telomerase
72. Gowin KL, Kosiorek HE, Dueck AC, et al. Final analysis of a multicenter inhibitor imetelstat for myelofibrosis. N Engl J Med. 2015;373:908-919.
pilot phase 2 study of ruxolitinib and danazol in patients with mye- 83. Durrant ST, Nagler A, Vannucchi AM, et al. An open-label, multicenter,
lofibrosis. Blood. 2015;126 (suppl; abstr 1618). 2-arm, dose-finding, phase 1b study of the combination of ruxolitinib
73. Daver N, Garcia-Manero G, Cortes JE, et al. 5-Azacytidine (AZA) in and buparlisib (BKM120) in patients with myelofibrosis: results from
combination with ruxolitinib (RUX) as therapy for patients (pts) with HARMONY Study. Blood. 2015;126 (suppl; abstr 827).
myelodysplastic/myeloproliferative neoplasms (MDS/MPNs). Blood. 84. Gupta V, Harrison CN, Hasselbalch H, et al. Phase 1b/2 study of the
2015;126 (suppl; abstr 823). efficacy and safety of sonidegib (LDE225) in combination with rux-
74. Stegelmann F, Bangerter M, Heidel FH, et al. A phase-Ib/II study of ruxolitinib olitinib (INC424) in patients with myelofibrosis. Blood. 2015;126 (suppl;
plus pomalidomide in myelofibrosis. Blood. 2015;126 (suppl; abstr 826). abstr 825).

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e335


HEMATOLOGIC MALIGNANCIESLEUKEMIA,
MYELODYSPLASTIC SYNDROMES, AND
ALLOTRANSPLANT

Why Are Myelodysplastic


Syndromes So Difficult to Cure?

CHAIR
Stephen Nimer, MD
Sylvester Comprehensive Cancer Center
Miami, FL

SPEAKERS
Gregory A. Abel, MD, MPH
Dana-Farber Cancer Institute
Boston, MA

Rami S. Komrokji, MD
Moffitt Cancer Center
Tampa, FL
FRAILTY, COMORBIDITY, AND QOL IN MDS

Integrating Frailty, Comorbidity, and Quality of Life in the


Management of Myelodysplastic Syndromes
Gregory A. Abel, MD, MPH, and Rena Buckstein, MD, FRCPC

OVERVIEW

Myelodysplastic syndromes (MDS) are a group of acquired hematopoietic stem cell disorders that manifest with progressive
bone marrow failure and have a propensity to transform into leukemia. Although an increase in biologic understanding of
MDS has led to improved patient risk stratification and prognostication, advances in treatment have lagged behind. While
hematopoietic cell transplantation (HCT) is a potentially curative option for some, most affected patients continue to be
treated with supportive care or with drugs that offer temporary palliation such as hematopoietic growth factors, DNA
hypomethylating agents, or immunomodulatory therapy. For several groups, such as those with intermediate-risk disease as
classified by the Revised International Prognostic Scoring System (IPSS-R) or those with higher-risk disease for whom
hypomethylating agents have failed, optimal treatment remains uncertain. Inclusion of patient-related factors such as frailty
and comorbid conditions into risk assessment can improve prognostication beyond the disease-associated variables in-
cluded in systems such as the IPSS-R. This article focuses on approaches to assessing and integrating frailty, comorbidities,
and quality of life into the treatment of patients with MDS.

M yelodysplastic syndromes are a group of acquired


hematopoietic stem cell disorders that manifest with
progressive bone marrow failure and have a propensity to
except with allogeneic HCT, treatment decisions often focus
on improving QOL.
Determining the optimal treatment strategy is difficult for
transform into acute myeloid leukemia.1 Although sup- patients with MDS due to the advanced age of affected
portive care had long been the standard for management of patients and heterogeneity in disease outcomes.10 Several
MDS,2 there are now several medications widely used for its scoring systems have been developed to help guide clinical
treatment and many others under investigation.3 Still, only decision making. These include the 1997 IPSS11 and its 2012
about 50% of patients diagnosed with MDS are alive 3 years revision, the IPPS-R,12 the World Health Organization (WHO)
after diagnosis, a figure even worse for those who develop Prognostic Scoring System (WPSS),13 and the MD Anderson
MDS related to prior treatment of cancer.4,5 The majority of Cancer Center MDS Model (MDACC Model).14 These sys-
patients with MDS ultimately die of causes related to their tems use various combinations of histologic classification,
disease, such as infection, progression to acute myeloid bone marrow blast percentage, cytogenetics, number and
leukemia, and hemorrhage.6 severity of cytopenias, performance status, age, and red cell
For people living with MDS, the burden of disease can be transfusion dependence to assign patients to different risk
highly troublesome. For example, one study estimated the categories used by clinicians to tailor treatment approaches.
3-year incidence of anemia, neutropenia, and thrombocy- These include observation alone, supportive care with
topenia to be 81%, 25%, and 41%, respectively, and 3-year transfusions, hypomethylating agents, immunomodulatory
rates of hospitalizations (for infection or bleeding associated drugs, HCT, and enrollment in clinical trials.
with low blood counts), emergency department visits, and There are two groups of patients for whom current scoring
transfusion to be 62%, 42%, and 45%.7 In addition, health systems are of limited utility. First, the systems do not help
care costs are high, estimated at $51,066 yearly (2009 US$) determine when patients with lower-risk disease at pre-
for patients who are transfusion-dependent (even without sentation should start therapy. Second, for patients in the
accounting for the cost of current drug therapies).8 More- intermediate-risk categories, it is often unclear whether
over, patients with MDS suffer from impairment to their treatments tailored to lower-risk or higher-risk patients are
quality of life (QOL),9 and, because the disease is incurable more likely to be useful. Increasingly, molecular genetic

From the Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA; Department of Medical Oncology/Hematology, Odette Cancer and
Sunnybrook Health Sciences Center, Toronto, ON, Canada.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Gregory A. Abel, MD, MPH, Dana-Farber Cancer Institute, 450 Brookline Ave., Dana 1106, Boston, MA 02215; email: gregory_abel@dfci.harvard.edu.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e337


ABEL AND BUCKSTEIN

testing can aid in risk stratification15 beyond the systems developed by Rockwood and associates, known as the Clin-
mentioned above, and, in some cases, can identify candi- ical Frailty Scale (CFS), patients were assigned scores of 1 to 9.
dates for specific clinical trials. Although such genomic A score 1 indicates very fit, 4 indicates vulnerable, and 8
advances are promising, this article will address how three indicates very severely frail (Fig. 1). The median age of en-
additional concepts not specifically addressed in the risk rolled patients was 73 (similar to the median age that MDS is
models abovefrailty, comorbidity, and QOLcan be for- diagnosed in the United States),5 and 79% of patients had
mally assessed and potentially help with treatment decisions IPSS-R scores of intermediate or lower. Frailty was found to
for patients with MDS. correlate only modestly with Eastern Cooperative Oncology
Group (ECOG) performance status (r = 0.39; p , .0001), less
with comorbidity as assessed by the MDS-specific comor-
FRAILTY bidity index (MDS-CI28; r = 0.33; p , .0001), and minimally
Although many definitions exist, geriatricians largely agree
with age-adjusted IPSS-R (r = 0.12; p = .03). Frailty improved
that frailty is a state of high vulnerability for adverse health
the prognostication of the IPSS-R in all but the highest-risk
outcomes, including disability, dependency, falls, need for
group. In a multivariate analysis that included IPSS-R and
long-term care, and mortality.16 Frailty is common in older
comorbidity scores, frailty was independently associated
patients with cancer and has been associated with treatment-
with survival (hazard ratio [HR] 2.7; 95% CI, 1.74.2).
related toxicity, poor response to therapy, and worse overall
Moreover, incorporation of frailty improved MDS risk strat-
survival.17,18 Given the synergistic effects of chemotherapy
ification by 30%.
and underlying malignancy on the immune and hematopoietic
The goal of frailty screening is to estimate a patients
systems, older patients with blood cancers such as MDS are at
physiological age when considering treatment options and
particular risk for the sequelae of frailty. For example, pre-
goals of care. It can also help predict practical outcomes,
liminary work in older patients with lymphoma,19 acute myeloid
such as a tendency toward falls or hospital admission. Frailty
leukemia,20 myeloma,21 and MDS22 has revealed that markers
screening is not easily achieved with a self-administered
of frailty are independent prognostic factors for morbidity and
questionnaire, as cognitive issues may preclude completion,
death in models that included conventional risk factors such
and several aspects of frailty are best captured through
as disease risk group, comorbidity, and performance status.
in-person functional examination. Many frailty assessment
Importantly, older patients with MDS have been shown to
instruments are based on Frieds phenotype model,29 which
be less likely to receive active treatment23 and are also less
assesses physiologic vulnerability: weight loss, poor grip
likely to receive high-quality care such as baseline marrow
strength, slow gait speed, low physical activity, and self-
cytogenetic testing.24 Although such decisions may be
reported exhaustion. An alternative model defines frailty as
reasonable for elderly patients who are frail (e.g., a di-
the cumulative effect of individual deficits and considers
agnostic bone marrow assessment with cytogenetic test-
more than 30 additional factors (e.g., self-reported health,
ing may not be appropriate if a patient is too frail for
questions about functioning in the home, and presence of
treatment), they are not reasonable for the elderly who are
chronic illnesses), creating a continuous index.30 A cumu-
robust. Stated another way, age is an imprecise proxy for
lative frailty index may have improved discriminatory ability
frailty.
compared with categorical measures and can incorporate
A recent study assessed the impact of frailty on survival
readily available clinical data.
in a large cohort of patients with MDS and chronic myelo-
Given its potential contribution to mortality, we suggest
monocytic leukemia (445 patients).25 Frailty was assessed by
that frailty be assessed in a rigorous manner for older
clinical judgment and also with a combination of physical
patients with MDS. Although the gold standard is com-
measures such as hand grip strength26 and ability to get out
prehensive geriatric assessment with a trained geriatri-
of a chair 10 times and walk 4 meters.27 Using a schema
cian,31 such an approach is not practical at most MDS
treatment facilities. The above mentioned 9-point CFS is
assessed based on clinical judgment and is highly corre-
KEY POINTS lated with the more comprehensive Canadian Study of
Health and Aging Frailty Index (which assesses between
Frailty is distinct from age, can be measured with 30-100 deficits).32 For those patients with MDS who are
precision, and is associated with survival. of advanced age, or those who seem to lack physiologic
The MDS-specific comorbidity index is a scale that capacity, we suggest that clinicians consider patients as
reliably measures comorbidity for patients with MDS. fitting within one of three larger categories of the CFS: 1-3
Depth of comorbidity can be integrated into decision
(robust to prefrail), 4 (vulnerable), and 5-8 (frail). Clinicians
making for all levels of disease risk.
Worse quality of life predicts poorer survival in MDS,
can assign a score by reviewing the patients history and
even when accounting for disease risk. soliciting input directly from patients and caregivers,
MDS-specific quality of life can be measured with asking Which do you think best describes you? and then
precision, but how to best integrate these data into reading the descriptions from the most likely categories
clinical decisions remains to be discovered. (Fig. 1; we do not suggest specifying that this is for a frailty
assessment).

e338 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


FRAILTY, COMORBIDITY, AND QOL IN MDS

FIGURE 1. The Clinical Frailty Scale

Reproduced with permission from Rockwood et al.32

Routine geriatric screening assessment of newly pre- disease itself and cannot be attributed to additional
senting elderly patients with blood cancers by a trained medical diagnoses. Thus, a patient with MDS and with no
nonphysician clinic assistant has been shown to be feasible comorbid conditionsor several comorbidities that are
and to correlate with the aggressiveness of subsequent well-managedmay still be considered frail due to the
treatment decisions.33 Unfortunately, there are currently no effects of MDS alone (such as advanced anemia and/or
guidelines regarding the incorporation of frailty assessment recurrent infections). Indeed, in the study of frailty among
into MDS clinical practice, and consensus recommenda- patients with MDS cited above,25 both frailty and Charlson
tions are needed. For example, elderly, robust patients Comorbidity Index (CCI)38 were independently prognostic
with intermediate-risk MDS may be referred for reduced- of overall survival in models that accounted for MDS
intensity conditioning HCT, while vulnerable and frail disease risk.
patients might be referred for comprehensive geriatrician Analyses of MDS-related administrative data have dem-
management. By addressing activities of daily living, mobility/ onstrated that comorbidities are associated with worse
fall risk, nutrition, poly-pharmacy, and mood, geriatric con- survival. In one study using registry data linked to Medicare
sultation has been shown to not only impact oncologic claims (SEER-Medicare; 1,708 patients), patients with
treatment decisions but also address many other comorbid comorbid conditions had greater risk of death than those
issues for older patients with cancer.34 Moreover, in a re- without comorbid conditions. Compared with patients with
cent systematic review of 18 publications assessing geriatric a CCI of 0 (no comorbidity), those with a CCI of 1 or 2
domains for patients with blood cancers, physical capacity, (HR 1.19; 95% CI, 1.051.36) and those with a CCI of 3 or
nutritional status, and comorbidity were found to have in- greater (HR 1.77; 95% CI, 1.502.08) had worse overall
dependent predictive value for survival and chemotherapy- survival.39 Another study characterized the impact of
related nonhematological toxicity and often performed better comorbidities on overall survival for patients with MDS
than age and performance status.35 using a clinical database,40 assessing comorbidity with
the Comorbidity Evaluation-27 (ACE-27) scale,41 a measure
originally developed for patients with head and neck
COMORBIDITY cancer. Among the 600 patients with MDS, 77% had at
Patients with MDS are often of advanced age at diagnosis least one comorbid condition, and, although there was no
such that the majority have at least one additional comorbid significant association between level of comorbidity and
condition.24,28 Although frailty is associated with comorbid leukemic transformation, patients with the highest level of
burden,36,37 it is felt to be an overlapping but distinct comorbidity had a 50% decrease in survival compared with
concept.16 One explanation is that much of the frailty seen in those with none, independent of age and IPSS risk group. A
patients with a syndrome such as MDS may be due to the follow-up study with the same cohort stratified by the

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e339


ABEL AND BUCKSTEIN

revised IPSS found similar results.42 In both analyses, systems.46-49 For example, one study (318 patients) found
comorbidity significantly impacted the median survival of the MDS-CI to be more effective than the HCT-CI in prog-
patients age 65 or younger but did not impact those older nostication even when accounting for age, but only for
than age 65. patients with lower-risk disease on the IPSS.46 Another study
One of the most important clinical situations in which (60 patients) found that the MDS-CI was able to identify
to formally assess comorbidity for patients with MDS is patients with improved outcomes after azacitidine,48 and
when considering fitness for HCT. Many clinicians use the yet another (363 patients) found that the MDS-CI refined the
HCT-comorbidity index (HCT-CI) for this purpose. The risk stratification of the IPSS-R.49
HCT-CI is a measure developed by Sorror et al43 that has We suggest integrating the MDS-CI or the HCT-CI with
been shown to have clinical utility specifically for patients one of the commonly used MDS scoring systems such as the
with MDS and acute myeloid leukemia.44 The HCT-CI was IPSS-R or WPSS. A schema for practically doing so with the
developed to improve on the CCI, as some elements of the WPSS is shown in Fig. 2.28 For example, for patients with
CCIsuch as presence of AIDS, comorbid leukemia, and lower WPSS who are high risk on the MDS-CI, the focus
comorbid lymphomawere not relevant for many pa- should be on preventing or ameliorating anemia, which
tients who underwent HCT. The HCT-CI is comprised of might exacerbate comorbid conditions. For patients who
17 groups of disease, each weighted with points from 1 to are low risk on the MDS-CIparticularly those lacking
3. For example, the presence of diabetes merits 1 point, cardiac conditionsmore severe anemia might be toler-
and the presence of severe pulmonary disease merits ated. As another example, for patients with higher-risk
3 points. The resulting scores on the HCT-CI are: low WPSS, clinical decisions regarding HCT can be helped with
(0 points), intermediate (1 to 2 points), and high ($ 3 the HCT-CI. If HCT is not available due to patient prefer-
points). ence or lack of suitable donor, the MDS-CI may also
In a retrospective analysis of patients with MDS from an help in addressing potential benefits of hypomethylating
Austrian registry that used the HCT-CI (616 patients),45 agents, although more research is needed to evaluate
the highest frequencies of specific comorbidities were how comorbidity scores can predict clinical outcomes and
cardiovascular disease (28%), diabetes (12%), and prior tolerance after therapy.
solid tumors (10%). Comorbidities were more frequent in
men and patients older than age 65; moreover, in mul-
tivariable models including age and IPSS, patients in higher QUALITY OF LIFE
HCT-CI groups had worse overall survival (HR 1.26; 95% CI, Quality of life is defined by the WHO as the net conse-
1.11.5). Building on the success of the HCT-CI for MDS quence of life characteristics on a persons perception of
and other blood cancers, Della Porta et al created a their position in life, in the context of the culture and value
time-dependent, MDS-specific comorbidity index, the systems in which they live, and in relation to their goals,
MDS-CI.28 expectations, standards, and concerns.50,51 The key to
To create the MDS-CI, a learning cohort of 840 Italian understanding patient-reported outcomes such as QOL is
patients with MDS was assigned comorbidities using that they are subjective and dynamic. Given that many
HCT-CI definitions. Next, multivariable models were created patients with MDS have a chronic illness with a rela-
to assess risk of nonleukemic death, and each comorbidity tively long survival and opportunity for many lived experi-
was given a score proportional to the regression coeffi- ences with the disease and its consequence, QOL has been
cient of the resulting multivariable Cox proportional haz- demonstrated to be impaired for many affected pa-
ards model. Only five of the 17 HCT-CI variables were tients.52,53 In a study of patients age 60 or older with acute
significant: cardiac disease (2 points), moderate to severe myeloid leukemia or advanced MDS (43 patients), although
hepatic disease (1 point), severe pulmonary disease initial QOL assessment was not associated with treatment
(1 point), renal disease (1 point), and current or prior history choice, 97% of patients reported that QOL was more im-
of solid tumor (1 point). Points were added together to portant to them than length of life.54 Moreover, rigorous
create low risk (0 points), intermediate risk (1 to 2 points), or measurement of QOL has been recognized as a priority for
high risk (. 2 points) scores. These categories identified MDS research.55-57
three groups that showed significantly different probabili- As an example, in one of the large clinical trials of aza-
ties of nonleukemic death and survival (p , .002 and citidine in MDS, QOL was found to be improved among
p = .005, respectively), a finding that was also repro- patients receiving therapy when assessed with a cancer-
duced in a validation cohort of German patients with MDS related measure,58 data that undoubtedly helped lead to
(504 patients). regulatory approval of that medication. More recent work
In a multivariable analysis including age, sex, WHO cate- by Efficace et al found that among patients with higher-risk
gories, cytogenetics, and transfusion-dependency, the MDS- MDS (280 patients), self-reported fatigue (measured from
CI was an independent predictor of nonleukemic death (HR the cancer-specific EORTC QLQ-C30) had prognostic value
1.89; p , .001) and survival (HR 1.67; p , .001).28 In ad- beyond standard MDS risk classification systems.59 Pa-
ditional data sets, the MDS-CI has been shown to add tients scoring equal or higher than the median on that
prognostic information beyond established risk scoring measures fatigue scale (34 or greater out of 100; higher

e340 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


FRAILTY, COMORBIDITY, AND QOL IN MDS

FIGURE 2. Schematic Representation of the Potential of Combined Use of WPSS and MDS-CI in Clinical Decision
Making in MDS

Reproduced with permission from Della Porta et al.28

scores or seen with more fatigue) had a worse median measures such as the Short-Form Health Survey (SF-36)63
overall survival than patients scoring lower than the median or cancer-specific scales such as the EOTC QLQ-C30
(HR 1.6; p = .0013). In a multivariable analysis, high-risk IPSS (EORTC-30)64 or the Functional Assessment of Cancer
score (HR 2.5 vs. intermediate-2 risk; p = .004) and higher Therapy-Anemia (FACT-An).65 Although these are useful,
fatigue score (HR 1.1 for every 10 points; p = .0007) were they may not be specific enough to capture all of the
both associated with poorer overall survival, a result also elements important to MDS-related QOL. Indeed, several
seen when risk stratifying patients with the IPSS-R and the important questions about QOL in MDS remain unanswered.
WPSS. For example, the impact of the erythropoiesis-stimulating
Studies such as these argue that patient-reported out- agents on patient-reported outcomes in MDS is still
comes should be a paramount consideration when making unknown, with much contradictory data.66 In addition,
treatment decisions for patients with MDS, not only because despite the existence of many associated studies, it is not
they can help determine if patients QOL will potentially clear whether red cell transfusions improve MDS-related
improve (or be harmed) by treatment, but also because they QOL.67
may have prognostic value. Another question that arises is Moreover, given the many ways that fatigue may affect
whether QOL should be a primary consideration when de- patient functioning, it has been shown that physicians
ciding on potentially curative therapy such as HCT. One informal QOL assessment may not reveal what patients
reason it might be a primary consideration is that pre-HCT actually experience. In a study pairing a total of 280 pa-
QOL likely adds prognostic information. Indeed, recent data tients with higher-risk MDS with 68 physicians, patient-
suggest that QOL prior to HCT for blood cancers is associated physician concordance regarding assessments of overall
with post-HCT mortality, even when controlling for per- health on a seven-point scale was found only 28% of the
formance status and comorbidity.60,61 Still, another study time (weighted kappa, 0.270).68 With no consistent way of
demonstrated that among patients with higher-risk MDS age measuring QOL in MDS clinical studies, and clinicians
60 to 75 with good performance status, scores on several imperfect assessments of patients well-being and experi-
QOL measures were not associated with ultimate receipt of ences, an MDS-specific QOL instrument is needed to cap-
reduced-intensity allogeneic HCT.62 Given that the best ture the experiences of patients with MDS in the modern
treatment of HCT-eligible older adults with MDS is not treatment era.
known, these results suggest that QOL is either not well- The Quality of Life in Myelodysplasia Scale (QUALMS)69
integrated into HCT treatment decisions or not rigorously may help address this need. Developed after a series of
assessed. focus groups with patient with MDS and providers, and
This may be because most studies assessing QOL in validated by an international group of MDS researchers
MDSincluding most of the ones aboveuse general (including the authors of this review),70 the QUALMS is a

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e341


ABEL AND BUCKSTEIN

between administrations (both p , .01). Principal com-


SIDEBAR 1. Topics Addressed in the QUALMS, an ponents analysis revealed physical burden, benefit-finding,
MDS-Specific Measure of Quality of Life70 and emotional burden subscales. Additional validation ef-
forts are ongoing to determine the utility of the mea-
Too tired for prior responsibilities
Low energy changes schedule sure and subscales both for clinical trials and for clinical
Feeling weak decision-making.
Unable to participate in activities We suggest that patients with MDS be asked specifically
Take into account fatigue when planning about their QOLat diagnosis and after treatment has
Worry about becoming burden begunusing a validated general, cancer, or MDS-specific
Felt hopelessness scale. If QOL is impaired, these data can be used together
Change in bowels with disease-specific risk factors and molecular genetic
Shortness of breath studies to help inform treatment decisions. For example,
Changing long-term plans due to health for patients with MDS who have low-risk-disease and
Trouble concentrating
might otherwise be observed, reduced QOL may sig-
Life organized around medical appointments
nify the need to start erythropoiesis-stimulating agents,
Nauseated
No energy for routine tasks transfusions, or disease-modifying treatments earlier
Family relationships strained than would otherwise be considered. Alternatively, for
Feeling grateful patients with higher-risk disease who are being treated
Getting quality information with hypomethylating agents but do not seem to be
Feel gratitude when prior took for granted responding in terms of counts, formal documentation
Bruising of improved QOL may argue in favor of continuing
Avoiding crowds treatment.
Could not do anything about disease
Disease feels unpredictable
Lack of concrete answers
CONCLUSION
No clear information
Myelodysplastic syndromes are a group of diseases that
Afraid of dying
Difficulty explaining MDS to others shorten life expectancy and negatively impact QOL. The
Worry about progressing/leukemia disease-related factors that predict survival and progres-
Anxious about tests or laboratory results sion to acute myeloid leukemia are becoming increasingly
Angry about diagnosis known but remain imprecise when patient-related factors
Worried about infection are excluded. Frailty and comorbidity have been con-
Feel limited emotional support available vincingly shown to further refine current clinical prog-
Worried about bleeding nostic risk scores. These factors are inextricably linked. We
Concerned about being a financial burden believe a baseline frailty assessment does the best job at
Concerned about losing job staging the aging in MDS, and should be considered by all
Too tired to drive
clinicians at diagnosis and before initiating treatment. The
Afraid to have sex due to blood counts
CFS is quick and simple to deploy in clinic. Patients deemed
Worried MDS treatment will stop working
Too tired to take care of family vulnerable (score = 4) or mildly frail (score = 5) should have
poorly controlled comorbidities addressed, as these may be
contributing to frailty. Frankly frail patients (score = 6 or
higher) should be referred for comprehensive geriatric as-
brief (less than 10 minute) questionnaire consisting of sessment and management, which includes comorbidity
38 items (Sidebar 1). Scored on a scale of 0 to 100 (higher assessment.
score is correlated with improved MDS-specific QOL), the We realize that it is not practical to assess every patient
QUALMS recently underwent validation in United States, with MDS formally for frailty and comorbidity, but since a
Canada, and Italy (255 patients), where patients were good medical history and physical examination will solicit
assessed twice with the instrument and several others. The most comorbidity data, given the choice, we would start
measure was internally consistent (a = 0.92) and mod- with frailty assessment. On the other hand, patients with
erately correlated with both the QLQ-C30 and FACT-An MDS who are considered for allogeneic HCT should have
scales. Moreover, patients with hemoglobin levels of both frailty and comorbidity formally assessed (CFS and
8 g/dL or lower scored lower than those with hemoglobin HCT-CI or MDS-CI), regardless of age. Finally, MDS clinicians
higher than 10 g/dL (61.8 vs. 71.1; p , .001), and must aim to both extend life as well as to improve its
transfusion-dependent patients scored lower than quality. In addition to guiding therapeutic decisions as
transfusion-independent patients (62.4 vs. 69.7; p , .01). discussed above, periodic rigorous assessment of QOL has
There was good overall test-retest reliability among pa- been shown to enhance the experiences of patients
tients with stable hemoglobin (r = 0.81), and significant with cancer71 and promises to do the same for patients
changes for patients hospitalized or with infections with MDS.

e342 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


FRAILTY, COMORBIDITY, AND QOL IN MDS

References
1. Jaffe ES, Harris NL, Stein H, et al. World Health Organization Classifi- 22. Fega KR, Abel GA, Motyckova G, et al. Non-hematologic predictors of
cation of Tumours: Pathology and Genetics, Tumors of Haematopoietic mortality improve the prognostic value of the international prognostic
and Lymphoid Tissues. Lyon, France: International Agency for Research scoring system for MDS in older adults. J Geriatr Oncol. 2015;6:288-298.
on Cancer Press; 2001. 23. Gattermann N, Kundgen A, Kellermann L, et al. The impact of age on the
2. Greenberg PL, Baer MR, Bennett JM, et al. Myelodysplastic syndromes clinical diagnosis and therapy of myelodysplastic syndromes: results from a
practice guidelines in oncology. J Natl Compr Canc Netw. 2006;4:58-77. retrospective multicenter analysis in Germany. Eur J Haematol. 2013;
3. Stone RM. How I treat patients with myelodysplastic syndromes. Blood. 91:473-482.
2009;113:6296-6303. 24. Abel GA, Cronin AM, Odejide OO, et al. Influence of patient and provider
4. Cogle CR, Craig BM, Rollison DE, et al. Incidence of the myelodysplastic characteristics on quality of care for the myelodysplastic syndromes. Br
syndromes using a novel claims-based algorithm: high number of uncap- J Haematol. Epub 2016 Feb 23.
tured cases by cancer registries. Blood. 2011;117:7121-7125. 25. Buckstein R, Wells R, Zhu N, et al. Patient-related factors independently
5. Ma X, Does M, Raza A, et al. Myelodysplastic syndromes: incidence and impact overall survival in patients with myelodysplastic syndromes: an
survival in the United States. Cancer. 2007;109:1536-1542. MDS-CAN prospective study. Br J Haematol. Epub 2016 March 15.
6. Dayyani F, Conley AP, Strom SS, et al. Cause of death in patients with 26. Bohannon RW. Reference values for extremity muscle strength ob-
lower-risk myelodysplastic syndrome. Cancer. 2010;116:2174-2179. tained by hand-held dynamometry from adults aged 20 to 79 years.
7. Lindquist KJ, Danese MD, Mikhael J, et al. Health care utilization and Arch Phys Med Rehabil. 1997;78:26-32.
mortality among elderly patients with myelodysplastic syndromes. Ann 27. Guralnik JM, Ferrucci L, Simonsick EM, et al. Lower-extremity function in
Oncol. 2011;22:1181-1188. persons over the age of 70 years as a predictor of subsequent disability.
8. Frytak JR, Henk HJ, De Castro CM, et al. Estimation of economic costs N Engl J Med. 1995;332:556-561.
associated with transfusion dependence in adults with MDS. Curr Med 28. Della Porta MG, Malcovati L, Strupp C, et al. Risk stratification based on
Res Opin. 2009;25:1941-1951. both disease status and extra-hematologic comorbidities in patients
9. Thomas ML. The impact of myelodysplastic syndromes on quality of life: with myelodysplastic syndrome. Haematologica. 2010;96:441-449.
lessons learned from 70 voices. J Support Oncol. 2012;10:37-44. 29. Fried LP, Tangen CM, Walston J, et al; Cardiovascular Health Study
10. Klepin HD, Rao AV, Pardee TS. Acute myeloid leukemia and myelo- Collaborative Research Group. Frailty in older adults: evidence for a
dysplastic syndromes in older adults. J Clin Oncol. 2014;32:2541-2552. phenotype. J Gerontol A Biol Sci Med Sci. 2001;56:M146-M156.
11. Greenberg P, Cox C, LeBeau MM, et al. International scoring system for 30. Rockwood K, Mitnitski A. Frailty in relation to the accumulation of
evaluating prognosis in myelodysplastic syndromes. Blood. 1997;89: deficits. J Gerontol A Biol Sci Med Sci. 2007;62:722-727.
2079-2088. 31. Dale W. Staging the aging when considering androgen deprivation
12. Greenberg PL, Tuechler H, Schanz J, et al. Revised international therapy for older men with prostate cancer. J Clin Oncol. 2009;27:
prognostic scoring system for myelodysplastic syndromes. Blood. 2012; 3420-3422.
120:2454-2465. 32. Rockwood K, Song X, MacKnight C, et al. A global clinical measure of
13. Malcovati L, Germing U, Kuendgen A, et al. Time-dependent prog- fitness and frailty in elderly people. CMAJ. 2005;173:489-495.
nostic scoring system for predicting survival and leukemic evolution in 33. Abel G, Hshieh T, Condron N, et al. Feasibility of routine frailty screening
myelodysplastic syndromes. J Clin Oncol. 2007;25:3503-3510. assessment for patients in a hematologic oncology clinic: Results from a
14. Kantarjian H, OBrien S, Ravandi F, et al. Proposal for a new risk model in pilot study. Presented at: 57th ASH Annual Meeting and Exposition;
myelodysplastic syndrome that accounts for events not considered in 2015; Orlando, FL.
the original International Prognostic Scoring System. Cancer. 2008;113: 34. Hamaker ME, Schiphorst AH, ten Bokkel Huinink D, et al. The effect of a
1351-1361. geriatric evaluation on treatment decisions for older cancer patients
15. Bejar R, Stevenson K, Abdel-Wahab O, et al. Clinical effect of point a systematic review. Acta Oncol. 2014;53:289-296.
mutations in myelodysplastic syndromes. N Engl J Med. 2011;364: 35. Hamaker ME, Prins MC, Stauder R. The relevance of a geriatric assessment
2496-2506. for elderly patients with a haematological malignancya systematic re-
16. Fried LP, Ferrucci L, Darer J, et al. Untangling the concepts of disability, view. Leuk Res. 2014;38:275-283.
frailty, and comorbidity: implications for improved targeting and care. 36. Garca-Garca FJ, Carcaillon L, Fernandez-Tresguerres J, et al. A new
J Gerontol A Biol Sci Med Sci. 2004;59:255-263. operational definition of frailty: the Frailty Trait Scale. J Am Med Dir
17. Handforth C, Clegg A, Young C, et al. The prevalence and outcomes of frailty Assoc. 2014;15:371.e7-371.e13.
in older cancer patients: a systematic review. Ann Oncol. 2014;26:1091-1101. 37. Gharacholou SM, Roger VL, Lennon RJ, et al. Comparison of frail patients
18. Puts MT, Tapscott B, Fitch M, et al. A systematic review of factors versus nonfrail patients $65 years of age undergoing percutaneous
influencing older adults decision to accept or decline cancer treatment. coronary intervention. Am J Cardiol. 2012;109:1569-1575.
Cancer Treat Rev. 2015;41:197-215. 38. Charlson ME, Pompei P, Ales KL, et al. A new method of classifying
19. Tucci A, Ferrari S, Bottelli C, et al. A comprehensive geriatric assessment prognostic comorbidity in longitudinal studies: development and val-
is more effective than clinical judgment to identify elderly diffuse large idation. J Chronic Dis. 1987;40:373-383.
cell lymphoma patients who benefit from aggressive therapy. Cancer. 39. Wang R, Gross CP, Halene S, et al. Comorbidities and survival in a large
2009;115:4547-4553. cohort of patients with newly diagnosed myelodysplastic syndromes.
20. Sherman AE, Motyckova G, Fega KR, et al. Geriatric assessment in older Leuk Res. 2009;33:1594-1598.
patients with acute myeloid leukemia: a retrospective study of asso- 40. Naqvi K, Garcia-Manero G, Sardesai S, et al. Association of comorbidities
ciated treatment and outcomes. Leuk Res. 2013;37:998-1003. with overall survival in myelodysplastic syndrome: development of a
21. Palumbo A, Bringhen S, Mateos MV, et al. Geriatric assessment predicts prognostic model. J Clin Oncol. 2011;29:2240-2246.
survival and toxicities in elderly myeloma patients: an International 41. Paleri V, Wight RG. Applicability of the adult comorbidity evaluation - 27
Myeloma Working Group report. Blood. 2015;125:2068-2074. and the Charlson indexes to assess comorbidity by notes extraction in a

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e343


ABEL AND BUCKSTEIN

cohort of United Kingdom patients with head and neck cancer: a ret- adults: recommendations from the European LeukemiaNet. Blood.
rospective study. J Laryngol Otol. 2002;116:200-205. 2013;122:2943-2964.
42. Daver N, Naqvi K, Jabbour E, et al. Impact of comorbidities by ACE-27 in 58. Kornblith AB, Herndon JE II, Silverman LR, et al. Impact of azacytidine on
the revised-IPSS for patients with myelodysplastic syndromes. Am J the quality of life of patients with myelodysplastic syndrome treated
Hematol. 2014;89:509-516. in a randomized phase III trial: a Cancer and Leukemia Group B study.
43. Sorror ML, Maris MB, Storb R, et al. Hematopoietic cell transplantation J Clin Oncol. 2002;20:2441-2452.
(HCT)-specific comorbidity index: a new tool for risk assessment before 59. Efficace F, Gaidano G, Breccia M, et al. Prognostic value of self-reported
allogeneic HCT. Blood. 2005;106:2912-2919. fatigue on overall survival in patients with myelodysplastic syndromes:
44. Sorror ML, Sandmaier BM, Storer BE, et al. Comorbidity and disease a multicentre, prospective, observational, cohort study. Lancet Oncol.
status based risk stratification of outcomes among patients with acute 2015;16:1506-1514.
myeloid leukemia or myelodysplasia receiving allogeneic hematopoi- 60. Hamilton BKR, Abounder L, Dabney DM, et al. Prognostic significance
etic cell transplantation. J Clin Oncol. 2007;25:4246-4254. of quality of life in patients undergoing allogeneic hematopoietic cell
45. Bammer C, Sperr WR, Kemmler G, et al. Clustering of comorbidities is transplantation. Presented at: 56th ASH Annual Meeting and Exposi-
related to age and sex and impacts clinical outcome in myelodysplastic tion; 2014; San Francisco, CA.
syndromes. J Geriatr Oncol. 2014;5:299-306. 61. Wood WAL-R, Fei J, Logan MBR, et al. Patient-reported quality of life is
46. Balleari E, Salvetti C, Del Corso L, et al. Age and comorbidities deeply an independent predictor of survival after allogeneic hematopoietic
impact on clinical outcome of patients with myelodysplastic syndromes. cell transplantation: a secondary analysis from the Blood and Marrow
Leuk Res. 2015;39:846-852. Transplant Clinical Trials Network (BMT CTN) 0902. Presented at: 56th
47. Breccia M, Federico V, Latagliata R, et al. Evaluation of comorbidities at ASH Annual Meeting and Exposition; 2014; San Francisco, CA.
diagnosis predicts outcome in myelodysplastic syndrome patients. Leuk 62. El-Jawahri A, Kim H, Steensma D, et al. Does quality of life impact the
Res. 2011;35:159-162. decision to pursue stem cell transplantation for elderly patients with
48. Breccia M, Salaroli A, Loglisci G, et al. MDS-specific comorbidity index is advanced MDS? BMT. Epub 2016 March 21.
useful to identify myelodysplastic patients who can have better out- 63. McHorney CA, Ware JE Jr, Lu JF, et al. The MOS 36-item Short-Form
come with 5-azacitidine. Haematologica. 2012;97:e2. Health Survey (SF-36): III. Tests of data quality, scaling assumptions,
49. van Spronsen MF, Ossenkoppele GJ, Holman R, et al. Improved risk and reliability across diverse patient groups. Med Care. 1994;32:
stratification by the integration of the revised international prognostic 40-66.
scoring system with the myelodysplastic syndromes comorbidity index. 64. Aaronson NK, Ahmedzai S, Bergman B, et al. The European Organization
Eur J Cancer. 2014;50:3198-3205. for Research and Treatment of Cancer QLQ-C30: a quality-of-life in-
50. World Health Organization. Study protocol for the World Health Or- strument for use in international clinical trials in oncology. J Natl Cancer
ganization project to develop a Quality of Life assessment instrument Inst. 1993;85:365-376.
(WHOQOL). Qual Life Res. 1993;2:153-159. 65. Cella D. The Functional Assessment of Cancer Therapy-Anemia (FACT-
51. World Health Organization. The World Health Organization Quality An) Scale: a new tool for the assessment of outcomes in cancer anemia
of Life assessment (WHOQOL): position paper from the World Health and fatigue. Semin Hematol. 1997;34(3, Suppl 2)13-19.
Organization. Soc Sci Med. 1995;41:1403-1409. 66. Rizzo JD, Somerfield MR, Hagerty KL, et al. Use of epoetin and dar-
52. Efficace F, Gaidano G, Breccia M, et al. Prevalence, severity and cor- bepoetin in patients with cancer: 2007 American Society of Hematology/
relates of fatigue in newly diagnosed patients with myelodysplastic American Society of Clinical Oncology clinical practice guideline update.
syndromes. Br J Haematol. 2015;168:361-370. Blood. 2008;111:25-41.
53. Steensma DP, Heptinstall KV, Johnson VM, et al. Common troublesome 67. Pinchon DJ, Stanworth SJ, Doree C, et al. Quality of life and use of red cell
symptoms and their impact on quality of life in patients with myelo- transfusion in patients with myelodysplastic syndromes. A systematic
dysplastic syndromes (MDS): results of a large internet-based survey. review. Am J Hematol. 2009;84:671-677.
Leuk Res. 2008;32:691-698. 68. Caocci G, Voso MT, Angelucci E, et al. Accuracy of physician assessment
54. Sekeres MA, Stone RM, Zahrieh D, et al. Decision-making and quality of of treatment preferences and health status in elderly patients with
life in older adults with acute myeloid leukemia or advanced myelo- higher-risk myelodysplastic syndromes. Leuk Res. 2015;39:859-865.
dysplastic syndrome. Leukemia. 2004;18:809-816. 69. Abel GA, Klaassen R, Lee SJ, et al. Patient-reported outcomes for the
55. Caocci G, La Nasa G, Efficace F. Health-related quality of life and myelodysplastic syndromes: a new MDS-specific measure of quality of
symptom assessment in patients with myelodysplastic syndromes. Expert life. Blood. 2014;123:451-452.
Rev Hematol. 2009;2:69-80. 70. Abel GA, Efficace F, Buckstein RJ, et al. Prospective international val-
56. Estey E. Acute myeloid leukemia and myelodysplastic syndromes in idation of the Quality of Life in Myelodysplasia Scale (QUALMS).
older patients. J Clin Oncol. 2007;25:1908-1915. Haematologica. Epub 2016 March 4.
57. Malcovati L, Hellstrom-Lindberg E, Bowen D, et al; European Leukemia 71. Cancer Quality Council of Ontario. Symptom Assessment and Man-
Net. Diagnosis and treatment of primary myelodysplastic syndromes in agement. Toronto, Canada: Cancer Quality Council of Ontario; 2016.

e344 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


BEYOND HYPOMETHYLATING AGENTS IN MDS

Searching for a Light at the End of the Tunnel? Beyond


Hypomethylating Agents in Myelodysplastic Syndromes
Rami S. Komrokji, MD

OVERVIEW

Hypomethylating agents (HMAs) are the mainstay treatment of patients with myelodysplastic syndromes. Hypomethylating
agents remain the only treatment, other than allogeneic hematopoietic stem cell transplantation (AHSCT), that improves
overall survival (OS) for patients with higher-risk myelodysplastic syndromes. It is crucial to maximize the benefit of HMAs by
selecting the appropriate dosing and schedule and continuing therapy until clear evidence of lack of response or failure of
therapy. Strategies to improve outcome with HMAs include identifying tools and biomarkers for better patient selection,
namely the ability to identify those who achieve complete response (CR) or long duration of response, combination
strategies with HMAs aim to improve response rate or its duration. The outcome of patients with myelodysplastic syndromes
after HMA failure is poor for both patients with higher-risk and lower-risk disease and represents an unmet medical need.
The best outcomes after HMA failure are reported with AHSCT or novel agents in clinical trials. This article discusses the
maximizing benefit of HMAs, offers strategies to improve outcome with HMAs, and, finally, reviews selected novel agents in
development after HMA failure.

M yelodysplastic syndromes are a heterogeneous group


of neoplastic hematopoietic stem cell disorders. The
common features of the disease include (1) clinical pre-
with expected OS measured in years and the goal of therapy
being alleviation of cytopenia, or higher-risk myelodysplastic
syndromes, with a short expected OS and therapy attempting
sentation with variant cytopenias and resultant complica- to alter the natural history of disease. The treatment options
tions; (2) the presence of dysplastic morphologic cytology for myelodysplastic syndromes remain limited. AHSCT is the
features; (3) evidence of clonal hematopoiesis for the ma- only curative option, which is offered to patients with higher-
jority of patients, either by detection of chromosomal ab- risk myelodysplastic syndromes.9 The HMAs azacitidine and
normalities, often copy number variation changes, or by decitabine are the only approved drugs for nondeletion 5q
identification of somatic gene mutations most commonly myelodysplastic syndrome.
involving epigenetic deregulation or splicing machinery; and Epigenetic gene silencing is a well-described mechanism
(4) a tendency to progress to acute myeloid leukemia by which tumor cells downregulate expression of tumor
(AML).1-3 suppression genes.10 Silencing is often accomplished by
The first step in managing myelodysplastic syndromes is methylation or histone deacylation. Hypomethylating agents
risk stratification after ensuring the proper diagnosis. The are a class of nucleoside analogs that induce hypomethylation,
disease staging not only provides the needed prognostic allowing re-expression of epigenetically silenced genes. These
information for patients and caregivers, but it also allows agents inhibit DNA methyl transferase by forming direct ad-
physicians to tailor therapy accordingly. The International ducts after incorporation into DNA, resulting in global hypo-
Prognostic Scoring System (IPSS) had been the most widely methylation. Both agents are pyrimidine analogs; however,
used tool for risk assessment in myelodysplastic syndromes. decitabine as a deoxy-analog is incorporated solely into DNA,
A lump score is based on the percentage of myeloblasts, whereas azacitidine has a ribose sugar structure and is
cytopenia(s), and karyotype.4 New risk models including the incorporated into RNA.11,12
revised IPSS5 and MD Anderson models6,7 refine the staging This article aims to briefly summarize the current data
system in myelodysplastic syndromes. The integration of using HMAs for treatment of myelodysplastic syndromes,
somatic gene mutations information adds independent discuss possible strategies to improve outcome using HMAs,
prognostic value to the clinical risk models.8 Patients are and focus on options for managing myelodysplastic syn-
classified as having lower-risk myelodysplastic syndromes, dromes after HMA failure.

From the Department of Malignant Hematology, Moffitt Cancer Center, Tampa, FL.

Disclosures of potential conflicts of interest provided by the author are available with the online article at asco.org/edbook.

Corresponding author: Rami S. Komrokji, MD, Department of Malignant Hematology, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612; email: rami.komrokji@moffitt.org.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e345


RAMI S. KOMROKJI

HOW TO OPTIMIZE CURRENT USE OF In lower-risk myelodysplastic syndromes, the goal of HMA
HYPOMETHYLATING AGENTS FOR TREATMENT use is to alleviate cytopenia. The 5-day regimen of azaciti-
OF MYELODYSPLASTIC SYNDROMES dine every 28 days emerged as an acceptable regimen for
Hypomethylating agents are currently the standard of care patients with lower-risk myelodysplastic syndromes. Lyons
for treating patients with higher-risk myelodysplastic syn- et al19 randomly assigned patients with less than 10% my-
dromes. Azacitidine demonstrated an improvement in OS eloblasts to treatment with azacitidine at a dose of 75 mg/m2
compared with conventional care regimens in a randomized for 5 days followed by 2 days of rest and then an additional
phase III clinical trial. In the AZA-001 study, the median OS 2 days of treatment (5-2-2), 50 mg/m2 for 5 days followed by
was significantly prolonged with azacitidine treatment: 2 days of rest and then another 5 days (5-2-5), or 75 mg/m2
24.4 months versus 15 months with conventional care for 5 consecutive days. Among the 151 patients enrolled in
regimens (p = .0001). The regimen consisted of 75 mg/m2 the study, no difference in response was observed among
of azacitidine subcutaneously for 7 days every 28 days.13 the three regimens. Grade 3 or 4 hematologic toxicity was
For decitabine use in higher-risk myelodysplastic syn- lower among patients treated with the 5-day consecutive
dromes, a phase III study comparing the U.S. Food and regimen.19
Drug Administrationapproved decitabine inpatient reg-
imen with best supportive care failed to show an OS POTENTIAL STRATEGIES TO IMPROVE OUTCOME
advantage.14 The median OS with the commonly used WITH HYPOMETHYLATING AGENTS IN
20 mg/m2 intravenously for a 5-day regimen every MYELODYSPLASTIC SYNDROMES
28 days in higher-risk myelodysplastic syndromes is 19 Biomarker-Driven Patient Selection
to 20 months. 15,16 There are no clinical parameters that can clearly predict
The key points in optimizing HMA use for patients with response to HMAs. The MDS Clinical Research Consortium
higher-risk myelodysplastic syndromes include using a 7-day explored the prognostic utility of the IPSS, the IPSS-R, the
azacitidine regimen, assessing initial response after four to MD Anderson Prognostic Scoring System, the World Health
six cycles of therapy, and continuing therapy for patients Organizationbased Prognostic Scoring System, and the
with stable disease or better response. The HMA emerged French Prognostic Scoring System among 632 patients who
as an acceptable alternative to intensive chemotherapy presented with higher-risk myelodysplastic syndromes and
prior to AHSCT.17 The role of HMA maintenance or treat- were treated with azanucleosides as the first-line ther-
ment of post-AHSCT relapse is being explored and could be a apy.20 No prognostic tool predicted the probability of
promising strategy.18 achieving an objective response. Nonetheless, all five tools
Objective clinical responses are seen for only approxi- were associated with OS. The Groupe-Francophone des
mately one-half of the patients treated with HMAs, and the My e lodysplasies (GFM) proposed a prognostic score for
CR rate is only 10% to 20%.13,15 Even among patients with azacitidine-treated patients with higher-risk myelodys-
initial responses to HMA, the median response duration plastic syndromes. Among 282 patients, the study sug-
is only 9 to 15 months, with the vast majority of patients gested that prior treatment with low-dose cytarabine
losing response within 2 years. (p = .009), bone marrow blasts greater than 15% (p = .004),
and abnormal karyotype (p = .03) independently predicted
lower response rates. Complex karyotype predicted shorter
KEY POINTS responses (p = .0003).21
Detection of certain somatic gene mutations may affect
Hypomethylating agents are the only drugs that response to HMAs. The presence of TET-2, DNMT3A, and
potentially alter the natural history of myelodysplastic IDH-1/IDH-2 gene mutations was reported to be associated
syndromes and improve overall survival in higher-risk with a higher response to HMAs. Among 213 patients with
disease. myelodysplastic syndromes treated with HMAs, Bejar et al22
Key points in maximizing benefit of HMAs include reported that clonal TET-2 mutations (allele fraction . 10%)
using a 7-day regimen in higher-risk myelodysplastic predicted response (odds ratio, 1.99; p = .036). Response
syndromes and continuing treatment for patients with rates were highest in the subset of patients with a TET-2
stable disease or better response.
mutant without clonal ASXL1 mutations (odds ratio, 3.65;
Strategies to improve outcome with HMAs include
biomarker-driven patient selection, combination
p = .009). Mutations of TP53 (hazard ration [HR] 2.01;
strategies to improve response rates or duration, and p = .002) and PTPN11 (HR 3.26; p = .006) were associated
exploration of alternate doses. with shorter OS but not drug response.22
The outcome after HMA failure is poor and represents
an unmet medical need.
The best outcomes after HMA failure are reported Combination Strategies
among patients who proceed to allogeneic stem cell The goals of this strategy would be to increase the fre-
transplantation or enroll in clinical trials studying novel quency, quality, and duration of clinical responses and ul-
agents. timately delay/prevent the development of resistance and
prolong survival.23

e346 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


BEYOND HYPOMETHYLATING AGENTS IN MDS

A promising overall response rate (ORR) of 72% was reported There is unmet need for treating patients with myelo-
in an early phase trial of 36 patients with higher-risk myelo- dysplastic syndromes with TP53 mutation. The outcome for
dysplastic syndromes who received azacitidine at a standard these patients is poor.33,34 The median OS for patients with
dose of 75 mg/m2 per day concurrently with lenalidomide.24 detectable TP53 mutation at the time of AHSCT is less than
The North American Intergroup Study SWOG S1117 un- 1 year.35 Responses to HMAs, although not different, are
fortunately did not confirm those findings. In this phase II often short lived.22 APR-246 covalently binds to cysteines in
study, 277 patients were randomly assigned to azacitidine, mutant TP53 or TP63. The drug reconstitutes wild-type
azacitidine/lenalidomide, or azacitidine plus vorinostat. No conformation and function in mutant proteins by stabilizing
differences in response rate were observed; a tendency of protein folding.36 Preclinical data suggest intrinsic and ad-
longer duration of response (. 6 on therapy) was noted for the ditive in vitro schedule-dependent cytotoxicity with azaci-
azacitidine/lenalidomide arm. Overall response rate for pa- tidine when administered in sequential fashion (unpublished
tients with chronic myelomonocytic leukemia (CMML) was data). The MDS Clinical Research Consortium is launching a
significantly higher for lenalidomide plus azacitidine versus phase I/II study of APR-246 and azacitidine among patients
azacitidine (63% vs. 29%; p = .04), with a trend for longer with higher-risk myelodysplastic syndromes with TP53
response duration for combinations (p = .06).25 mutation. In addition, TP53 mutation is associated with
Histone deacetylase (HDAC) inhibitors are another group overexpression of PD-L1 and CTLA-4, which provides the
of agents that have been combined with azacitidine. As rationale to explore PD-L1 inhibitors and CTLA-4 in-
mentioned, the azacitidine/vorinostat combination did not hibitors in this population (unpublished data).
improve the response rate or OS over azacitidine mono- Other selected current HMA combination clinical trials in
therapy.25 In a randomized phase II trial of 150 patients, myelodysplastic syndromes are summarized in Table 1.
azacitidine/entinostat also did not improve outcomes.26
Finally, a randomized phase II study combining pracinostat
with azacitidine did not improve response rates.27 It is clear Alternative Dosing and Schedules
that concomitant use of HMAs and HDAC inhibitors does not Recent data presented on the 3-day use of azacitidine or
improve responses and an alternative schedule or sequence decitabine in lower-risk myelodysplastic syndromes are
must be pursued if such a combination is to be further encouraging. Among 91 patients with lower-risk myelo-
explored. dysplastic syndromes who were treated with either azaci-
There are ongoing or planned studies for other agents in tidine or decitabine in a 3-day regimen every 28 days, the
combination with HMAs. Vadastuximab talirine (SGN-CD33 ORR was 59%, and 29% of patients became RBC transfusion
A), a conjugated monoclonal antibody with a highly stable independent. Treatment was well tolerated and 1-year OS
linker, is a promising agent. An AML combination with was 86%.37 A randomized clinical trial comparing a 5-day
azacitidine or decitabine was explored in a phase I study in azacitidine regimen, a 3-day azacitidine regimen, or a 3-day
which six of 23 patients achieved CR and nine other patients decitabine is currently ongoing. The study also includes
had complete remission with incomplete count recovery randomly assigning nontransfusion-dependent patients to
(CRi). Seventy-two percent of patients were alive at a me- observation versus treatment.
dian 7.7 months of follow-up. Prolonged myelosuppression
was observed with treatment.28 In subtypes of myelodys- TABLE 1. Selected Hypomethylating Agent
plastic syndromes, PD-L1 expression on myeloblasts has Combination Strategies
been independently associated with myelodysplastic syn-
drome transformation to AML.29 Hypomethylating agents Drug Target Study Identifier
can enhance the expression of CTLA-4, PD-1, and PD-L1 Nivolumab PD-1 NCT02530463
among patients with myelodysplastic syndromes and AML.30
Lirilumab and nivolumab KIR and PD-1 NCT02599649
Ongoing and planned studies are exploring combining PD-L1
Atezolizumab PD-L1 NCT02508870
and PD-1 inhibitors in higher-risk myelodysplastic syn-
Deferasirox (ICL670) NF-kB NCT02038816
dromes. A study examining oral azacitidine combined with
darvalumab is ongoing. Pevonedistat (MLN4924; TAK-924) Eltrombopag TPO NCT02158936
is a first-in-class small molecule inhibitor of the neural Ibrutinib BTK NCT02553941
precursor cell expressed developmentally downregulated PF-04449913 Hedgehog pathway NCT02367456
protein 8activating enzyme.31 Pevonedistat had reported LDE255 Hedgehog pathway NCT02323139
single-agent clinical activity in a phase I study of patients Erismodegib Hedgehog pathway NCT02129101
with relapsed/refractory AML.31 Based on nonclinical Rigosertib PLK-1 NCT01926587
studies in AML models that demonstrated a synergistic le- Volasertib PLK-1 NCT01957644
thality in cell lines and tumor regression in murine xeno-
Sirolimus mTOR NCT01869114
grafts when pevonedistat was combined with azacitidine,
Vosaroxin Topoisomerase II NCT01913951
pevonedistat is being studied in combination with azaciti-
Abbreviations: HMA, hypomethylating agent; KIR, killer-cell immunoglobulin-like
dine in treatment-naive elderly patients with AML and receptor; NF-kB, nuclear factor-kappa B; TPO, thrombopoietin; BTK, Bruton tyrosine
higher-risk myelodysplastic syndromes.32 kinase; PLK, polo-like kinase.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e347


RAMI S. KOMROKJI

DEFINITION OF HYPOMETHYLATING AGENT Acute myeloid leukemia arising from myelodysplastic


FAILURE AND THE EXPECTED OUTCOME syndrome is also characterized by a particularly poor prog-
For higher-risk myelodysplastic syndromes, outcome after nosis, especially if prior HMA has been received.38,45 The
HMA failure is poor.38-40 The median OS ranges from 4 to median OS among 74 patients with higher-risk myelodys-
6 months. Approximately 20% to 30% of patients progress to plastic syndromes who developed secondary AML after
AML. Hypomethylating agent treatment failure can be di- azacitidine failure was 34 months with a 1-year survival
vided into primary or secondary HMA failure. Primary failure probability of only 8%.38 In another report, prior therapy with
is defined as lack of initial response in which there is clear HMAs for patients with myelodysplastic syndromes who
evidence of progressive disease. Stable disease is more progressed to AML was independently associated with in-
controversial where no definitive increase in myeloblasts is ferior response and survival after intensive chemotherapy.45
observed, but on the other hand no hematologic im-
provement is achieved by the International Working Group
(IWG 2006) criteria. In a landmark analysis of the AZA-001 HOW TO TREAT PATIENTS WITH
study, patients who achieved hematologic improvement or MYELODYSPLASTIC SYNDROMES AFTER
better response at 3, 6, and 9 months had better OS while HYPOMETHYLATING AGENT FAILURE WHEN
receiving azacitidine treatment compared with conventional THERE IS NO CLINICAL TRIAL
care regimens. For patients with stable disease, OS was the There are no standard treatment options for patients with
same for azacitidine and conventional care regimens. myelodysplastic syndromes after HMA failure. In lower-risk
However, 19% of patients treated with azacitidine who had myelodysplastic syndromes, lenalidomide may be used for
stable disease at 3 months achieved hematologic im- treatment of anemia; however, responses are much lower
provement or better at 6 months compared with 13% for when lenalidomide is used as a second-line therapy after
patients treated with conventional care regimens. At HMA failure rather than as first-line treatment after ery-
9 months, only 14% of patients who had stable disease with throid stimulating agents in nondeletion 5q myelodysplastic
azacitidine at 6 months achieved hematologic improvement syndrome.46
or better, whereas none of the patients who received In higher-risk myelodysplastic syndromes, few studies
conventional care regimens did.41 Among 291 patients have addressed sequential use of HMAs. In a study of 14
treated with HMA, Nazha et al42 reported that 55% of pa- patients, Borthakur et al47 reported that 28% responded to
tients achieved their best response at 4 to 6 months. Among decitabine after azacitidine failure. In another study, 21
patients with stable disease at 4 to 6 months, 29 (20%) patients (19%) responded to decitabine after azacitidine
achieved a better response at a later treatment time point. failure and 10 (40%) responded to azacitidine after deci-
Younger patients with lower bone marrow blast percentages tabine failure.48 Harel et al reported response to decitabine
and intermediate risk per the IPSS-R were more likely to after azacitidine treatment among 7 of 37 patients (19%).49
achieve a better response after stable disease at 4 to Five of those responses were marrow CR or stable disease.49
6 months. Patients with stable disease who subsequently These studies are limited by their retrospective nature, lack
achieved CR had superior OS compared with patients who of standard definition of HMA failure, and inclusion of pa-
remained with stable disease (28.1 vs. 14.4 months, re- tients who were nontolerant to the first agent.
spectively; p = .04).42 The GFM reported no response to low-dose chemotherapy
The median OS for primary HMA failure was 5.5 months after azacitidine failure among 18 evaluable patients. AHSCT
in the rigosertib randomized clinical study.43 Second- offers the best outcome for patients after HMA failure. An
ary HMA failure is defined as a clear loss of initial re- analysis of 435 patients with higher-risk myelodysplastic
sponse (hematologic improvement or better by IWG 2006 syndromes for whom azacitidine failed demonstrated that
criteria). participants who were offered an AHSCT or investigational
Outcome after HMA failure is also reported to be poor for agents had better survival compared with those offered
patients with lower-risk myelodysplastic syndromes. The supportive care or conventional chemotherapy, whether
MDS Clinical Research Consortium reported median OS of low or intensive dose. The median OS after best supportive
17 months after HMA failure.44 The definition of treatment care, low-dose chemotherapy, intensive-dose chemother-
failure in lower-risk myelodysplastic syndromes is strictly apy, investigational therapy, or AHSCT was 4.1, 7.3, 8.9, 13.2,
based on hematologic improvement, where there is lack of and 19.5 months, respectively.38
hematologic improvement in primary failure, and hemato-
logic response is lost after treatment in secondary failure.
Primary failure should be assessed after at least four to six NOVEL AGENTS FOR TREATMENT OF
cycles, in which treatment can be discontinued if there is no MYELODYSPLASTIC SYNDROMES AFTER
clear evidence of hematologic improvement. There is HYPOMETHYLATING AGENT FAILURE
probably no value of achieving stable disease without he- A comprehensive review of all novel agents tested after HMA
matologic improvement in lower-risk myelodysplastic syn- failure is beyond the scope of this review. The following are
dromes, especially if myeloblasts are not increased. selected examples of current agents in development.

e348 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


BEYOND HYPOMETHYLATING AGENTS IN MDS

Oral Azacitidine Among patients with HMA failure, 40% CRi/partial response
50
An oral formulation of azacitidine has been evaluated in was reported.57,58 In another study among patients with
three clinical studies to date. Results from a pilot study AML receiving tosedostat in combination with low-dose
indicated that azacitidine in an oral formulation was bio- cytarabine, a CR rate of 48.5% was reported for 16 of 33
available.51 This was confirmed when evaluating several patients; 33% of patients were still in remission after a
different formulations of the drug in a phase I study. Results median follow-up of 506 days.
from another phase I study of CC-486 among patients with Analysis of these patients cells identified a molecular
myelodysplastic syndrome, CMML, or AML indicated that signature associated with clinical response (clinical re-
administration on different treatment schedules (7 , 14 , and sponse vs. no clinical response). Results suggested that
21 days once a day and 14 and 21 days twice a day) was differentially expressed genes were associated with clinical
feasible and generally well tolerated and exhibited biologic response, including among biologically relevant pathways
and clinical activity.52 Responses for patients previously such as beta-catenin, tumor necrosis factor-alpha, nuclear
treated with subcutaneous/intravenous azacitidine were re- factor-kappa B, erythroblastosis oncogene B (ERB2), in-
ported. Oral azacitidine alone or in combination with a PD-L1 flammatory response, and epithelial-mesenchymal transi-
inhibitor is being tested for patients with HMA stable disease tion pathways.59
or after failure.
Thrombopoietin Stimulants
SGI-110 Thrombocytopenia treatment is challenging in myelodys-
SGI-110 is a dinucleotide of decitabine and deoxyguanosine plastic syndromes. Severe thrombocytopenia is more
that protects it from deamination. In a phase I study that commonly encountered in higher-risk myelodysplastic
included 14 patients with myelodysplastic syndromes after syndromes.60 Hypomethylating agents are currently the only
HMA failure, SGI-110 had a 4.5-fold longer half-life than available therapy. Romiplostim is an Fc-fusion dipeptide
decitabine. An equivalent or higher area under the curve was ligand of the thrombopoietin receptor. In a double-blind,
reached with lower Cmax compared with reference levels placebo-controlled study, 167 and 83 patients with low-
from intravenous decitabine (20 mg/m2). A dose-dependent or intermediate-risk myelodysplastic syndromes and
increase in demethylation was observed up to 60 mg/m2 thrombocytopenia were treated with 750 mg of romiplostim
daily for 5 days.53 In the phase II part of the study for per week or placebo, respectively. Results showed a sig-
treatment-naive elderly patients with AML or refractory/ nificant decrease in clinically relevant bleeding events for
relapsed AML, 43% and 16% remission rates were reported, romiplostim-treated patients with a baseline platelet count
respectively.54 of 20 to 50 3 109 cells/L (relative risk 0.35; p , .0001). In
addition, romiplostim produced a significant decrease in
Rigosertib platelet transfusion events versus placebo for patients with a
Rigosertib is a novel small molecule that targets pathways baseline platelet count of less than 20 3 109 cells/L (relative
including phosphoinositide-3 kinase and polo-like kinase. risk 0.71; p , .0001). Originally there was a concern about
Initial studies with both oral and intravenous rigosertib
indicated clinical activity in myelodysplastic syndromes and TABLE 2. Novel Agents in Myelodysplastic Syndromes
AML.55,56 Results from ONTIME, a phase III randomized After HMA Failure
clinical study comparing intravenous rigosertib with best
supportive care after HMA failure, were recently reported. Drug Target Study Identifier
ONTIME enrolled 299 patients and rigosertib was admin- Durvalumab PD-L1 NCT02281084
istered as 1,800 mg/24 hours for 72 hours as a continuous Atezolizumab PD-L1 NCT02508870
intravenous ambulatory infusion. The median OS was
Ipilimumab CTLA-4 NCT01757639
8.2 months for patients who received rigosertib compared
Nivolumab PD-1 NCT02530463
with 5.9 months for best supportive care (p = .33). For
patients with primary HMA failure, the median OS was Lirilumab and KIR and PD-1 NCT02599649
Nivolumab
8.6 months with rigosertib compared with 5.3 months for
FT-1101 BET NCT02543879
best supportive care (p = .04).43 Further studies are being
Omacetaxine NCT02159872
conducted specifically for patients with higher-risk disease
and primary HMA failure. Rigosertib in combination with CPI-613 Ketoglutarate dehydrogenase NCT01520805
azacitidine is being explored. TEN-010 Bromodomain NCT02308761
Selinexor Selective nuclear export NCT02228525

Tosedostat ASTX727 Oralcytidinedeaminaseinhibitor NCT02103478


E7727 with oral decitabine
Tosedostat is an aminopeptidase inhibitor that leads to
Vosaroxin Topoisomerase II NCT01980056
cellular amino acid deprivation. In a phase I study with 44
Abbreviations: MDS, myelodysplastic syndrome; HMA, hypomethylating agent; KIR,
participants, the maximum tolerated dose was reached at killer-cell immunoglobulin-like receptor; BET, bromodomain and extra-terminal family
180 mg with hepatic toxicity as the dose-limiting toxicity. of proteins.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e349


RAMI S. KOMROKJI

increased risk of AML transformation; however, this was not pattern. At the plenary session of 2015 American Society of
confirmed on longer follow-up.61 Results of a multicenter, Hematology Annual Meeting, Lee et al66 presented data
randomized, double-blind, placebo-controlled phase II study demonstrating that the presence of SRSF2 mutation with the
of romiplostim with azacitidine have been reported.62 Pa- second allele knockout was lethal in mice. The presence of a
tients with IPSS low-risk and intermediate-risk myelodys- mutated allele along with wild copy translates in developing
plastic syndromes were randomly assigned to receive the hematologic malignancy. This provides a therapeutic op-
following in addition to azacitidine treatment: 500 mg of portunity in which using splicing inhibitors leads to synthetic
romiplostim per week, 750 mg of romiplostim per week, or lethality for patients with splicing mutations. Mice with
placebo. The incidence of clinically relevant thrombocyto- SRSF-2 mutation treated with E7101, a splicing inhibitor,
penic events for patients receiving 500 mg of romiplostim, lived longer.66 H3B-8800 is a potent oral and selective SF3b
750 mg of romiplostim, or placebo was 62%, 71%, and 85%, complex pan modulator. Phase I/II studies of patients with
respectively. The rate of platelet transfusions was 46%, 36%, myelodysplastic syndromes/AML with splicing mutations
and 69%, respectively. are planned to start in 2016.
The second approved thrombopoietin receptor agonist is
eltrombopag, an orally bioavailable agent that binds at a site
distinct from romiplostim.63 Preliminary results from a CPX-351
phase II randomized study of eltrombopag versus placebo In secondary AML from myelodysplastic syndromes after
for 70 patients with lower-risk myelodysplastic syndromes HMA failure, responses to standard induction chemotherapy
were reported at the 2015 American Society of Hematology is low with 14% to 29% CR rates reported.38,45 CPX-351 is a
Annual Meeting. Among 41 evaluable patients treated with liposomal formulation of daunorubicin and cytarabine in
eltrombopag, 21 responded at week 8; nine of these patients fixed molar ratio (5:1) optimizing delivery and cytotoxicity.
achieved CR (platelets . 100 3 109 cells/L). Twenty-four Promising results were reported in a phase IIB clinical trial, in
patients completed 24 weeks of eltrombopag treatment at which the median OS for secondary AML was 12.1 months
the time of data reporting, in which 13 patients were still for the CPX351 arm compared with 6.1 months for the
responders (eight achieved CR).64 standard induction (p = .01).67 A randomized phase III clinical
A phase I study of heavily treated patients with higher-risk trial finished accrual of patients with secondary AML.
disease who were resistant to HMAs noted a transition to Table 2 summarizes other potential agents for myelo-
transfusion independence for 30% of patients receiving dysplastic syndrome treatment after HMA failure not dis-
eltrombopag, with a median duration of response of cussed in this review.
3.3 months. In higher-risk myelodysplastic syndromes,
concerns about increased myeloblasts, leukocytosis, and CONCLUSION
bone marrow fibrosis were raised, particularly in CMML.65 Hypomethylating agents remain the current standard
therapy for the majority of patients with myelodysplastic
H3B-8800 syndromes. An international and collaborative effort is ur-
Splicing mutations are frequent among patients with gently needed to conduct clinical trials to improve outcomes
myelodysplastic syndromes. The splicing machinery muta- with HMAs and discover novel active new agents for
tions occur in an exclusive manner and in a heterozygous treatment of myelodysplastic syndromes after HMA failure.

References
1. Komrokji RS, Zhang L, Bennett JM. Myelodysplastic syndromes classi- 6. Kantarjian H, OBrien S, Ravandi F, et al. Proposal for a new risk model in
fication and risk stratification. Hematol Oncol Clin North Am. 2010;24: myelodysplastic syndrome that accounts for events not considered in
443-457. the original International Prognostic Scoring System. Cancer. 2008;113:
2. Komrokji R, Bennett JM: What is WHO?: myelodysplastic syndrome 1351-1361.
classification and prognosis. Am Soc Clin Oncol Educ Book. 2009;29: 7. Garcia-Manero G, Shan J, Faderl S, et al. A prognostic score for patients
413-419. with lower risk myelodysplastic syndrome. Leukemia. 2008;22:538-543.
3. Bennett JM, Komrokji RS. The myelodysplastic syndromes: di- 8. Bejar R, Stevenson K, Abdel-Wahab O, et al. Clinical effect of point
agnosis, molecular biology and risk assessment. Hematology. mutations in myelodysplastic syndromes. N Engl J Med. 2011;364:
2005;10:258-269. 2496-2506.
4. Greenberg P, Cox C, LeBeau MM, et al. International scoring system for 9. Cutler CS, Lee SJ, Greenberg P, et al. A decision analysis of allogeneic
evaluating prognosis in myelodysplastic syndromes. Blood. 1997;89: bone marrow transplantation for the myelodysplastic syndromes:
2079-2088. delayed transplantation for low-risk myelodysplasia is associated with
5. Greenberg PL, Tuechler H, Schanz J, et al. Revised international improved outcome. Blood. 2004;104:579-585.
prognostic scoring system for myelodysplastic syndromes. Blood. 2012; 10. Issa JP. Epigenetic variation and human disease. J Nutr. 2002;132:
120:2454-2465. 2388S-2392S.

e350 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


BEYOND HYPOMETHYLATING AGENTS IN MDS

11. Kumar A, List AF, Hozo I, et al. Decitabine versus 5-azacitidine for the acute myeloid leukemia with myelodysplasia-related changes: results of
treatment of myelodysplastic syndrome: adjusted indirect meta- the US Leukemia Intergroup trial E1905. J Clin Oncol. 2014;32:
analysis. Haematologica. 2010;95:340-342, author reply 343-344. 1242-1248.
12. Qin T, Castoro R, El Ahdab S, et al. Mechanisms of resistance to dec- 27. Garcia-Manero G, Berdeja JG, Komrokji RS, et al. A randomized,
itabine in the myelodysplastic syndrome. PLoS One. 2011;6:e23372. placebo-controlled, phase II study of pracinostat in combination with
13. Fenaux P, Mufti GJ, Hellstrom-Lindberg E, et al; International Vidaza azacitidine (AZA) in patients with previously untreated myelodysplastic
High-Risk MDS Survival Study Group. Efficacy of azacitidine compared syndrome (MDS). Blood. 2015;126 (abstr 911).
with that of conventional care regimens in the treatment of higher-risk 28. Fathi AT, Erba HP, Lancet JE, et al. SGN-CD33A plus hypomethylating
myelodysplastic syndromes: a randomised, open-label, phase III study. agents: a novel, well-tolerated regimen with high remission rate in
Lancet Oncol. 2009;10:223-232. frontline unfit AML. Blood. 2015;126 (abstr 454).
14. Lubbert M, Suciu S, Baila L, et al. Low-dose decitabine versus best 29. Ogata K, Kakumoto K, Matsuda A, et al. Differences in blast immu-
supportive care in elderly patients with intermediate- or high-risk nophenotypes among disease types in myelodysplastic syndromes:
myelodysplastic syndrome (MDS) ineligible for intensive chemother- a multicenter validation study. Leuk Res. 2012;36:1229-1236.
apy: final results of the randomized phase III study of the European 30. Yang H, Bueso-Ramos CE, Parmar S, et al. Induction of PD-1 and PD-1
Organisation for Research and Treatment of Cancer Leukemia Group ligand expression by hypomethylating agents (HMA) in myelodysplastic
and the German MDS Study Group. J Clin Oncol. 2011;29:1987-1996. syndromes and acute myelogenous leukemia suggest a role for T cell
15. Kantarjian H, Issa JP, Rosenfeld CS, et al. Decitabine improves patient function in clinical resistance to Hmas. Blood. 2012;120 (abstr 3810).
outcomes in myelodysplastic syndromes: results of a phase III ran- 31. Swords RT, Erba HP, DeAngelo DJ, et al. Pevonedistat (MLN4924), a first-
domized study. Cancer. 2006;106:1794-1803. in-class NEDD8-activating enzyme inhibitor, in patients with acute
16. Steensma DP, Baer MR, Slack JL, et al. Multicenter study of decitabine myeloid leukaemia and myelodysplastic syndromes: a phase 1 study. Br
administered daily for 5 days every 4 weeks to adults with myelo- J Haematol. 2015;169:534-543.
dysplastic syndromes: the alternative dosing for outpatient treatment 32. Swords RT, Savona MR, Maris MB, et al. Pevonedistat (MLN4924), an
(ADOPT) trial. J Clin Oncol. 2009;27:3842-3848. investigational, first-in-class NAE Inhibitor, in combination with azaci-
17. Damaj G, Duhamel A, Robin M, et al. Impact of azacitidine before al- tidine in elderly patients with acute myeloid leukemia (AML) considered
logeneic stem-cell transplantation for myelodysplastic syndromes: unfit for conventional chemotherapy: updated results from the phase 1
a study by the Societe Franaise de Greffe de Moelle et de Therapie- C15009 trial. Blood. 2014;124 (abstr 2313).
Cellulaire and the Groupe-Francophone des Myelodysplasies. J Clin 33. Sallman DA, Komrokji R, Vaupel C, et al. Impact of TP53 mutation variant
Oncol. 2012;30:4533-4540. allele frequency on phenotype and outcomes in myelodysplastic syn-
18. de Lima M, Giralt S, Thall PF, et al. Maintenance therapy with low-dose dromes. Leukemia. Epub 2015 Oct 30.
azacitidine after allogeneic hematopoietic stem cell transplantation for 34. Bejar R, Papaemmanuil E, Haferlach T, et al. Somatic mutations in MDS
recurrent acute myelogenous leukemia or myelodysplastic syndrome: patients are associated with clinical features and predict prognosis
a dose and schedule finding study. Cancer. 2010;116:5420-5431. independent of the IPSS-R: analysis of combined datasets from the
19. Lyons RM, Cosgriff TM, Modi SS, et al. Hematologic response to three International Working Group for Prognosis in MDS-Molecular Com-
alternative dosing schedules of azacitidine in patients with myelo- mittee. Blood. 2015;126 (abstr 907).
dysplastic syndromes. J Clin Oncol. 2009;27:1850-1856. 35. Bejar R, Stevenson KE, Caughey B, et al. Somatic mutations predict poor
20. Zeidan AM, Sekeres MA, Garcia-Manero G, et al. Comparison of risk outcome in patients with myelodysplastic syndrome after hemato-
stratification tools in predicting outcomes of patients with higher-risk poietic stem-cell transplantation. J Clin Oncol. 2014;32:2691-2698.
myelodysplastic syndromes treated with azanucleosides. Leukemia. 36. Zawacka-Pankau J, Selivanova G. Pharmacological reactivation of p53
Epub 2015 Oct 14. as a strategy to treat cancer. J Intern Med. 2015;277:248-259.
21. Itzykson R, Thepot S, Quesnel B, et al; Groupe Francophone des 37. Short NJ, Garcia-Manero G, Montalban Bravo G, et al. Low-dose
Myelodysplasies (GFM). Prognostic factors for response and overall hypomethylating agents (HMAs) are effective in patients (Pts) with
survival in 282 patients with higher-risk myelodysplastic syndromes low- or intermediate-1-risk myelodysplastic syndrome (MDS): a report
treated with azacitidine. Blood. 2011;117:403-411. on behalf of the MDS Clinical Research Consortium. Blood. 2015;126
22. Bejar R, Lord A, Stevenson K, et al. TET2 mutations predict response to (abstr 94).
hypomethylating agents in myelodysplastic syndrome patients. Blood. 38. Pre bet T, Gore SD, Esterni B, et al. Outcome of high-risk myelodysplastic
2014;124:2705-2712. syndrome after azacitidine treatment failure. J Clin Oncol. 2011;29:
23. Zeidan AM, Kharfan-Dabaja MA, Komrokji RS. Beyond hypomethylating 3322-3327.
agents failure in patients with myelodysplastic syndromes. Curr Opin 39. Jabbour E, Garcia-Manero G, Batty N, et al. Outcome of patients with
Hematol. 2014;21:123-130. myelodysplastic syndrome after failure of decitabine therapy. Cancer.
24. Sekeres MA, Othus M, List AF, et al. A randomized phase II study of 2010;116:3830-3834.
azacitidine combined with lenalidomide or with vorinostat vs. azaci- 40. Duong VH, Lin K, Reljic T, et al. Poor outcome of patients with mye-
tidine monotherapy in higher-risk myelodysplastic syndromes (MDS) lodysplastic syndrome after azacitidine treatment failure. Clin Lym-
and chronic myelomonocytic leukemia (CMML): North American In- phoma Myeloma Leuk. 2013;13:711-715.
tergroup Study SWOG S1117. Blood. 2014;124 (abstr LBA-5). 41. Gore SD, Fenaux P, Santini V, et al. A multivariate analysis of the re-
25. Sekeres MA, Othus M, List AF, et al. Additional analyses of a randomized lationship between response and survival among patients with higher-
phase II study of azacitidine combined with lenalidomide or with risk myelodysplastic syndromes treated within azacitidine or conven-
vorinostat vs. azacitidine monotherapy in higher-risk myelodysplastic tional care regimens in the randomized AZA-001 trial. Haematologica.
syndromes (MDS) and chronic myelomonocytic leukemia (CMML): 2013;98:1067-1072.
North American Intergroup Study SWOG S1117. Blood. 2015;126 (abstr 42. Nazha A, Sekeres MA, Garcia-Manero G, et al; MDS Clinical Research
908). Consortium. Outcomes of patients with myelodysplastic syndromes
26. Prebet T, Sun Z, Figueroa ME, et al. Prolonged administration of aza- who achieve stable disease after treatment with hypomethylating
citidine with or without entinostat for myelodysplastic syndrome and agents. Leuk Res. 2016;41:43-47.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e351


RAMI S. KOMROKJI

43. Garcia-Manero G, Fenaux P, Al-Kali A, et al. Overall survival and sub- myelodysplastic syndrome and correlation with overall survival. Blood.
group analysis from a randomized phase III study of intravenous rig- 2013;122 (abstr 3822).
osertib versus best supportive care (BSC) in patients (pts) with higher- 56. Komrokji RS, Raza A, Lancet JE, et al. Phase I clinical trial of oral rigosertib
risk myelodysplastic syndrome (HR-MDS) after failure of hypo- in patients with myelodysplastic syndromes. Br J Haematol. 2013;162:
methylating agents (HMAs). Blood. 2014;124 (abstr 163). 517-524.
44. Jabbour EJ, Garcia-Manero G, Strati P, et al. Outcome of patients with 57. Cortes JE, Feldman EJ, Yee K, et al. Results of the OPAL study: a phase II
low-risk and intermediate-1-risk myelodysplastic syndrome after study to evaluate the efficacy, safety and tolerability of tosedostat
hypomethylating agent failure: a report on behalf of the MDS Clinical (CHR-2797) in elderly subjects with treatment refractory or relapsed
Research Consortium. Cancer. 2015;121:876-882. acute myeloid leukemia. Blood. 2011;118 (abstr 767).
45. Bello C, Yu D, Komrokji RS, et al. Outcomes after induction chemo- 58. Lowenberg B, Morgan G, Ossenkoppele GJ, et al. Phase I/II clinical
therapy in patients with acute myeloid leukemia arising from myelo- study of tosedostat, an inhibitor of aminopeptidases, in patients with
dysplastic syndrome. Cancer. 2011;117:1463-1469. acute myeloid leukemia and myelodysplasia. J Clin Oncol. 2010;28:
46. Zeidan AM, Al Ali NH, Padron E, et al. Lenalidomide treatment for lower 4333-4338.
risk nondeletion 5q myelodysplastic syndromes patients yields higher 59. Visani G, Loscocco F, Fuligni F, et al. Tosedostat plus low dose cytarabine
response rates when used before azacitidine. Clin Lymphoma Myeloma induces a high rate of responses that can be predicted by genetic
Leuk. 2015;15:705-710. profiling in elderly AML. Blood. 2015;126 (abstr 329).
47. Borthakur G, Ahdab SE, Ravandi F, et al. Activity of decitabine in patients 60. Kantarjian H, Giles F, List A, et al. The incidence and impact of
with myelodysplastic syndrome previously treated with azacitidine. thrombocytopenia in myelodysplastic syndromes. Cancer. 2007;109:
Leuk Lymphoma. 2008;49:690-695. 1705-1714.
48. Komrokji RS, Apuri S, Al Ali N, et al. Evidence for selective benefit of 61. Kantarjian HM, Mufti GJ, Fenaux P, et al. Treatment with the throm-
sequential treatment with azanucleosides in patients with myelodys- bopoietin (TPO)-receptor agonist romiplostim in thrombocytopenic
plastic syndromes (MDS). J Clin Oncol 2013;31 (suppl; abstr 7113). patients (Pts) with low or intermediate-1 (int-1) risk myelodysplastic
49. Harel S, Cherait A, Berthon C, et al. Outcome of patients with high risk syndrome (MDS): follow-up AML and survival results of a randomized,
myelodysplastic syndrome (MDS) and advanced chronic myelomono- double-blind, placebo (PBO)-controlled study. Blood. 2012;120 (abstr
cytic leukemia (CMML) treated with decitabine after azacitidine failure. 421).
Leuk Res. 2015;39:501-504. 62. Kantarjian HM, Giles FJ, Greenberg PL, et al. Phase 2 study of romi-
50. Laille E, Shi T, Garcia-Manero G, et al. Pharmacokinetics and phar- plostim in patients with low- or intermediate-risk myelodysplastic
macodynamics with extended dosing of CC-486 in patients with he- syndrome receiving azacitidine therapy. Blood. 2010;116:3163-3170.
matologic malignancies. PLoS One. 2015;10:e0135520. 63. Bussel JB, Cheng G, Saleh MN, et al. Eltrombopag for the treatment of
51. Garcia-Manero G, Stoltz ML, Ward MR, et al. A pilot pharmacokinetic chronic idiopathic thrombocytopenic purpura. N Engl J Med. 2007;357:
study of oral azacitidine. Leukemia. 2008;22:1680-1684. 2237-2247.
52. Garcia-Manero G, Gore SD, Kambhampati S, et al. Efficacy and safety of 64. Oliva EN, Santini V, Alati C, et al. Eltrombopag for the treatment of
extended dosing schedules of CC-486 (oral azacitidine) in patients with thrombocytopenia of low and intermediate-1 IPSS risk myelodysplastic
lower-risk myelodysplastic syndromes. Leukemia. Epub 2015 Oct 7. syndromes: interim results on efficacy, safety and quality of life of an
53. Kantarjian HM, Roboz GJ, Rizzieri DA, et al. Results from the dose international, multicenter prospective, randomized, trial. Blood. 2015;
escalation phase of a randomized phase 1-2 first-in-human (FIH) study 126 (abstr 91).
of SGI-110, a novel low volume stable subcutaneous (SQ) second 65. Ramadan H, Duong VH, Al Ali NH, et al. Eltrombopag use in chronic
generation hypomethylating agent (HMA) in patients with relapsed/ myelomonocytic leukemia (CMML) patients: a cautionary tale. Blood.
refractory MDS and AML. Blood. 2012;120 (abstr 414). 2015;126 (abstr 2897).
54. Jabbour E, Yee K, Kropf P, et al. First clinical results of a randomized 66. Lee SC-W, Dvinge H, Kim E, et al. Therapeutic targeting of spliceosomal
phase 2 study of SGI-110, a novel subcutaneous (SQ) hypomethylating mutant myeloid leukemias through modulation of splicing catalysis.
agent (HMA), in adult patients with acute myeloid leukemia (AML). Blood. 2015;126 (abstr 4).
Blood. 2013;122 (abstr 497). 67. Lancet JE, Cortes JE, Hogge DE, et al. Phase 2 trial of CPX-351, a fixed 5:1
55. Raza A, Greenberg PL, Olnes MJ, et al. Final phase I/II results of rig- molar ratio of cytarabine/daunorubicin, vs cytarabine/daunorubicin in
osertib (ON 01910.Na) hematological effects in patients with older adults with untreated AML. Blood. 2014;123:3239-3246.

e352 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


HEMATOLOGIC MALIGNANCIESLYMPHOMA
AND CHRONIC LYMPHOCYTIC LEUKEMIA

Current Management Concepts:


Primary Central Nervous System
Lymphoma, Natural Killer T-Cell
Lymphoma Nasal Type,
Post-transplant
Lymphoproliferative Disorder

CHAIR
Thomas M. Habermann, MD
Mayo Clinic
Rochester, MN

SPEAKERS
Soon Thye Lim, MD
National Cancer Centre Singapore
Sinagpore

Tracy T. Batchelor, MD, MPH


Massachusetts General Hospital
Boston, MA
BATCHELOR, THYE, AND HABERMANN

Current Management Concepts: Primary Central Nervous


System Lymphoma, Natural Killer T-Cell Lymphoma Nasal
Type, and Post-transplant Lymphoproliferative Disorder
Tracy T. Batchelor, MD, MPH, Lim Soon Thye, MD, and Thomas M. Habermann, MD

OVERVIEW

Primary central nervous system lymphoma, natural killer T-cell lymphoma nasal type, and post-transplant lymphoproli-
ferative disorder are uncommon and complex lymphoproliferative disorders. These disorders present with different risk
factors, have complex tumor characteristics, and require unique therapeutic interventions. These diseases require a
multidisciplinary complex team approach. This article will update current management approaches and concepts.

P rimary central nervous system lymphoma (PCNSL) is


an extranodal non-Hodgkin lymphoma confined to
the brain, leptomeninges, eyes, or spinal cord. Natural
molecular mechanisms underlying transformation and lo-
calization to the CNS are poorly understood.4

killer (NK)/T-cell lymphoma disorders, more prevalent in Diagnosis and Prognostic Factors
Asian countries and parts of Latin America, are categorized as Neurocognitive symptoms are the most common presenting
extranodal NK T-cell lymphoma nasal type and as aggressive clinical features of PCNSL. The International PCNSL Collab-
NK-cell leukemia. Post-transplant lymphoproliferative disorder orative Group has developed guidelines to determine extent
is applied to a group of lymphoproliferative disorders arising in of disease.5 A gadolinium-enhanced brain MRI scan is the
a pharmacologically immunocompromised host after solid most sensitive radiographic study for the detection of PCNSL
organ or allogeneic stem cell transplantation. In this article, (Fig. 1). Most patients with PCNSL present with a single brain
we will update discuss current management approaches mass. The diagnosis of PCNSL is typically established by a
and concepts for these diseases. stereotactic brain biopsy, cerebrospinal fluid (CSF) analysis,
or analysis of vitreous aspirate from patients with ocular
PRIMARY CENTRAL NERVOUS SYSTEM involvement. Given the possible delay in diagnosis and
LYMPHOMA treatment with the latter two methods, prompt stereotactic
Epidemiology biopsy is advised in almost all cases that are surgically ac-
Primary central nervous system lymphoma accounts for cessible. Secondary CSF and ocular involvement occurs
approximately 2% of all primary central nervous system among approximately 15%20% and 5%20% of patients
(CNS) tumors, with median age 65 at diagnosis.1,2 Since with PCNSL, respectively. Presenting symptoms of ocular
2000, there has been an increase in the overall incidence of involvement include eye pain, blurred vision, and floaters.6
PCNSL, especially in elderly individuals. B symptoms such as weight loss, fevers, and night sweats are
infrequent in PCNSL. A thorough diagnostic evaluation is
needed to establish the extent of the lymphoma and to
Pathology confirm localization to the CNS. A lumbar puncture should
Approximately 90% of PCNSL cases are diffuse large B-cell be performed if not contraindicated, and CSF should be
lymphomas (DLBCLs), with the remainder consisting of T-cell assessed by flow cytometry, cytology, and immunoglobulin
lymphomas, poorly characterized low-grade lymphomas, heavy chain gene rearrangement. Because extraneural
or Burkitt lymphomas.3 More than 90% of primary CNS disease must be excluded to establish a diagnosis of pri-
DLBCL cases consist of the activated B-celllike subtype. The mary CNS lymphoma, CT/PET scans of the chest, abdomen,

From the Division of Hematology/Oncology, Departments of Neurology and Radiation Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston,
MA; Department of Medical Oncology, National Cancer Centre, Duke-National University of Singapore Medical School, Singapore; Division of Hematology, Department of Medicine,
Mayo Clinic, Rochester, MN.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Thomas M. Habermann, MD, Division of Hematology, Department of Medicine, Mayo Clinic, 200 First St. SW, Rochester, MN 55905;
email: habermann.thomas@mayo.edu.

2016 by American Society of Clinical Oncology.

e354 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


POST-TRANSPLANT LYMPHOPROLIFERATIVE DISORDER

FIGURE 1. MRIs From a Patient With Primary Central Nervous System Lymphoma

A T1-weighted, postcontrast, sequence (left) demonstrates homogenous enhancement of the tumor in the region of the left caudate nucleus. A T2/fluid-attenuated inversion
recovery sequence (right) demonstrates a hyperintense signal surrounding the tumor, reflecting vasogenic cerebral edema.
Courtesy of Priscilla Brastianos, MD.

and pelvis and a bone marrow biopsy and aspirate analysis Cooperative Oncology Group performance status greater
should be performed to exclude occult systemic disease. than 1, elevated serum LDH level, elevated CSF protein
Involvement of the optic nerve, retina, or vitreous humor concentration, and involvement of deep regions of the brain.
should be excluded with a comprehensive eye evaluation by For patients with zero to one factor, two to three factors, and
an ophthalmologist that includes a slit-lamp examination. four to five factors, the 2-year survival proportions were
Blood tests should include serum lactate dehydrogenase 80%, 48%, and 15%, respectively. In another prognostic
(LDH) and HIV serology.5 model, patients with PCNSL were divided into three groups
Two prognostic scoring systems have been developed based on age and performance status: (1) age younger than
specifically for PCNSL.7,8 In a retrospective review of 105 50, (2) age 50 and older with a Karnofsky performance score
patients with PCNSL, the International Extranodal Lym- (KPS) of 70 or greater, and (3) age 50 and older with a KPS less
phoma Study Group identified the following as independent than 70. Based on these three divisions, significant differ-
predictors of poor prognosis: age older than 60, Eastern ences in overall survival (OS) and failure-free survival were
observed (p = .0001).

KEY POINTS Treatment


Defining response to treatment in PCNSL requires assess-
The prognostic scoring systems in PCNSL, ENKL nasal ment of all sites involved by the disease. The International
type, and PTLD are different than standard non-Hodgkin PCNSL Collaborative Group has established response criteria
lymphomas. that have been adopted into most prospective clinical trials
The most effective treatment in PCNSL is high-dose (Table 1).5
methotrexate in combination with other agents, but Corticosteroids decrease tumor-associated edema and
standardized induction and consolidation regimens may result in partial radiographic regression of tumors. An
have yet to be defined. initial response to corticosteroids is associated with a fa-
ENKL is an Epstein-Barr virusrelated disorder. The vorable outcome in PCNSL.9 However, after they have an
incorporation of radiation therapy with chemotherapy
initial response to corticosteroids, almost all patients quickly
is crucial.
Advanced ENKL should be treated with L-asparaginase
experience relapse. Corticosteroids should be avoided prior
regimens. to a biopsy if possible, given the risk of disrupting cellular
Immunosuppression reduction and rituximab are key morphology, which can result in a nondiagnostic pathologic
management approaches in the initial approach to specimen.
PTLD. Surgical resection is not part of the standard treatment
approach for PCNSL, given the multifocal nature of this

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e355


BATCHELOR, THYE, AND HABERMANN

TABLE 1. International PCNSL Collaborative Group Consensus Guidelines for the Assessment of Response in
Primary Central Nervous System Lymphoma5

Steroid
Response Brain Imaging Dose Ophthalmologic Examination CSF Cytology
Complete Response No contrast-enhancing disease None Normal Negative
Unconfirmed Complete No contrast-enhancing disease Any Normal Negative
Response
Minimal enhancing disease Any Minor RPE abnormality Negative
Partial Response 50% decrease in enhancement NA Normal or minor RPE abnormality Negative
No contrast-enhancing disease NA Decrease in vitreous cells or retinal Persistent or
infiltrate suspicious
Progressive Disease 25% increase in enhancing disease NA Recurrent or new disease Recurrent or positive
Any new site of disease
Stable Disease All scenarios not covered by responses
above
Abbreviations: PCNSL, primary central nervous system lymphoma; NA, not applicable; RPE, retinal pigment epithelium; CSF, cerebral spinal fluid.

tumor and potential long-term morbidities.10 The role of administered to patients who had achieved a complete re-
neurosurgery in PCNSL is to establish a diagnosis via ste- sponse (CR) to induction chemotherapy including HD-MTX.16
reotactic biopsy. However, longer neuropsychologic follow-up of these pa-
Standardized induction and consolidation treatment of tients is necessary to definitively assess the safety of this
PCNSL has yet to be defined. Historically, PCNSL was treated regimen because numerous studies have demonstrated the
only with whole-brain radiotherapy (WBRT) at doses ranging delayed neurotoxic effects of WBRT in the PCNSL population
from 36 to 45 Gy, which resulted in a high proportion of and the reduced risk of neurotoxicity in regimens consisting
radiographic responses but early relapse. In a multicenter of chemotherapy alone.17,18 Given the risk of clinical neu-
phase II trial, 41 patients were treated with WBRT to 40 Gy rotoxicity, other studies have assessed whether WBRT can be
plus a 20-Gy tumor boost and achieved a median OS of only eliminated from the initial management of PCNSL. In a
12 months.11 Given the lack of durable responses to radiation multicenter phase III trial, patients were randomly assigned
and the risk of neurotoxicity associated with this therapeutic to receive HD-MTXbased chemotherapy with or without
modality, WBRT alone is no longer a recommended initial WBRT.19 The trial enrolled 551 patients; of these, 318 were
treatment for most patients with PCNSL. treated per protocol. Intent-to-treat analysis revealed that
The most effective treatment of PCNSL is high-dose patients treated in the combined modality arm (chemo-
methotrexate (HD-MTX) administered intravenously at therapy plus WBRT) achieved prolonged progression-free
variable doses (18 g/m2), typically used in combination with survival (PFS) but had no improvement in OS, demonstrat-
other chemotherapeutic agents and/or WBRT. However, ing that the elimination of WBRT from the treatment regimen
there is no consensus on the optimal dose of HD-MTX or on did not compromise OS. This has led to deferral of WBRT and
the role of radiation in combination with methotrexate in chemotherapy-alone approaches for patients with newly
the management of PCNSL. A number of randomized trials diagnosed PCNSL. These approaches are based on a foun-
are ongoing to address these issues. Methotrexate doses of dation of HD-MTX. Variable doses and schedules of HD-MTX
3 g/m2 or greater result in therapeutic concentrations in have been used, but in general, a dose of 3 g/m2 or greater
the brain parenchyma and CSF and lead to more durable delivered as an initial bolus followed by an infusion over
treatment responses when combined with WBRT.12-14 In a 3 hours administered every 1021 days is recommended.20
phase II trial, 79 patients with PCNSL were randomly assigned Multiple phase II studies have reported on the safety, ef-
to receive either HD-MTX (3.5 g/m2, day 1) or HD-MTX ficacy, and relatively preserved cognition of HD-MTXbased
(3.5 g/m2, day 1) plus cytarabine (2 g/m2 twice a day, days chemotherapy regimens.21,22 Moreover, a longer duration
23). Each chemotherapy cycle was 21 days. All patients of induction chemotherapy with HD-MTX (more than six
underwent consolidative WBRT after induction chemo- cycles) results in higher CR proportions.16,21
therapy. The HD-MTX plus cytarabine arm had a higher Several first-generation chemotherapy regimens for
proportion of complete radiographic responses and a su- PCNSL included intrathecal chemotherapy. However, in non-
perior 3-year OS.14 However, it is now widely recognized randomized studies that included intrathecal chemotherapy,
that there is a high incidence of neurotoxicity with com- there was no improvement in outcomes versus regimens that
bined modality treatment that includes WBRT.15 The latter did not include intrathecal injections of chemotherapy.23,24
observation prompted studies utilizing lower doses of Rituximab is being incorporated in combination regimens
WBRT. In a multicenter phase II study, no significant for PCNSL. When rituximab is administered intravenously at
neurocognitive decline was observed after consolidative doses of 375800 mg/m2, CSF levels from 0.1% to 4.4% of
reduced-dose WBRT (23.4 Gy) and cytarabine were serum levels are achieved. Despite limited CSF penetration,

e356 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


POST-TRANSPLANT LYMPHOPROLIFERATIVE DISORDER

radiographic responses have been observed for patients with A PET/CT scan is the preferred imaging modality.29-31 Bone
relapsed PCNSL treated with rituximab monotherapy.25 In a marrow biopsy is recommended, and EBER-ISH should be
cooperative group phase II study, 44 patients with PCNSL were considered to detect occult marrow involvement.32,33 Quan-
treated with induction chemotherapy consisting of 8 g/m2 of titative measurement of Epstein-Barr virus (EBV) DNA in cell-
HD-MTX (day 1), 375 mg/m2 of rituximab (day 3), and 150 mg/m2 free plasma, which has been shown to correlate with tumor
of temozolomide (days 711), all drugs with demonstrated load and adverse outcomes, is recommended.34,35
efficacy as monotherapy in PCNSL.22 This induction chemo-
therapy was followed by consolidation chemotherapy con- Prognostic Factors
sisting of 5 mg/kg of intravenous etoposide as a continuous Several prognostic models for ENKL based on pretreatment
infusion over 96 hours and 2 g/m2 of cytarabine every 12 hours characteristics have been developed, including the In-
for 8 doses. Sixty-six percent of these patients achieved CR, the ternational Prognostic Index (IPI) and the Korean Prognostic
median PFS of the entire group was 2.4 years, and the esti- Index.36-39 Nonetheless, these models were based on data
mated 4-year OS was 65%. These results are comparable to from patients treated with cyclophosphamide, doxorubicin,
regimens that include WBRT. vincristine, and prednisolone (CHOP) or CHOP-like regimens.
Given the limited durability of responses observed in many Because prognostic factors are dependent on the efficacy
studies of PCNSL, there is increasing interest in high-dose of the regimen used, they may not be applicable to pa-
chemotherapy (HDT) followed by autologous stem cell tients who are increasingly treated with nonanthracycline-
transplantation (ASCT) as first-line consolidative therapy containing regimens. Recently, combined evaluation of the
for PCNSL. Conditioning regimens including thiotepa have post-treatment Deauville score on a PET/CT scan as well as
demonstrated the most encouraging results. In a multi- EBV DNA was shown to be very effective in predicting risk of
center phase II study, 79 patients were treated with in- treatment failure and may be more relevant.40
duction HD-MTX, cytarabine, rituximab, and thiotepa, followed
by carmustine and thiotepa conditioning prior to ASCT. The Management of Localized Nasal NK/T-Cell Lymphoma
overall response rate (ORR) was 91%, 2-year OS was 87%, and Radiotherapy is a crucial treatment modality in localized ENKL,
treatment-related deaths occurred among less than 10% of and a dose in excess of 50 Gy is recommended.41 This requires
enrolled patients. The toxicities, mostly cytopenias, were careful planning to cover the entire nasal cavity and involved
manageable.26 There are three ongoing multicenter ran- areas.42,43 Radiotherapy alone is insufficient to achieve high
domized trials comparing the efficacy of consolidative cure rates, owing to both local and systemic relapses.44,45
HDT/ASCT versus chemotherapy or WBRT for newly diagnosed However, results from early studies incorporating standard
PCNSL (Table 2). CHOP and dose-intensified CHOP were disappointing.46,47
This high failure rate is attributed to the expression of the
multidrug resistance (MDR) gene in NK cells, a feature that is
EXTRANODAL NATURAL KILLER/T-CELL recapitulated in neoplastic NK cells.
LYMPHOMA NASAL TYPE Subsequent approaches incorporated non-MDRrelated
Natural killer/T-cell lymphoma is more prevalent in Asian agents and etoposide with radiotherapy (Table 3).48-52
countries and parts of Latin America.27 The World Health Or- However, the optimal sequence of chemotherapy and ra-
ganization (WHO) classification categorized these disorders as diotherapy remains to be determined.
extranodal NK/T-cell lymphoma nasal type (ENKL) and as ag- In a phase II study of 27 patients with localized ENKL,
gressive NK-cell leukemia.27,28 Histologically and phenotypically, concurrent radiotherapy alone (50 Gy) with three cycles of
they are similar. The neoplastic cells are surface CD32 but 2/3 dexamethasone, etoposide, ifosfamide, and carboplatin
cytoplasmic CD3e+, CD56+, and cytotoxic molecule (granzyme B, (DeVIC) resulted in an ORR of 81% and a CR rate of 77%.53
TIA1, and perforin) positive. Epstein-Barr virusencoded RNA With a median follow-up of 67 months, the updated 5-year OS
(EBER) in situ hybridization (ISH) is invariably positive. and PFS were 73% and 67%, respectively.52 Acute toxicities
associated with this regimen were generally mild. In another
Clinical Presentation, Diagnosis, and Staging phase II study, 30 patients received concurrent radiotherapy
ENKL commonly involves the nasal cavity and upper aero- with cisplatin followed by three cycles of etoposide, ifosfa-
digestive tract.28 Rarely, patients may present with non- mide, cisplatin, and dexamethasone (VIPD).50 The CR and ORR
nasal lesions in the skin, gastrointestinal tract, and/or at best response were 80% and 83.3%, whereas the estimated
testis. In such cases, it is important to exclude occult na- 3-year PFS and OS were 85.2% and 86.3%, respectively. Acute
sal involvement using either more sensitive radiologic toxicities during concurrent chemotherapy were mild, but
modalities such as PET/CT or nasal panendoscopy.27,29,30 62% of patients developed grade 3/4 febrile neutropenia
This distinction is important because it affects staging and during VIPD therapy. A recent study of concurrent cisplatin
treatment. with radiotherapy followed by gemcitabine, dexamethasone,
The minimum diagnostic criteria for ENKL include ap- and cisplatin (GDP) chemotherapy reported comparable
propriate histopathologic features, surface CD32, cyto- treatment outcomes and less toxicity.49
plasmic CD3e+, CD56+, and EBER+.27,28 If CD56 is negative, Because arranging timely radiotherapy may be challeng-
both cytotoxic molecules and EBER should be positive. ing, sandwich protocols were explored.48,51 In one study,

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e357


BATCHELOR, THYE, AND HABERMANN

26 patients received two cycles of L-asparaginase, vincris- A risk-adapted approach to the treatment of patients with
tine, and prednisone (LVP) followed by radiotherapy (56 Gy) localized ENKL was proposed.56 Patients were classified as
and two to four additional cycles of LVP.48 The ORR and CR low or high risk using five independent prognostic factors:
rate after initial chemotherapy were 92% and 88.5%, re- age older than 60, Eastern Cooperative Oncology Group
spectively. The corresponding ORR and CR after completion performance status of 2 or more, stage II disease, elevated
of radiotherapy and subsequent chemotherapy were 92% LDH level, and presence of primary tumor invasion. For high-
and 88.5%, respectively. The reported 2-year OS and PFS risk patients (defined as the presence of one or more adverse
were 88.5% and 80.6%. Frontline use of sandwich gemci- factors), radiotherapy followed by chemotherapy was found
tabine, oxaliplatin, and L-asparaginase (GELOX) with radio- to be the most optimal sequence because it was associated
therapy yielded similar ORR and CR rates of 96.3% and with the best OS.56 For low-risk patients (defined as no
74.1%.51 At present, the most active regimen for patients adverse factors), radiotherapy alone achieved favorable
with advanced ENKL is the steroid, methotrexate, ifosfamide, long-term survival.56 Neither induction nor consolidation
54,55
L-asparaginase, and etoposide (SMILE) regimen. The ORR chemotherapy was beneficial. Although these results are in-
and CR rates for patients with untreated NK/T-cell lymphoma triguing, the conclusions are limited by the fact that more than
after two to three cycles of therapy were 81% and 60%, 80% of the patients were treated with anthracycline-based
respectively.54 The high induction response rate observed regimens, and these findings may not be applicable to patients
suggests that SMILE with sandwich radiotherapy may be an treated with contemporary regimens.
attractive approach but requires confirmation in a pro- In summary, patients with localized ENKL should re-
spective clinical trial. ceive combination chemotherapy and radiotherapy. Both

TABLE 2. Randomized Trials in Primary Central Nervous System Lymphoma


Completed Trials
Trial Type Phase No. of Patients (Age) Description
Medical Research Council39 Induction II 53 Stopped early; CHOP vs. WBRT followed by CHOP
IELSG 20 (NCT00210314)14 Induction II 79 (1875) Induction arm 1: methotrexate 1 cytarabine 0 WBRT
Induction arm 2: methotrexate 0 WBRT
ANOCEF-GOELAMS Induction II 95 ($ 60) Arm 1: methotrexate, procarbazine, vincristine, cytarabine
(NCT00503594)31
Arm 2: methotrexate, temozolomide
G-PCNSL-SG-1 (NCT00153530)19 Consolidation III 551 ($ 18) Arm 1: methotrexate 6 ifosfamide 0 WBRT
Arm 2: methotrexate 6 ifosfamide

Ongoing Trials
Trial Type Phase No. of Patients (Age) Description
IESLG 32 (NCT01011920) Induction II 200 (1870) Induction arm 1: methotrexate, cytarabine
Induction arm 2: methotrexate, cytarabine, rituximab
Induction arm 3: methotrexate, cytarabine, rituximab,
thiotepa
ALLG/HOVON (EudraCT 2009- Induction III 200 (1870) Arm 1: methotrexate, BCNU, teniposide, prednisone 0
014722-42) cytarabine, WBRT
Arm 2: methotrexate, BCNU, teniposide, prednisone 0
cytarabine, WBRT
IESLG 32 (NCT01011920) Consolidation II 104 (1870) Consolidation arm 1: WBRT
Consolidation arm 2: HDT/ASCT
ANOCEF-GOELAMS (NCT00863460) Consolidation II 100 (1860) R-MBVP 0
Consolidation arm 1: HDT/ASCT
Consolidation arm 2: WBRT
RTOG 1114 (NCT01399372) Consolidation II 84 ($ 18) Methotrexate, procarbazine, vincristine, rituximab 0
Consolidation arm 1: WBRT (lower dose) 0 cytarabine
Consolidation arm 2: cytarabine
Alliance 51101 (NCT01511562) Consolidation II 160 (1875) Methotrexate, temozolomide, rituximab, cytarabine 0
Consolidation arm 1: HDT/ASCT
Consolidation arm 2: etoposide, cytarabine
Abbreviations: CHOP, cyclophosphamide, doxorubicin, vincristine, and prednisolone; WBRT, whole-brain radiotherapy; BCNU, bis-chloroethylnitrosourea; HDT/ASCT, high-dose
chemotherapy and autologous stem cell transplantation.

e358 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


POST-TRANSPLANT LYMPHOPROLIFERATIVE DISORDER

TABLE 3. Combined Chemotherapy and Radiotherapy for Localized ENKL

No. of
Combination Therapy Reference Patients ORR, % CR, % Survival Outcomes
Concurrent RT (50 Gy) with 2/3 DeVIC alone Rubenstein et al and 27 81 77 5-year PFS, 67%; 5-year
Khan et al22,23 OS, 73%
30 mg/m2 of concurrent weekly intravenous cisplatin for 35 weeks with Sierra del Rio et al24 30 83.3 80 3-year PFS, 85.2%; 3-year
RT (4052.8 Gy) followed by 3 cycles of VIPD OS, 86.3%
30 mg/m2 of concurrent cisplatin weekly for 35 weeks with IMRT (56 Gy) Batchelor et al25 32 90.6 84.4 3-year PFS, 84.4%; 3-year
followed by 3 cycles of GDP OS, 87.5%
2 cycles of LVP RT (56 Gy) 24 cycles LVP Illerhaus et al26 26 92.0 88.5 2-year PFS, 80.6%; 2-year
OS, 88.5%
2 cycles of GELOX RT (56 Gy) 24 cycles GELOX Kwong et al27 27 96.3 74.1 2-year PFS, 86%; 2-year
OS, 86%
Abbreviations: ENKL, extranodal natural killer/T-cell lymphoma, nasal type; RT, radiotherapy; DeVIC, dexamethasone, etoposide, ifosfamide, and carboplatin; VIPD, etoposide,
ifosfamide, cisplatin, and dexamethasone; IMRT, intensive modulated radiotherapy; GDP, gemcitabine, dexamethasone, and cisplatin; LVP, L-asparaginase, vincristine, and
prednisolone; GELOX, gemcitabine, L-asparaginase, and oxaliplatin; ORR, overall response rate; CR, complete remission; PFS, progression-free survival; OS, overall survival.

concurrent chemoradiotherapy and sandwiched radiother- Intriguingly, among the patients with relapsed/refractory
apy approaches are appropriate (Table 4). Chemotherapy disease who attained CR after induction SMILE chemo-
regimens should incorporate non-MDRrelated agents and therapy, eight patients (28%) who went on to receive
etoposide. In comprehensive cancer centers where admin- consolidation HSCT remained in remission compared with
istering concurrent chemoradiation is feasible, radiotherapy 12 (41%) who received only chemotherapy. Consistent with
concurrent with 2/3 DeVIC is an attractive option based on its the SMILE regimen, the hematologic toxicities encountered
robust clinical evidence and toxicity profile. There is a trend to were significant, and care is required in its administration.
adopt sandwich radiotherapy or sequential approaches with One recent study reported that the combination of gem-
L-asparaginasebased regimens, which may be logistically less citabine, oxaliplatin, L-asparagine, and dexamethasone may
complex to administer. yield similar efficacious results with fewer toxicities.60
Although consolidation treatment with autologous or allo-
Management of Newly Diagnosed Advanced ENKL genic HSCT for patients with advanced ENKL after induction
Based on earlier studies57-59 that demonstrated the ef- chemotherapy is feasible, its role remains contentious.61-63
ficacy of L-asparaginase, a phase II trial involving the Recent data suggest that the results of autologous HSCT are
SMILE regimen (Table 2)55 was initiated among 38 pa- comparable or inferior to L-asparaginasecontaining regi-
tients with newly diagnosed or relapsed/refractory dis- mens.54,64 The Asian Lymphoma Study Group examined the
ease. The primary endpoint of the study was ORR after outcome of allogeneic HSCT among 18 patients, 14 of whom
two courses of SMILE. The ORR and CR rates after 2 cycles received SMILE prior to transplantation.65 With a median
of SMILE were 79% and 45%, respectively. Interestingly, follow-up of 20.5 months, 5-year OS and event-free survival
there were no differences in response rates between were 57% and 51%, respectively, comparable to the results
patients with untreated disease or those with a first re- observed with SMILE.54 Four patients (22%) died of treatment-
lapse. With a median follow-up of 24 months, the 1-year related infective complications. Interestingly, the survival rates
OS rate was 45%. Of the 28 patients who completed of patients with CR1 and CR2 were similar, implying that al-
at least two courses of SMILE treatment, four received logeneic HSCT may not be necessary for a significant pro-
further autologous hematopoietic stem cell transplantation portion of patients achieving CR1 with L-asparaginase therapy.
(HSCT), 17 received allogeneic HSCT, and seven received Taken together, patients with newly diagnosed advanced
chemotherapy alone. Although the 1-year OS and PFS of pa- ENKL should be treated with L-asparaginasebased regimens
tients who received autologous HSCT seemed better compared such as SMILE (Fig. 2). The role of consolidation autologous or
with those who received allogeneic HSCT or chemotherapy allogenic HSCT for patients attaining complete remission re-
alone, the differences were not statistically significant. Of note, mains debatable and best administered in the context of a
the regimen was associated with significant toxicities, with clinical trial.
grade 4 neutropenia occurring among 92% of patients.
Outside the context of a clinical trial, the efficacy and Management of Relapsed/Refractory NK/T-Cell
safety of the SMILE regimen was studied in 87 patients Lymphoma
with either untreated or relapsed/refractory disease.54 The Patients who experience relapse after receiving L-asparaginase
median number of courses of SMILE administered was based therapy are particularly difficult to treat. Gemcitabine-
three (range, 06). The ORR and CR rates were 81% and 66%, containing regimens may be potentially effective.66 Several agents
respectively. Of note, the estimated 5-year OS for newly are now approved for use in peripheral T-cell lymphoma
diagnosed patients was similar to the 5-year OS for patients not otherwise specified, including romidepsin, pralatrexate,
with relapsed/refractory disease (47.3% vs. 52.3%, p = .17). and brentuximab vedotin.67-69 Their roles in ENKL remain to

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e359


BATCHELOR, THYE, AND HABERMANN

TABLE 4. Selected Chemotherapy Regimens for ENKL can identify patients who are at risk for treatment failure.40
Future studies should examine the best consolidation ap-
Drug Regimen proaches in this group of patients. Finally, recent molecular
VIPD (repeat every Days 13: 100 mg/m2 of intravenous etoposide studies have identified aberrations involving the JAK/STAT
21 days)24 cascade and histone modificationrelated genes that may
Days 13: 1,200 mg/m2 of intravenous
ifosfamide offer opportunities for targeted therapy in future.71,72
Days 13: 33 mg/m2 of intravenous cisplatin
Days 13: 40 mg of intravenous or oral
dexamethasone POST-TRANSPLANT LYMPHOPROLIFERATIVE
GDP (repeat every Day 1 and 8: 1,000 mg/m2 of intravenous DISORDERS
21 days)25 gemcitabine The term post-transplant lymphoproliferative disorder (PTLD)
Days 14: 40 mg of oral dexamethasone is applied to a group of lymphoproliferative disorders arising
Day 1: 75 mg/m2 of cisplatin in a pharmacologically immunocompromised host after solid
LVP (repeat every Days 15: 6,000 IU/m2 of intravenous organ or allogeneic stem cell transplantation.73 PTLD is re-
21 days)26 L-asparaginase lated to EBV, but the number of EBV cases has increased from
Day 1: 1.4 mg/m2 of intravenous vincristine 10% (1990 to 1995) to 48% (2008 to 2013).74 Lymphoma after
Days 15: 100 mg of oral prednisolone solid organ transplantation represents about 21% of all
GELOX (repeat every Day 1 and 8: 1,000 mg/m2 of intravenous malignancies, whereas lymphoma represents 5% of all ma-
21 days)27 gemcitabine lignancies among the normal population. The biology, di-
Day 1: 130 mg/m2 of intravenous oxaliplatin agnosis, and management of this heterogeneous group of
Days 17: 6,000 U/m2 of intravenous disorders is different from other lymphoproliferative disor-
L-asparaginase ders. There are many variables in PTLD presentation, including
SMILE (repeat every Day 1: 2 g/m2 of intravenous methotrexate EBV serostatus prior to organ transplantation in the donor and
21 days)28,29 the recipient, histology, location of disease, stage of disease,
Days 24: 40 mg of oral dexamethasone (days
14) types and doses of immunosuppression agents in the course
Days 24: 15 mg 3 4 of intravenous or oral of the transplant, initial management strategies (surgery,
leucovorin immunosuppression reduction strategies), types of treat-
Days 24: 1,500 mg/m2 of intravenous ment (rituximab monotherapy or chemotherapy), presentation
ifosfamide from the time of transplantation, locations of extranodal dis-
Days 24: 100 mg/m2 of intravenous etoposide ease, comorbid conditions (other vital organ involvement),
Days 8, 10, 12, 14, 16, 18, and 20: 6,000 U/m2 of performance score, and active infections (cytomegalovirus,
intravenous L-asparaginase
fungal, etc.). International consensus development meetings
GOLD (repeat every Days 1: 1,000 mg/m2 of gemcitabine held in 1997 and 1998 addressed PTLD in working groups and
14 days)34
Day 1: 100 mg/m2 of intravenous oxaliplatin recommended management guidelines that set the stage for
Days 15: 10,000 U/m2 of intravenous future directions.75
L-asparaginase

Days 14: 20 mg of intravenous or oral dexa- Risk Factors for the Development of PTLD
methasone twice a day
PTLD has been highly associated with immunosuppression,
Abbreviations: ENKL, extranodal natural killer/T-cell lymphoma nasal type; VIPD,
etoposide, ifosfamide, cisplatin, and dexamethasone; LVP, L-asparaginase, vincristine,
which creates a state of impaired T-cell immunity and EBV
and prednisone; GELOX, gemcitabine, oxaliplatin, and L-asparaginase; SMILE, steroid, infection. EBV has been implicated in 48%93% of all cases of
methotrexate, ifosfamide, L-asparaginase, and etoposide; GOLD, emcitabine,
oxaliplatin, L-aspariginase, and dexamethasone.
PTLD. Early PTLD occurs in the first 612 months, and late
PTLD occurs after 12 months. ISH studies on paraffin section
tissues determine EBV status. Late PTLD is associated with
be determined, and caution is required when used outside the negative EBV status and has an inferior prognosis.
context of a clinical trial. In one pilot study, romidepsin increased The spectrum of PTLDs in the WHO classification include
the risk of EBV reactivation in patients with ENKL.70 Allogeneic early-type PTLD lesions (reactive plasmacytic hyperplasia
transplantation for patients achieving CR2 may be potentially that is infectious mononucleosislike), polymorphic PTLD,
curable, but its role has not been confirmed in prospective trials. monomorphic PTLD (DLBCL, Burkitt lymphoma, plasmablastic
An individual patient-based decision approach is suggested. lymphoma, T-cell lymphomas, and composite lymphomas),
and Hodgkin lymphoma. Immunochemotherapy approaches
Future Directions are histology dependent.76 There may be histologic overlap,
Current management strategies are summarized (Table 4). and there may be progressive transition from early PTLD
Significant progress has been achieved over the years with the through polymorphic PTLD to monomorphic PTLD.
adoption of combined modality approaches and intensified
L-asparaginasebased therapy. However, disease progression The Risk Factors for Surviving PTLD
remains a risk. Recent data showed that the post-treatment The risk factors for surviving PTLD have been reported by
Deauville score on a PET/CT scan and the presence of EBV DNA different groups. Leblond et al reported that a performance

e360 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


POST-TRANSPLANT LYMPHOPROLIFERATIVE DISORDER

FIGURE 2. Management of Newly Diagnosed Natural Killer/T-Cell Lymphoma

score of 2 or more, number of extranodal sites (one vs. more Immunosuppression reduction. The hallmark of the initial
than one), primary CNS location, T-cell origin, monoclonality, management of PTLD has been to reduce immunosup-
EBV-negative status, and treatment with chemotherapy pression with a goal of restoring EBV-specific cellular im-
were poor prognostic factors for 61 patients.77 Ghobrial et al munity without inducing graft rejection. The initial
reported on 107 patients in a multivariate analysis and consideration for all patients should be a reduction in im-
identified monomorphic disease, graft organ involvement, munosuppression, irrespective of EBV status. Regression of
and a performance score of 3 to 4 as adverse; patients with a monoclonal and polyclonal lymphoproliferative disorders
score of 0 to 1 versus 2 to 3 had superior outcomes (hazard after reduction of the doses of immunosuppressive agents
ratio 5.31; p , .0001).78 CNS involvement, bone marrow has been reported to be 20%85%.83 The approaches to
involvement, and hypoalbuminemia were predictive of a immunosuppression reduction have been empirical, pro-
negative outcome among 81 patients.79 The PTLD prog- spective clinical trials have not been reported, and approaches
nostic index was predictive of outcome. In a series of 70 are dependent on the organ transplanted. Consensus guide-
patients from the PTLD-1 trial, an IPI score less than 3, lung lines recommended the following and have evolved.75 For
transplant, and response to rituximab were the strongest critically ill patients with excessive disease, prednisone should
predictors of outcome.80 The PTLD-2 trial and organ- be decreased to 7.510 mg/day and all other immunosup-
specific studies will define risk groups based on response pressive agents should be discontinued. If there is no re-
to rituximab monotherapy, IPI score, and the type of organ sponse (objective reduction in tumor mass within a period of
transplanted.81,82 1020 days), then further interventions must be considered. In
less critically ill patients (the majority of patients), the initial
management strategy should include reduction of cyclospor-
Management ine, tacrolimus, prednisone, and related immunosuppressive
The treatment of PTLD is complex and may include multiple agents by at least 50%, and azathioprine or mycophenolate
approaches. mofetil should be discontinued. After a 14-day trial of de-
creased immunosuppression, a further decrease can be con-
Local therapy. A minority of patients with very limited sidered. This is one of the few examples in lymphoproliferative
disease might be treated with surgical extirpation of lo- disorders that patients are restaged with scans in such a short
calized radiation and minor immunosuppression reduction. period of time. Serial biopsies or other forms of graft organ
Emergent or palliative local radiation therapy has been function monitoring are performed during this time period of
reported. immunosuppression reduction to allow early detection of

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e361


BATCHELOR, THYE, AND HABERMANN

allograft rejection. This is especially important in the rejection combination therapy (R-CHOP-21) with granulocyte-colony
of vital organs such as the heart, liver, and lung, which may stimulating factor for four cycles. There were 111 patients
carry a risk of death. Clinical improvement occurs within who received R-CHOP consolidation, with an ORR of 85% (78 of
1.54 weeks. EBV-negative disease is less responsive to im- 92) and a CR rate of 60% (55 of 92). The 3-year TTP was 73% in
munosuppression reduction, but successful outcomes have the PTLD-1 trial among patients who were refractory to rit-
been reported. In a series of 42 patients treated with uximab induction. The treatment-related mortality was 7%.91
immunosuppression reduction alone or in conjunction with
surgical excision, the complete remission rate was 74%.84 Chemotherapy. Immunosuppression reduction should be
The response rate was 89% if no risk factors were present considered, but, in late PTLD, doses are low. Indications for
(elevated LDH, organ dysfunction, or multiorgan disease), initial treatment with chemotherapy include specific histolo-
which is in contrast with two to three risk factors and a 0% gies, such as peripheral T-cell lymphoma not otherwise
response rate. The long-term remission rates in two series specified, Hodgkin lymphoma, late PTLD, and other uncommon
were 20% and 25%.78,84 lymphomas. These lymphomas must be treated with the
standard-of-care approaches for these specific histologies.
Rituximab. Rituximab after reduction in immunosuppres-
sion is the standard of care for most CD20+ PTLD subtypes, Stem cell transplantation. Patients who have failed other
including polymorphic and monomorphic DLBCL disease. approaches have been in long-term remission with autol-
The ORRs to rituximab therapy, in conjunction with opti- ogous or allogeneic stem cell transplantation.92
mization of immunosuppression agents, are 44%75%, with
CR rates of 35%69%.85-88 The variable response rates relate Cellular immunotherapy. The transfer of selected or un-
to whether the PTLD was early of late, the presence of EBV in selected EBV-specific cytotoxic T cells to patients with
the tissue, LDH level, the type of organ transplanted, his- EBV-positive PTLD to restore EBV-specific cellular immunity
tology, other comorbid conditions, retrospective versus pro- as prevention or treatment has been under investigation. Of
spective reports, and duration of follow-up. There are no 114 patients who received infusions of EBV-specific cyto-
randomized clinical trials comparing rituximab with other in- toxic T lymphocytes, none of 101 patients who received
terventions. Monotherapy trials have reported a median OS prophylaxis developed PTLD, and 11 of 13 patients with
of 2.4 years. An international phase II trial, PTLD-1, enrolled biopsy-proven or probable PTLD achieved a sustained
152 patients to evaluate rituximab consolidation among patients CR.93-95
who had a CR after rituximab consolidation introducing risk-
stratified sequential treatment to PTLD management in a third Unique PTLD Scenarios
amendment to the trial. Patients were treated with 375 mg/m2 Burkitt lymphoma PTLD is rare and may be MYC negative, but it
of intravenous rituximab on days 1, 8, 15, and 22. After restaging, is associated with 11q- gain/loss abnormality.95,96 In a retro-
patients in CR were treated with 3-weekly courses of rituximab spective review, 25 of 26 evaluable patients had a proven MYC
monotherapy. The 70 patients who were treated with rituximab rearrangement. Intensive therapy was associated with toxicity-
followed by cyclophosphamide, adriamycin, vincristine, and related deaths in three of five patients.96 The German Study
prednisone (CHOP-21) served as the control population. Of Group on PTLD reported that four of five patients reached a
148 patients, 37 (25%) achieved a CR with four cycles of rit- complete remission, and the authors concluded that sequential
uximab and went on to rituximab consolidation.89 The median immunochemotherapy was safe and effective.97
time to progression (TTP) was significantly longer than in the Primary CNS PTLD represents a distinct clinicopatho-
corresponding group in the PTLD-1 trial (37 patients vs. 14 logic entity that occurs in 7%15% of patients with PTLD.
patients; p , .05). The addition of four doses of rituximab was a An international collaboration reported on 84 patients
predictor of OS, TTP, and PFS. Rituximab consolidation among treated over a 14-year period between 1997 and 2010.98
early rituximab responders resulted in significantly better This entity is more commonly a late PTLD with median
disease control and fewer toxicities compared with CHOP time of solid organ transplantation to PTLD of 54 months;
consolidation. The PD-1 trial provides information to tailor 94% of patients had EBV-positive disease the histology
treatment.90 was monomorphic for 83% (DLBCL in 66), and 79% had
received renal transplants. In addition, 37% of patients
Immunochemotherapy in DLBCL PTLD. The response rates had multifocal disease, and deep brain involvement was
to systemic chemotherapy during the 1980s were reported to seen in 33% of patients. Immunosuppression was decreased
be less than 20%. This has changed over time. The PTLD-1 trial in 93% of patients. Additional first-line treatment consisted
demonstrated the efficacy and safety of 375 mg/m2 of rit- of HD-MTX (48%), high-dose cytarabine (33%) alone or in
uximab weekly for four doses followed by four cycles of CHOP- combination with 20% receiving after methotrexate, brain
21 with a median OS of 6.6 years.91 This study underwent three radiation (24%), and/or rituximab with 10% rituximab
amendments, and the protocol was amended. All patients alone. Methotrexate and rituximab was the most common
were initially treated with 375 mg/m2 of rituximab on days 1, 8, regimen administered to 12 patients (10%). The ORR was
15, and 22. Patients who were not in a complete remis- 60%. The median PFS was 8 months and median OS was
sion were subsequently treated with rituximab and CHOP 17 months. At a median follow-up of 43 months, the 3-year

e362 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


POST-TRANSPLANT LYMPHOPROLIFERATIVE DISORDER

FIGURE 3. Proposed Initial Management of PTLD

PFS and OS were 32% and 43%, respectively. In a multi- 13%. Proposed initial management strategies for PTLD are
variate analysis, lack of response to first-line therapy outlined in Fig. 3. Dierickx et al have published a proposed
(p = .0005) and increased LDH were associated with an treatment algorithm PTLD following hematopoietic stem
inferior OS (p = .02). The treatment-related mortality was cell transplantation.81

References
1. Dolecek TA, Propp JM, Stroup NE, et al. CBTRUS statistical report: 8. Abrey LE, Ben-Porat L, Panageas KS, et al. Primary central nervous
primary brain and central nervous system tumors diagnosed in the system lymphoma: the Memorial Sloan-Kettering Cancer Center
United States in 2005-2009. Neuro Oncol. 2012;14:v1-v49. prognostic model. J Clin Oncol. 2006;24:5711-5715.
2. Villano JL, Koshy M, Shaikh H, et al. Age, gender, and racial differences in 9. Mathew BS, Carson KA, Grossman SA. Initial response to glucocorti-
incidence and survival in primary CNS lymphoma. Br J Cancer. 2011;105: coids. Cancer. 2006;106:383-387.
1414-1418. 10. Bellinzona M, Roser F, Ostertag H, et al. Surgical removal of primary
3. Swerdlow SH, Campo E, Harris NL, et al (eds). WHO Classification of central nervous system lymphomas (PCNSL) presenting as space
Tumours of the Haematopoietic and Lymphoid Tissues. Lyon, France: occupying lesions: a series of 33 cases. Eur J Surg Oncol. 2005;31:
International Agency for Research on Cancer; 2008. 100-105.
4. Ponzoni M, Issa S, Batchelor TT, et al. Beyond high-dose methotrexate 11. Nelson DF, Martz KL, Bonner H, et al. Non-Hodgkins lymphoma of the
and brain radiotherapy: novel targets and agents for primary CNS brain: can high dose, large volume radiation therapy improve survival?
Report on a prospective trial by the Radiation Therapy Oncology Group
lymphoma. Ann Oncol. 2014;25:316-322.
(RTOG): RTOG 8315. Int J Radiat Oncol Biol Phys. 1992;23:9-17.
5. Abrey LE, Batchelor TT, Ferreri AJ, et al; International Primary CNS
12. Glantz MJ, Cole BF, Recht L, et al. High-dose intravenous methotrexate
Lymphoma Collaborative Group. Report of an international workshop
for patients with nonleukemic leptomeningeal cancer: is intrathecal
to standardize baseline evaluation and response criteria for primary
chemotherapy necessary? J Clin Oncol. 1998;16:1561-1567.
CNS lymphoma. J Clin Oncol. 2005;23:5034-5043. 13. DeAngelis LM, Seiferheld W, Schold SC, et al; Radiation Therapy
6. Chan CC, Rubenstein JL, Coupland SE, et al. Primary vitreoretinal Oncology Group Study 93-10. Combination chemotherapy and ra-
lymphoma: a report from an International Primary Central Nervous diotherapy for primary central nervous system lymphoma: Radia-
System Lymphoma Collaborative Group symposium. Oncologist. 2011; tion Therapy Oncology Group Study 93-10. J Clin Oncol. 2002;20:
16:1589-1599. 4643-4648.
7. Ferreri AJ, Blay JY, Reni M, et al. Prognostic scoring system for primary 14. Ferreri AJ, Reni M, Foppoli M, et al; International Extranodal Lymphoma
CNS lymphomas: the International Extranodal Lymphoma Study Group Study Group (IELSG). High-dose cytarabine plus high-dose metho-
experience. J Clin Oncol. 2003;21:266-272. trexate versus high-dose methotrexate alone in patients with primary

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e363


BATCHELOR, THYE, AND HABERMANN

CNS lymphoma: a randomised phase 2 trial. Lancet. 2009;374: 32. Ito Y, Kimura H, Maeda Y, et al. Pretreatment EBV-DNA copy number is
1512-1520. predictive of response and toxicities to SMILE chemotherapy for
15. Correa DD, Maron L, Harder H, et al. Cognitive functions in primary extranodal NK/T-cell lymphoma, nasal type. Clin Cancer Res. 2012;18:
central nervous system lymphoma: literature review and assessment 4183-4190.
guidelines. Ann Oncol. 2007;18:1145-1151. 33. Lee J, Suh C, Huh J, et al. Effect of positive bone marrow EBV in situ
16. Morris PG, Correa DD, Yahalom J, et al. Rituximab, methotrexate, hybridization in staging and survival of localized extranodal natural
procarbazine, and vincristine followed by consolidation reduced-dose killer/T-cell lymphoma, nasal-type. Clin Cancer Res. 2007;13:3250-3254.
whole-brain radiotherapy and cytarabine in newly diagnosed primary 34. Au WY, Pang A, Choy C, et al. Quantification of circulating Epstein-Barr
CNS lymphoma: final results and long-term outcome. J Clin Oncol. 2013; virus (EBV) DNA in the diagnosis and monitoring of natural killer cell and
31:3971-3979. EBV-positive lymphomas in immunocompetent patients. Blood. 2004;
17. Doolittle ND, Korfel A, Lubow MA, et al. Long-term cognitive function, 104:243-249.
neuroimaging, and quality of life in primary CNS lymphoma. Neurology. 35. Suzuki R, Yamaguchi M, Izutsu K, et al; NK-cell Tumor Study Group.
2013;81:84-92. Prospective measurement of Epstein-Barr virus-DNA in plasma and
18. Juergens A, Pels H, Rogowski S, et al. Long-term survival with favorable peripheral blood mononuclear cells of extranodal NK/T-cell lymphoma,
cognitive outcome after chemotherapy in primary central nervous nasal type. Blood. 2011;118:6018-6022.
system lymphoma. Ann Neurol. 2010;67:182-189. 36. Chim CS, Ma SY, Au WY, et al. Primary nasal natural killer cell lymphoma:
19. Thiel E, Korfel A, Martus P, et al. High-dose methotrexate with or long-term treatment outcome and relationship with the International
without whole brain radiotherapy for primary CNS lymphoma (G- Prognostic Index. Blood. 2004;103:216-221.
PCNSL-SG-1): a phase 3, randomised, non-inferiority trial. Lancet 37. Lee J, Suh C, Park YH, et al. Extranodal natural killer T-cell lymphoma,
Oncol. 2010;11:1036-1047. nasal-type: a prognostic model from a retrospective multicenter study.
20. Ferreri AJ, Guerra E, Regazzi M, et al. Area under the curve of meth- J Clin Oncol. 2006;24:612-618.
otrexate and creatinine clearance are outcome-determining factors in 38. Suzuki R, Suzumiya J, Yamaguchi M, et al; NK-cell Tumor Study Group.
primary CNS lymphomas. Br J Cancer. 2004;90:353-358. Prognostic factors for mature natural killer (NK) cell neoplasms: ag-
21. Batchelor T, Carson K, ONeill A, et al. Treatment of primary CNS gressive NK cell leukemia and extranodal NK cell lymphoma, nasal type.
lymphoma with methotrexate and deferred radiotherapy: a report of Ann Oncol. 2010;21:1032-1040.
NABTT 96-07. J Clin Oncol. 2003;21:1044-1049. 39. Yang Y, Zhang YJ, Zhu Y, et al. Prognostic nomogram for overall
22. Rubenstein JL, Hsi ED, Johnson JL, et al. Intensive chemotherapy and survival in previously untreated patients with extranodal NK/T-cell
immunotherapy in patients with newly diagnosed primary CNS lym- lymphoma, nasal-type: a multicenter study. Leukemia. 2015;29:
phoma: CALGB 50202 (Alliance 50202). J Clin Oncol. 2013;31: 1571-1577.
3061-3068. 40. Kim SJ, Choi JY, Hyun SH, et al; Asia Lymphoma Study Group. Risk
23. Khan RB, Shi W, Thaler HT, et al. Is intrathecal methotrexate necessary in stratification on the basis of Deauville score on PET-CT and the presence
the treatment of primary CNS lymphoma? J Neurooncol. 2002;58: of Epstein-Barr virus DNA after completion of primary treatment for
175-178. extranodal natural killer/T-cell lymphoma, nasal type: a multicentre,
24. Sierra del Rio M, Ricard D, Houillier C, et al. Prophylactic intrathecal retrospective analysis. Lancet Haematol. 2015;2:e66-e74.
chemotherapy in primary CNS lymphoma. J Neurooncol. 2012;106: 41. Li YX, Yao B, Jin J, et al. Radiotherapy as primary treatment for stage IE
143-146. and IIE nasal natural killer/T-cell lymphoma. J Clin Oncol. 2006;24:
25. Batchelor TT, Grossman SA, Mikkelsen T, et al. Rituximab monotherapy 181-189.
for patients with recurrent primary CNS lymphoma. Neurology. 2011; 42. Isobe K, Uno T, Tamaru J, et al. Extranodal natural killer/T-cell lym-
76:929-930. phoma, nasal type: the significance of radiotherapeutic parameters.
26. Illerhaus G, Fritsch K, Egerer G, et al. Sequential high dose immuno- Cancer. 2006;106:609-615.
chemotherapy followed by autologous peripheral blood stem cell 43. Koom WS, Chung EJ, Yang WI, et al. Angiocentric T-cell and NK/T-cell
transplantation for patients with untreated primary central nervous lymphomas: radiotherapeutic viewpoints. Int J Radiat Oncol Biol Phys.
system lymphoma - a multicentre study by the Collaborative PCNSL 2004;59:1127-1137.
Study Group Freiburg. Blood. 2012;120 (abstr 302). 44. Cheung MM, Chan JK, Lau WH, et al. Early stage nasal NK/T-cell lym-
27. Kwong YL, Anderson BO, Advani R, et al; Asian Oncology Summit. phoma: clinical outcome, prognostic factors, and the effect of treat-
Management of T-cell and natural-killer-cell neoplasms in Asia: con- ment modality. Int J Radiat Oncol Biol Phys. 2002;54:182-190.
sensus statement from the Asian Oncology Summit 2009. Lancet Oncol. 45. Kim GE, Cho JH, Yang WI, et al. Angiocentric lymphoma of the head and
2009;10:1093-1101. neck: patterns of systemic failure after radiation treatment. J Clin Oncol.
28. Chan JKC, Jaffe ES, Ralfkiaer E, et al. Aggressive NK-cell leukemia. In 2000;18:54-63.
Swerdlow SH, Campo E, Harris NL, et al (eds). WHO Classification of 46. Kim WS, Song SY, Ahn YC, et al. CHOP followed by involved field ra-
Tumours of Haematopoietic and Lymphoid Tissues. Lyon, France: In- diation: is it optimal for localized nasal natural killer/T-cell lymphoma?
ternational Agency for Research on Cancer, 2008; 276-277. Ann Oncol. 2001;12:349-352.
29. Chan WK, Au WY, Wong CY, et al. Metabolic activity measured by F-18 47. Lee SH, Ahn YC, Kim WS, et al. The effect of pre-irradiation dose intense
FDG PET in natural killer-cell lymphoma compared to aggressive B- and CHOP on anthracyline resistance in localized nasal NK/T-cell lymphoma.
T-cell lymphomas. Clin Nucl Med. 2010;35:571-575. Haematologica. 2006;91:427-428.
30. Khong PL, Pang CB, Liang R, et al. Fluorine-18 fluorodeoxyglucose 48. Jiang M, Zhang H, Jiang Y, et al. Phase 2 trial of sandwich
positron emission tomography in mature T-cell and natural killer cell L-asparaginase, vincristine, and prednisone chemotherapy with radio-
malignancies. Ann Hematol. 2008;87:613-621. therapy in newly diagnosed, stage IE to IIE, nasal type, extranodal natural
31. Moon SH, Cho SK, Kim WS, et al. The role of 18F-FDG PET/CT for initial killer/T-cell lymphoma. Cancer. 2012;118:3294-3301.
staging of nasal type natural killer/T-cell lymphoma: a compari- 49. Ke QH, Zhou SQ, Du W, et al. Concurrent IMRT and weekly cisplatin
son with conventional staging methods. J Nucl Med. 2013;54: followed by GDP chemotherapy in newly diagnosed, stage IE to IIE,
1039-1044. nasal, extranodal NK/T-Cell lymphoma. Blood Cancer J. 2014;4:e267.

e364 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


POST-TRANSPLANT LYMPHOPROLIFERATIVE DISORDER

50. Kim SJ, Kim K, Kim BS, et al. Phase II trial of concurrent radiation and 67. Coiffier B, Pro B, Prince HM, et al. Results from a pivotal, open-label,
weekly cisplatin followed by VIPD chemotherapy in newly diagnosed, phase II study of romidepsin in relapsed or refractory peripheral T-
stage IE to IIE, nasal, extranodal NK/T-cell lymphoma: Consortium for cell lymphoma after prior systemic therapy. J Clin Oncol. 2012;30:
Improving Survival of Lymphoma study. J Clin Oncol. 2009;27: 631-636.
6027-6032. 68. OConnor OA, Pro B, Pinter-Brown L, et al. Pralatrexate in patients with
51. Wang L, Wang ZH, Chen XQ, et al. First-line combination of gemcitabine, relapsed or refractory peripheral T-cell lymphoma: results from the
oxaliplatin, and L-asparaginase (GELOX) followed by involved-field ra- pivotal PROPEL study. J Clin Oncol. 2011;29:1182-1189.
diation therapy for patients with stage IE/IIE extranodal natural killer/T- 69. Younes A, Bartlett NL, Leonard JP, et al. Brentuximab vedotin (SGN-35)
cell lymphoma. Cancer. 2013;119:348-355. for relapsed CD30-positive lymphomas. N Engl J Med. 2010;363:
52. Yamaguchi M, Tobinai K, Oguchi M, et al. Concurrent chemo- 1812-1821.
radiotherapy for localized nasal natural killer/T-cell lymphoma: an 70. Kim SJ, Kim JH, Ki CS, et al. Epstein-Barr virus reactivation in extranodal
updated analysis of the Japan clinical oncology group study JCOG0211. natural killer/T-cell lymphoma patients: a previously unrecognized
J Clin Oncol. 2012;30:4044-4046. serious adverse event in a pilot study with romidepsin. Ann Oncol. Epub
53. Yamaguchi M, Tobinai K, Oguchi M, et al. Phase I/II study of concurrent 2015 Dec 9.
chemoradiotherapy for localized nasal natural killer/T-cell lymphoma: 71. Koo GC, Tan SY, Tang T, et al. Janus kinase 3-activating mutations
Japan Clinical Oncology Group Study JCOG0211. J Clin Oncol. 2009;27: identified in natural killer/T-cell lymphoma. Cancer Discov. 2012;2:
5594-5600. 591-597.
54. Kwong YL, Kim WS, Lim ST, et al. SMILE for natural killer/T-cell lym- 72. Lee S, Park HY, Kang SY, et al. Genetic alterations of JAK/STAT cascade
phoma: analysis of safety and efficacy from the Asia Lymphoma Study and histone modification in extranodal NK/T-cell lymphoma nasal type.
Group. Blood. 2012;120:2973-2980. Oncotarget. 2015;6:17764-17776.
55. Yamaguchi M, Kwong YL, Kim WS, et al. Phase II study of SMILE 73. Murray JE, Wilson RE, Tilney NL, et al. Five years experience in renal
chemotherapy for newly diagnosed stage IV, relapsed, or re- transplantation with immunosuppressive drugs: survival, function,
fractory extranodal natural killer (NK)/T-cell lymphoma, nasal type: complications, and the role of lymphocyte depletion by thoracic duct
the NK-Cell Tumor Study Group study. J Clin Oncol. 2011;29: fistula. Ann Surg. 1968;168:416-435.
4410-4416. 74. Luskin MR, Heil DS, Tan KS, et al. The impact of EBV status on char-
56. Yang Y, Zhu Y, Cao JZ, et al. Risk-adapted therapy for early-stage acteristics and outcomes of posttransplantation lymphoproliferative
extranodal nasal-type NK/T-cell lymphoma: analysis from a multicen- disorders. Am J Transplant. 2015;15:2665-2673.
ter study. Blood. 2015;126:1424-1432, quiz 1517. 75. Paya CV, Fung JJ, Nalesnik MA, et al. Epstein-Barr virus-induced post-
57. Matsumoto Y, Nomura K, Kanda-Akano Y, et al. Successful treatment transplant lymphoproliferative disorders. AST/ASTP EBV-PTLD Task
with Erwinia L-asparaginase for recurrent natural killer/T cell lym- Force and the Mayo Clinic Organized International Consensus Devel-
phoma. Leuk Lymphoma. 2003;44:879-882. opment Meeting. Transplantation. 1999;68:1517-1525.
58. Nagafuji K, Fujisaki T, Arima F, et al. L-asparaginase induced durable 76. Swerdlow SH, Webber SA, Chadbum A, et al. Post-transplant lym-
remission of relapsed nasal NK/T-cell lymphoma after autologous phoproliferative disorders. In Swerdlow SH, Campo E, Harris NL, et al
peripheral blood stem cell transplantation. Int J Hematol. 2001;74: (eds). WHO Classification of Tumours of Haematopoietic and Lymphoid
447-450. Tissues. Lyon, France: International Agency for Research on Cancer
59. Obama K, Tara M, Niina K. L-Asparaginase-based induction therapy for Press, 2008; 343-349.
advanced extranodal NK/T-cell lymphoma. Int J Hematol. 2003;78: 77. Leblond V, Dhedin N, Mamzer Bruneel MF, et al. Identification of
248-250. prognostic factors in 61 patients with posttransplantation lympho-
60. Guo HQ, Liu L, Wang XF, et al. Efficacy of gemcitabine combined with proliferative disorders. J Clin Oncol. 2001;19:772-778.
oxaliplatin, L-asparaginase and dexamethasone in patients with newly- 78. Ghobrial IM, Habermann TM, Maurer MJ, et al. Prognostic analysis for
diagnosed extranodal NK/T-cell lymphoma. Mol Clin Oncol. 2014;2: survival in adult solid organ transplant recipients with post-
1172-1176. transplantation lymphoproliferative disorders. J Clin Oncol. 2005;23:
61. Kim HJ, Bang SM, Lee J, et al. High-dose chemotherapy with autologous 7574-7582.
stem cell transplantation in extranodal NK/T-cell lymphoma: a retro- 79. Evens AM, David KA, Helenowski I, et al. Multicenter analysis of 80 solid
spective comparison with non-transplantation cases. Bone Marrow organ transplantation recipients with post-transplantation lympho-
Transplant. 2006;37:819-824. proliferative disease: outcomes and prognostic factors in the modern
62. Murashige N, Kami M, Kishi Y, et al. Allogeneic haematopoietic stem cell era. J Clin Oncol. 2010;28:1038-1046.
transplantation as a promising treatment for natural killer-cell neo- 80. Dierickx D, Tousseyn T, Sagaert X, et al. Single-center analysis of biopsy-
plasms. Br J Haematol. 2005;130:561-567. confirmed posttransplant lymphoproliferative disorder: incidence,
63. Suzuki R, Suzumiya J, Nakamura S, et al; NK-cell Tumor Study Group. clinicopathological characteristics and prognostic factors. Leuk Lym-
Hematopoietic stem cell transplantation for natural killer-cell lineage phoma. 2013;54:2433-2440.
neoplasms. Bone Marrow Transplant. 2006;37:425-431. 81. Dierickx D, Tousseyn T, Gheysens O. How I treat posttransplant lym-
64. Kwong YL. High-dose chemotherapy and hematopoietic SCT in the phoproliferative disorders. Blood. 2015;126:2274-2283.
management of natural killer-cell malignancies. Bone Marrow Trans- 82. Hourigan MJ, Doecke J, Mollee PN, et al. A new prognosticator for post-
plant. 2009;44:709-714. transplant lymphoproliferative disorders after renal transplantation. Br
65. Tse E, Chan TS, Koh LP, et al. Allogeneic haematopoietic SCT for J Haematol. 2008;141:904-907.
natural killer/T-cell lymphoma: a multicentre analysis from the Asia 83. Habermann TM. Posttransplant lymphoproliferative disorders. Cancer
Lymphoma Study Group. Bone Marrow Transplant. 2014;49: Treat Res. 2008;142:273-292.
902-906. 84. Tsai DE, Hardy CL, Tomaszewski JE, et al. Reduction in immunosup-
66. Ahn HK, Kim SJ, Hwang DW, et al. Gemcitabine alone and/or containing pression as initial therapy for posttransplant lymphoproliferative dis-
chemotherapy is efficient in refractory or relapsed NK/T-cell lymphoma. order: analysis of prognostic variables and long-term follow-up of 42
Invest New Drugs. 2013;31:469-472. adult patients. Transplantation. 2001;71:1076-1088.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e365


BATCHELOR, THYE, AND HABERMANN

85. Oertel SH, Verschuuren E, Reinke P, et al. Effect of anti-CD 20 antibody 91. Trappe R, Oertel S, Leblond V, et al; German PTLD Study Group; Eu-
rituximab in patients with post-transplant lymphoproliferative disorder ropean PTLD Network. Sequential treatment with rituximab followed by
(PTLD). Am J Transplant. 2005;5:2901-2906. CHOP chemotherapy in adult B-cell post-transplant lymphoproliferative
86. Blaes AH, Peterson BA, Bartlett N, et al. Rituximab therapy is ef- disorder (PTLD): the prospective international multicentre phase 2
fective for posttransplant lymphoproliferative disorders after solid PTLD-1 trial. Lancet Oncol. 2012;13:196-206.
organ transplantation: results of a phase II trial. Cancer. 2005;104: 92. Komrokji RS, Oliva JL, Zand M, et al. Mini-BEAM and autologous he-
1661-1667. matopoietic stem-cell transplant for treatment of post-transplant
87. Choquet S, Leblond V, Herbrecht R, et al. Efficacy and safety of rit- lymphoproliferative disorders. Am J Hematol. 2005;79:211-215.
uximab in B-cell post-transplantation lymphoproliferative disorders: 93. Choquet S, Varnous S, Deback C, et al. Adapted treatment of Epstein-
results of a prospective multicenter phase 2 study. Blood. 2006;107: Barr virus infection to prevent posttransplant lymphoproliferative
3053-3057. disorder after heart transplantation. Am J Transplant. 2014;14:857-866.
88. Gonzalez-Barca E, Domingo-Domenech E, Capote FJ, et al; GEL/TAMO 94. Omar H, Hagglund H, Gustafsson-Jernberg A, et al. Targeted monitoring
(Grupo Espan ~ol de Linfomas); GELCAB (Grupo para el Estudio de los of patients at high risk of post-transplant lymphoproliferative disease by
Linfomas Catalano-Balear); GOTEL (Grupo Oncologico para el Trata- quantitative Epstein-Barr virus polymerase chain reaction. Transpl In-
miento y Estudio de los Linfomas). Prospective phase II trial of extended fect Dis. 2009;11:393-399.
treatment with rituximab in patients with B-cell post-transplant lym- 95. Heslop HE, Slobod KS, Pule MA, et al. Long-term outcome of EBV-
phoproliferative disease. Haematologica. 2007;92:1489-1494. specific T-cell infusions to prevent or treat EBV-related lymphoproli-
89. Trappe RU, Dierickx D, Zimmermann H, et al. Response to rituximab ferative disease in transplant recipients. Blood. 2010;115:925-935.
induction is a predictive biomarker in post-transplant lymphoproli- 96. Ferreiro JF, Morscio J, Dierickx D, et al. Post-transplant molecularly defined
ferative disorder (PTLD) and allows successful treatment stratification Burkitt lymphomas are frequently MYC-negative and characterized by the
in an international phase II trial including 152 patients. Blood. 2015;126 11q-gain/loss pattern. Haematologica. 2015;100:e275-e279.
(abstr 816). 97. Xicoy B, Ribera JM, Esteve J, et al. Post-transplant Burkitts leukemia or
90. Trappe RU, Choquet S, Dierickx D, et al; German PTLD Study Group and lymphoma. Study of five cases treated with specific intensive therapy
the European PTLD Network. International prognostic index, type of (PETHEMA ALL-3/97 trial). Leuk Lymphoma. 2003;44:1541-1543.
transplant and response to rituximab are key parameters to tailor 98. Evens AM, Choquet S, Kroll-Desrosiers AR, et al. Primary CNS post-
treatment in adults with CD20-positive B cell PTLD: clues from the PTLD- transplant lymphoproliferative disease (PTLD): an international report
1 trial. Am J Transplant. 2015;15:1091-1100. of 84 cases in the modern era. Am J Transplant. 2013;13:1512-1522.

e366 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


HEMATOLOGIC MALIGNANCIESLYMPHOMA
AND CHRONIC LYMPHOCYTIC LEUKEMIA

PET-Directed Strategies in
Lymphoma

CHAIR
John A. Radford, MD
Christie Hospital NHS Foundation Trust
Manchester, United Kingdom

SPEAKERS
Franco Cavalli, MD
Oncology Institute of Southern Switzerland
Bellinzona, Switzerland

Ranjana H. Advani, MD
Stanford University School of Medicine
Stanford, CA
CAVALLI, CERIANI, AND ZUCCA

Functional Imaging Using 18-Fluorodeoxyglucose PET in the


Management of Primary Mediastinal Large B-Cell Lymphoma:
The Contributions of the International Extranodal Lymphoma
Study Group
Franco Cavalli, MD, Luca Ceriani, MD, and Emanuele Zucca, MD

OVERVIEW

Primary mediastinal large B-cell lymphoma (PMLBCL) is recognized as a distinct disease entity. Treatment outcomes appear better
than in other diffuse large B-cell lymphoma (DLBCL) types, partly because of their earlier stage at presentation and the younger
age of most patients. If initial treatment fails, however, the results of salvage chemotherapy and myeloablative treatment are
poor. The need to avoid relapses after initial therapy has led to controversy over the extent of front-line therapy, particularly
whether consolidation radiotherapy to the mediastinum is always required and whether the 18-fluorodeoxyglucose (18F-FDG)
uptake detected by PET-CT scan can be used to determine its requirements. Functional imaging using PET-CT generally allows
distinguishing of residual mediastinal masses containing active lymphoma from those with only sclerotic material remaining. The
International Extranodal Lymphoma Study Group (IELSG) conducted the prospective IELSG-26 study, which showed that a five-
point visual scale can be used to define metabolic response after immunochemotherapy and that a cut point based on liver uptake
discriminates effectively between high or low risk of failure, with 5-year progression-free survival (PFS) of 99% versus 68% and 5-
year overall survival (OS) of 100% versus 83%. This study also showed that a baseline quantitative PET parameter, namely the total
lesion glycolysis describing the metabolic tumor burden, can be a powerful predictor of PMLBCL outcomes and warrants further
validation as a biomarker. The ongoing IELSG-37 randomized study addresses the need for consolidation mediastinal radiotherapy
in patients in whom a complete metabolic response (CMR) can be seen on PET scans after standard immunochemotherapy.

P rimary mediastinal large B-cell lymphoma is recognized


by the World Health Organizations classification of
hematopoietic and lymphoid tissue tumors as a distinct
Cell of Origin and Immunophenotype
Immunohistochemical characterization and molecular studies
strongly suggest that PMLBCL is of germinal center or post-
entity on clinicopathologic1,2 and molecular3-5 criteria. It germinal center origin.2,5 Phenotypic analysis shows positivity
accounts for less than 5% of non-Hodgkin lymphomas and for CD45, CD79a, CD22, CD19, and CD20, with negativity for
comprises approximately 5%10% of the DLBCL.1,2,6 Patients CD3, CD10, CD21, and class I/II major histocompatibility
with PMLBCL tend to be younger than the other DLBCL cases antigens. Expression of surface immunoglobulins is lost
with a median age at diagnosis in the third to fourth decade despite the presence of isotype-switched immunoglobulin
and a female preponderance.2,7 This lymphoma is clinically genes.1,8,9 CD30 staining is observed in most cases but is
characterized by a rapidly progressive anterior mediastinal weaker and less homogeneous than in Hodgkin lymphoma
mass, often with local invasion and compressive syndromes, or anaplastic large cell lymphoma1,5; BCL2 and BCL6 proteins
and by recurrence at unusual sites, such as the liver, kidneys, are often expressed.1
and central nervous system.6
Molecular Genetics
BIOLOGIC PECULIARITIES OF PMLBCL Primary mediastinal large B-cell lymphoma is characterized
Primary mediastinal large B-cell lymphoma is considered to by peculiar genetic features that make this entity closer to
typically arise from a small population of B cells within the classic Hodgkin lymphoma than to DLBCL.3-5,10 The main
thymus and, thymic components such as Hassall corpuscles characteristics are represented by deregulated immune
may be identified in the pathology specimen.2 checkpoint and JAK/STAT signaling, via DNA gains and

From the Oncology Institute of Southern Switzerland, Lymphoma UnitOspedale San Giovanni, Bellinzona, Switzerland.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Franco Cavalli, MD, Ospedale Regionale Bellinzona e Valli, Bellinzona, CH-6500, Switzerland; email: franco.cavalli@eoc.ch.

2016 by American Society of Clinical Oncology.

e368 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


ROLE OF PET IN THE MANAGEMENT OF PMLBCL

amplifications at 9p, affecting PDL2 and JAK2, and recurrent PMLBCL after first-generation (standard CHOP or CHOP-like
somatic mutations of the STAT6 and SOCS1 genes.1,5,9,11-15 regimens), third-generation (dose-intensive/alternating reg-
There are also recurrent point mutations of the XPO1 gene, imens such as MACOP-B, VACOP-B, ProMACE, CytaBOM), and
coding for the Exportin-1 cargo protein that mediates the high-dose chemotherapy strategies with autologous stem cell
nuclear export of multiple tumor suppressor proteins, in- transplant, frequently including adjuvant radiation therapy. A
cluding p53. Although their biologic significance is still under total of 426 previously untreated patients with confirmed
investigation, XPO1 mutations appear to be of potential diagnosis were enrolled by 20 institutions. Projected 10-year
clinical relevance in terms of outcome prediction and dif- PFS rates were 35%, 67%, and 78%, after first generation, third
ferential diagnosis with classic Hodgkin lymphoma and gray- generation, and high-dose chemotherapy strategies, re-
zone lymphomas.16,17 spectively (p , .001). Projected 10-year OS rates were 44%,
71%, and 77%, respectively (p , .001), after median follow-
ups from diagnosis of 52.3 months, 54.9 months, and
CURRENT CLINICAL CONTROVERSIES 35.8 months, respectively. Very similar results were reported
The outcome of therapy for patients with PMLBCL is gen- by other retrospective studies from Italy27 and North
erally better than for those with other DLBCL types, possibly America.18,28,29
also as a result of the younger age of the patients and earlier In the last decade, the inclusion of rituximab as part of
stage at presentation. In untreated PMLBCL, the response initial therapy for CD20+ B-cell lymphomas has clearly im-
to combination chemotherapy is very good and is commonly proved response rates and OS in several DLBCL randomized
followed by consolidation involved-field radiotherapy (IFRT). trials.30,31 Although direct comparison between different
After the addition of rituximab to doxorubicin, cyclophos- nonrandomized clinical studies with or without rituximab
phamide, vincristine, and prednisone (CHOP) or CHOP-like is problematic, the rituximab regimen of CHOP (R-CHOP)
regimens, the reported PFS and OS rates at 3 years are is likely to have also improved the outcome in PMLBCL.
approximately 75%80% and 85%90%, respectively.18,19 Indeed, in a Canadian retrospective study, the addition of
However, there is a subset of patients in whom early re- rituximab to CHOP chemotherapy resulted in the same 5-year
fractory disease develops, often during the initial courses of OS obtained with more intensive regimens of MACOP-B/
treatment, with a high failure rate2,7: in the case this initial VACOP-B.18 In the subgroup analysis of the MINT interna-
treatment fails, the results of salvage chemotherapy and tional randomized trial, the subgroup of patients with PMLBCL
myeloablative treatment are often very poor.20 The need to treated with rituximab plus chemotherapy (CHOP in most
maximize cure rates with initial therapy has led to contro- cases) showed a significantly better 3-year PFS than did pa-
versy over the utility of consolidation radiotherapy to the tients treated with chemotherapy alone (87.7% vs. 64.1%;
mediastinum. The optimal chemotherapy regimen also re- p = .005).32 Conversely, in a phase II Italian trial on MACOP-B/
mains controversial. VACOP-B plus rituximab followed by mediastinal IFRT, the PFS
The outcomes reported in the prerituximab era by several improvement was not significant compared with historical
European groups have suggested that third-generation controls.33
regimens (e.g., MACOP-B or VACOP-B) may be superior to Overall, the combination of immunochemotherapy plus
the CHOP regimen.21-25 Improved PFS and OS with more IFRT results in a 3-year PFS rate of 80%85%.32-34 Recent
aggressive chemotherapy regimens compared with CHOP- guidelines35 recommended chemotherapy with a CHOP/
like regimens were confirmed by a large European survey MACOP-Blike regimen plus rituximab, followed by medi-
conducted by the IELSG.26 This multinational retrospective astinal IFRT as standard front-line treatment of PMLBCL.
study (IELSG-9) compared the outcomes of patients with However, the role of adjuvant mediastinal irradiation in
patients who achieve CMR with chemotherapy remains
unclear. Retrospective series suggest that the best outcomes
KEY POINTS are seen when consolidation radiotherapy is given to the
mediastinum,26,27 particularly in the large proportion of
PMLBCL is a distinct clinicopathologic and biologic type patients with a residual mediastinal mass at completion of
of large-cell lymphoma. chemotherapy.36,37 Indeed, before rituximab introduction,
Survival is usually excellent with intensive front-line the use of consolidation radiotherapy for PMLBCL was a
immunochemotherapy regimens, but the few patients historical standard of care, based on poor results after CHOP
in whom it fails have a particularly dismal outcome. chemotherapy alone compared with the excellent results in
18
F-FDG PET-CT is a very important staging tool for series where almost all patients underwent irradiation.2,7
patients with PMLBCL.
This combined modality strategy was also preferred by many
The metabolic activity on PET scans is a powerful
predictor of PMLBCL outcome.
to maximize cures at the first attempt because of the dismal
An ongoing randomized study is addressing the need of outcomes for patients in whom recurrent disease develops.7
consolidation mediastinal radiotherapy in patients who However, concerns about long-term effects of mediastinal
have shown a complete metabolic response on PET radiotherapy, including coronary heart disease, valvular
scans. disorders, heart failure, and risk of second tumors (mainly
cancers of the breast, lung, and thyroid), justify studies

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e369


CAVALLI, CERIANI, AND ZUCCA

aiming to investigate scenarios where radiotherapy may be FIGURE 1. IELSG-37 Study Design
omitted.38-40 Because modern radiotherapy techniques may
minimize the morbidity to other tissues and/or reduce the
risk of long-term side effects,41-43 but do not completely
address the risks of late effects, safety remains a central
issue.44,45 For this reason, in a patient group dominated by
young women, some investigators have elected to exclude
consolidation radiotherapy. In a phase II study at the Na-
tional Cancer Institute, the use of dose-adjusted REPOCH
combined with rituximab seemed to allow omission of ra-
diotherapy without compromising cure rates (the event-free
survival rate was 93%, and the OS rate was 97%),46 and in the
BC Cancer Agency, the decision to spare radiotherapy in
patients with a negative 18F-FDG PET-CT at the completion of
R-CHOP, reduced the use of radiotherapy in patients from
80% to 38%, apparently with good outcomes.47 Unfortunately,
to date there are no published randomized trials in which this
question has been definitely answered.

THE IELSG-37 RANDOMIZED TRIAL


The ongoing IELSG-37 (NCT01599559) study was designed to
address the issue of the role of irradiation in the current
rituximab era offering randomization to standard radio-
This is a prospective, randomized, noninferiority phase III trial that requires the
therapy versus no further treatment for patients who were random assignment of 376 patients. Patients are treated with standard
found on PET scan to be tumor free at the end of initial immunochemotherapy regimens currently in use for PMLBCL (e.g., R-CHOP, DA-
EPOCH-R, R- ACVBP, R-VACOP-B, or R-MACOP-B). Restaging PET-CT scans will be
rituximab-chemotherapy treatment. The study design is performed at 56 weeks after the last immunochemotherapy administration. Central
summarized in Fig. 1. review of PET-CT scans is mandatory before randomization. All patients with
a negative PET-CT scan, either with complete or partial radiologic regression of the
This is the first study on a large international basis that mediastinal mass, will be randomly assigned to receive consolidation IFRT (30 Gy) or
aimed to define in a prospective, controlled way the role of observation. Based on the results of the previous IELSG-26 study, a negative PET scan
is defined using the liver uptake as the cutoff point (Figs. 2 and 3).
radiotherapy in first-line treatment of these patients. This Abbreviation: IFRT, involved-field radiotherapy.
very ambitious study, to which more than 500 patients will
be enrolled (more than half of them have been enrolled so
Gallium scans24 or, more recently, 18F-FDG PET-CT scans60
far), aims at personalizing treatment and, it is hoped, will
suggest that it may be possible to distinguish residual me-
determine whether radiotherapy can be safely spared in
diastinal masses, which contain active lymphoma, from
patients achieving CMR indicated on PET scans after in-
those in which only sclerotic material remains. However,
duction immunochemotherapy. The use of PET in this trial is
they have not clarified whether radiotherapy could be
based on the result achieved by the IELSG-26 study, which
avoided solely on the basis of a negative PET-CT scan.59,61
will be described in the following section.
In a retrospective study of 54 patients with PMLBCL who
were treated with the R-CHOP/ICE dose-dense regimen
THE ROLE OF FUNCTIONAL IMAGING: without mediastinal radiotherapy, the Memorial Sloan
CONTRIBUTIONS FROM THE IELSG-26 STUDY Kettering Cancer Center group reported 3-year OS and PFS
PET-CT is currently considered the best imaging tool for rates of 88% and 78%, respectively, in patients who had
staging and response assessment in 18F-FDGavid lympho- negative PET-CT scans at the end of the chemotherapy
mas,48 as defined by the recommendations of the Lugano regimen. Furthermore, an abnormal result in an interim PET-
Classification for initial evaluation, staging, and response CT scan was still evident in 47% of patients and did not
assessment of Hodgkin lymphoma and non-Hodgkin lym- predict a worse PFS rate.62
phoma.49 PET-CT scans are increasingly used as prognostic In a BC Cancer Agency retrospective study of 196 patients
indicators in classic Hodgkin lymphoma50-53 and, to a minor with DLBCL treated with R-CHOP (25 with PMLBCL) with a
extent, in DLBCL.54-57 Although PMLBCL is an 18F-FDGavid residual mass of greater than 2 cm, post-therapy PET-CT scan
lymphoma,48,49,58 very few studies on the use of PET-CT have was negative in 62% of patients who were then only ob-
focused specifically on PMLBCL. served, whereas 34% had a positive post-therapy PET-CT
Establishing whether residual masses, which are almost scan and received radiotherapy (IFRT, 3040 Gy) to sites
invariably visible on CT scans at the completion of initial noted as positive on PET-CT scan. No differences were
chemotherapy, represent viable lymphoma or merely fi- observed in terms of 3-year PFS (80% vs. 75%; p = .41)
brotic scar tissue is a major problem in the clinical man- between the patients with negative PET-CT versus positive
agement of PMLBCL.59 Studies of functional imaging using PET-CT scans.63

e370 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


ROLE OF PET IN THE MANAGEMENT OF PMLBCL

PET-CT Response After Immunochemotherapy FIGURE 2. Deauville Criteria for the Visual
Currently, the introduction of rituximab has changed the Interpretation of PET/CT Scan Using a Five-Point Scale
therapeutic scenario, whereas the general use of 18F-FDG (Deauville Score)
PET-CT to evaluate mediastinal residual disease after che-
motherapy has led to the assumption that a lymphoma can
also be considered in complete remission in the presence
of a negative PET-CT scan showing residual lesion.64
Only scanty and mainly retrospective studies on the validity
of PET in response evaluation were specifically focused on
PMLBCL.65 The IELSG-26 study of PMLBCL represents a first,
and thus far unique, attempt to obtain data on this issue
in a prospective manner.66 This is the largest study on the
results of 18F-FDG PET-CT scanning in PMLBCL. The outcome
after induction treatment in this series, in which a majority of
patients received consolidation radiotherapy of treatment
with rituximab and anthracycline-containing chemotherapy, Interim and end-of-treatment PET/CT scans are scored according to uptake in
sites involved at baseline PET/CT by lymphoma as: (1) no uptake, (2) 18F-FDG
is very good, with more than 90% of patients projected to uptake equal to or below the mediastinum blood pool uptake, (3) 18F-FDG uptake
equal to or below the liver uptake, (4) 18F-FDG uptake moderately higher than the
be alive and progression-free at 5 years despite a lower liver uptake, or (5) 18F-FDG uptake markedly higher (i.e., more than two-fold) than the
proportion of patients classified as having CMR. Among 125 uptake in the liver and/or new lesions.67 At the end of treatment, a score of 13 is
currently considered to indicate a complete metabolic response in most instances,48
patients prospectively enrolled in this study, 115 were eligible whereas a score of 4 or 5 is considered to indicate lymphoma persistence or
for central review of PET-CT scans at the completion of recurrence. When new areas of uptake are considered unlikely to be caused by
lymphoma, they are usually reported as Score X.48
standard immunochemotherapy, using the Deauville five- Abbreviations: SUVmax, maximum standardized uptake value;18FDG,
point scale (Fig. 2).67 Consolidation radiotherapy was given 18-fluorodeoxyglucose.
to 102 patients. Fifty-four patients (47%) achieved CMR,
defined as a completely negative scan or with residual the treatment of DLBCL and Hodgkin lymphoma.67 A cutoff
18
F-FDG activity equal to or below the mediastinal blood point for PET positivity below the uptake in the liver dis-
pool (MBP) uptake. In the remaining 61 patients (53%), the criminates most effectively between groups of PMLBCL at
residual uptake was higher than the MBP but below the high or low risk of treatment failure.
liver uptake in 27 (23%), slightly higher than the liver uptake
in 24 (21%), and markedly higher in 10 (9%). Complete
The Problem of False-Positive PET-CT Scans After
metabolic response after immunochemotherapy predicted Immunochemotherapy
higher 5-year PFS (98% vs. 82%; p = .0044) and OS (100% vs. Although the outcome of treatment was very good, after
91%; p = .0298). Patients with residual uptake higher than immunochemotherapy a persistently positive PET-CT scan
the MBP but below liver uptake had equally good outcomes, was seen in more than half of the patients (53%, with
without any recurrence. Using the liver uptake as the cutoff Deauville score 35), which contributed to a positive
for PET positivity (boundary of score 34) discriminated predictive value substantially lower than that reported for
most effectively between high or low risk of failure, with other types of DLBCL and Hodgkin lymphoma. With the use
5-year PFS of 99% versus 68% (p , .0001) and 5-year OS of of the liver cutoff point to define a CMR, the rate of patients
100% versus 83% (p = .0003). Moreover, moving the cut with PET-positive scans decreases (30% with score 4 or 5).
point for the definition of CMR from the MBP to the liver The results of IELSG-26 appear to be in line with those
uptake increased the specificity, and the positive predictive observed in a series of patients treated with the infusional
value rose from 18% to 32%, without loss of sensitivity (Fig. 3). doseadjusted R-EPOCH (DA-R-EPOCH) regimen, which
The use of liver uptake as the cutoff for PET positivity resulted showed 50% PET positivity, defined by 18F-FDG uptake
in a clearer distinction between risk subgroups, both in terms greater than the MBP at the completion of chemotherapy
of PFS (p , .0001) and OS (p = .0003). The International but only 3 of 18 progressions without the use of consoli-
Prognostic Index (IPI) and age-adjusted IPI at diagnosis and dation radiotherapy in this group.46 Because all of the cases
the chemotherapy regimen did not significantly correlate with that recurred had a standardized uptake value (SUV) of at
the PET response, whereas initial bulky disease (. 10 cm) was least 5, the authors suggested that radiotherapy might
significantly associated with a persistent postchemotherapy reasonably be omitted for nearly all patients treated with
residual uptake above the liver cutoff (p = .005). This cor- this regimen.46 The IELSG-26 study likely carries a similarly
relation with tumor bulk was only of borderline significance high false-positive rate, although PMLBCL is clearly ra-
for the MBP cut point (p = .05). diosensitive, so it is also possible that the almost universal
This study is also the first attempt to validate the use of use of radiotherapy in this study may have eliminated
PET-CT scanning in restaging an aggressive lymphoma at the residual lymphoma in some instances. However, the IELSG-
completion of chemotherapy using the Deauville criteria 26 study design did not allow us to draw any conclusions
originally developed for interim PET-CT evaluation during on the benefit of radiotherapy.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e371


CAVALLI, CERIANI, AND ZUCCA

FIGURE 3. IELSG-26 Study Results Using the Five-Point of most patients, thymic rebound should also be considered
Scale (Deauville Score) for the Visual Interpretation of as a possible explanation of a positive PET scan.
PET-CT Scans Obtained at 34 Weeks After
Immunochemotherapy Utility of Baseline 18F-FDG PET-CT Functional
Parameters in Defining Prognosis
Because a successful primary treatment is crucial for
PMLBCL, the early identification of high-risk patients at risk
for relapse is critical to allow for alternative therapeutic
strategy. Valuable prognosis factors are therefore warranted
for tailoring therapy and possibly reducing treatment tox-
icity, as IPI and age-adjusted IPI have proved not useful for
this purpose. Other proposed clinical predictors of survival
have not been validated prospectively, and biomarkers
useful for risk stratification are not yet available.58
In this sense, the IELSG-26 study also aimed at exploring
the potential of quantitative 18F-FDG PET-CT as a biomarker
in PMLBCL by investigating whether the main quantitative
baseline PET-CTderived metabolic parameters can predict
prognosis and aimed to compare them with the commonly
used clinical indices.69
The metabolic activity defined by maximum SUV (SUVmax),
the metabolic tumor volume, and the total lesion glycolysis
(TLG), an index of metabolic tumor burden, were measured in
103 patients on baseline 18F-FDG PET-CT using a practical and
reproducible method to segment the mediastinal mass, based
on the 25% thresholding of the tumor SUVmax.69 This method
was developed considering the specific presentation features
of PMLBCL, where the technical difficulties of quantitative
PET-CT are reduced because the tumor burden is mainly
(A) PET-CT interpretation in a blind central review performed on 115 evaluable
limited to a unique bulky mass.58
patients and change in the positive predictive value induced by a shift of the All patients received combination immunochemotherapy
cutoff point for the definition of CMR from Deauville score 2 (MBP uptake) to
score 3 (liver uptake). (B) Kaplan-Meier estimates of OS in PMLBCL according to
with doxorubicin and rituximab-based regimens; 93 received
positive PET using the MBP uptake (Deauville score 35) or the liver uptake as consolidation radiotherapy. Cutoff points were determined
a cutoff point (Deauville score 45).
Abbreviations: MBP, mediastinal blood pool; NPV, negative predictive value; PPV,
using the receiver operating characteristic curve.69
positive predictive value; OS, overall survival; CMR, complete metabolic response. At a median follow-up of 36 months, PFS and OS rates were
Modified from Martelli et al 201466.
87% and 94%, respectively. In univariate analysis, elevated
metabolic tumor volume and TLG were significantly associ-
PET-CT Response After Consolidation Radiotherapy ated with worse PFS and OS. Only TLG retained statistical
The evaluation of postirradiation PET-CT scans in the IELSG- significance for both OS (p = .001) and PFS (p , .001) in
26 study has thus far been reported only as a meeting multivariate analysis. At 5 years, OS was 100% for patients
abstract.68 In this study, 88 of 125 patients were eligible for with low TLG versus 80% for those with high TLG (p = .0001),
central review of PET-CT scans at 8 weeks after the com- whereas PFS was 99% versus 64%, respectively (p , .0001).69
pletion of radiotherapy. The CMR (using the liver cutoff This study represents the largest prospective series of
point) rate increased from 74% after immunochemotherapy PMLBCL investigated with central review of PET-CT scans, in
to 89% after irradiation. At a median follow-up of 60 months, which the quantitative functional parameters were ana-
no patients with CMR after radiotherapy have relapsed. lyzed. The PET-CT imaging was obtained by following a
Notably, in this study the few patients with a Deauville score strictly defined technical procedure, although detailed cross-
of 4 also had an excellent outcome, suggesting that they do calibration for equipment at the different centers was not
not necessarily require additional therapy because the re- possible. Total lesion glycolysis on baseline PET-CT appeared
sidual 18F-FDG uptake may be caused by an inflammatory to be a powerful predictor of PMLBCL outcomes and war-
reaction. In this context, it seems worth mentioning that the rants further validation as a biomarker. Indeed, this easily
18
F-FDG uptake in untreated PMLBCL is likely a result of the and early accessible parameter can help identify the few
lymphoma cells that are the main component of the tumor, patients who will eventually experience relapse and may be
even though a contributing microenvironment uptake cannot useful for designing trials aimed at improving their poor
be excluded58 given its relevant role in PMLBCL.5 prognosis (e.g., using TLG to distinguish patients being
In post-treatment evaluation of PMLBCL, because of their considered for elective front-line consolidation with myeloa-
thymic derivation and anatomic localization and young age blative therapies). However, despite the very high sensitivity

e372 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


ROLE OF PET IN THE MANAGEMENT OF PMLBCL

(92%) of TLG with a 98% negative predictive value for PFS, the value without a detrimental effect on negative predictive
positive predictive value was only 36%. Hence, a better se- value.72 However, because early stratification is preferred,
lection of high-risk patients would be useful to choose those to the integration of other prognostic indicators (such as in-
submit to intensive treatment.58 terim PET-CT data or baseline molecular and clinical fea-
tures) with baseline quantitative 18F-FDG PET-CT should be
Integrative PET: A Novel Promising Tool addressed by future studies to provide an earlier and clin-
Integrative 18F-FDG PET in lymphoma is a new concept that ically meaningful selection of patients with high-risk PMLBCL
can be defined as a method of prognostic imaging, where suitable for risk-adapted therapeutic strategies.58
imaging data obtained with different techniques are com-
bined together or with data from other fields (clinical, bi-
ologic, molecular) to improve the risk stratification of ACKNOWLEDGMENT
patients.70 In classic Hodgkin lymphoma, it has been sug- We thank Rita Gianascio Gianocca and Sarah Ortelli Giannakis
gested that baseline metabolic tumor volume may be for the secretarial and editorial assistance; the chairpersons
combined with the interim PET-CT score to identify patients (Prof. Peter Johnson and Prof. Maurizio Martelli), data
at different levels of risk.71 Preliminary data from the IELSG- managers and investigators of the IELSG-26 and IELSG-37
26 series of PMLBCL showed that the combination of studies on PET-CT use in PMLBCL; Dr. Francesco Bertoni for his
baseline TLG with end-treatment PET-CT more accurately critical suggestions; and Swiss Cancer Research for the sup-
identified patients at risk, improving the positive predictive port to the IELSG activities.

References
1. Gaulard P, Harris NL, Pileri SA, et al. Primary mediastinal (thymic) large 13. Weniger MA, Gesk S, Ehrlich S, et al. Gains of REL in primary mediastinal
B-cell lymphoma. In Swerdlow S, Campo E, Harris NL, et al (eds). B-cell lymphoma coincide with nuclear accumulation of REL protein.
WHO Classification of Tumours of Haematopoietic and Lymphoid Tis- Genes Chromosomes Cancer. 2007;46:406-415.
sues. Lyon: IARC, 2008;250-253. 14. Joos S, Otan~o-Joos MI, Ziegler S, et al. Primary mediastinal (thymic) B-cell
2. Johnson PW, Davies AJ. Primary mediastinal B-cell lymphoma. Hema- lymphoma is characterized by gains of chromosomal material including
tology (Am Soc Hematol Educ Program). 2008;2008:349-358. 9p and amplification of the REL gene. Blood. 1996;87:1571-1578.
3. Savage KJ, Monti S, Kutok JL, et al. The molecular signature of medi- 15. Melzner I, Bucur AJ, Bruderlein S, et al. Biallelic mutation of SOCS-1
astinal large B-cell lymphoma differs from that of other diffuse large B- impairs JAK2 degradation and sustains phospho-JAK2 action in the
cell lymphomas and shares features with classical Hodgkin lymphoma. MedB-1 mediastinal lymphoma line. Blood. 2005;105:2535-2542.
Blood. 2003;102:3871-3879. 16. Mareschal S, Dubois S, Viailly PJ, et al. Whole exome sequencing of relapsed/
4. Rosenwald A, Wright G, Leroy K, et al. Molecular diagnosis of primary refractory patients expands the repertoire of somatic mutations in diffuse
mediastinal B cell lymphoma identifies a clinically favorable subgroup of large B-cell lymphoma. Genes Chromosomes Cancer. 2016;55(3):251-267.
diffuse large B cell lymphoma related to Hodgkin lymphoma. J Exp Med. 17. Jardin F, Pujals A, Pelletier I, et al. Whole exome sequencing of refractory
2003;198:851-862. aggressive B-cell lymphomas identified recurrent mutations of the
5. Steidl C, Gascoyne RD. The molecular pathogenesis of primary medi- exportin 1 gene (XPO1) in primary mediastinal B-cell lymphoma subtype, a
astinal large B-cell lymphoma. Blood. 2011;118:2659-2669. LYSA study. Hematol Oncol. 2015;33:124-125 (suppl; abstr 048).
6. van Besien K, Kelta M, Bahaguna P. Primary mediastinal B-cell lymphoma: 18. Savage KJ, Al-Rajhi N, Voss N, et al. Favorable outcome of primary
mediastinal large B-cell lymphoma in a single institution: the British
a review of pathology and management. J Clin Oncol. 2001;19:1855-1864.
Columbia experience. Ann Oncol. 2006;17:123-130.
7. Martelli M, Di Rocco A, Russo E, et al. Primary mediastinal lymphoma:
19. Rieger M, Osterborg A, Pettengell R, et al; MabThera International Trial
diagnosis and treatment options. Expert Rev Hematol. 2015;8:173-186.
(MInT) Group. Primary mediastinal B-cell lymphoma treated with
8. Pileri SA, Gaidano G, Zinzani PL, et al. Primary mediastinal B-cell
CHOP-like chemotherapy with or without rituximab: results of the
lymphoma: high frequency of BCL-6 mutations and consistent ex-
Mabthera International Trial Group study. Ann Oncol. 2011;22:664-670.
pression of the transcription factors OCT-2, BOB.1, and PU.1 in the
20. Kuruvilla J, Pintilie M, Tsang R, et al. Salvage chemotherapy and au-
absence of immunoglobulins. Am J Pathol. 2003;162:243-253.
tologous stem cell transplantation are inferior for relapsed or refractory
9. Pileri SA, Zinzani PL, Gaidano G, et al; International Extranodal Lym-
primary mediastinal large B-cell lymphoma compared with diffuse large
phoma Study Group. Pathobiology of primary mediastinal B-cell lym-
B-cell lymphoma. Leuk Lymphoma. 2008;49:1329-1336.
phoma. Leuk Lymphoma. 2003;44(suppl 3):S21-S26. 21. Cazals-Hatem D, Lepage E, Brice P, et al. Primary mediastinal large B-cell
10. Feuerhake F, Kutok JL, Monti S, et al. NFkappaB activity, function, and lymphoma. A clinicopathologic study of 141 cases compared with 916
target-gene signatures in primary mediastinal large B-cell lymphoma and nonmediastinal large B-cell lymphomas, a GELA (Groupe dEtude des
diffuse large B-cell lymphoma subtypes. Blood. 2005;106:1392-1399. Lymphomes de lAdulte) study. Am J Surg Pathol. 1996;20:877-888.
11. Ritz O, Guiter C, Castellano F, et al. Recurrent mutations of the STAT6 22. Bieri S, Roggero E, Zucca E, et al. Primary mediastinal large B-cell
DNA binding domain in primary mediastinal B-cell lymphoma. Blood. lymphoma: the need for prospective controlled clinical trials. Leuk
2009;114:1236-1242. Lymphoma. 1999;35:537-544.
12. Hao Y, Chapuy B, Monti S, et al. Selective JAK2 inhibition specifically 23. Lazzarino M, Orlandi E, Paulli M, et al. Primary mediastinal B-cell
decreases Hodgkin lymphoma and mediastinal large B-cell lymphoma lymphoma with sclerosis: an aggressive tumor with distinctive clini-
growth in vitro and in vivo. Clin Cancer Res. 2014;20:2674-2683. cal and pathologic features. J Clin Oncol. 1993;11:2306-2313.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e373


CAVALLI, CERIANI, AND ZUCCA

24. Zinzani PL, Martelli M, Magagnoli M, et al. Treatment and clinical 37. De Sanctis V, Finolezzi E, Osti MF, et al. MACOP-B and involved-field
management of primary mediastinal large B-cell lymphoma with radiotherapy is an effective and safe therapy for primary mediastinal
sclerosis: MACOP-B regimen and mediastinal radiotherapy monitored large B cell lymphoma. Int J Radiat Oncol Biol Phys. 2008;72:1154-1160.
by (67)Gallium scan in 50 patients. Blood. 1999;94:3289-3293. 38. Hancock SL, Hoppe RT. Long-term complications of treatment and
25. Zinzani PL, Martelli M, Bendandi M, et al. Primary mediastinal large B- causes of mortality after Hodgkins disease. Semin Radiat Oncol. 1996;6:
cell lymphoma with sclerosis: a clinical study of 89 patients treated with 225-242.
MACOP-B chemotherapy and radiation therapy. Haematologica. 2001; 39. van Leeuwen FE, Klokman WJ, Veer MB, et al. Long-term risk of second
86:187-191. malignancy in survivors of Hodgkins disease treated during adoles-
26. Zinzani PL, Martelli M, Bertini M, et al; International Extranodal Lym- cence or young adulthood. J Clin Oncol. 2000;18:487-497.
phoma Study Group (IELSG). Induction chemotherapy strategies for 40. Henderson TO, Amsterdam A, Bhatia S, et al. Systematic review: sur-
primary mediastinal large B-cell lymphoma with sclerosis: a retro- veillance for breast cancer in women treated with chest radiation for
spective multinational study on 426 previously untreated patients. childhood, adolescent, or young adult cancer. Ann Intern Med. 2010;
Haematologica. 2002;87:1258-1264. 152:444-455.
27. Todeschini G, Secchi S, Morra E, et al. Primary mediastinal large B-cell 41. Yoon M, Ahn SH, Kim J, et al. Radiation-induced cancers from modern
lymphoma (PMLBCL): long-term results from a retrospective multi- radiotherapy techniques: intensity-modulated radiotherapy versus
centre Italian experience in 138 patients treated with CHOP or MACOP- proton therapy. Int J Radiat Oncol Biol Phys. 2010;77:1477-1485.
B/VACOP-B. Br J Cancer. 2004;90:372-376. 42. Bucci MK, Bevan A, Roach M III. Advances in radiation therapy: con-
28. Hamlin PA, Portlock CS, Straus DJ, et al. Primary mediastinal large B-cell ventional to 3D, to IMRT, to 4D, and beyond. CA Cancer J Clin. 2005;55:
lymphoma: optimal therapy and prognostic factor analysis in 141 117-134.
consecutive patients treated at Memorial Sloan Kettering from 1980 to 43. Tsang RW, Gospodarowicz MK. Radiation therapy for localized low-
1999. Br J Haematol. 2005;130:691-699. grade non-Hodgkins lymphomas. Hematol Oncol. 2005;23:10-17.
29. Dunleavy K, Pittaluga S, Janik J, et al. Primary mediastinal large B-cell 44. van Nimwegen FA, Schaapveld M, Cutter DJ, et al. Radiation Dose-
lymphoma (PMBL) outcome is significantly improved by the addition of response relationship for risk of coronary heart disease in survivors of
rituximab to dose adjusted (DA)-EPOCH and overcomes the need for Hodgkin lymphoma. J Clin Oncol. 2016;34:235-243.
radiation. Blood. 2005;106:929. 45. Nieder C, Schill S, Kneschaurek P, et al. Comparison of three different
30. Pfreundschuh M, Schubert J, Ziepert M, et al; German High-Grade Non- mediastinal radiotherapy techniques in female patients: Impact on
Hodgkin Lymphoma Study Group (DSHNHL). Six versus eight cycles of heart sparing and dose to the breasts. Radiother Oncol. 2007;82:
bi-weekly CHOP-14 with or without rituximab in elderly patients with 301-307.
aggressive CD20+ B-cell lymphomas: a randomised controlled trial 46. Dunleavy K, Pittaluga S, Maeda LS, et al. Dose-adjusted EPOCH-
(RICOVER-60). Lancet Oncol. 2008;9:105-116. rituximab therapy in primary mediastinal B-cell lymphoma. N Engl J
31. Coiffier B, Thieblemont C, Van Den Neste E, et al. Long-term outcome of Med. 2013;368:1408-1416.
patients in the LNH-98.5 trial, the first randomized study comparing 47. Savage KJ, Yenson PR, Shenkier T, et al. The outcome of primary me-
rituximab-CHOP to standard CHOP chemotherapy in DLBCL patients: diastinal large B-cell lymphoma (PMLBCL) in the R-CHOP treatment era.
a study by the Groupe dEtudes des Lymphomes de lAdulte. Blood. Blood. 2012;120:303.
2010;116:2040-2045. 48. Barrington SF, Mikhaeel NG, Kostakoglu L, et al. Role of imaging in the
32. Trneny M, Osterborg A, Pettengel R, et al. The addition of rituximab staging and response assessment of lymphoma: consensus of the In-
eliminates the negative prognostic impact of PMLBCL compared to ternational Conference on Malignant Lymphomas Imaging Working
DLBCL in young patients with CD20-positive aggressive lymphomas Group. J Clin Oncol. 2014;32:3048-3058.
receiving a CHOP-like chemotherapy: results of a subgroup analysis of 49. Cheson BD, Fisher RI, Barrington SF, et al; Alliance, Australasian Leu-
the Mabthera International Trial Group (MInT) study. Blood. 2008;112: kaemia and Lymphoma Group; Eastern Cooperative Oncology Group;
839. European Mantle Cell Lymphoma Consortium; Italian Lymphoma
33. Zinzani PL, Stefoni V, Finolezzi E, et al. Rituximab combined with Foundation; European Organisation for Research; Treatment of Cancer/
MACOP-B or VACOP-B and radiation therapy in primary mediastinal Dutch Hemato-Oncology Group; Grupo Espan ~ol de Medula Osea;

large B-cell lymphoma: a retrospective study. Clin Lymphoma Myeloma. German High-Grade Lymphoma Study Group; German Hodgkins Study
2009;9:381-385. Group; Japanese Lymphorra Study Group; Lymphoma Study Associa-
34. Vassilakopoulos TP, Angelopoulou MK, Galani Z, et al. Rituximab-CHOP tion; NCIC Clinical Trials Group; Nordic Lymphoma Study Group;
(R-CHOP) and radiotherapy (RT) for primary mediastinal large B-cell Southwest Oncology Group; United Kingdom National Cancer Research
lymphoma (PMLBCL): an update. Haematologica. 2006;91:260 (abstr Institute. Recommendations for initial evaluation, staging, and response
0704). assessment of Hodgkin and non-Hodgkin lymphoma: the Lugano class-
35. Zinzani PL, Martelli M, Poletti V, et al; Italian Society of Hematology; ification. J Clin Oncol. 2014;32:3059-3068.
Italian Society of Experimental Hematology; Italian Group for Bone 50. Adams HJ, Nievelstein RA, Kwee TC. Prognostic value of interim FDG-PET
Marrow Transplantation. Practice guidelines for the management of in Hodgkin lymphoma: systematic review and meta-analysis. Br J
extranodal non-Hodgkins lymphomas of adult non-immunodeficient Haematol. 2015;170:356-366.
patients. Part I: primary lung and mediastinal lymphomas. A project of 51. Sickinger MT, von Tresckow B, Kobe C, et al. Positron emission
the Italian Society of Hematology, the Italian Society of Experimental tomography-adapted therapy for first-line treatment in individuals
Hematology and the Italian Group for Bone Marrow Transplantation. with Hodgkin lymphoma. Cochrane Database Syst Rev. 2015;1:
Haematologica. 2008;93:1364-1371. CD010533.
36. Mazzarotto R, Boso C, Vianello F, et al. Primary mediastinal large B- 52. Gallamini A, Hutchings M, Rigacci L, et al. Early interim 2-[18F]fluoro-2-
cell lymphoma: results of intensive chemotherapy regimens deoxy-D-glucose positron emission tomography is prognostically su-
(MACOP-B/VACOP-B) plus involved field radiotherapy on 53 pa- perior to international prognostic score in advanced-stage Hodgkins
tients. A single institution experience. Int J Radiat Oncol Biol Phys. lymphoma: a report from a joint Italian-Danish study. J Clin Oncol. 2007;
2007;68:823-829. 25:3746-3752.

e374 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


ROLE OF PET IN THE MANAGEMENT OF PMLBCL

53. Gallamini A, Barrington SF, Biggi A, et al. The predictive role of interim 63. Sehn LH, Hoskins P, Klasa R, et al. FDG-PET scan guided consolidative
positron emission tomography for Hodgkin lymphoma treatment radiation therapy optimizes outcome In patients with advanced-stage
outcome is confirmed using the interpretation criteria of the Deauville diffuse large B-cell lymphoma (DLBCL) with residual abnormalities on CT
five-point scale. Haematologica. 2014;99:1107-1113. scan following R-CHOP. Blood. 2010;-116:854.
54. Mamot C, Klingbiel D, Hitz F, et al. Final Results of a prospective 64. Cheson BD, Pfistner B, Juweid ME, et al; International Harmonization
evaluation of the predictive value of interim positron emission to- Project on Lymphoma. Revised response criteria for malignant lym-
mography in patients with diffuse large B-cell lymphoma treated with phoma. J Clin Oncol. 2007;25:579-586.
R-CHOP-14 (SAKK 38/07). J Clin Oncol. 2015;33:2523-2529. 65. Zinzani PL, Broccoli A, Casadei B, et al. The role of rituximab and positron
55. Mikhaeel NG, Timothy AR, ODoherty MJ, et al. 18-FDG-PET as a emission tomography in the treatment of primary mediastinal large B-
prognostic indicator in the treatment of aggressive Non-Hodgkins cell lymphoma: experience on 74 patients. Hematol Oncol. 2015; 33:
Lymphoma-comparison with CT. Leuk Lymphoma. 2000;39:543-553. 145-150.
56. Spaepen K, Stroobants S, Dupont P, et al. Prognostic value of positron 66. Martelli M, Ceriani L, Zucca E, et al. [18F]fluorodeoxyglucose positron
emission tomography (PET) with fluorine-18 fluorodeoxyglucose ([18F]FDG) emission tomography predicts survival after chemoimmunotherapy for
after first-line chemotherapy in non-Hodgkins lymphoma: is [18F]FDG-PET a primary mediastinal large B-cell lymphoma: results of the International
valid alternative to conventional diagnostic methods? J Clin Oncol. 2001;19: Extranodal Lymphoma Study Group IELSG-26 Study. J Clin Oncol. 2014;
414-419. 32:1769-1775.
57. Zinzani PL, Fanti S, Battista G, et al. Predictive role of positron emission 67. Meignan M, Gallamini A, Meignan M, et al. Report on the First In-
tomography (PET) in the outcome of lymphoma patients. Br J Cancer. ternational Workshop on Interim-PET-Scan in Lymphoma. Leuk Lym-
2004;91:850-854. phoma. 2009;50:1257-1260.
58. Meignan M. Quantitative FDG-PET: a new biomarker in PMBCL. Blood. 68. Ceriani L, Martelli M, Zinzani PL, et al. Use of the Lugano classification
2015;126:924-926. criteria for PET/CT assessment of primary mediastinal B-cell lymphoma
59. Johnson PWM. IV. Masses in the mediastinum: primary mediastinal after immunochemotherapy and irradiation in the IELSG-26 study.
lymphoma and intermediate types. Hematol Oncol. 2015;33(suppl 1): Hematol Oncol. 2015;33:143 (abstr 081).
29-32. 69. Ceriani L, Martelli M, Zinzani PL, et al. Utility of baseline 18FDG-PET/CT
60. Zinzani PL, Tani M, Trisolini R, et al. Histological verification of positive functional parameters in defining prognosis of primary mediastinal
positron emission tomography findings in the follow-up of patients with (thymic) large B-cell lymphoma. Blood. 2015;126:950-956.
mediastinal lymphoma. Haematologica. 2007;92:771-777. 70. Meignan M, Cottereau AS. Integrative PET: a new concept for outcome
61. Kahn ST, Flowers C, Lechowicz MJ, et al. Value of PET restaging after prediction in lymphoma. Clin Transl Imaging. 2015;3:343-344.
chemotherapy for non-Hodgkins lymphoma: implications for consol- 71. Meignan M, Itti E, Gallamini A, et al. FDG PET/CT imaging as a biomarker
idation radiotherapy. Int J Radiat Oncol Biol Phys. 2006;66:961-965. in lymphoma. Eur J Nucl Med Mol Imaging. 2015;42:623-633.
62. Moskowitz C, Hamlin PA Jr, Maragulia J, et al. Sequential dose-dense 72. Zucca E, Martelli M, Zinzani PL, et al. Prognostic models for primary
RCHOP followed by ICE consolidation (MSKCC protocol 01-142) without mediastinal B-cell lymphoma derived from 18-FDG PET/CT quantitative
radiotherapy for patients with primary mediastinal large B cell lym- parameters in the IELSG-26 study. Haematologica. 2014;99:524-525
phoma. Blood. 2010;116:420. (suppl; abstr S1346).

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e375


LYNCH AND ADVANI

Risk-Adapted Treatment of Advanced Hodgkin Lymphoma


With PET-CT
Ryan C. Lynch, MD, and Ranjana H. Advani, MD

OVERVIEW

Although patients with advanced-stage classic Hodgkin lymphoma have excellent outcomes with contemporary therapy,
the outcomes of patients with refractory disease is suboptimal. Identification of these high-risk patients at diagnosis is
challenging as the differences in outcomes using clinical criteria are less marked using current modern therapy. Data suggest
that an interim PET-CT may be a powerful tool in risk-stratifying patients. Retrospective studies show that a negative interim
PET-CT after two to four cycles of ABVD (doxorubicin, bleomycin, vinblastine, dacarbazine) is predictive of favorable outcome
independent of IPS score. Currently, there are several ongoing trials that aim to determine whether early-response as-
sessment can be used to select patients who might benefit from modifications of subsequent therapy, either by intensifying
or abbreviating regimens and/or omitting radiotherapy with promising early results. Longer follow-up is required to assess
whether this strategy impacts overall survival (OS). Herein, we review the results of recent trials using interim PET-CT-based
adaptive design in the treatment of advanced HL.

P atients with advanced-stage classic Hodgkin lymphoma


(defined in North America as clinical stage IIIIV disease
and early-stage disease with bulk or B symptoms) have
Therefore, there is an ongoing debate regarding how to best
achieve a balance between aggressively curing the disease
with front-line therapy versus a gentler approach that may be
excellent outcomes with contemporary therapy with an OS slightly less efficacious in the short term but with no long-term
of greater than 80%.1,2 OS difference. Attempts to identify patients who are defined
In the National Comprehensive Cancer Network guide- as high risk and thus might potentially benefit from a more
lines, therapy choices for advanced-stage disease include aggressive initial approach is also an evolving concept.
ABVD, Stanford V regimen (a combined modality approach A commonly used index is the International Prognostic
including mechlorethamine, doxorubicin, vincristine, vin- Score (IPS), which has been used in some trials to stratify
blastine, bleomycin, etoposide, and prednisone), and the patients (score $ 3 identifies a high-risk group10). However,
German Hodgkin Study Group (GHSG) regimen escalated as the benefit of escBEACOPP has been observed across all
BEACOPP ( escBEACOPP; bleomycin, etoposide, doxorubicin, IPS risk groups, its utility in patient selection is limited.11
cyclophosphamide, vincristine, procarbazine, and predni- Additionally, the range of differences in outcomes between
sone) followed by radiotherapy given to PET-positive areas groups in the contemporary era has also narrowed, and
greater than 2.5 cm after completion of chemotherapy.3 In other methods for patient selection are required.12,13
North America, ABVD is the most commonly used regimen 18-Fluorodeoxyglucose-(FDG)-PET/CT (or PET-CT) is an
based on a balance of efficacy and toxicity, and escBEACOPP important tool in the management of Hodgkin lymphoma
is used more commonly in Europe. The latter has not been and has a well-established role in staging as well as response
widely accepted in North America largely because of increased assessment at the completion of therapy.14,15 Several studies
toxicity as well as an increased risk of sterility and secondary also suggest that a negative interim PET-CT after two to four
cancers.1,4,5 Several randomized clinical trials have compared cycles of ABVD is predictive of favorable outcomes in-
some version of escBEACOPP to ABVD and have shown an dependent of the IPS score.16-20 The initial studies used
improved progression-free survival (PFS) without any impact variable criteria to define a positive scan with a sensitivity of
on OS.6-8 A recent long-term follow-up from the HD2000 trial 67% to 100% and a specificity of 95% to 100% because of
failed to confirm the PFS benefit with escBEACOPP mainly inter-reader variability.14,21,22 Efforts to standardize response
because of higher mortality rates resulting from second criteria led to the development of the numeric five-point scale
malignancies observed after treatment with escBEACOPP.9 (5PS), also known as the Deauville criteria. In this scale, FDG

From the Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Ranjana H. Advani, MD, Stanford University Medical Center, 875 Blake Wilbur Dr., Suite CC-2338, Stanford, CA 94305-5821; email: radvani@stanford.edu.

2016 by American Society of Clinical Oncology.

e376 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


RISK-ADAPTED TREATMENT OF ADVANCED HL

uptake at sites of disease is compared with uptake in the liver treated with ABVD. Therefore, an adaptive treatment strategy
and the mediastinal blood pool. The specific criteria consist based on interim PET-CT results may help distinguish patients
of a score of the following: (1) no uptake; (2) uptake higher with high-risk disease who could potentially benefit from a
than or equal to mediastinal blood pool; (3) uptake higher change in therapy to an escalated regimen despite the in-
than mediastinal blood pool, but uptake is lower than or equal creased toxicity. This was evaluated in a retrospective analysis
to liver; (4) uptake moderately higher than liver; (5) uptake of prospectively collected data in a study in which all patients
markedly higher than liver and/or new lesions; (X) new areas with PET-2positive disease after two cycles of ABVD received
of uptake unlikely to be related to lymphoma.23 A score of 13 is four escBEACOPP plus four standard BEACOPP cycles, whereas
considered to represent a complete metabolic response in patients with PET-2negative disease continued on ABVD (total
advanced Hodgkin lymphoma. The prognostic utility value of this of six cycles) followed by consolidative radiotherapy.27 At a
score was confirmed in an international study that showed a median follow-up of 34 months and a central radiology review
3-year failure-free survival (FFS) of 28% and 95% for patients of PET-2 data using the Deauville criteria, the FFS in PET-
with early PET-positive versus PET-negative disease respectively 2negative versus PET-2positive group was 95% versus
(p , .0001).24 Additionally, the interobserver agreement was 62%, respectively (p , .001). The outcomes of patients with
also high between six independent reviewers. The reproducibility PET-2positive disease (approximately 20% of patients) was
has resulted in the Deauville criteria being endorsed as the better than historical controls using ABVD.20
current recommended method for response assessment.25,26 The efficacy of intensifying therapy to escBEACOPP for
This has resulted in numerous trials in Hodgkin lymphoma patients with PET-2positive disease has also been tested in
based on interim PET-CTadapted strategies. These studies aim several prospectively designed trials in advanced Hodgkin
to determine whether early-response assessment can be used lymphoma described below and summarized in Table 1.
to select patients who might benefit from modifications of Although most of the interim PET-positive arms do not
subsequent therapy, either by intensifying or abbreviating have a control group without escalated therapy, the sta-
regimens and/or omitting radiotherapy. Herein, we review the tistical design is essentially based on the historical PFS data
results of recent trials using interim PET-CTbased adaptive described above.
design in the treatment of advanced Hodgkin lymphoma. The U.S. intergroup phase II trial S0816 led by the
Southwest Oncology Group (SWOG) reported preliminary
results in 357 patients with a median follow-up 28 months in
STUDIES EVALUATING THERAPY ESCALATION IN
patients with advanced Hodgkin lymphoma. Patients re-
PATIENTS WITH A POSITIVE INTERIM PET-CT
ceived two cycles of ABVD followed by a PET-CT with central
SCAN
review.28 Those with a PET-2negative scan (82% of pa-
Results of an early interim PET-CT is now a widely accepted
tients) defined as Deauville 13 continued with four more
prognostic tool for patients with advanced Hodgkin lymphoma
cycles of ABVD, whereas patients with PET-2positive dis-
ease (Deauville 45) had a change in therapy to six cycles
of escBEACOPP. An interim analysis presented in 2013
KEY POINTS reported a 1-year PFS for the PET-2positive group of 72%
(95% CI, 55%84%) and 85% in the PET-2negative group
Retrospective studies in advanced Hodgkin lymphoma (95% CI, 79%90%).
suggest that a negative interim PET-CT (variably defined) The U.K. RATHL trial included 1,214 patients and tested
after two to four cycles of ABVD is predictive of a similar concept as the U.S. intergroup study. Patients with
a favorable outcome.
advanced-stage Hodgkin lymphoma (stages IIBIV, or IIA
Several prospective trials aim to determine whether
early response assessment after two cycles of ABVD
with bulk or three or more involved sites) received two
using the five-point scale can be used to select patients cycles of ABVD followed by a PET-CT. Patients with PET-
who might benefit from modifications of subsequent 2negative disease (defined as Deauville 13) based on a
therapy. central review were randomly assigned to either ABVD or
Results of several trials suggest that an interim AVD for four additional cycles.29,30 Patients with PET-
PETadapted approach of escalating therapy to a more 2positive disease (Deauville 45) received intensified
aggressive regimen is feasible and possibly associated therapy with four cycles of BEACOPP-14 or three cycles of
with better progression-free survival in patients with escBEACOPP followed by another PET-CT. Patients whose
PET-2positive disease after ABVD compared with disease remained PET-3positive (performed after four
historical controls. additional cycles of BEACOPP-14 or three of escBEACOPP)
A major consideration for the escalation strategies is the
were taken off study and given salvage treatment. Patients
lack of a control arm in which ABVD therapy is continued
regardless of interim PET-CT results.
with PET-3negative disease received two additional cycles
Interim results of the RATHL trial suggest that for of BEACOPP-14 or one more cycle of escBEACOPP. Patients
patients with a negative PET-CT after two cycles of who had a positive PET-2 scan (16% of all patients) had a
ABVD, bleomycin can be safely omitted from 3-year PFS and OS of 68% and 86%, respectively, with no
subsequent cycles. difference in the nonrandomized comparison between
escBEACOPP and BEACOPP-14.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e377


LYNCH AND ADVANI

TABLE 1. Comparison of PET-Adapted Prospective Clinical Trials in Advanced Hodgkin Lymphoma


Results
Median
Study Trial Design PET-2 Results Clinical Stage Follow-up PFS/EFS/FFS OS
Trials With Treatment Escalated If PET-2 Positive
RATHL Two cycles ABVD interim PET-CT 16.1% positive IIBIV, IIA with 32 months 3-year PFS 3-year OS
(Johnson bulk or $ 3
et al,29 2015, involved
1,214 sites
patients)
PET2+: six cycles BEACOPP-14 or four PET2+: 68% PET2+: 86%
cycles escBEACOPP (taken off study
if PET+ after four cycles BEACOPP-14
or three cycles escBEACOPP)
AHL 2011 LYSA Standard arm 12.4% positive IIBIV 16.3 months 2-year PFS Not reported
Study
(Casanovas
et al,41 2015,
823 patients)
Six cycles escBEACOPP (regardless of Standard arm: 91.6%
result of interim PET-CT)
Experimental arm Experimental arm:
88.3%
Two cycles escBEACOPP interim Overall study cohort
PET-CT PET2+: 72.9%
PET2+: four cycles escBEACOPP Overall study cohort
PET22: 92.8%
(p , .0001)*
PET22: four cycles ABVD Standard PET22:
75.1%
Experimental
PET22: 70.8%
Israeli H2 (Dann IPS $ 3 14.2% positive IB, IIB, IIIIV 36 months 3-year PFS (all ad- Not reported
et al,35 2014, vanced HL regard-
183 patients) less of IPS)
Two cycles escBEACOPP, interim PET2+: 75%
PET-CT
PET2+: four cycles escBEACOPP + RT to
bulky mediastinal disease at EOT
IPS , 3
Two cycles ABVD interim PET-CT
PET2+: four cycles escBEACOPP + RT to
bulky mediastinal disease at EOT
Israeli Group Two cycles escBEACOPP interim 24.6% positive IIBIV (IPS $ 3) 5.6 years 5-year PFS 5-year OS
(Kedmi PET-CT
et al,39 2015,
69 patients;
24 patients
were treated
on the same
protocol
off-study)
PET2+: four cycles escBEACOPP PET2+: 60% PET2+: 79%
PET22: four cycles ABVD PET22: 80% PET22: 98%
HD18 (Borch- Two cycles escBEACOPP interim 40.0% positive IIB with bulky 35 months 3-year PFS 3-year OS
mann et al,34 PET-CT mediastinal
2014, 1,100 mass or EN
patients) involvement,
IIIIV
PET2+: randomized to four cycles PET2+: escBEACOPP, PET2+: escBEACOPP
escBEACOPP vs. one cycle escBEA- 91.4%; 96.5%,
COPP, then four cycles R-BEACOPP
R-BEACOPP, 93% R-BEACOPP 94.4%

Continued

e378 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


RISK-ADAPTED TREATMENT OF ADVANCED HL

TABLE 1. Comparison of PET-Adapted Prospective Clinical Trials in Advanced Hodgkin Lymphoma


(Contd)
Results
Median
Study Trial Design PET-2 Results Clinical Stage Follow-up PFS/EFS/FFS OS
U.S. Intergroup Two cycles ABVD, interim PET-CT 77.7% negative IIIIV 28 months 1-year PFS Not reported
S0816 (Press
et al,28 2013,
357 patients)
PET2+: six cycles escBEACOPP PET2+: 72%
PET22: four cycles ABVD PET22: 85%
Trials With Treatment De-escalated If PET-2 Negative

RATHL Two cycles ABVD interim PET-CT 83.9% negative IIBIV, IIA with 32 months 3-year PFS 3-year OS
(Johnson bulk or three
et al,29 2015; or more in-
1,214 volved sites
patients)
PET22: randomized to four cycles PET22: ABVD, PET22: ABVD 97%,
ABVD vs. four cycles AVD 85.5%;
AVD, 84.5% AVD 97.5%
HD 0607 Two cycles ABVD interim PET-CT 80.5% negative IIBIV 34.3 months No interim data on No interim data on
(Gallamini randomized arms randomized arms
et al,31 2015,
773 patients)
PET2+: randomized to escBEACOPP + Overall study cohort Overall study cohort
BEACOPP (four + four) vs. four + four
and rituximab
PET22: four cycles ABVD. If CR, then 4-year FFS 4-year OS
randomized to RT vs. observation
PET2+: 62% PET2+: 86%
PET22: 85% PET22: 95%
HD 0801 Two cycles ABVD interim PET-CT 79.9% negative IIBIV Not reported PET2+: 78.6% Not reported
(Zinzani in abstract (81/103 patients)
32
et al, 2015, received
519 patients) HDC-ASCT,
PET2+: ifosfamide-containing salvage 2-year PFS
regimen HDC-ASCT
PET22: four cycles ABVD PET2+: 74%
PET22: 81%
Israeli H2 (Dann IPS $ 3 85.8% negative IB, IIB, IIIIV 36 months 3-year PFS (All Not reported
et al,35 2014, advanced HL
183 patients) regardless of IPS)
Two cycles escBEACOPP, interim PET22: 86%
PET-CT (p = .012)
PET22: four cycles ABVD, omit RT to
bulky sites
IPS , 3
Two cycles ABVD interim PET-CT
PET22: four cycles ABVD, omitting RT
to bulky sites
NCT013048490 Two cycles ABVD interim PET-CT 83.7% negative IIBIV 24.7 months 2-year EFS Not reported
(Ganesan
et al,36 2015,
50 patients)
PET2+: four cycles escBEACOPP PET2+: 50%
PET22: four cycles ABVD PET22: 82%
(p = .013)

Continued

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e379


LYNCH AND ADVANI

TABLE 1. Comparison of PET-Adapted Prospective Clinical Trials in Advanced Hodgkin Lymphoma


(Contd)
Results
Median
Study Trial Design PET-2 Results Clinical Stage Follow-up PFS/EFS/FFS OS
HD18 Two cycles escBEACOPP interim 60.0% negative IIB with bulky 35 months 3-year PFS 3-year OS
(Borchmann PET-CT mediastinal
et al,34 2014, mass or EN
1,100 involvement,
patients) IIIIV
PET22: randomized to Two cycles PET22: no data PET22: no data
escBEACOPP vs. four cycles available available
escBEACOPP
Abbreviations: ABVD, doxorubicin, bleomycin, vinblastine, and dacarbazine; AVD, doxorubicin, vinblastine, and dacarbazine; BEACOPP, bleomycin, etoposide, doxorubicin,
cyclophosphamide, vincristine, procarbazine, and prednisone; CR, complete remission; EFS, event-free survival; EN, extranodal; EOT, end of therapy; escBEACOPP, escalated-dose
BEACOPP; FFS, failure-free survival; HDC-ASCT, high-dose chemotherapy-autologous stem cell transplantation; HL, Hodgkin lymphoma; IPS, International Prognostic Score; LYSA,
Lymphoma Study Association; NCT, National Clinical Trial number; OS, overall survival; PET2+, interim PET-CT positive after 2 cycles of therapy; PET22, interim PET-CT negative after
two cycles of therapy; PFS, progression-free survival; R-BEACOPP, rituxumab plus BEACOPP; RT, radiotherapy.
* If comparison is statistically significant, p value is given.

A phase II multicenter Italian HD 0607 trial evaluated of escBEACOPP (four or six total). No data from the PET-
773 patients with advanced Hodgkin lymphoma who received 2negative arm have been presented to date. In the PET-
two cycles of ABVD followed by an interim PET-CT. Patients 2positive group, patients were randomly assigned to four
with PET-2negative disease (Deauville 13) continued with additional cycles of escBEACOPP versus one cycle of esc-
four additional cycles of ABVD.31 Those in complete remission BEACOPP followed by four cycles of rituximab-escBEACOPP
(CR) after six cycles of ABVD were subsequently randomly (R-escBEACOPP). Patients with PET-CTpositive disease
assigned to radiotherapy to sites of initial bulky disease versus greater than 2.5 cm at the end of treatment received ra-
observation. Patients with PET-2positive disease (Deauville diotherapy. Interim results of this trial reported no difference
45) were randomly assigned to either alternating cycles of in outcomes between the two arms. At a median follow-up of
standard BEACOPP and escBEACOPP (four plus four) or the 35 months, the 3-year PFS for escBEACOPP was 91.4% (95%
four-plus-four regimen plus rituximab. Although there are no CI, 87.0%95.7%) versus 93% for R-escBEACOPP (95% CI,
data currently available comparing the randomized arms, at a 89.4%96.6%). Moreover, 3-year OS was not significantly
median follow-up 34.3 months, the overall 4-year FFS and OS different between escBEACOPP and R-escBEACOPP (96.5% vs.
were 62% and 86% for patients with PET-2positive disease 94.4%; p = .31).
and 85% and 95% for patients with PET-2negative disease The Israeli H2 study included 183 patients and stratified initial
respectively. treatment of patients with advanced-stage Hodgkin lymphoma
The Italian HD 0801 trial tested an alternative aggressive or early-stage Hodgkin lymphoma with B symptoms based
strategy in 519 patients. Treatment of patients with PET- on the IPS score. 35 Patients with IPS scores lower than 3
2positive disease after two cycles of ABVD was changed (106 patients) were treated with two cycles of ABVD fol-
to IGEV (ifosfamide, gemcitabine, vinorelbine) in preparation lowed by interim PET-CT. Patients with PET-2positive dis-
for transplant.32 Patients in CR by PET-CT after IGEV received ease had therapy escalated to escBEACOPP. In contrast,
an autologous transplant, whereas those not in CR received a patients with IPS scores of 3 or lower started out with two
tandem autologous-allogeneic transplant. Of the patients cycles of escBEACOPP, with patients with PET-2positive
with PET-2positive disease 78.6% (81 of 103) received a disease continuing with escBEACOPP and patients with
salvage regimen containing at least one autologous trans- PET-2negative disease deescalating therapy to four cycles
plant; 14.6% (15 of 103) of patients with PET-2positive of ABVD (discussed in the next section). Among the 162
disease continued on ABVD per investigator decision due patients with advanced-stage disease, 26 had a positive PET-
to a minimally positive PET-CT. There was no significant 2 scan. Interim results report a 3-year PFS for PET-2 negative
difference in 2-year PFS in the PET-2positive intention-to- and PET-2 positive disease of 86% and 75%, respectively
treat group (76%) and the patients with PET-2positive (p = .012), with no difference in PFS based on IPS score.
disease who subsequently underwent salvage chemother- An Indian phase II study evaluated two cycles of ABVD
apy and transplant (74%) with a 2-year PFS for patients with followed by interim PET-CT in 50 patients with stage IIB-IV
PET-2negative disease of 81%, which was similar to that Hodgkin lymphoma.36 Patients with PET-2positive dis-
observed in patients with PET-2positive disease. ease were escalated to escBEACOPP and patients with
The GHSG HD18 study evaluated 1,100 patients and PET-2negative disease continued therapy with four addi-
also used a PET-adaptive strategy intensifying therapy tional cycles of ABVD. At a median follow-up 24.7 months,
based on interim PET results after two initial cycles the 2-year EFS for patients with PET-2negative disease
of escBEACOPP.33,34 Patients with PET-2negative disease and patients with PET-2positive disease was 82% and 50%,
were randomly assigned to two versus four additional cycles respectively (p = .013).

e380 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


RISK-ADAPTED TREATMENT OF ADVANCED HL

In addition to the above trials, there is an ongoing study of interim PET after two cycles versus an experimental arm that
examining very early PET-adapted escalation with interim evaluated a PET-adapted de-escalation strategy.41 Patients with
PET-CT after one cycle of ABVD.37 No interim results have PET-2positive disease continued with four additional cycles of
been presented so far. escBEACOPP, whereas for patients with PET-2negative dis-
Cumulatively, results of all of the above trials suggest that ease, therapy was de-escalated to four cycles of ABVD. At a
an interim PET-adapted approach of escalating therapy to a median follow-up of 16.3 months, the 2-year PFS was similar in
more aggressive regimen is feasible and possibly associated the standard and experimental arms (91.6% vs. 88.3%; p = .79).
with better outcomes in patients with PET-2positive dis- Overall in both arms, patients with PET-2positive disease
ease after ABVD compared with historical controls, albeit (97 patients) had a lower 2-year PFS than patients with
with more toxicity. PET-2negative disease (72.9% vs. 92.8%; p , .0001). Standard-
A major issue with most of these trials is the lack of a control arm patients with PET-2negative disease had a similar 2-year
arm in which ABVD therapy is continued regardless of interim PFS as the de-escalated experimental-arm patients with
PET-CT results. Additionally, as discussed previously, the Deau- PET-2negative disease (75.1% vs. 70.8%; not significant).
ville scoring system has led to less inter-reader variability and The U.K. RATHL trial randomly assigned patients with
differences in outcomes between patients with PET-CTnegative Hodgkin lymphoma with a negative PET-CT after two cycles
or PET-CTpositive disease are not as disparate as in the original of ABVD to continue with four additional cycles of ABVD
report by Gallamini et al.20,21,38 versus AVD (omitting bleomycin).29 Interim results of the
Therefore, long-term follow-up is critical to determine if PET-2negative group (944 patients) with a median follow-up
treatment modification translates into an OS advantage or of 32 months showed similar 3-year PFS for ABVD (85.45%;
not. Until that is clearer, escalating therapy based on in- 95% CI, 83.4289.70) versus AVD (84.48%; 95% CI,
terim PET-CT results should ideally be done in the con- 82.4788.97) with no difference in 3-year OS (ABVD, 97.0%;
text of a clinical trial with strict definitions of PET-CT 95% CI, 94.598.4; AVD, 97.5%; 95% CI, 95.198.7). In ad-
interpretation. dition, there were more grade 3/4 clinical pulmonary events
in the ABVD arm (12 total doses of bleomycin; 13 of 468
patients, 3%) versus the AVD arm (four total doses of
STUDIES EVALUATING DE-ESCALATION OF bleomycin; 6 of 440 patients, 1%).42 There was also a greater
CHEMOTHERAPY IN PATIENTS WITH A NEGATIVE decrease in lung diffusing capacity in patients who received
INTERIM PET-CT SCAN additional doses of bleomycin (8.7%; 95% CI, 6.1011.36).
An alternate strategy for patients with advanced Hodgkin As described earlier, the GHSG HD18 trial also randomly
lymphoma is to de-escalate chemotherapy in patients with assigned patients with PET-2negative disease to two versus
a negative interim PET-CT to mitigate some short- and long- four additional cycles of escBEACOPP (four or six total).33,34
term toxicities. Although there have been no data presented yet from the
The Israeli group was among the first to report on a de- PET-2negative arm, the 3-year PFS for patients with PET-
escalation strategy. Patients with stage IIBIV disease with 2positive disease in this study exceeded 90%, suggesting
IPS of 3 or higher started therapy with two cycles of esc- that the PET-2negative cohort is at lower risk with room for
BEACOPP.39,40 For patients who had PET-2negative disease, de-escalation.
chemotherapy was de-escalated to four additional cycles of Cumulatively, the results of studies on continuing with
ABVD, whereas patients with PET-2positive disease con- standard therapy (i.e., ABVD) after a negative interim PET-CT
tinued on escBEACOPP. Radiotherapy was administered to scan report excellent outcomes and also support the
initial sites of bulk ($ 10 cm) or mediastinal bulk at physician omission of bleomycin.
discretion. The results of their prospective phase II study
were combined with additional patients who were treated
on the same protocol off-study (24 patients) for a total of 69 END OF THERAPY PET: WHEN SHOULD
patients. With this de-escalation strategy, an interim PET-CT RADIOTHERAPY BE GIVEN
predicted 5-year OS (PET-2 positive, 79%, vs. PET-2 negative, Another ongoing debate is regarding the role of radiotherapy
98%; p = .015). The same group is currently evaluating in the H2 in advanced Hodgkin lymphoma specifically because of concern
trial a similar strategy as used in the earlier study, in which regarding late effects.43-46 Although the risk of late effects with
patients with PET-2negative disease after two cycles of esc- contemporary radiotherapy are lower, attempts to eliminate
BEACOPP had therapy de-escalated to four additional cycles of radiotherapy are ongoing.1,47-49 Although radiotherapy is well
ABVD without radiotherapy.35 Interim results for 183 patients studied in the management of early-stage disease, its role in
at a median follow-up of 36 months show a 3-year PFS for patients with advanced Hodgkin lymphoma is less precise.
patients with PET-2negative disease and those with PET- Historic data suggest that, with consolidative radiotherapy,
2positive disease of 86% and 75%, respectively (p = .012), with patients who achieve a partial remission (PR) using standard
no difference in PFS based on IPS score. definitions based on CT scans to assess response have out-
The AHL 2011 study randomly assigned 823 patients with comes similar to those in CR.50-52 Therefore, radiotherpy has
stage IIBIV Hodgkin lymphoma to a standard arm that commonly been used to consolidate sites of initial bulky dis-
received six cycles total of escBEACOPP regardless of results ease especially in the mediastinum with improved outcomes

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e381


LYNCH AND ADVANI

in some series.52 Because current response criteria are now unfavorable patients with PET-2negative disease with
based on PET-CT, the role of radiotherapy needs to be re- and without radiotherapy was 97.3% and 94.9%, respectively
evaluated. The only trial that has prospectively studied this (p = .026), with no difference in OS.
is the GHSG HD15 trial, in which patients with a residual mass
larger than 2.5 cm mass and PET positivity after six cycles of CONCLUSION
escBEACOPP were treated with 30 Gy.1 The addition of PET- Interim PET-CT has emerged as a surrogate to better select
CT to response assessment decreased the total number of patients for therapy intensification or de-escalation. The
patients receiving radiotherapy when compared with the prior goal of these trials is to limit intensive chemotherapy/
GHSG HD9 trial (11% vs. 66%).53 The 4-year PFS curves for PET- radiotherapy to a subset of patients who might benefit
negative PR and CR/CRu post-treatment were similar at 92.1%. from an aggressive approach and spare the vast majority of
Patients with residual PET positivity had excellent outcomes patients the associated toxicities. Early PFS and FFS data
with a PFS of 86.2% after radiotherapy. suggest that there may be fewer earlier treatment failures in
When ABVD is used as the chemotherapy backbone, the patients with PET-2positive disease with escalation of
data are less clear as there are no trials that have evaluated therapy compared with historical controls, although longer
radiotherapy versus no radiotherapy. The GITL/IIL study follow-up is required to assess whether these strategies
used radiotherapy at the completion of treatment25.2 Gy actually translate to an OS advantage. Results with escala-
in all patients to sites of initial bulk and 30.6 Gy to areas of tion or de-escalation strategies appear largely similar, and
residual disease.6 However, the use of PET was not man- the two have not been compared. The U.K. RATHL trial
dated by the protocol. No patients in the RATHL or U.S. supports the omission of bleomycin in patients with a
Intergroup trials received radiotherapy.28,29 negative PET-CT after two cycles of ABVD.
The ongoing HD 0607 trial is randomly assigning patients Although patients with interim or end-of-therapy PET-
with PET-2negative disease who have received a total of six 2positive disease and PET-2negative disease have dif-
cycles of ABVD to radiotherapy to initially bulky sites versus ferent outcomes with standard therapy, these are likely a
observation, and results are awaited.31 Since July 2005, the reflection of variable biology at diagnosis. The limitations of
BC Cancer Agency has adopted a treatment strategy of six clinical prognostic models have led to the development of
cycles of ABVD for patients with advanced-stage Hodgkin molecular and immune-based markers.56 Gene expression
lymphoma (including stage I with bulk and stage II with B profiling identified a signature of tumor-associated CD68+
symptoms and/or bulk) followed by observation with ra- macrophages, whose frequency correlated with treatment
diotherapy used only for patients with PET-positive disease failure and OS and outperformed the IPS as a prognostic
at the end of therapy.54 Recent results suggest that post- biomarker.57 Increase in CD68+ macrophages at diagnosis
treatment patients with PET-negative disease had superior has also been shown to correlate with positive interim PET-
5-year freedom-from-treatment failure (FFTF) compared CT using the Deauville scale.58,59 Correlative data from the
with those with PET-positive disease (89% vs. 56%; p , .00001). E2496 trial found that tumor-associated macrophages on
In addition, there was no difference in 5-year FFTF in the initial pathology was associated with a worse FFS and OS.56
PET-negative group between bulky (112 patients) and non- An analysis of a cohort of GHSG patients found that ele-
bulky (152 patients; 89% vs. 88.5%; p = .50) with similar OS vated serum levels of thymus and activation-regulated
(96% vs. 94%, p = .51). Although the BC Cancer Agency data chemokine/CCL17 at diagnosis were associated with an
support the lack of a benefit of radiotherapy in patients increased risk of chemotherapy failure with multivariate
who achieve CR by PET-CT at completion of chemotherapy, analysis (odds ratio, 3.052; 95% CI, 1.6055.804, p , .007)
the outcomes of patients with PET-positive disease after when treated based on standard of care at the time of
ABVD is suboptimal with use of radiotherapy. Every attempt diagnosis.60 A 23-gene panel has been developed that may
should be made to rebiopsy such patients and consider salvage identify at diagnosis patients who are at increased risk of
therapy followed by autologous stem cell transplantation if treatment failure and death.61 All of these biomarkers hold
feasible. promise but require prospective validation. Additional
For patients with stage III unfavorable disease (often treated studies are also required to assess how they compare with
as advanced disease in the United States if bulky disease or B prognosis based on interim PET results.
symptoms), the EORTC/GELA/IIT H10 trial randomly assigned Although the trials discussed in the risk-adapted strategies
patients to a standard arm (four total cycles of ABVD followed by described have largely used ABVD and escBEACOPP, there
involved-node radiotherapy [INRT]), or an experimental arm are other novel combinations that are currently being
based on interim PET results.55 After two cycles of ABVD, patients evaluated in phase III randomized trials due to excellent
with PET-2positive disease received two cycles of escBEACOPP initial results. A phase I pilot study showed an excellent CR
followed by INRT. Patients with PET-2negative disease received rate with the anti-CD30 antibody-drug conjugate, brentux-
four additional cycles of ABVD (six total) without radiotherapy. imab vedotin plus ABVD (21 of 22 patients, 95%) or for
Interim analysis found an increase in events in the experimental brentuximab plus AVD (24 of 25 patients, 95%) when used
arm (16 vs. seven), which met prespecified significance for up front for stage IIAX, IIBIVB Hodgkin lymphoma.62 Be-
futility, and therefore the experimental arm omitting radiotherapy cause of increased pulmonary toxicity seen when bleomycin
was closed to further accrual. The 1-year PFS for early-stage was used in conjunction with brentuximab vedotin,

e382 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


RISK-ADAPTED TREATMENT OF ADVANCED HL

the ongoing phase III multicenter study is evaluating setting.65-67 Whether these novel combinations will result in
brentuximab plus AVD versus ABVD in Hodgkin lymphoma.63 better outcomes than standard therapies or interim PET-
A variant of BEACOPP called BrECADD has been tested adapted strategies is unknown. Additionally, the utility of
in an early-phase clinical trial with excellent results.64 the Deauville criteria in the setting of these agents is largely
BrECADD eliminates bleomycin, vincristine, procarbazine, unknown. It is likely that some modification may be required
and prednisone and replaces them with brentuximab to account for tumor flare associated with checkpoint
vedotin, dacarbazine, and dexamethasone. BrECADD has an inhibitors to eliminate over interpretation due to a pseudo-
18-month PFS of 89% (95% CI, 77%100%) with 18-month progression.68
OS of 100% in an early-phase trial. BrECADD is cur- Careful attention will need to be made toward late
rently being compared with escBEACOPP in a randomized follow-up with these regimens to assess for OS benefit,
frontline trial for advanced Hodgkin lymphoma (HD21 trial) as well as secondary malignancies. The future will likely
by the GHSG. Emerging data suggest that immune incorporate biologic and clinical factors along with tumor
checkpoint inhibitors are very effective in the relapsed/ burden defined by PET-CT to better tailor treatment to
refractory setting, and trials are underway in the frontline each individual patient.

References
1. Engert A, Haverkamp H, Kobe C, et al; German Hodgkin Study Group; 11. Engert A, Diehl V, Franklin J, et al. Escalated-dose BEACOPP in the
Swiss Group for Clinical Cancer Research; Arbeitsgemeinschaft Medi- treatment of patients with advanced-stage Hodgkins lymphoma: 10
kamentose Tumortherapie. Reduced-intensity chemotherapy and years of follow-up of the GHSG HD9 study. J Clin Oncol. 2009;27:
PET-guided radiotherapy in patients with advanced stage Hodgkins 4548-4554.
lymphoma (HD15 trial): a randomised, open-label, phase 3 non-inferiority 12. Moccia AA, Donaldson J, Chhanabhai M, et al. International Prognostic
trial. Lancet. 2012;379:1791-1799. Score in advanced-stage Hodgkins lymphoma: altered utility in the
2. Gordon LI, Hong F, Fisher RI, et al. Randomized phase III trial of ABVD modern era. J Clin Oncol. 2012;30:3383-3388.
versus Stanford V with or without radiation therapy in locally extensive 13. Diefenbach CS, Li H, Hong F, et al. Evaluation of the International Prognostic
and advanced-stage Hodgkin lymphoma: an intergroup study co- Score (IPS-7) and a Simpler Prognostic Score (IPS-3) for advanced Hodgkin
ordinated by the Eastern Cooperative Oncology Group (E2496). J Clin lymphoma in the modern era. Br J Haematol. 2015;171:530-538.
Oncol. 2013;31:684-691. 14. Juweid ME, Stroobants S, Hoekstra OS, et al; Imaging Subcommittee of
3. Hoppe RT, Advani RH, Ai WZ, et al. Hodgkin lymphoma, version 2.2015. International Harmonization Project in Lymphoma. Use of positron
J Natl Compr Canc Netw. 2015;13:554-586. emission tomography for response assessment of lymphoma: con-
4. Wongso D, Fuchs M, Plutschow A, et al. Treatment-related mortality in sensus of the Imaging Subcommittee of International Harmonization
patients with advanced-stage hodgkin lymphoma: an analysis of the Project in Lymphoma. J Clin Oncol. 2007;25:571-578.
german hodgkin study group. J Clin Oncol. 2013;31:2819-2824. 15. El-Galaly TC, dAmore F, Mylam KJ, et al. Routine bone marrow biopsy
5. Behringer K, Mueller H, Goergen H, et al. Gonadal function and fertility in has little or no therapeutic consequence for positron emission
survivors after Hodgkin lymphoma treatment within the German Hodgkin tomography/computed tomography-staged treatment-naive patients
Study Group HD13 to HD15 trials. J Clin Oncol. 2013;31:231-239. with Hodgkin lymphoma. J Clin Oncol. 2012;30:4508-4514.
6. Viviani S, Zinzani PL, Rambaldi A, et al; Michelangelo Foundation; 16. Cerci JJ, Pracchia LF, Linardi CC, et al. 18F-FDG PET after 2 cycles of ABVD
Gruppo Italiano di Terapie Innovative nei Linfomi; Intergruppo Italiano predicts event-free survival in early and advanced Hodgkin lymphoma.
Linfomi. ABVD versus BEACOPP for Hodgkins lymphoma when high- J Nucl Med. 2010;51:1337-1343.
17. Hutchings M, Loft A, Hansen M, et al. FDG-PET after two cycles of
dose salvage is planned. N Engl J Med. 2011;365:203-212.
chemotherapy predicts treatment failure and progression-free survival
7. Federico M, Luminari S, Iannitto E, et al; HD2000 Gruppo Italiano per lo
in Hodgkin lymphoma. Blood. 2006;107:52-59.
Studio dei Linfomi Trial. ABVD compared with BEACOPP compared with
18. Hutchings M, Mikhaeel NG, Fields PA, et al. Prognostic value of interim
CEC for the initial treatment of patients with advanced Hodgkins
FDG-PET after two or three cycles of chemotherapy in Hodgkin lym-
lymphoma: results from the HD2000 Gruppo Italiano per lo Studio dei
phoma. Ann Oncol. 2005;16:1160-1168.
Linfomi Trial. J Clin Oncol. 2009;27:805-811.
19. Hutchings M, Kostakoglu L, Zaucha JM, et al. In vivo treatment sensi-
8. Carde PP, Karrasch M, Fortpied C, et al. ABVD (8 cycles) versus
tivity testing with positron emission tomography/computed tomog-
BEACOPP (4 escalated cycles =. 4 baseline) in stage III-IV high-risk
raphy after one cycle of chemotherapy for Hodgkin lymphoma. J Clin
Hodgkin lymphoma (HL): First results of EORTC 20012 Intergroup Oncol. 2014;32:2705-2711.
randomized phase III clinical trial. J Clin Oncol. 2012;30 (suppl; abstr 20. Gallamini A, Hutchings M, Rigacci L, et al. Early interim 2-[18F]fluoro-2-
8002). deoxy-D-glucose positron emission tomography is prognostically superior
9. Merli F, Luminari S, Gobbi PG, et al. Long-term results of the HD2000 to international prognostic score in advanced-stage Hodgkins lymphoma:
trial comparing ABVD versus BEACOPP versus COPP-EBV-CAD in un- a report from a joint Italian-Danish study. J Clin Oncol. 2007;25:3746-3752.
treated patients with advanced Hodgkin lymphoma: a study by Fon- 21. Le Roux PY, Gastinne T, Le Gouill S, et al. Prognostic value of interim FDG
dazione Italiana Linfomi. J Clin Oncol. Epub 2015 Dec 28. PET/CT in Hodgkins lymphoma patients treated with interim response-
10. Hasenclever D, Diehl V. A prognostic score for advanced Hodgkins adapted strategy: comparison of International Harmonization Project
disease. International Prognostic Factors Project on Advanced Hodg- (IHP), Gallamini and London criteria. Eur J Nucl Med Mol Imaging. 2011;
kins Disease. N Engl J Med. 1998;339:1506-1514. 38:1064-1071.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e383


LYNCH AND ADVANI

22. Terasawa T, Lau J, Bardet S, et al. Fluorine-18-fluorodeoxyglucose study. Paper presented at: 56th American Society of Hematology Annual
positron emission tomography for interim response assessment of Meeting and Exposition; December 2014; San Francisco, CA.
advanced-stage Hodgkins lymphoma and diffuse large B-cell lym- 36. Ganesan P, Rajendranath R, Kannan K, et al. Phase II study of interim
phoma: a systematic review. J Clin Oncol. 2009;27:1906-1914. PET-CT-guided response-adapted therapy in advanced Hodgkins
23. Meignan M, Gallamini A, Meignan M, et al. Report on the First In- lymphoma. Ann Oncol. 2015;26:1170-1174.
ternational Workshop on Interim-PET-Scan in Lymphoma. Leuk Lym- 37. Waxman JH, Terry YA, Wrigley PF, et al. Gonadal function in Hodgkins
phoma. 2009;50:1257-1260. disease: long-term follow-up of chemotherapy. Br Med J (Clin Res Ed).
24. Biggi A, Gallamini A, Chauvie S, et al. International validation study for 1982;285:1612-1613.
interim PET in ABVD-treated, advanced-stage Hodgkin lymphoma: in- 38. Gallamini A, Barrington SF, Biggi A, et al. The predictive role of interim
terpretation criteria and concordance rate among reviewers. J Nucl positron emission tomography for Hodgkin lymphoma treatment
Med. 2013;54:683-690. outcome is confirmed using the interpretation criteria of the Deauville
25. Cheson BD, Fisher RI, Barrington SF, et al; Alliance, Australasian Leu- five-point scale. Haematologica. 2014;99:1107-1113.
kaemia and Lymphoma Group; Eastern Cooperative Oncology Group; 39. Kedmi M, Apel A, Davidson T, et al. High-risk, advanced-stage Hodgkin
European Mantle Cell Lymphoma Consortium; Italian Lymphoma lymphoma: the impact of combined escalated BEACOPP and ABVD
Foundation; European Organisation for Research; Treatment of Cancer/ treatment in patients who rapidly achieve metabolic complete remission on
Dutch Hemato-Oncology Group; Grupo Espan ~ol de Medula Osea;
interim FDG-PET/CT scan. Acta Haematol. 2016;135:156-161.
German High-Grade Lymphoma Study Group; German Hodgkins Study 40. Dann EJ, Blumenfeld Z, Bar-Shalom R, et al. A 10-year experience with
Group; Japanese Lymphorra Study Group; Lymphoma Study Associa- treatment of high and standard risk Hodgkin disease: six cycles of
tion; NCIC Clinical Trials Group; Nordic Lymphoma Study Group; tailored BEACOPP, with interim scintigraphy, are effective and female
Southwest Oncology Group; United Kingdom National Cancer Research fertility is preserved. Am J Hematol. 2012;87:32-36.
Institute. Recommendations for initial evaluation, staging, and re- 41. Casasnovas O, Brice P, Bouabdallah R, et al. Randomized phase III study
sponse assessment of Hodgkin and non-Hodgkin lymphoma: the Lugano comparing an early PET driven treatment de-escalation to a not PET-
classification. J Clin Oncol. 2014;32:3059-3068. monitored strategy in patients with advanced stages Hodgkin lym-
26. Barrington SF, Mikhaeel NG, Kostakoglu L, et al. Role of imaging in the phoma: interim analysis of the AHL2011 Lysa study. Paper presented at:
staging and response assessment of lymphoma: consensus of the In- 57th American Society of Hematology Annual Meeting and Exposition;
ternational Conference on Malignant Lymphomas Imaging Working December 2015; Orlando, FL.
Group. J Clin Oncol. 2014;32:3048-3058. 42. Hague C, Johnson P, Federico M, et al. Pulmonary function and grade 3/
27. Gallamini A, Patti C, Viviani S, et al; Gruppo Italiano Terapie Innovative 4 clinical events in PET negative patients taking part in the international
nei Linfomi (GITIL). Early chemotherapy intensification with BEACOPP in rathl trial (CRUK/07/033): a comparison of 12 vs 4 doses of bleomycin.
advanced-stage Hodgkin lymphoma patients with a interim-PET posi- Paper presented at: 13th International Conference on Malignant
tive after two ABVD courses. Br J Haematol. 2011;152:551-560. Lymphoma; June 2015; Lugano, Switzerland.
28. Press OW, LeBlanc M, Rimsza LM, et al. A phase II trial of response-adapted 43. van Leeuwen FE, Klokman WJ, Stovall M, et al. Roles of radiation dose,
therapy of stage III-IV Hodgkin lymphoma using early interim FDG-PET chemotherapy, and hormonal factors in breast cancer following
imaging: U.S. Intergroup S0816. Paper presented at: 12th International Hodgkins disease. J Natl Cancer Inst. 2003;95:971-980.
Conference on Malignant Lymphoma; June 2013; Lugano, Switzerland. 44. Aleman BM, van den Belt-Dusebout AW, Klokman WJ, et al. Long-term
29. Johnson PW, Federico M, Fossa A, et al. Response-adapted therapy cause-specific mortality of patients treated for Hodgkins disease. J Clin
based on interim FDA-PET scans in advanced Hodgkin lymphoma: first Oncol. 2003;21:3431-3439.
analysis of the safety of de-escalation and safety of escalation in the 45. Myrehaug S, Pintilie M, Tsang R, et al. Cardiac morbidity following
international RATHL study. Paper presented at: 13th International modern treatment for Hodgkin lymphoma: supra-additive cardiotox-
Conference on Malignant Lymphoma; June 2015; Lugano, Switzerland. icity of doxorubicin and radiation therapy. Leuk Lymphoma. 2008;49:
30. Barrington SF, Kirkwood AA, Franceschetto A, et al. PET-CT for staging and 1486-1493.
early response: results from Response Adapted Therapy in Advanced 46. Hodgson DC, Grunfeld E, Gunraj N, et al. A population-based study of follow-
Hodgkin Lymphoma (RATHL) (CRUK/07/033). Blood. Epub 2016 Jan 8. up care for Hodgkin lymphoma survivors: opportunities to improve sur-
31. Gallamini A, Rossi A, Patti C, et al. Interim PET-adapted chemotheapy in veillance for relapse and late effects. Cancer. 2010;116:3417-3425.
advanced Hodgkin lymphoma (HL): Results of the second interim analysis 47. De Bruin ML, Sparidans J, vant Veer MB, et al. Breast cancer risk in
of the Italian GITIL/FIL DH0607 trial. Paper presented at: 13th International female survivors of Hodgkins lymphoma: lower risk after smaller ra-
Conference on Malignant Lymphoma; June 2015; Lugano, Switzerland. diation volumes. J Clin Oncol. 2009;27:4239-4246.
32. Zinzani PL, Broccoli A, Gioia D, et al. Interim PET response-adapted 48. Picardi M, De Renzo A, Pane F, et al. Randomized comparison of
therapy in advanced stage Hodgkins lymphoma: final results of the consolidation radiation versus observation in bulky Hodgkins lym-
Phase II part of the Fondazione Italiana Linfomi (FIL) HD0801 Study. phoma with post-chemotherapy negative positron emission tomog-
Paper presented at: 13th International Conference on Malignant raphy scans. Leuk Lymphoma. 2007;48:1721-1727.
Lymphoma; June 2015; Lugano, Switzerland. 49. Hodgson DC. Long-term toxicity of chemotherapy and radiotherapy in
33. Borchmann P, Eichenauer DA, Engert A. State of the art in the treatment lymphoma survivors: optimizing treatment for individual patients. Clin
of Hodgkin lymphoma. Nat Rev Clin Oncol. 2012;9:450-459. Adv Hematol Oncol. 2015;13:103-112.
34. Borchmann P, Haverkamp H, Lohri A, et al. Addition of rituximab to BEA- 50. Aleman BM, Raemaekers JM, Tirelli U, et al; European Organization for
COPPescalated to improve the outcome of early interim PET positive ad- Research and Treatment of Cancer Lymphoma Group. Involved-field
vanced stage Hodgkin lymphoma patients: second planned interim analysis radiotherapy for advanced Hodgkins lymphoma. N Engl J Med. 2003;
of the HD18 study. Paper presented at: 56th American Society of Hema- 348:2396-2406.
tology Annual Meeting and Exposition; December 2014; San Francisco, CA. 51. Aleman BM, Raemaekers JM, Tomisic R, et al; European Organization for
35. Dann EJ, Bairer O, Bar-Shalom R, et al. Tailored therapy in Hodgkin lym- Research and Treatment of Cancer (EORTC) Lymphoma Group.
phoma, based on predefined risk factors and early interim PET/CT: Israeli H2 Involved-field radiotherapy for patients in partial remission after

e384 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


RISK-ADAPTED TREATMENT OF ADVANCED HL

chemotherapy for advanced Hodgkins lymphoma. Int J Radiat Oncol positron emission tomography assessment. Leuk Lymphoma. 2015;56:
Biol Phys. 2007;67:19-30. 332-341.
52. Johnson PW, Sydes MR, Hancock BW, et al. Consolidation radiotherapy 60. Sauer M, Plutschow A, Jachimowicz RD, et al. Baseline serum TARC
in patients with advanced Hodgkins lymphoma: survival data from the levels predict therapy outcome in patients with Hodgkin lymphoma. Am
UKLG LY09 randomized controlled trial (ISRCTN97144519). J Clin Oncol. J Hematol. 2013;88:113-115.
2010;28:3352-3359. 61. Scott DW, Chan FC, Hong F, et al. Gene expression-based model using
53. Diehl V, Franklin J, Hasenclever D, et al. BEACOPP, a new dose-escalated formalin-fixed paraffin-embedded biopsies predicts overall survival in
and accelerated regimen, is at least as effective as COPP/ABVD in advanced-stage classical Hodgkin lymphoma. J Clin Oncol. 2013;31:692-700.
patients with advanced-stage Hodgkins lymphoma: interim report 62. Younes A, Connors JM, Park SI, et al. Brentuximab vedotin combined
from a trial of the German Hodgkins Lymphoma Study Group. J Clin with ABVD or AVD for patients with newly diagnosed Hodgkins lym-
Oncol. 1998;16:3810-3821. phoma: a phase 1, open-label, dose-escalation study. Lancet Oncol.
54. Savage KJ, Connors JM, Villa DR, et al. Advanced stage classical Hodgkin 2013;14:1348-1356.
63. National Institutes of Health. Phase 3 Frontline Therapy Trial in Patients
lymphoma patients with a negative PET-scan following treatment with
With Advanced Classical Hodgkin Lymphoma. https://clinicaltrials.gov/
ABVD have excellent outcomes without the need for consolidative
ct2/show/nct01712490?Term=c25003&rank=1. Accessed January 6,
radiotherapy regardless of disease bulk at presentation. Paper pre-
2016.
sented at: 57th American Society of Hematology Annual Meeting and
64. Borchmann P, Eichenauer DA, Plutschow A, et al. Targeted BEACOPP
Exposition; December 2015; Orlando, FL.
variants in patients with newly diagnosed advanced stage classical
55. Raemaekers JM, Andre MP, Federico M, et al. Omitting radiotherapy in
Hodgkin lymphoma: final analysis of a randomized phase II study. Paper
early positron emission tomography-negative stage I/II Hodgkin lym-
presented at: 57th American Society of Hematology Annual Meeting
phoma is associated with an increased risk of early relapse: clinical
and Exposition; December 2015; Orlando, FL.
results of the preplanned interim analysis of the randomized EORTC/ 65. Ansell SM, Lesokhin AM, Borrello I, et al. PD-1 blockade with nivolumab
LYSA/FIL H10 trial. J Clin Oncol. 2014;32:1188-1194. in relapsed or refractory Hodgkins lymphoma. N Engl J Med. 2015;372:
56. Tan KL, Scott DW, Hong F, et al. Tumor-associated macrophages predict 311-319.
inferior outcomes in classic Hodgkin lymphoma: a correlative study 66. National Institutes of Health. A Study of Brentuximab Vedotin Com-
from the E2496 Intergroup trial. Blood. 2012;120:3280-3287. bined With Nivolumab for Relapsed or Refractory Hodgkin Lymphoma.
57. Steidl C, Lee T, Shah SP, et al. Tumor-associated macrophages and survival in https://clinicaltrials.gov/ct2/show/study/NCT02572167. Accessed Janu-
classic Hodgkins lymphoma. N Engl J Med. 2010;362:875-885. ary 6, 2016.
58. Cuccaro A, Annunziata S, Cupelli E, et al. CD68+ cell count, early 67. National Institutes of Health. ACP-196 in Combination With Pem-
evaluation with PET and plasma TARC levels predict response in Hodgkin brolizumab, for Treatment of Hematologic Malignancies (KEYNOTE145).
lymphoma. Cancer Med. Epub 2016 Jan 13. https://clinicaltrials.gov/ct2/show/NCT02362035. Accessed January 6,
59. Touati M, Delage-Corre M, Monteil J, et al. CD68-positive tumor- 2016.
associated macrophages predict unfavorable treatment outcomes in 68. Chiou VL, Burotto M. Pseudoprogression and Immune-Related Re-
classical Hodgkin lymphoma in correlation with interim fluorodeoxyglucose- sponse in Solid Tumors. J Clin Oncol. 2015;33:3541-3543.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e385


HEMATOLOGIC MALIGNANCIESLYMPHOMA
AND CHRONIC LYMPHOCYTIC LEUKEMIA

Chemoimmunotherapy Versus
Targeted Treatment in Chronic
Lymphocytic Leukemia: When,
How Long, How Much, and in
Which Combination?

CHAIR
John M. Pagel, MD, PhD
Fred Hutchinson Cancer Research Center
Seattle, WA

SPEAKERS
Jennifer R. Brown, MD, PhD
Dana-Farber Cancer Institute
Boston, MA

Michael J. Hallek, MD
University Hospital Cologne
Cologne, Germany
MANAGEMENT OF CLL IN THE ERA OF NOVEL TARGETED THERAPIES

Chemoimmunotherapy Versus Targeted Treatment in Chronic


Lymphocytic Leukemia: When, How Long, How Much, and in
Which Combination?
Jennifer R. Brown, MD, PhD, Michael J. Hallek, MD, and John M. Pagel, MD, PhD

OVERVIEW

During the past 5 years, rapid therapeutic advances have changed the landscape of chronic lymphocytic leukemia (CLL)
therapy. This disease has traditionally been treated using cytotoxic chemotherapy regimens in combination with anti-
CD20 antibody treatment, and recent long-term follow-up data from multiple centers suggest that fit patients with CLL
with favorable disease featuresparticularly mutated immunoglobulin heavy chain variable region (IGHV) genes
derive very long-term benefit from the most potent of these regimens, namely the fludarabine, cyclophosphamide, and
rituximab (FCR) regimen. The advent of oral targeted therapies, particularly ibrutinib and idelalisib, has provided
generally well-tolerated and highly effective additional options that have come into widespread use in the relapsed
setting. Additional agents are advancing in clinical development, with the BCL-2 inhibitor venetoclax likely to be ap-
proved by the U.S. Food and Drug Administration (FDA) in 2016. With the development of these novel therapies for
patients with relapsed CLL, many unanswered questions remain, including the optimal sequence (first vs. second line),
duration, discontinuation, and combination of these agents. In addition, recent publications show the emergence of a
pattern of treatment resistance in certain subgroups of patients with del(17p) and complex karyotype that needs further
study and improvement. Because the field of CLL management has become much more complex, we focus here on
understanding the recent data and discuss many of the questions and controversies important for how we approach
patients with CLL.

C hronic lymphocytic leukemia is a heterogeneous neo-


plasm. Some patients never require treatment, whereas
others face an aggressive treatment course similar to
USE OF CHEMOIMMUNOTHERAPY FOR PATIENTS
WITH FRONT-LINE AND RELAPSED DISEASE
Potential Advantages of Chemoimmunotherapy
acute leukemia. In general practice, newly diagnosed Currently there remains a general consensus that no curative
patients with asymptomatic early- or intermediate-stage treatment of CLL exists, with the exception of allogeneic
disease (Rai 0, I-II, Binet A or B) should be monitored stem cell transplantation.4 Chemoimmunotherapy achieves
without therapy unless they have evidence of disease disease control and survival prolongation and has therefore
progression or show clear symptoms caused by CLL.1 been the current standard treatment for patients with
Several studies on early-stage CLL, as well as a meta- previously untreated CLL.5-7 Additionally, though the outcome
analysis,2 have not been able to demonstrate a potential following chemoimmunotherapy may be highly variable, im-
benefit of early intervention therapy to date, and thus the portant subgroups of patients show an excellent outcome,
International Workshop on CLL guidelines1 remain the corroborating its use as standard first-line therapy. On the
standard determinant of therapy initiation. In CLL, the other hand, secondary malignancies are one of the most
absolute lymphocyte count should not be used as the sole concerning unwanted effects following chemoimmunotherapy
indicator for treatment because even markedly elevated and the true long-term incidence in comparison with patients
leukocyte counts rarely cause symptoms or complications. with CLL not receiving chemoimmunotherapy needs to be
It should be stressed that these recommendations have better defined.
not changed in the era of new targeted agents. Clinical The German CLL Study Group has previously shown in their
trials that test the early use of novel inhibitors are cur- CLL8 study that the addition of the anti-CD20 monoclonal
rently underway.3 antibody rituximab (R) to fludarabine and cyclophosphamide

From the Dana-Farber Cancer Institute, Boston, MA; University Hospital of Cologne, Cologne, Germany; Swedish Cancer Institute, Seattle, WA.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: John M. Pagel, MD, PhD, Swedish Cancer Institute, 1221 Madison St., Suite 1000, Seattle, WA 98104; email: john.pagel@swedish.org.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e387


BROWN, HALLEK, AND PAGEL

(FC) improved both the progression-free survival (PFS) and the 12.8-year PFS was 53.9% for patients with mutated IGHV
overall survival (OS) of physically fit patients with previously (IGHV-M) and 8.7% for patients with unmutated IGHV (IGHV-
untreated symptomatic CLL.5 On the basis of these findings, UM). A plateau was seen on the PFS curve for patients
FCR has become a standard therapy for physically fit patients with IGHV-M, with no relapses beyond 10.4 years in 42
with previously untreated CLL. In an updated report of this trial, patients (total follow-up, 105.4 patient-years). On multi-
Fischer et al presented the results of an extended observation variable analysis, IGHV-UM (hazard ratio [HR] 3.37; 95% CI,
time with a median follow-up of 5.9 years, with particular 2.185.21; p , .001) and del(17p) by conventional kar-
emphasis on long-term follow-up for survival and adverse yotyping (HR 7.96; 95% CI, 1.0261.92; p = .048) were sig-
events.8 This follow-up analysis of the CLL8 study continues to nificantly associated with inferior PFS. In a retrospective
demonstrate an improvement in PFS and OS in physically fit report on 404 patients with CLL receiving front-line FCR,
patients treated with FCR when compared with FC. Patients Rossi et al also found that patients with CLL with mutated
with del(17p) showed a significantly shorter OS than those in all IGHV and neither del(11q) nor del(17p) deletion had a very
of the other cytogenetic subgroups. Conversely, of the patients high rate of long-term PFS, with OS similar to an age-
with del(11q), which is considered an adverse prognostic matched normal control population at 5 years.18
group,9 patients with IGHV mutation responded very well to Taken together, most patients with IGHV-mutated CLL
FCR and, as a consequence, showed an outcome similar to seem to benefit substantially from FCR chemoimmunotherapy,
other patients without del(17p). FCR induced a higher rate of resulting in long-term control of the disease. This observed
prolonged neutropenia during the first year after the end of benefit of FCR for the majority of patients with IGHV mutation
treatment. may announce the emergence of a therapeutic approach that
In multivariate analyses including the most commonly is guided by the IGHV mutational status. Secondly, given the
used clinical and biologic prognostics, several predicted excellent PFS following first-line FCR therapy, it may be more
OS. Each of these factors were shown to correlate with OS difficult to replace first-line chemoimmunotherapy with FCR
in previous publications.9-16 The PFS for patients with with novel agents in this specific subset of patients with CLL.
IGHV-mutated disease showed a significant difference in To test the value of newer targeted therapies against FCR
favor of the IGHV mutation subgroup compared with un- in previously untreated, fit patients with CLL, several phase
mutated disease. Most importantly, the IGHV mutation III trials have started that compare the potential of
subgroup treated with FCR showed a significantly longer PFS chemotherapy-free treatment strategies with FCR or other
and OS than those treated with FC. The median OS for chemoimmunotherapies.19,20
patients with IGHV-mutated CLL was not reached. More than
83% of the patients with an IGHV mutation treated with FCR
were still alive after almost 6 years of observation time. Potential Disadvantages of Chemoimmunotherapy
Notably, cytogenetic subgroup analysis of patients with One of the potential risks of chemoimmunotherapy is myelo-
IGHV mutation treated with FCR confirmed this excellent and immunosuppression and, in its consequence, the onset
outcome for all cytogenetic subgroups including del(11q), of myeloid malignancies (acute myeloid leukemia [AML] and
except for patients with del(17p) and normal karyotype. myelodysplastic syndrome [MDS]). Moreover, other cancers
In a similar way, the University of Texas MD Anderson may also be increased in CLL. Therefore, Benjamini et al
Cancer Center group reviewed the phase II FCR study to investigated the characteristics, incidence, outcomes, and
identify long-term disease-free survivors among the 300 factors associated with second cancers for 234 patients
patients included in the trial.17 At the median follow-up of receiving FCR-based regimens in the front-line setting.21
12.8 years, PFS was 30.9% (median PFS, 6.4 years). The They showed that the risk of second cancers was 2.38 times
higher than the expected risk for the general population.
Ninety-three patients (40%) had other cancers before FCR,
and 66 patients (28%) had them after FCR. Rates of therapy-
KEY POINTS related acute AML/MDS (t-AML/MDS; 5.1%) and Richter
transformation (9%) were high, while solid tumors were not
Chemoimmunotherapy remains an appropriate option increased.
for many patients with CLL and in particular those with In some contrast, in their follow-up report on the CLL8
most favorable features in the front-line setting. protocol, Fischer et al were not able to report increased rates
The use of targeted novel agents has changed the of t-AML/MDS and Richter transformation following FCR
therapeutic landscape for treating relapsed, refractory chemoimmunotherapy when compared with FC.22,23 The
CLL.
frequency of secondary malignancies was also similar in both
Many unanswered questions remain for further
investigation, including those assessing the optimal
arms. An increased frequency of Richter transformations
length of therapy, the sequencing of agents, and was noted in the FC but not FCR arm, in line with the cur-
understanding responses with class switching, as well as rent literature.24,25 In addition, achievement of a response
questions focusing on the mechanisms of overcoming seemed to correlate with a lower incidence of secondary
resistance. malignancies and Richter transformations.26 As CLL itself
appears to increase the risk of second malignancies, very

e388 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


MANAGEMENT OF CLL IN THE ERA OF NOVEL TARGETED THERAPIES

long-term follow-up studies of patients treated with NOVEL TARGETED THERAPIES IN CHRONIC
chemoimmunotherapy or with novel agents will be required LYMPHOCYTIC LEUKEMIA
to accurately assess how either type of therapy influences The recent development of targeted therapies has revolu-
this risk. tionized the treatment landscape for patients with CLL,
particularly relapsed disease. The most advanced of these
molecules are FDA approved and include the first-generation
PARAMETERS TO BE CONSIDERED FOR THE Bruton tyrosine kinase (BTK) inhibitor, ibrutinib, and the PI3K
CHOICE OF THE BEST THERAPY delta inhibitor, idelalisib. The BCL-2 inhibitor venetoclax has
Given the impressive number of choices, the selection of the been also expected to receive FDA approval in the first half of
optimal treatment of a given CLL patient has become a task 2016. To understand how we might use these agents and
that requires experience, good clinical judgment, and ap- others as the field evolves, we will consider each target and
propriate use of diagnostic tools. The following parameters inhibitor in turn (Table 1 and Fig. 1).
should be considered before recommending a treatment of
CLL: (1) the clinical stage of the disease, (2) the symptoms of
the patient, (3) the fitness of the patient, (4) the genetic risk BTK Inhibitors: Ibrutinib
of the leukemia, and (5) the treatment situation (first vs. Ibrutinib is the first-in-class inhibitor of BTK, which is critical
second line, response vs. nonresponse to the last treat- for B-cell survival and function but whose loss otherwise
ment).27 Using the assessment of these five parameters, the causes few problems for patients with Bruton X-linked
following recommendations can be given. agammaglobulinemia. Ibrutinib is a covalent inhibitor that
binds to Cys481 in BTK, resulting in prolonged target in-
hibition despite a short circulating half-life.32,33 Ibrutinib is a
First-Line Treatment potent inhibitor but not a specific one, inhibiting 19 other
Treatment should be initiated for patients with advanced kinases, several covalently, with IC50 greater than 100 nM.33
(Binet C, Rai III-IV) or active symptomatic disease. In this In fact, ibrutinib has been shown to covalently inhibit ITK,34 a
situation, patients must be evaluated for their physical kinase important for T and natural killer cell activity that may
condition (or comorbidity). For patients in good physical lead to impaired antibody-dependent cellular cytotoxicity.35
condition (go-go) as defined by a normal creatinine clear- Nonetheless, the clinical activity of ibrutinib in CLL is
ance and a low score on the cumulative illness rating scale impressive. In the initial CLL-specific phase IB/II study,
(CIRS),28 patients should be offered combination therapies nodal responses were noted to be very high initially, often
such as FCR. Patients with a somewhat impaired physical with increase in lymphocytosis, which then resolved
condition (slow-go) may be offered either chlorambucil in slowly over time.29 This observation, which is universal
combination with an anti-CD20 antibody (with obinutuzu- with B-cell receptortargeting therapies, led to the defi-
mab being the most active agent). The aim of therapy in this nition of a new response categorypartial response (PR)
situation is symptom control. For patients with symptomatic with lymphocytosisfor patients who meet all criteria for
disease and with del(17p) or TP53 mutations, it has been PR except that their lymphocyte count is persistently el-
generally agreed that patients should receive ibrutinib as evated.36 Over time as the lymphocyte count decreases,
first-line treatment.29,30 these responses have evolved to PR by standard criteria.1
Thus, at the time of the first single-agent report, the overall
response rate (ORR) was 71%, with an estimated 26-month
Second-Line Treatment PFS of 75% for patients with a median of four prior ther-
As a general rule, the first-line treatment may be repeated, if apies.29 This study led to accelerated FDA approval in
the duration of the first remission exceeds 24 to 36 months. February 2014 of ibrutinib for patients with CLL after at
The choice is more difficult in treatment-refractory CLL (as least one prior therapy. The confirmatory registration trial,
defined by an early relapse within 612 months after the RESONATE, randomly assigned patients with relapsed CLL
last treatment) or in cases with the chromosomal aber- to ibrutinib or ofatumumab and demonstrated a significant
ration del(17p). In principle, the initial regimen should be prolongation of both PFS (HR 0.22; p , .001) and OS (HR
changed. The following treatment options should be dis- 0.43; p = .005) in the ibrutinib arm.37 The RESONATE-17
cussed: (1) kinase inhibitors such as idelalisib or ibrutinib, study was a phase II registration trial focused on patients
(2) experimental protocols using other nonapproved drugs, with relapsed del(17p) with one to four prior therapies and
and (3) cellular therapies (e.g., chimeric antigen recep- early results demonstrated an estimated 12-month PFS of
tor [CAR] T-cell therapy) and allogeneic stem cell trans- 79%.38 Based on these data, full FDA approval was granted
plantation with curative intent in select patients.31 The in July 2014 for patients with CLL after at least one prior
choice of one of these options may depend on the fitness of therapy, or patients with CLL with del(17p) in any line of
the patient, the availability of the drugs, and the molecular therapy.
cytogenetics. Finally, it is important to emphasize that Longer-term 3-year follow-up of the original phase IB/II
patients with refractory disease should be treated within study was recently published and demonstrated that the
clinical trials whenever possible. ORR has increased in the relapsed refractory cohort to 90%

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e389


BROWN, HALLEK, AND PAGEL

TABLE 1. Current Novel Agents for Chronic Lymphocytic Leukemia


Agent Unique Features Select Key Trials Progression-Free Survival
BTK Inhibitors
Ibrutinib First-in-class BTK RESONATE, R/R CLL, ibrutinib vs. ofatumumab, FDA R/R CLL single-agent: 26 mos = 75%39
inhibitor approved37
RESONATE-17, R/R CLL and front-line 17p deletion del17p R/R CLL single-agent:
under FDA review38 12 mos = 79%38
RESONATE-2, first-line, ibrutinib vs. chlorambucil First-line single-agent:
FDA approved49 18 mos = 94%49
Acalabrutinib Lacks targeting of Phase I-II single-agent trial with ORR of 95%51 R/R CLL single-agent with median
ITK or TEC kinase 3 prior regimens: 12 mos = 100%51
BGB-3111 Targets BTK and TEC Phase I trial in first 14 patients with ORR of 93%52 N/A
but not ITK kinase
Pi3K Inhibitors
Idelalisib First-in-class PI3K Study 116, R/R CLL, idelalisib/rituximab vs. R/R CLL with rituximab (45% with
inhibitor targeting rituximab, FDA approved.57 del(17p)): median 19.4 mos57
d isoform
Study 115, R/R CLL BR/idelalisib vs. BR under FDA R/R CLL with BR: median 23.1 mos59
review59
Phase II first line, idelalisib and rituximab in 64 First line with rituximab: 36
patients older than age 65 with ORR of 97%64 mos = 83%64
Duvelisib Dual PI3K d and g Phase III R/R CLL, duvelisib vs. ofatumumab, R/R CLL phase I single-agent (52%
inhibitor under FDA review with del(17p)): 24 mos = 59%.66
TGR-1202 PI3K d inhibitor with Phase I, single-agent TGR-1202 in 20 patients N/A
once-daily dosing with R/R CLL with 88% nodal response67
Bcl-2 Inhibitor
Venetoclax Lacks targeting of Phase I/expansion R/R CLL, venetoclax single R/R CLL single agent: median
Bcl-xL agent, under FDA review72 25 mos72
Phase Ib R/R CLL, venetoclax and rituximab in R/R CLL with rituximab (33% with
49 patients with ORR of 86%73 del(17p)): 24 mos = 83%.73
Monoclonal Antibodies/Engineered Antibody-Like Molecules
Obinutuzumab Humanized anti- CLL11 first line, R/R CLL, obinutuzumab and First line with chlorambucil: median
CD20 antibody chlorambucil, FDA approved7 26.7 mos.7
Ofatumumab Humanized anti- First line, ofatumumab and chlorambucil First line with chlorambucil: median
CD20 antibody vs. chlorambucil, FDA approved87 22.4 mos.87
CLL refractory to fludarabine (F) and alemtuzumab Refractory to FA duration of
(A), FDA approved88 response = 6.5 mos.88
Ublituximab IgG1 antibody with Phase II R/R (40 patients) CLL with ibrutinib with N/A
unique binding ORR = 88% and 95% in high risk (20 patients)67
sequence
Phase III R/R CL, ublituximab and TGR-1202
vs. obinutuzumab and chlorambucil ongoing
Otlertuzumab Humanized anti- Phase I, single agent in 83 patients with R/R CLL R/R CLL with bendamustine:
CD37 Ig fusion with 23% ORR89 median 16.1 mos90
protein
Phase II R/R CLL, otlertuzumab and bendamustine
vs. bendamustine90
Abbreviations: N/A, not available; R/R, relapsed/refractory; BR, bendamustine and rituximab; BTK, Bruton tyrosine kinase; CLL, chronic lymphocytic leukemia; FDA, U.S. Food and Drug
Administration; mos, months; ORR, overall response rate.

with 7% complete remissions (CR) with a median time on in PFS has been seen between patients with and without
treatment of 23 months and 53% of patients remaining del(17p).40 However, among patients with disease that re-
on treatment.39 The estimated PFS at 30 months was lapsed on ibrutinib, both del(17p) and complex karyotype are
69%, indicating that although most patients remain in PR, quite common, with the latter suggested to be more important
nonetheless they can maintain long-term remissions. In in two studies.41,42 Among patients with relapsing disease,
addition, cytogenetic aberrations were also profoundly Richter transformation had a cumulative incidence of 4.5%
predictive of PFS. Those patients with del(17p) had a median at 1 year in the largest study to date and was seen in 18 of
PFS of 28 months, while the PFS for those with del(11q) was 31 patients with relapsed disease with median survival
74% at 30 months and for those with neither high-risk of 3.5 months.41 Chronic lymphocytic leukemia relapse
deletion, it was 87%. In the RESONATE study, at present tended to occur later but was also aggressive with median
with 16-month follow-up on the ibrutinib arm, no difference OS of 17.5 months. In a second study, median OS after

e390 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


MANAGEMENT OF CLL IN THE ERA OF NOVEL TARGETED THERAPIES

FIGURE 1. Schematic Representation of CLL Lymphocyte Depicting the B-Cell Signaling Cascade and Novel
Therapeutic Agents and Their Associated Targets

discontinuing ibrutinib was only 3.1 months.43 Thus, disease registration trial for patients with previously untreated CLL
progression on ibrutinib has become a new unmet need for older than age 65 without del(17p) who were randomly
patients showing aggressive disease that may be very dif- assigned to either single-agent ibrutinib or chlorambucil.
ficult to treat. Ongoing work has focused on elucidating the Ibrutinib significantly improved PFS (HR 0.16; p = .001), with
mechanism(s) of relapse; among patients with relapsing CLL, an estimated 18-month PFS of 94%, as well as ORR and OS,
mutations have been identified in the target Cys residue in and 87% of patients continue to take ibrutinib.49 Notable
BTK that results in a change to Ser that abrogates covalent serious adverse events among the 136 patients in the
binding, as well as activating mutations in PLCg, the im- ibrutinib arm included five major hemorrhages, two un-
mediate downstream target of BTK.44,45 Little is yet known explained sudden deaths, hypertension in 14%, and atrial
about the biology of the Richter transformations. fibrillation in 6%. This study led to FDA approval of single-
Ibrutinib has been generally well tolerated with the most agent ibrutinib for patients with previously untreated CLL in
common adverse events including mild diarrhea, nausea, early March 2016.
ecchymosis, and arthralgia; in most clinical trials, the rate of
discontinuation for adverse events has been 10%12%.37,39
However, this risk has been shown to be age-dependent, Next-Generation BTK Inhibitors
with higher rates among those over age 70 and even more-so Given the success of ibrutinib, much interest has naturally
among patients over age 80.41 Medically important adverse focused on efforts for continued improvement. To date, this
events include atrial fibrillation that was increased in the attention has largely been aimed at developing an equally
ibrutinib arm in RESONATE and occurred at 5%10% fre- potent inhibitor that may be more specific for inhibition of
quency in most studies,37,38 as well as risk of major hem- BTK in hopes of reducing the bleeding and cardiac toxicities
orrhage, which may be related, at least in part, to inhibition that appear to be because of ibrutinib-mediated off-target
of collagen-induced platelet aggregation by ibrutinib through effects. The first more specific inhibitor was CC-292, which
its effect on BTK and possibly TEC kinase.46,47 Because of the showed lower response rates and less durability of response
latter risk, ibrutinib should be cautiously combined with despite escalating to a high-dose and achieving high-levels of
anticoagulation, if at all. BTK occupancy.50 The reason for this reduced activity has
Increasing data have become available on the use of been unclear but seems likely due to reduced bioavailability
ibrutinib as initial therapy for CLL. In the initial phase IB/II or delivery of the drug to tumor. Within the past year,
study, 31 treatment-naive patients over age 65 with gen- however, two additional specific BTK inhibitors that bind
erally low-risk disease were enrolled, and, at the most recent covalently to the same Cys target as ibrutinib have shown
30-month follow-up, 81% remained on study treatment with promising results. Acalabrutinib (ACP-196) targets BTK but
an ORR of 84% and 23% patients were in CR at this longer not ITK or TEC and has been studied in a phase I and II
follow-up.48 The PFS was 96% at 30 months with one relapse relapsed CLL single-arm study among patients with a me-
and death in a patient with del(17p).39 RESONATE-2 was a dian of three prior regimens and showed an ORR of 95%,

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e391


BROWN, HALLEK, AND PAGEL

including 10% PR with lymphocytosis (PR-L) at 14.3 months, Idelalisib shows a characteristic pattern of toxicity that
with only two PFS events thus far (one death from pneumonia includes transaminitis at weeks 4 to 8, delayed colitis oc-
and one CLL progression).51 With current early follow-up, no curring at a median of 7 months after starting treatment,
events of major hemorrhage or atrial fibrillation have occurred. and a low-rate of drug-related pneumonitis occurring at a
Acalabrutinib has entered two registration trials, including a trial median of 4 months. A recent summary of toxicity data
head-to-head against ibrutinib in relapsed high-risk CLL. The across eight trials of patients primarily with relapsed disease
second drug, BGB-3111, targets BTK and TEC but not ITK. In a demonstrated that grade 3 or greater transaminitis and
phase I study across B-cell malignancies presented at the 2015 colitis were each seen in 14% of patients, and pneumonitis
American Society of Hematology (ASH) Annual Meeting, 39 pa- was seen in 3%. These toxicities have been responsive to
tients have been enrolled, and complete BTK occupancy has been drug hold and to corticosteroids, and recent data have
demonstrated in lymph node biopsies.52 The overall response demonstrated CD8+ T-cell infiltrates in the liver60 and co-
rate was 93% (13/14 patients) to date, and this study is ongoing. lon61,62 of affected patients. An immunologic mechanism
of toxicity may not be surprising given that the delta iso-
form of PI3K has been shown to be critical for the survival
PI3K Inhibitors: Idelalisib and function of regulatory T cells,63 and recent data have
The other critical target that led to an FDA-approved drug in demonstrated that early hepatotoxicity is associated with a
CLL has been the delta isoform of PI3 kinase (PI3K) that is decrease in T regulatory cells 1 month after starting idela-
targeted by idelalisib. Expression of the delta isoform has lisib.60 These toxicities do resolve with drug hold and/or
been shown to be limited to hematopoietic cells, and its steroid use, and, following that, the majority of patients are
primary physiologic function appears to be in B cells based able to resume dosing at the same or lower dose. Colitis can
on the phenotype of the knockout mouse.53 Most down- prove most troublesome but can be commonly managed by
stream PI3K signaling in CLL cells has been shown to go oral budesonide with a prolonged slow taper.
through delta54,55 and can be inhibited in vitro and in vivo by Use of idelalisib in the up-front setting has been more
idelalisib.54-56 In the phase I relapsed CLL study, idelalisib limited than ibrutinib. The study with the longest follow-up
had a 72% ORR, including 33% PR-L.56 Median PFS across all was a phase II study of 64 patients older than age 65 treated
cohorts in this heavily pretreated population (median of five front line with idelalisib with rituximab. These results
prior regimens) was 15.8 months, while the median PFS at showed a 97% ORR with 19% CR and 83% PFS at 36 months.64
the recommended phase II dose of 150 mg or more two Among nine patients with del(17p)/TP53 mutation, the ORR
times per day was 32 months. Because of the persistence of and PFS were both 100%. However, toxicity was also higher,
lymphocytosis over time in the phase I patients and the with rates of 42% for grade 3 or higher diarrhea with long-
ongoing evolution of the response criteria, it was decided to term follow-up and 23% for grade 3-4 transaminitis. These
study combination therapies with idelalisib in registration higher rates of toxicity appear consistent with an immu-
trials. Three phase III randomized trials were designed for nologic mechanism and recently reported high rates of grade
patients with relapsed CLL: idelalisib/rituximab versus rit- 3-4 transaminitis among younger patients treated up front
uximab (study 116), idelalisib/ofatumumab versus ofatu- with idelalisib.60 Randomized registration trials in previously
mumab (study 119), and idelalisib/bendamustine/rituximab untreated patients were recently stopped due to excess
(BR) versus bendamustine/rituximab (BR) (study 115). Study infectious deaths on the idelalisib arms, albeit without
116 enrolled patients with relapsed CLL with increased mandatory prophylaxis.
numbers of comorbidities, 45% of whom had del(17p), and
demonstrated significant improvements in both PFS (HR
0.25; median, 19.4 vs. 7.3 months at the second interim Other PI3K Inhibitors
analysis following crossover; p , .0001) and OS.57 No dif- Two other PI3K inhibitors are in advanced clinical devel-
ference in PFS was seen based on the adverse prognostic opment. Duvelisib is a dual PI3K delta and gamma inhibitor.
factors of unmutated IGHV or del(17p)/TP53 mutation. The gamma isoform of PI3K is expressed in CLL cells but also
These results led to the FDA approval of idelalisib with in cells of the microenvironment, including T cells and
rituximab in July 2014 for patients with relapsed CLL for macrophages.65 The phase I study of duvelisib enrolled 55
whom rituximab may be appropriate therapy. Subsequently, patients with CLL with a median of four prior regimens, 89%
study 119 results showed improved PFS for idelalisib with unmutated IGHV, and 52% with del(17p) or TP53 mutation.66
ofatumumab (median, 16.3 months) versus ofatumumab The ORR was 58% with an estimated 24-month PFS of 59%.
alone (median, 8 months; HR 0.27; p , .0001).58 The results The recommended phase II dose was selected as 25 mg two
of idelalisib plus BR versus BR (study 115) were reported at times per day, which reaches the IC90 for delta and the IC50
the ASH Annual Meeting in December 2015 and showed for gamma inhibition in vivo. A phase III registration trial in
significant improvement in median PFS 11.1 to 23.1 months relapsed CLL that randomly assigned patients to duvelisib or
(HR 0.33; p , .0001).59 In this patient population with 33% of ofatumumab has now completed accrual. TGR-1202 is a
patients with del(17p) and 30% of patients with refractory next-generation PI3K delta inhibitor that has a different
CLL, OS was also significantly improved. Both of these studies chemistry and has been postulated to reduce the trans-
are expected to lead to additional indications for idelalisib. aminitis seen with other delta inhibitors. A phase I study

e392 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


MANAGEMENT OF CLL IN THE ERA OF NOVEL TARGETED THERAPIES

using TGR-1202 enrolled patients across multiple B-cell Venetoclax has also been investigated in very high-risk CLL
malignancies, including 20 patients with CLL.67 Among the groups. A phase II potential registration study focused on
patients with CLL, the nodal response rate was 88% (14/16) patients with relapsed disease with del(17p) who received
and the PR rate was 63%. Thus far, the rates of grade 3-4 continuous single-agent venetoclax. The study enrolled 107
transaminitis (2%) and colitis (1%) have been much lower patients with a median of two prior regimens, and PFS at
than with the other delta inhibitors, but follow-up remains 12 months was estimated at 72% with OS estimated at 87%.
shorter. TGR-1202 is being further evaluated in many on- These data appear comparable to those of ibrutinib in a
going phase I and II combination trials, with recent initiation similar study.38
of a phase III registration trial.
Few studies of pan-PI3K inhibitors have been per-
formed in CLL. Pilaralisib has been reported to have a
UNANSWERED QUESTIONS
Despite the seminal advances seen to date with these novel
similar pattern of response as delta inhibitors and a 50%
agents in CLL therapy, many practical questions remain. The
ORR in CLL, with PFS of 7.4 to 22 months and a 20% overall
appropriate sequencing of these agents remains in question,
response in lymphoma.68 This level of activity may not
and the utility of class switching after drug failure has not
be as high as the delta-specific inhibitors described
been fully elucidated. Questions also persist about the
above. Voxtalisib had activity in follicular lymphoma but
unknown ability to stop one of these novel agents, partic-
was limited in other histologies. 69 This more limited
ularly in more favorable, less-heavily pretreated patients
activity may reflect the pharmacologic properties of
who may not require indefinite long-term therapy. At the
these compounds in comparison with the delta-specific
other end of the disease spectrum, will these agents be
inhibitors, so additional studies of pan-PI3K inhibi-
meaningful options for patients with very aggressive dis-
tors with better pharmacodynamic properties are still
ease, such as those with Richter transformation? Or will use
warranted.
of these drugs result in increased rates of aggressive Richter
transformation? Recently there has also been keen interest
in combinations of targeted agents with standard chemo-
BCL-2 Inhibition
immunotherapeutic regimens. Will these combination ap-
The BCL-2 specific inhibitor venetoclax is expected to receive
proaches be critical to overcome drug resistance and lead to
FDA approval in the first half of 2016. This drug represents
major improvements in survival? What about combinations
the culmination of 2 decades of structure-based design
of two (or even more) targeted agents? Furthermore, how
targeted at BCL-2. Its predecessor navitoclax had clinical
will these important advances translate from the major
activity in CLL70 but showed dose-limiting thrombocytopenia
academic centers, where there has been the vast majority of
because of the direct effect of navitoclax on BCL-XL in cir-
experience with these agents, to the community setting and
culating platelets, leading to their rapid clearance from the
standard clinical practice? Lastly, given this new landscape of
circulation.71 Venetoclax only has activity against BCL-2 and
exciting agents, where do we use cellular therapies and
has not shown significant clinical thrombocytopenia.72 The
potentially curative stem cell transplantation as we move
most substantial adverse event with venetoclax has been
forward? We should recognize, moreover, that follow-up
tumor lysis syndrome, which led to several revisions of the
observation after beginning use with a kinase inhibitor has
dosing schema during the phase I study. Ultimately, the
been short relative to their projected duration of therapy,
selected dosing schedule that has moved forward into all
and, thus, the full toxicity profiles of these agents have not
other trials includes a risk stratification for tumor lysis that
yet become completely known.
dictates whether patients are admitted to the hospital for
first dose and starts all patients at a low dose of 20 mg daily
for a week, with increases to 50 mg, 100 mg, 200 mg, and Can We Stop Therapy With the New Targeted Agents?
eventually to 400 mg daily, which is the recommended phase How long should patients remain on therapy with a kinase or
II dose for patients with CLL.72 The phase I and expansion BCL-2 inhibitor? Although patients universally favor the use
cohort study enrolled 116 patients with a median of four of an oral regimen, the use of these agents indefinitely may
prior regimens and showed an ORR of 79% with 20% CR. be less appealing to a large proportion of patients, especially
Median PFS was 25 months in the dose-escalation cohorts. those who are treatment-naive or relatively early in their
The CR rate suggests that venetoclax can more effectively treatment encounters. We previously acknowledged that
clear blood and bone marrow than the kinase inhibitors, and many patients with high-risk disease who have failed mul-
this observation has been further supported by data from tiple lines of therapy will relapse very quickly after stopping
the venetoclax combination study with rituximab, which one of the kinase inhibitors. However, for those who may
enrolled less heavily pretreated patients with a median of have lower-risk disease and particularly those who might be
two prior regimens.73 The overall response rate was 86% receiving single agents as first-line therapy, can, or should,
with 47% CR, and, perhaps most impressively, the estimated we stop therapy at some point and observe? Specifically, is
24-month PFS was 83%. Furthermore, half of both CR and PR there a clinical marker such as the absence of any evidence of
were minimal residual disease (MRD)negative in bone MRD that would suggest appropriate discontinuation of a
marrow, for an overall rate of 55%. novel drug therapy?

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e393


BROWN, HALLEK, AND PAGEL

Stopping therapy has the advantage of reducing toxicity ultimately depend on whether the increased early CR rate
and reducing the selective pressure for resistance. It has translates into improved PFS.
become widely accepted that assessment of MRD can be a With the use of new targeted agents and combinations
highly sensitive measure of disease burden, especially fol- with cytotoxic regimens such as BR, however, it is well
lowing CLL-directed chemoimmunotherapy, and has been understood that there are increased associated treatment-
commonly defined as having less than one CLL cell in 10,000 related toxicities. Perhaps a chemotherapy-free regimen,
leukocytes using standard four-color flow cytometric anal- even in the front-line setting, may be an attractive option for
ysis.1 Compared with patients with MRD-positive disease, patients who are older and who may have multiple comor-
achieving MRD negativity after treatment correlates with bidities. The long-term complication of developing an MDS and
longer disease-free intervals.74 If a patients disease be- acute leukemia from the use of cytotoxic chemotherapy reg-
comes MRD negative, would this result allow for discon- imens must also be considered. Therefore, specifically in
tinuation of the targeted drug therapy? Data addressing this patients with a low to moderate tumor burden and nonlife-
question are limited, but Ma et al presented a 2015 ASH threatening CLL, the improved responses and important
report in a small number of patients using venetoclax in advances with combination therapies must result in large
combination with rituximab where patients were permitted improvements in long-term survival; otherwise, a more ag-
to stop the BCL-2 inhibitor at any point desired. The data gressive regimen may not be warranted. Studies of combi-
demonstrated that 27 of 49 treated patients became MRD nations of two novel agents have just begun, so neither the
negative in the bone marrow, and nine of those patients efficacy nor toxicity of those regimens is yet known. Whether
stopped venetoclax between 10 and 36 months after beginning concurrent combination therapy truly represents an ad-
therapy.73 All nine patients with MRD-negative disease remain vancement over sequential therapy with the multiple novel
in CR with a median follow-up of 16 months. Of note, two targeted agents discussed here remains unclear, although it
patients in CR but who had evidence of MRD in the marrow also has the theoretical benefit of reducing selection pressure for
stopped treatment between 5 and 9 months, but both had resistance and may increase the rate of MRD negativity,
asymptomatic progression while off of therapy. Interestingly, thereby allowing consideration of stopping therapy.
at least one of these patients responded to rechallenge with
the same regimen. These data clearly set the foundation for
early stopping studies and in particular for patients with How Do We Sequence These Novel Agents?
favorable-risk disease, although it must be noted that ven- Is switching drug class required or is an alternative kinase
etoclax has been much more potent in inducing MRD nega- inhibitor appropriate when a patient fails a targeted agent?
tivity than the kinase inhibitors that may require combination Recent data suggest that patients who discontinue a tar-
therapy to achieve appreciable rates of MRD negativity. geted agent for adverse events have better outcomes than
those who progress, but because of the rapid evolution of
the targeted agents, little data are available addressing
Can Further Advances Be Made With Combinations of approaches to sequencing these therapeutic agents once a
Therapies Using Targeted Agents? patient may have failed treatment. Limited data exist re-
Combining established therapies with new complementary, garding practice patterns following ibrutinib or idelalisib
perhaps synergistic, approaches to improve response rates discontinuation. The response to subsequent therapies is
and deepen the quality of remissions remains a common also largely unknown, particularly outside the context of
theme in oncology care. The promise of inducing more CR or clinical research. A phase II, open-label, two-arm study
perhaps becoming MRD negative may translate to longer evaluating venetoclax monotherapy for patients with CLL
durations of remission and improvements in survival. The that had relapsed after or was refractory to either ibrutinib
conclusions are not clear, but the rationale may be solid, or idelalisib, was presented at the 2015 ASH Annual
albeit primarily based on preclinical models. For example, Meeting.76 In this early report, 41 patients had failed prior
venetoclax was demonstrated in lymphoma xenograft ibrutinib, and 13 failed treatment with idelalisib, with three
models to significantly control tumor growth and provide patients who had failed both of these kinase inhibitors.
synergy when combined with BR.71 In clinical practice, Approximately one-third of patients discontinued treatment
several single-arm combination studies have been reported because of kinase inhibitor intolerance. About 8 weeks after
with these novel agents and anti-CD20 antibodies, including starting venetoclax, the majority of patients had resolution
rituximab and ofatumumab or with chemoimmunotherapy, of B-symptoms, with approximately 80% of patients having
particularly BR. The HELIOS trial randomly assigned patients normalization of their absolute lymphocyte count, which
with relapsed CLL without del(17p) to BR versus BR with occurred quickly at a median less than 1 month after starting
ibrutinib and reported a significant improvement in PFS (HR the BCL-2 inhibitor. In addition, a 50% or greater reduction in
0.20; p , .0001) in the ibrutinib arm, but no difference in nodal mass was achieved in almost three-quarters of pa-
OS.75 The CR rate was 21% by investigator assessment in tients after switching drug class. Overall, venetoclax mono-
the investigational arm as compared with 5.9% with BR therapy demonstrated an ORR of 61% in the ibrutinib
alone. Whether BR with ibrutinib will prove to be signifi- cohort and 50% in the idelalisib cohort, albeit with small
cantly better than ibrutinib alone remains unclear and may numbers of patients studied to date. Importantly, responses

e394 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


MANAGEMENT OF CLL IN THE ERA OF NOVEL TARGETED THERAPIES

were similar for patients who discontinued kinase inhibitors discontinuation. Reasons for discontinuation included 57%
for refractory disease compared with intolerance. Clearly with Richter, 25% with CLL progression, and 18% with other
continued treatment and extended follow-up will be re- events. The high rate of Richter appears reminiscent of the
quired to fully determine response rates and durability of reports to date of patients progressing on ibrutinib where
responses after class switching. there is also a 30%50% rate of transformation.41,43
Can switching to an alternative kinase inhibitor provide Whether these Richter events are the natural history of the
efficacy after failure from the first kinase inhibitor? A disease, if we are able to keep patients alive long enough or
multicenter retrospective cohort analysis among patients they represent a biologic evolution under strong selection
with CLL who discontinued ibrutinib or idelalisib therapy was pressure from the drugs remains to be seen. Survivals re-
also recently reported at the 2015 ASH Annual Meeting.77 A ported with venetoclax were somewhat better than pre-
total of 143 patients with CLL who had previously taken viously reported with ibrutinib, however, with 1-year OS of
ibrutinib and another 35 who had failed idelalisib switched 69% for those progressing with CLL and a median 1-year OS
to the alternate kinase inhibitor. Two-thirds of these pa- for those progressing with Richter.
tients had unmutated IGHV and more than one-third had
del(17p). More than 50% who failed either drug dis-
Will Novel Immunotherapies Overcome Resistance in
continued the kinase inhibitor because of toxicity, and
Patients With CLL?
another one-third of patients stopped therapy because of
Treatment of relapsed disease continues to be clinically
disease progression, while 6%8% discontinued because of
challenging, primarily because of eventual drug resistance.
Richter transformation. The ORR was 50% (all PR) after
There has been tremendous recent interest in novel im-
switching to the alternative kinase inhibitor. Therefore,
munotherapeutic approaches to treat patients with ad-
patients who discontinue a kinase inhibitor because of
vanced disease and overcome resistance. Blocking the
toxicity can achieve a durable response with an alternate
immune checkpoint PD-1 receptor to activate cytotoxic
kinase inhibitor, although one shorter than the response
T cells has been shown to have remarkable clinical results
durations reported when these drugs are used first; the
in a variety of advanced malignancies but as yet limited
median PFS from the start of the alternative kinase in-
evaluation in CLL.80-82 PD-1 has been shown to be highly
hibitor was 11.9 months, and the alternative kinase in-
expressed in CLL cells and is also expressed in the CLL mi-
hibitor choice did not affect PFS. Importantly the median
croenvironment.83 Preclinical data have demonstrated that
PFS was only 7 months for those who had failed the first
blocking PD-L1 in Tcl-1Tg mice can delay CLL progression.84
kinase inhibitor because of CLL progression, while a median
Data from an early efficacy report from a phase II trial
PFS was not reached after 6 months of median follow-up
presented at the 2015 ASH Annual Meeting explored PD-1
for those who failed because of kinase inhibitor in-
blockade in 20 patients with relapsed/refractory CLL, in-
tolerance. These data suggest that turning to an alternate
cluding seven with Richter transformation, of which five had
kinase inhibitor therapy following kinase inhibitor dis-
progressed after ibrutinib therapy.85 The ORR was limited at
continuation can be efficacious, particularly for patients
about 25%, yet interestingly, one patient with Richter
who discontinue kinase inhibitor because of intolerance. A
achieved a CR and two achieved a PR, suggesting that further
clinical trial targeting the kinase inhibitorintolerant pa-
assessment of PD-1 blockade may be appropriate for pa-
tient population will be undertaken to validate these
tients with highly aggressive disease and probably drug
findings prospectively.
resistance.
In addition, the use of CAR T cells has recently been in-
vestigated in an attempt to provide long-term immune
Will This New Area of Targeted Therapy Lead to the
surveillance and decrease relapse in aggressive B-cell ma-
Development of an Increased Rate of Richter
lignancies.86 Briefly, the CAR has been introduced into T cells
Syndrome?
by genetic modification designed to redirect T-cell specificity
An obvious unmet need remains how to treat Richter
to the CAR-targeted antigen. In particular, CD19-specific
transformation, which occurs in about 5%10% of patients
CAR-modified T cells have shown activity in aggressive
with CLL and transforms into a fast-growing lymphoma,
B-cell lymphoma and leukemia, although toxicity can be
typically diffuse large B-cell lymphoma.78 It is well un-
high. Currently, however, given the other options in CLL, CAR
derstood that Richter transformation carries a very poor
T-cell therapy remains an investigational approach that
prognosis and is commonly highly refractory to standard
should be limited to patients with aggressive relapsed, re-
treatments. Unfortunately, there is a paucity of data to help
fractory disease.
understand how these new targeted drugs will impact the
incidence of Richter transformation. At the 2015 ASH Annual
Meeting, the first report of outcomes after failure of vene- CONCLUSION
toclax was reported in 28 patients with CLL who had a Many unresolved and controversial issues remain with
median of four prior regimens and 59% with deletion of regard to the treatment of patients with CLL, including the
17p.79 Discontinuations occurred after a median 7.5 months identification of appropriate candidates for each category of
on venetoclax, with a median 12-month follow-up after therapy. Patients with low-risk IGHV-mutated disease in the

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e395


BROWN, HALLEK, AND PAGEL

front-line setting derive long-term benefit from chemo- It appears likely that patients of all stages in disease evo-
immunotherapy (e.g., FCR). Chemoimmunotherapy in gen- lution may benefit from targeted kinase or BCL-2 inhibitor
eral has the benefit of planned short duration therapy with agents, but questions regarding the use of these agents with
long remission, in contrast to the targeted agents that to regard to sequencing, class switching, length of therapy, and
date have largely been studied as continuous single agents how to avoid or overcome resistance remain at the fore-
or continuously in combination with chemoimmunotherapy. front. Nonetheless, the exciting clinical trial data with these
Randomized trials are comparing targeted agents with rational targeted therapies will soon translate to clinical
chemoimmunotherapy, particularly in the front-line setting, practice experience and set the stage for continued pro-
while in the relapse setting, targeted agents have been found improvements in outcomes for patients with CLL, with
preferred for most patients, although it remains unclear major promise for eventual long-term improvements in
whether single-agent therapy or combinations are optimal. survival.

References
1. Hallek M, Cheson BD, Catovsky D, et al; International Workshop on 15. Hallek M, Wanders L, Ostwald M, et al. Serum beta(2)-microglobulin
Chronic Lymphocytic Leukemia. Guidelines for the diagnosis and and serum thymidine kinase are independent predictors of progression-
treatment of chronic lymphocytic leukemia: a report from the In- free survival in chronic lymphocytic leukemia and immunocytoma. Leuk
ternational Workshop on Chronic Lymphocytic Leukemia updating the Lymphoma. 1996;22:439-447.
National Cancer Institute-Working Group 1996 guidelines. Blood. 2008; 16. Hamblin TJ, Orchard JA, Ibbotson RE, et al. CD38 expression and im-
111:5446-5456. munoglobulin variable region mutations are independent prognostic
2. Chemotherapeutic options in chronic lymhocytic leukemia. CLL Trialists variables in chronic lymphocytic leukemia, but CD38 expression may
Collaborative Group. J Natl Cancer Inst. 1999;91:861-868. vary during the course of the disease. Blood. 2002;99:1023-1029.
3. Langerbeins P, Gro-Ophoff-Muller C, Herling CD. Risk-adapted therapy in 17. Thompson PA, Tam CS, OBrien SM, et al. Fludarabine, cyclophos-
early-stage chronic lymphocytic leukemia. Oncol Res Treat. 2016;39:18-24. phamide, and rituximab treatment achieves long-term disease-free
4. Dreger P. Allotransplantation for chronic lymphocytic leukemia. ASH survival in IGHV-mutated chronic lymphocytic leukemia. Blood. 2016;
Education Book. 2009;2009:602-609. 127:303-309.
5. Hallek M, Fischer K, Fingerle-Rowson G, et al; International Group of 18. Rossi D, Terzi-di-Bergamo L, De Paoli L, et al. Molecular prediction of
Investigators; German Chronic Lymphocytic Leukaemia Study Group. durable remission after first-line fludarabine-cyclophosphamide-
Addition of rituximab to fludarabine and cyclophosphamide in patients rituximab in chronic lymphocytic leukemia. Blood. 2015;126:
with chronic lymphocytic leukaemia: a randomised, open-label, phase 3 1921-1924.
trial. Lancet. 2010;376:1164-1174. 19. Hallek M. Signaling the end of chronic lymphocytic leukemia: new
6. Fischer K, Cramer P, Busch R, et al. Bendamustine in combination with frontline treatment strategies. Blood. 2013;122:3723-3734.
rituximab for previously untreated patients with chronic lymphocytic 20. Cramer P, Hallek M, Eichhorst B. State-of-the-art treatment and novel
leukemia: a multicenter phase II trial of the German Chronic Lym- agents in chronic lymphocytic leukemia. Oncol Res Treat. 2016;39:
phocytic Leukemia Study Group. J Clin Oncol. 2012;30:3209-3216. 25-32.
7. Goede V, Fischer K, Busch R, et al. Obinutuzumab plus chlorambucil in 21. Benjamini O, Jain P, Trinh L, et al. Second cancers in patients with
patients with CLL and coexisting conditions. N Engl J Med. 2014;370: chronic lymphocytic leukemia who received frontline fludarabine, cy-
1101-1110. clophosphamide and rituximab therapy: distribution and clinical out-
8. Fischer K, Bahlo J, Fink AM, et al. Long-term remission after FCR comes. Leuk Lymphoma. 2015;56:1643-1650.
chemotherapy in previously untreated patients with CLL: updated 22. Strati P, Wierda W, Burger J, et al. Myelosuppression after frontline
results of the CLL8 trial. Blood. 2016;127:208-215. fludarabine, cyclophosphamide, and rituximab in patients with chronic
9. Dohner H, Stilgenbauer S, Benner A, et al. Genomic aberrations and lymphocytic leukemia: analysis of persistent and new-onset cytopenia.
survival in chronic lymphocytic leukemia. N Engl J Med. 2000;343: Cancer. 2013;119:3805-3811.
1910-1916. 23. Gill S, Carney D, Ritchie D, et al. The frequency, manifestations, and
10. Zenz T, Eichhorst B, Busch R, et al. TP53 mutation and survival in chronic duration of prolonged cytopenias after first-line fludarabine combi-
lymphocytic leukemia. J Clin Oncol. 2010;28:4473-4479. nation chemotherapy. Ann Oncol. 2010;21:331-334.
11. Pflug N, Bahlo J, Shanafelt TD, et al. Development of a comprehensive 24. Tam CS, OBrien S, Wierda W, et al. Long-term results of the fludarabine,
prognostic index for patients with chronic lymphocytic leukemia. Blood. cyclophosphamide, and rituximab regimen as initial therapy of chronic
2014;124:49-62. lymphocytic leukemia. Blood. 2008;112:975-980.
12. Ghia EM, Jain S, Widhopf GF II, et al. Use of IGHV3-21 in chronic 25. Woyach JA, Ruppert AS, Heerema NA, et al. Chemoimmunotherapy with
lymphocytic leukemia is associated with high-risk disease and reflects fludarabine and rituximab produces extended overall survival and
antigen-driven, post-germinal center leukemogenic selection. Blood. progression-free survival in chronic lymphocytic leukemia: long-term
2008;111:5101-5108. follow-up of CALGB study 9712. J Clin Oncol. 2011;29:1349-1355.
13. Rassenti LZ, Jain S, Keating MJ, et al. Relative value of ZAP-70, CD38, and 26. Langerbeins P. Impact of first-line therapy on secondary malignancies in
immunoglobulin mutation status in predicting aggressive disease in CLL: a meta-analysis of four prospective trials of the German CLL Study
chronic lymphocytic leukemia. Blood. 2008;112:1923-1930. Group. Presented at: XVth International Workshop on CLL (IwCLL);
14. Stilgenbauer S, Schnaiter A, Paschka P, et al. Gene mutations and September 2013; Sydney, Australia.
treatment outcome in chronic lymphocytic leukemia: results from the 27. Cramer P, Hallek M. Prognostic factors in chronic lymphocytic leukemia-
CLL8 trial. Blood. 2014;123:3247-3254. what do we need to know? Nat Rev Clin Oncol. 2011;8:38-47.

e396 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


MANAGEMENT OF CLL IN THE ERA OF NOVEL TARGETED THERAPIES

28. Extermann M, Overcash J, Lyman GH, et al. Comorbidity and functional 47. Levade M, David E, Garcia C, et al. Ibrutinib treatment affects collagen
status are independent in older cancer patients. J Clin Oncol. 1998;16: and von Willebrand factor-dependent platelet functions. Blood. 2014;
1582-1587. 124:3991-3995.
29. Byrd JC, Furman RR, Coutre SE, et al. Targeting BTK with ibrutinib in 48. OBrien S, Furman RR, Coutre SE, et al. Ibrutinib as initial therapy for
relapsed chronic lymphocytic leukemia. N Engl J Med. 2013;369:32-42. elderly patients with chronic lymphocytic leukaemia or small lym-
30. Burger JA, Keating MJ, Wierda WG, et al. Safety and activity of ibrutinib phocytic lymphoma: an open-label, multicentre, phase 1b/2 trial.
plus rituximab for patients with high-risk chronic lymphocytic leu- Lancet Oncol. 2014;15:48-58.
kaemia: a single-arm, phase 2 study. Lancet Oncol. 2014;15:1090-1099. 49. Burger JA, Tedeschi A, Barr PM, et al; RESONATE-2 Investigators.
31. Dreger P, Schetelig J, Andersen N, et al; European Research Initiative on Ibrutinib as initial therapy for patients with chronic lymphocytic leu-
CLL (ERIC) and the European Society for Blood and Marrow Trans- kemia. N Engl J Med. 2015;373:2425-2437.
plantation (EBMT). Managing high-risk CLL during transition to a new 50. Harb WA, Hill BT, Gabrilove J, et al. Phase 1 study of single agent CC-
treatment era: stem cell transplantation or novel agents? Blood. 2014; 292, a highly selective Brutons tyrosine kinase (BTK) inhibitor, in
124:3841-3849. relapsed/refractory chronic lymphocytic leukemia (CLL). Blood. 2013;
32. Advani RH, Buggy JJ, Sharman JP, et al. Bruton tyrosine kinase inhibitor 122:1630.
ibrutinib (PCI-32765) has significant activity in patients with relapsed/ 51. Byrd JC, Harrington B, OBrien S, et al. Acalabrutinib (ACP-196) in re-
refractory B-cell malignancies. J Clin Oncol. 2013;31:88-94. lapsed chronic lymphocytic leukemia. N Engl J Med. 2016;374:323-332.
33. Honigberg LA, Smith AM, Sirisawad M, et al. The Bruton tyrosine kinase 52. Tam C, Grigg AP, Opat S, et al. The BTK inhibitor, Bgb-3111, is safe,
inhibitor PCI-32765 blocks B-cell activation and is efficacious in models tolerable, and highly active in patients with relapsed/refractory B-cell
of autoimmune disease and B-cell malignancy. Proc Natl Acad Sci USA. malignancies: initial report of a phase 1 first-in-human trial. Blood.
2010;107:13075-13080. 2015;126:832.
34. Dubovsky JA, Beckwith KA, Natarajan G, et al. Ibrutinib is an irreversible 53. Vanhaesebroeck B, Ali K, Bilancio A, et al. Signalling by PI3K isoforms:
molecular inhibitor of ITK driving a Th1-selective pressure in insights from gene-targeted mice. Trends Biochem Sci. 2005;30:
T lymphocytes. Blood. 2013;122:2539-2549. 194-204.
35. Kohrt HE, Sagiv-Barfi I, Rafiq S, et al. Ibrutinib antagonizes rituximab- 54. Herman SE, Gordon AL, Wagner AJ, et al. Phosphatidylinositol 3-kinase-
dependent NK cell-mediated cytotoxicity. Blood. 2014;123:1957-1960. d inhibitor CAL-101 shows promising preclinical activity in chronic
36. Cheson BD, Byrd JC, Rai KR, et al. Novel targeted agents and the need to lymphocytic leukemia by antagonizing intrinsic and extrinsic cellular
refine clinical end points in chronic lymphocytic leukemia. J Clin Oncol. survival signals. Blood. 2010;116:2078-2088.
2012;30:2820-2822. 55. Lannutti BJ, Meadows SA, Herman SE, et al. CAL-101, a p110delta
37. Byrd JC, Brown JR, OBrien S, et al; RESONATE Investigators. Ibrutinib selective phosphatidylinositol-3-kinase inhibitor for the treatment of
versus ofatumumab in previously treated chronic lymphoid leukemia. B-cell malignancies, inhibits PI3K signaling and cellular viability. Blood.
N Engl J Med. 2014;371:213-223. 2011;117:591-594.
38. OBrien S, Jones JA, Coutre S, et al. Efficacy and safety of ibrutinib in 56. Brown JR, Byrd JC, Coutre SE, et al. Idelalisib, an inhibitor of phos-
patients with relapsed or refractory chronic lymphocytic leukemia or phatidylinositol 3-kinase p110d, for relapsed/refractory chronic lym-
small lymphocytic leukemia with 17p deletion: results from the phase II phocytic leukemia. Blood. 2014;123:3390-3397.
RESONATE-17 trial. Blood. 2014;124 (abstr 327). 57. Sharman JP, Coutre SE, Furman RR, et al. Second interim analysis of a
39. Byrd JC, Furman RR, Coutre SE, et al. Three-year follow-up of treatment- phase 3 study of idelalisib (ZYDELIG) plus rituximab (R) for relapsed
nave and previously treated patients with CLL and SLL receiving single- chronic lymphocytic leukemia (CLL): efficacy analysis in patient sub-
agent ibrutinib. Blood. 2015;125:2497-2506. populations with Del(17p) and other adverse prognostic factors. Blood.
40. Brown JR, Hillmen P, OBrien S, et al. Updated efficacy including genetic 2014;124 (abstr 33).
and clinical subgroup analysis and overall safety in the phase 3 RES- 58. Jones JA, Wach M, Robak T, et al. Results of a phase III randomized,
ONATE trial of ibrutinib versus ofatumumab in previously treated controlled study evaluating the efficacy and safety of idelalisib (IDELA)
chronic lymphocytic leukemia/small lymphocytic lymphoma. Blood. in combination with ofatumumab (OFA) for previously treated chronic
2014;124 (abstr 3331). lymphocytic leukemia (CLL). J Clin Oncol. 2015;33:15 (suppl; abstr 7023).
41. Maddocks KJ, Ruppert AS, Lozanski G, et al. Etiology of ibrutinib therapy 59. Zelenetz AD, Robak T, Coiffier B, et al. Idelalisib plus bendamustine and
discontinuation and outcomes in patients with chronic lymphocytic rituximab (BR) is superior to BR alone in patients with relapsed/
leukemia. JAMA Oncol. 2015;1:80-87. refractory chronic lymphocytic leukemia: results of a phase 3 ran-
42. Thompson PA, OBrien SM, Wierda WG, et al. Complex karyotype is a domized double-blind placebo-controlled study. Blood. 2015;126 (abstr
stronger predictor than del(17p) for an inferior outcome in relapsed or LBA-5).
refractory chronic lymphocytic leukemia patients treated with 60. Lampson BL, Matos T, Kim HT, et al. Idelalisib given front-line for the
ibrutinib-based regimens. Cancer. 2015;121:3612-3621. treatment of chronic lymphocytic leukemia results in frequent and
43. Jain P, Keating M, Wierda W, et al. Outcomes of patients with chronic severe immune-mediated toxicities. Blood. 2015;126:497-497.
lymphocytic leukemia after discontinuing ibrutinib. Blood. 2015;125: 61. Louie CY, DiMaio MA, Matsukuma KE, et al. Idelalisib-associated en-
2062-2067. terocolitis: clinicopathologic features and distinction from other
44. Woyach JA, Furman RR, Liu TM, et al. Resistance mechanisms for the enterocolitides. Am J Surg Pathol. 2015;39:1653-1660.
Brutons tyrosine kinase inhibitor ibrutinib. N Engl J Med. 2014;370: 62. Weidner AS, Panarelli NC, Geyer JT, et al. Idelalisib-associated colitis:
2286-2294. histologic findings in 14 patients. Am J Surg Pathol. 2015;39:1661-1667.
45. Albitar A, Ma W, De Dios I, et al. High sensitivity testing shows mul- 63. Patton DT, Garden OA, Pearce WP, et al. Cutting edge: the phos-
ticlonal mutations in patients with CLL treated with BTK inhibitor and phoinositide 3-kinase p110 delta is critical for the function of
lack of mutations in ibrutinib-naive patients. Blood. 2015;126 (abstr CD4+CD25+Foxp3+ regulatory T cells. J Immunol. 2006;177:6598-6602.
716). 64. OBrien SM, Lamanna N, Kipps TJ, et al. A phase 2 study of idelalisib plus
46. Kamel S, Horton L, Ysebaert L, et al. Ibrutinib inhibits collagen-mediated rituximab in treatment-nave older patients with chronic lymphocytic
but not ADP-mediated platelet aggregation. Leukemia. 2014:1-5. leukemia. Blood. 2015;126:2686-2694.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e397


BROWN, HALLEK, AND PAGEL

65. Douglas M, Allen K, Sweeney J, et al. Serum chemokines and cytokines in discontinuation: results from a large multi-center study. Blood.
CLL patients treated with duvelisib, a PI3K-{delta},{gamma} inhibitor. 2015;126 (abstr 719).
J Clin Oncol. 2015;33:15s (suppl; abstr 7072). 78. Jain P, OBrien S. Richters transformation in chronic lymphocytic
66. OBrien SM, Patel M, Kahl BS, et al. Duvelisib (IPI-145), a PI3K-d,g in- leukemia. Oncology (Williston Park). 2012;26:1146-1152.
hibitor, is clinically active in patients with relapsed/refractory chronic 79. Tam C, Anderson MA, Ritchie DS, et al. Favorable patient survival after
lymphocytic leukemia. Blood. 2014;124 (abstr 334). failure of venetoclax (ABT-199/ GDC-0199) therapy for relapsed or
67. Burris HA, Patel MR, Fenske TS, et al. TGR-1202, a novel once daily PI3K- refractory chronic lymphocytic leukemia (CLL). Blood. 2015;126 (abstr
delta inhibitor, demonstrates clinical activity with a favorable safety 2939).
profile, lacking hepatotoxicity, in patients with CLL and B-cell lym- 80. Ansell SM, Lesokhin AM, Borrello I, et al. PD-1 blockade with nivolumab
phoma. Haematologica. 2015;100:154. in relapsed or refratory Hodgkins lymphoma. N Engl J Med. 2015;372:
68. Brown JR, Davids MS, Rodon J, et al. Phase I trial of the pan-PI3K in- 311-319.
hibitor pilaralisib (SAR245408/XL147) in patients with chronic lym- 81. Hamid O, Robert C, Daud A, et al. Safety and tumor responses with
phocytic leukemia (CLL) or relapsed/refractory lymphoma. Clin Cancer lambrolizumab (anti-PD-1) in melanoma. N Engl J Med. 2013;369:
Res. 2015;21:3160-3169. 134-144.
69. Brown JR, Hamadani M, Arnason J, et al. SAR245409 monotherapy in 82. Grzywnowicz M, Zaleska J, Mertens D, et al. Programmed death-1 and
relapsed/refractory follicular lymphoma: preliminary results from the its ligand are novel immunotolerant molecules expressed on leukemic
phase II ARD12130 study. Blood. 2013;122 (abstr 86). B cells in chronic lymphocytic leukemia. PLoS One. 2012;7:e35178.
70. Roberts AW, Seymour JF, Brown JR, et al. Substantial susceptibility of 83. Riches JC, Davies JK, McClanahan F, et al, T cells from CLL patients
chronic lymphocytic leukemia to BCL2 inhibition: results of a phase I exhibit features of T-cell exhaustion but retain capacity for cytokine
study of navitoclax in patients with relapsed or refractory disease. J Clin production. Blood. 2013;121:1612-1621.
Oncol. 2012;30:488-496. 84. McClanahan F, Riches JC, Miller S, et al. Mechanisms of PD-L1/PD-1-
71. Souers AJ, Leverson JD, Boghaert ER, et al. ABT-199, a potent and mediated CD8 T-cell dysfunction in the context of aging-related immune
selective BCL-2 inhibitor, achieves antitumor activity while sparing defects in the Em-TCL1 CLL mouse model. Blood. 2015;126:212-221.
platelets. Nat Med. 2013;19:202-208. 85. Ding W, Dong H, Call T, et al. PD-1 blockade with pembrolizumab (MK-
72. Roberts AW, Davids MS, Pagel JM, et al. Targeting BCL2 with venetoclax in 3475) in relapsed/refractory CLL including Richter transformation: an
relapsed chronic lymphocytic leukemia. N Engl J Med. 2016;374:311-322. early efficacy report from a phase 2 trial (MC1485). Blood. 2015;126
73. Ma S, Brander DM, Seymour JF, et al. Deep and durable responses (abstr 834).
following venetoclax (ABT-199/GDC-0199) combined with rituximab in 86. Turtle CJ, Hudecek M, Jensen MC, et al. Engineered T cells for anti-
patients with relapsed/refractory chronic lymphocytic leukemia: results cancer therapy. Curr Opin Immunol. 2012;24:633-639.
from a phase 1b study. Blood. 2015;126 (abstr 830). 87. Hillmen P, Robak T, Janssens A, et al; COMPLEMENT 1 Study In-
74. Moreton P, Kennedy B, Lucas G, et al. Eradication of minimal residual vestigators. Chlorambucil plus ofatumumab versus chlorambucil alone
disease in B-cell chronic lymphocytic leukemia after alemtuzumab in previously untreated patients with chronic lymphocytic leukaemia
therapy is associated with prolonged survival. J Clin Oncol. 2005;23: (COMPLEMENT 1): a randomised, multicentre, open-label phase 3 trial.
2971-2979. Lancet. 2015;385:1873-1883.
75. Chanan-Khan A, Cramer P, Demirkan F, et al. Ibrutinib combined with 88. Wierda WG, Kipps TJ, Mayer J, et al; Hx-CD20-406 Study Investigators.
bendamustine and rituximab (BR) in previously treated chronic lym- Ofatumumab as single-agent CD20 immunotherapy in fludarabine-
phocytic leukemia/small lymphocytic lymphoma (CLL/SLL): First results refractory chronic lymphocytic leukemia. J Clin Oncol. 2010;28:
from a randomized, double-blind, placebo-controlled, phase III study. J 1749-1755.
Clin Oncol. 2015;33:15s (suppl; LBA7005). 89. Byrd JC, Pagel JM, Awan FT, et al. A phase 1 study evaluating the safety
76. Jones J, Mato AR, Coutre S, et al. Preliminary results of a phase 2, open- and tolerability of otlertuzumab, an anti-CD37 mono-specific ADAPTIR
label study of venetoclax (ABT-199/GDC-0199) monotherapy in patients therapeutic protein in chronic lymphocytic leukemia. Blood. 2014;123:
with chronic lymphocytic leukemia relapsed after or refractory to 1302-1308.
ibrutinib or idelalisib therapy. Blood. 2015;126 (abstr 715). 90. Robak T, Hellmann A, Kloczko J, et al. Randomized phase 2 study of
77. Mato A, Nabhan C, Barr PM, et al. Favorable outcomes in CLL pts otlertuzumab (TRU-016) and bendamustine vs bendamustine in pa-
with alternate kinase inhibitors following ibrutinib or idelalisib tients with relapsed chronic lymphocytic leukemia. In press.

e398 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


HEMATOLOGIC MALIGNANCIESPLASMA CELL
DYSCRASIA

Consolidation and Maintenance:


Moving Beyond Autotransplant

CHAIR
Parameswaran Hari, MD, MRCP
Medical College of Wisconsin
Milwaukee, WI

SPEAKERS
Amrita Krishnan, MD
City of Hope
Duarte, CA

Ravi Vij, MBBS, MD


Washington University School of Medicine
St. Louis, MO
KRISHNAN ET AL

Moving Beyond Autologous Transplantation in Multiple


Myeloma: Consolidation, Maintenance, Allogeneic
Transplant, and Immune Therapy
Amrita Krishnan, MD, Ravi Vij, MD, MBA, Jesse Keller, MD, Binod Dhakal, MD, and Parameswaran Hari,
MD, MRCP

OVERVIEW

For multiple myeloma, introduction of novel agents as part of the front-line treatment followed by high-dose chemotherapy
and autologous hematopoietic stem cell transplantation (ASCT) induces deep responses in a majority of patients with this
disease. However, disease relapse is inevitable, and, with each relapse, the remission duration becomes shorter, ultimately
leading to a refractory disease. Consolidation and maintenance strategy after ASCT is one route to provide sustained disease
control and prevent repeated relapses. Though the consolidation strategy remains largely confined to clinical trials, sig-
nificant data support the efficacy of consolidation in improving the depth of response and outcomes. There are also in-
creasing rates of minimal residual diseasenegativity with additional consolidation therapy. On the other hand,
maintenance with novel agents post-transplant is well established and has been shown to improve both progression-free
and overall survival. Evolving paradigms in maintenance include the use of newer proteasome inhibitors, immunotherapy
maintenance, and patient-specific maintenancea concept that utilizes minimal residual disease as the primary driver of
decisions regarding starting or continuing maintenance therapy. The other approach to overcome residual disease is immune
therapeutic strategies. The demonstration of myeloma-specific alloimmunity from allogeneic transplantation is well
established. More sophisticated and promising immune approaches include adoptive cellular therapies, tumor vaccines, and
immune checkpoint manipulations. In the future, personalized minimal residual diseasedriven treatment strategies
following ASCT will help overcome the residual disease, restore multiple myelomaspecific immunity, and achieve sustained
disease control while minimizing the risk of overtreatment.

A utologous hematopoietic cell transplantation with high-


dose chemotherapy and autologous cell rescue is a
mainstay of therapy for patients with multiple myeloma and
ASCT (planned consolidation or maintenance), allo-HCT, and
emerging immune therapies, are discussed.

induces a response in a large proportion of patients. CONSOLIDATION THERAPY IN MULTIPLE


However, relapse is near universal as a result of either MYELOMA
measurable disease or minimal residual disease (MRD) after Consolidation therapy following ASCT for multiple myeloma
ASCT, and multiple myeloma remains incurable.1 Effective implies a short period of intensive treatment with a single-
therapies for sustained disease control after ASCT and agent or a combination of agents. Depth of response is
prevention of repeated relapses in high-risk subgroups are widely accepted as prognostic for outcomes with multiple
unmet needs. Consolidation and maintenance after ASCT is myeloma, and this strategy further reduces disease burden
one route to overcome residual disease, whereas immune following ASCT.5,6 Yet, compared with lower-dose long-term
approaches such as allogeneic transplantation (allo-HCT), maintenance, consolidation mostly remains confined to
adoptive cellular therapies, vaccines, or antibody-based clinical trials. Historically, efforts of post-transplant con-
immune manipulations represent another approach. Allo- solidation have ranged from aggressive attempts to eradi-
HCT, despite its toxicities, has been known to cure a cate disease with tandem autologous transplantation and
minority of patients by establishing myeloma-specific consolidation cytotoxic chemotherapy, to modern novel
alloimmunity.2-4 Herein, potential treatments beyond agentbased approaches6,7

From the Judy and Bernard Briskin Center for Myeloma, City of Hope Cancer Center, Duarte, CA; Division of Medical Oncology, Washington University School of Medicine in St. Louis,
St Louis, MO; Division of Hematology Oncology, Medical College of Wisconsin, Milwaukee, WI.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Parameswaran Hari, MD, MRCP, Froedtert Hospital and Medical College of Wisconsin, 9200 West Wisconsin Ave., Milwaukee, WI 53226; email: phari@mcw.
edu.

2016 by American Society of Clinical Oncology.

210 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


BEYOND AUTOLOGOUS TRANSPLANTATION IN MULTIPLE MYELOMA

TABLE 1. Lenalidomide and Bortezomib Monotherapy Consolidation

Consolidation Induction Comparison Before After


Study Regimen Regimen Arm Duration Consolidation Consolidation PFS OS
Attal et al11
Lenalidomide VAD (46%) None 2 cycles $ VGPR: 58% $ VGPR: 69% 4-year: 43% 4-year : 73%
(614 patients) (all treated) (p , .001) vs. 22% vs. 75%
VD (46%)
Uy et al13 Bortezomib Bortezomib-naive None 6 cycles CR + VGPR: 43% CR + VGPR: 43% NR 3-year: 63.1%
(40 patients)
Mellqvist et al14 Bortezomib Bortezomib-naive Placebo 6 cycles $ nCR: 20.1% $ nCR: 45.1% 27 vs. 3-year: 80%
(187 patients) 20 months vs. 80%
$ VGPR: 39.7% $ VGPR: 70.9%
Abbreviations: PFS, progression-free survival; OS, overall survival; VAD, vincristine/doxorubicin/dexamethasone; VD, bortezomib/dexamethasone; VGPR, very good partial response;
nCR, near CR; NR, not reported.

TANDEM TRANSPLANTATION AND CYTOTOXIC IMMUNOMODULATORY AGENT


CHEMOTHERAPY CONSOLIDATION THERAPY
Initial efforts of post-ASCT consolidation therapy evolved Novel agents such as proteasome inhibitors and immuno-
from the University of Arkansas Total Therapy trials and modulatory agents (IMIDs) have advanced all aspects of
mirrored prolonged treatment regimens in pediatric acute multiple myeloma treatment. Evaluation of consolidation
lymphocytic leukemia. Total Therapy 1 (TT1) (which did not therapy with IMID monotherapy was undertaken in a phase III
include consolidation) used aggressive induction chemo- study by Intergroupe Francophone du Myelome (IFM 05-02;
therapy and tandem ASCT combined with maintenance Table 1).11 Newly diagnosed patients with multiple myeloma
interferon (IFN) to produce results superior to contemporary who underwent post-ASCT consolidation therapy with lena-
standard therapy in a SWOG cooperative group trial.8 Total lidomide were randomly assigned to receive maintenance
Therapy 2 (TT2), which expanded on TT1, evaluated ag- lenalidomide versus placebo. Two cycles of lenalidomide
gressive induction chemotherapy, tandem ASCT followed (25 mg daily) improved the rate of complete response (CR) or
by four cycles of dexamethasone, cisplatin, doxorubicin, very good partial responses (PR) from 58% to 69% (p , .001),
cyclophosphamide, and etoposide (DPACE) consolidation respectively, among all participants, but maintenance was not
chemotherapy; patients were then randomly assigned to associated with improvement of overall survival (OS). A
receive maintenance therapy with IFN with or without similarly designed phase III U.S. study (CALGB 100104) lacked
thalidomide.9 Total Therapy 3 (TT3A) included the addition the lenalidomide consolidation phase, but randomly assigned
of bortezomib-based chemotherapy induction, tandem patients to receive maintenance lenalidomide versus placebo
ASCT followed by DPACE plus thalidomide and bortezomib and reported an OS benefit in the maintenance arm.12 One of
(V-DTPACE) consolidation and then maintenance.10 The role the possible explanations for the lack of OS benefit in the
of maintenance in these trials was evaluated in a non- former study is that a short period of consolidation given to all
randomized fashion, making its individual contribution dif- subjects in IFM 0502 may have abrogated the survival benefit
ficult to discern; however, they spawned further interest for for protracted maintenance therapy.
improved post-ASCT strategies.
PROTEASOME INHIBITORBASED
CONSOLIDATION
KEY POINTS Bortezomib
Uy et al13 evaluated pre- and post-ASCT bortezomib con-
Induction with novel agent combinations followed by high- solidation in a phase II study with 40 patients (all newly di-
dose chemotherapy ASCT is the standard of care in patients agnosed with multiple myeloma) who received two cycles of
with multiple myeloma who are eligible for transplant. intravenous bortezomib followed by ASCT, and six cycles of
Relapse is inevitable in the majority of the patients intravenous bortezomib after ASCT. Only 28 patients received
despite recent improvements in progression-free post-ASCT bortezomib, with two upgraded responsesone
survival. from very good PR to CR and one from PR to very good PR. The
Effective strategies to overcome residual disease and 3-year OS and disease-free survival (DFS) were 63.1% and
prevent relapse is an unmet need. 38.2%, respectively.13
Post-transplant consolidation and minimal residual
The NORDIC Myeloma Study Group randomly assigned
diseasedirected maintenance are promising new
avenues.
bortezomib-naive patients post-ASCT to receive either no
Emerging post-ASCT immunotherapy to establish further treatment or bortezomib consolidation for six cycles.
myeloma-specific immunity and allotransplant are tools With a median follow-up of 38 months, PFS for the bortezomib-
for long-term disease control, especially for young high- treatment group was 27 months compared with 20 months
risk patients. for the control group (p = .05), and no difference in OS.
Patients who experienced at least a very good PR

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 211


KRISHNAN ET AL

TABLE 2. Bortezomib and Immunomodulatory Combination Consolidation Therapy

Consolidation Induction Comparator Before After


Study Regimen Regimen Arm Duration Consolidation Consolidation PFS OS
72
Cavo et al VTD VTD x 3 TD 2 cycles CR: 48.7% CR: 60.6% 3-year: 60% 3-year: 90%
(160 patients) (161 patients)
$ VGPR: 86.2% $ VGPR: 91.9%
p = NS (VGPR)
72
Cavo et al TD TD x 3 VTD 2 cycles CR: 40.4% CR: 46.6% 3-year: 48% 3-year: 88%
(161 patients) (160 patients)
$ VGPR: 81.4% $ VGPR: 88.2%
p = NS (VGPR)
17
Leleu et al VTD VTD No consolidation 2 cycles CR: 33% CR: 52% NR 3-year: NR vs.
(121 patients) (96 patients) 22 months
$ VGPR: 43% $ VGPR: 31%
p , .001 (CR)
73
Roussel et al RVD RVD None 2 cycles CR: 47% CR: 50% 3-year: 77% 3-year: 100%
(31 patients)
$ VGPR: 70% $ VGPR: 87%
Nooka et al18 RVD RVD None 3 years sCR approx. 20% sCR: 51% 32 months 3-year: 93%
$ VGPR approx. 85% $ VGPR: 96%
Moreau et al19 RVD RVD No transplant 2 cycles $ VGPR:73% $ VGPR: 81% 34 vs. 4-year: 81%
43 months vs. 83%
Abbreviations: PFS, progression-free survival; OS, overall survival; CR, complete response; NR, not reported; NS, not significant; RVD, lenalidomide/bortezomib/dexamethasone; TD,
thalidomide/dexamethasone; VGPR, very good partial response; VTD, bortezomib/thalidomide/dexamethasone.

demonstrated a prolonged PFS irrespective of bortezomib were 77% and 100%, respectively, and CR rates were 47%
consolidation. The beneficial effect of bortezomib consoli- following ASCT and 50% after RVD consolidation. Therapy
dation was confined to patients who did not have at least a was tolerable, with no treatment-related mortality (TRM),
very good PR following ASCT. More importantly, PFS among and 97% completed the planned sequence.16 Leleu et al17
patients whose disease had a very good PR to therapy at retrospectively analyzed 121 patients with newly diagnosed
randomization was similar to patients whose disease re- multiple myeloma across nine IFM centers who had received
sponse improved to the very good PR category or better three cycles of VTD induction, followed by ASCT and two
during consolidation. These results provide substantial cycles of VTD consolidation. Complete response rates in-
support to the role of post-transplant consolidation to creased from 33% after induction and ASCT to 52% following
further deepen disease response.14 consolidation. Compared with 96 similar patients treated
with VTD induction and ASCT without consolidation, CR rates
Combination Therapy Bortezomib and were superior and relapse risk were lower in the consoli-
Immunomodulatory Agents dation arm.17
In a phase III trial, Cavo et al15 assessed the efficacy of Nooka et al18 evaluated prolonged therapy with RVD in
consolidation with thalidomide and dexamethasone (TD) 45 patients with high-risk multiple myeloma (p53 deletion,
compared with bortezomib, thalidomide, and dexametha- 1p deletion, immunoglobulin heavy chain gene trans-
sone (VTD; Table 2). The patients were randomly assigned at locations, or plasma cell leukemia). Patients received RVD
diagnosis to induction therapy with either VTD or TD, and all as consolidation/maintenance therapy post-ASCT for up to
underwent tandem ASCT followed by two 35-day consoli- 3 years, followed by single-agent lenalidomide maintenance
dation cycles with the same regimen as their induction thereafter. RVD consolidation/maintenance achieved at least
therapy (VTD vs. TD). CR rates after second ASCT was 48.7% very good PR status in 96% of patients, and a stringent CR
for the VTD group and 40.4% for the TD group, improving to (sCR) in 51%. Median PFS was 32 months with a 3-year OS of
60.6% and 46.6%, respectively, following consolidation. PFS 93%a promising improvement in PFS and OS in this high-risk
was superior for the VTD group compared with the TD group group. Despite its length and intensity, therapy was well
(60% vs. 48% at 3 years; p = .042).15 Notably, rates of CR and tolerated overall.18
at least very good PR favored VTD treatment, but were not More recently, the second interim analysis of the IFM 2009
significant. The isolated impact of consolidation is difficult to trial was reported.19 In this trial, RVD was administered for
determine from this study. eight cycles followed by 12 months of lenalidomide main-
Roussel et al16 investigated lenalidomide, bortezomib, and tenance or RVD induction with RVD consolidation post-ASCT
dexamethasone (RVD) induction and consolidation therapy followed by lenalidomide maintenance. Response to treat-
in 31 patients with newly diagnosed multiple myeloma who ment continued to deepen with progressive therapy, and at
received three RVD induction cycles, followed by ASCT and least very good PR rates improved from 73% to 81% after RVD
two RVD consolidation cycles with subsequent lenalidomide consolidation. This study also showed an improvement in PFS
maintenance for 1 year. The estimated 3-year PFS and OS (median of 34 vs. 43 months) in the ASCT arm.

212 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


BEYOND AUTOLOGOUS TRANSPLANTATION IN MULTIPLE MYELOMA

TABLE 3. Carfilzomib-Based Consolidation Therapy

Induction Comparator Before After


Consolidation Regimen Regimen Arm Duration Consolidation Consolidation PFS OS
20
KTd (Sonneveld et al ; KTd None 4 cycles CR: 33% CR: 63% 3-year: 60% 3-year: 90%
91 patients)
$ VGPR: 76% $ VGPR: 89%
KRd (Jakubowiak et al21; KRd None 4 cycles $ CR: 27% $ CR: 77% 11 months: 99% 11 months: 100%
71 patients)
$ sCR: 22% $ sCR: 70%
Abbreviations: PFS, progression-free survival; OS, overall survival; KTd, carfilzomib/thalidomide/dexamethasone; KRd, carfilzomib/lenalidomide/dexamethasone; CR, complete
response; VGPR, very good partial response; sCR, stringent complete response.

Carfilzomib of MRD-negative response was significantly associated


In a multicenter phase II study from the European Myeloma with improved median PFS (68 months vs. 23 months,
Network, the carfilzomib, thalidomide, and dexamethasone p , .0001).
(KTd) combination was investigated as induction and con- In the study by Roussel et al,16 the outcomes of patients
solidation therapy in patients with multiple myeloma who who were MRD-negative (evaluated by marrow flow cytom-
were previously untreated (Table 3). KTd was given in 28-day etry) were impressive with a 3-year PFS of 100%.16 MRD
cycles for up to four cycles and followed by ASCT. After ASCT negativity rates steadily increased through induction to the
patients received four cycles of KTd consolidation therapy. completion of consolidation. Jakubowiak et al21 assessed MRD
The at least very good PR rate increased from 68% after in a subset of patients whose disease had a CR using 10-color
induction to 76% after ASCT, and finally to 89% after flow cytometry and noted substantial improvements in MRD
four cycles of consolidation. Progression-free survival at negativity rates with consolidation therapy.21
36 months was 72%, and only 5% of patients discontinued
therapy.20 Pre- and post-transplant carfilzomib, lenalido- CONSOLIDATION VERSUS MAINTENANCE
mide, and dexamethasone (KRd) was studied in a phase II Current evidence does not enable a clear recommendation
trial. After KRd induction, ASCT and KRd consolidation (four of consolidation therapy for all patients after they have
cycles) were administered followed by KRd maintenance undergone ASCT. Substantial data support its efficacy in
for nine cycles, and then lenalidomide maintenance off- improving depth of response of multiple myeloma, which
protocol. Rates of CR increased from 27% following ASCT to has historically translated into improvements of survival
77% after consolidation. At the conclusion of KRd treatment, outcomes. The awaited results of the Blood and Marrow
90% of patients demonstrated an at least CR with a 3-year Transplant Clinical Trials Network (BMT CTN) 0702 STAMINA
PFS and OS of 79% and 100%.21 trial (NCT 1109004), compares three competing post-
ASCT strategies after initial ASCT: patients will be ran-
CONSOLIDATION THERAPY AND MINIMAL domly assigned to receive a second (tandem) ASCT, RVD
RESIDUAL DISEASE consolidation, or immediate initiation of lenalidomide
The role of MRD evaluation in prediction of outcomes and maintenance (Fig. 1). All arms will ultimately receive
duration of therapy is increasing. Ladetto et al22 enrolled lenalidomide maintenance based on the paradigm that
patients whose disease had a very good PR following ASCT continuous therapy or maintenance is sufficient to control
and monitored MRD after four cycles of VTD consolida- residual disease. Additionally, an ongoing multicenter study
tion using polymerase chain reactionbased techniques (NCT02253316) is evaluating the role of ixazomib-based
(Table 4).22 Molecular remission rates were 3% after ASCT consolidation therapy followed by maintenance with MRD
compared with 18% after VTD consolidation. Achievement post-ASCT (Fig. 2).

TABLE 4. Minimal Residual Disease With Consolidation Therapy


Before After
Consolidation Regimen Induction Regimen Comparator Arm Duration Consolidation Consolidation
VTD (Ladetto et al22; VAD None 4 cycles CR: 15% CR: 49%
39 patients)
MRD: 4.15log MRD: 10.09log
reduction* reduction*
VRD (Roussel et al16; VRD None 2 cycles MRD: 54% MRD: 58%
26 patients) negative negative
KRd (Jakubowiak et al21; KRd None 4 cycles MRD: 79% MRD: 90%
71 patients) negative negative
*Reduction from baseline levels.
Abbreviations: VTD, bortezomib/thalidomide/dexamethasone; VAD, vincristine/doxorubicin/dexamethasone; CR, complete response; MRD, minimal residual disease; KRd,
carfilzomib/lenalidomide/dexamethasone.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 213


KRISHNAN ET AL

FIGURE 1. CTN 0702 Study (STAMINA) Schema In the United States, therefore, interest shifted to lenali-
Mel 200 Transplant &
domide as a better-tolerated IMID maintenance strategy.25
Mel 200 Transplant
Lenalidomide Maintenance
Newly Diagnosed Lenalidomide. Three published phase III trials explored
VRD Consolidaon &
Transplant Eligible Mel 200 Transplant lenalidomide maintenance after ASCT (Table 6). The CALGB
Lenalidomide Maintenance
Mulple Myeloma
100104 Intergroup trial randomly assigned patients to re-
Mel 200 Transplant Lenalidomide ceive lenalidomide (10 mg daily escalated to 15 mg) or
Maintenance
observation after ASCT. The median time-to-progression
(TTP) was 46 months in the lenalidomide arm compared
with 27 months with placebo, which translated into an
MAINTENANCE THERAPY AFTER improved OS (88% vs. 80% at 3 years).12 A recently updated
TRANSPLANTATION: THE PAST AND THE FUTURE analysis showed a median TTP of 53 months compared with
The concept of sustained low intensity post-ASCT therapy 27 months as well as continued OS advantage.26 The phase III
(or maintenance) for patients with multiple myeloma dates IFM 05-02 trial, as noted previously, had the subtle, but
back to IFN, which was associated with improved PFS and important, difference that all patients received lenalidomide
OS in several trials.23 The advent of novel agents with im- consolidation followed by lenalidomide maintenance or
proved efficacy and toxicity supplanted IFN. Given emerging placebo.11 In this trial, although median TTP was superior for
immuno-oncologic approaches, this may also have been the lenalidomide arm (41 months vs. 23 months p , .001),
prescient in harnessing the immune system as the major there was no difference in OS. The lack of OS benefit may in
mechanism to maintain control against residual disease. part be attributed to lenalidomide consolidation and or an
imbalance of high-risk patients between arms. An Italian trial
(GIMEMA RV-MM-PI209) assigned patients to melphalan,
Maintenance Agents: Immunomodulatory Agents prednisone, and lenalidomide (MPR) or ASCT followed by
Thalidomide. Probably the largest body of clinical trial ex- second random assignment to maintenance lenalidomide or
perience has been with thalidomide maintenance therapy, no maintenance. The maintenance lenalidomide cohort in
although these trials differed with some having used tha- both the MPR and ASCT groups had superior PFS (but not
lidomide as part of induction continuing through mainte- OS).27 In all trials, the lenalidomide cohorts had a higher
nance, whereas others used it in conjunction with steroids, incidence of neutropenia. Even more important was the
and with major variation in thalidomide doses chosen. higher incidence of second primary malignancies (SPM) at
Table 5 outlines the major trials of thalidomide maintenance around 8% in the IFM and CALGB trials compared with 3% to
following ASCT. All studies of thalidomide maintenance 4% in the placebo arm. Strategies to minimize SPM risk
suggest improved PFS, but toxicity and tolerability remain without losing the benefit of maintenance are areas of study,
issues. In the IFM 99 trial, 39% of patients discontinued such as limiting the duration of maintenance, pre-assessing
thalidomide mainly due to neuropathy.24 In the MRC IX trial SPM risk, and MRD-based discontinuation.
with thalidomide maintenance after intensive (transplant) or A comparison of the recently reported IFM DFCI 2009 trial
nonintensive (chemotherapy only) pathways, 52% stopped with the ongoing DFCI (Dana-Farber Cancer Institute) BMT
therapy due to side effects, and thalidomide maintenance CTN Determination trial is expected to shed light on the
did not prolong PFS or OS in those with adverse cytogenetics. duration of maintenance lenalidomide and the impact of
MRD. As described previously, the IFM version of this phase
III trial incorporated 1 year of lenalidomide maintenance.19
FIGURE 2. Design of NCT02253316: Ixazomib,
In contrast, the DFCI version (DFCI10-106; NCT1208662)
Lenalidomide, and Dexamethasone Consolidation
follows the same overall schema, but lenalidomide main-
Therapy and Minimal Residual Disease
tenance continues until disease progression. The incidence
Autologous Stem of SPM as well as sequential evaluation of MRD by flow
Cell Transplant Completion of
Consolidation cytometry and next-generation sequencing is being evalu-
ated in both trials.28
Day 80-120:
MRD Assessment
Randomized to Randomized to
Maintenance Agents: Proteasome Inhibitors
Lenalidomide Ixazomib Bortezomib. Although there is a large body of evidence
Maintenance Maintenance
Consolidation with surrounding bortezomib maintenance (Table 5), most pro-
IRD Begins tocols used bortezomib administered intravenously, which
renders the toxicity data less relevant in the era of sub-
Assess Tolerability, PFS, OS cutaneous dosing. A large Dutch-German trial randomly
4 Cycles of IRD:
MRD Assessment assigned patients to receive bortezomib, doxorubicin, and
dexamethasone (PAD) or vincristine, doxorubicin, and
Abbreviations: MRD, minimal residual disease; IRD, ixazomib, lenalidomide, and
dexamethasone (VAD)29 induction followed by ASCT. The
dexamethasone; PFS, progression-free survival; OS, overall survival. PAD induction arm subsequently received bortezomib

214 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


BEYOND AUTOLOGOUS TRANSPLANTATION IN MULTIPLE MYELOMA

TABLE 5. Thalidomide and Bortezomib Maintenance Studies

Schedule Control CR PFS/EFS/TTP Reference


PAM + Thal 400 mg/day PAM or OBS NR 52% (3-year) Attal et al24
Thal 400 mg/day None 50% 72% (6-year) Barlogie et al74
Thal 200 mg/day + PSE 50 mg QOD PSE NR 42% (3-year) Spencer et al75
Thal 50 mg/day IFN 31% 50% (34-month) Lokhorst et al76
Thal 100 mg/day OBS NR 50% (30-month) Morgan et al25
Thal 200 mg/day + PSE 50 mg QOD OBS NR 32% (4-year) Stewart et al77
2
Bort 1.3 mg/m Q2 weeks Thal NR 50% (35-month) Sonneveld et al29
Bort + Thal Thal or IFN 19% 50% (45-month) Rosinol et al31
Abbreviations: CR, complete response; PFS, progression-free survival; EFS, event-free survival; TTP, time-to-progression; PAM, pamidronate; Thal, thalidomide; OBS, observation, NR,
not reported; PSE, prednisone; QOD, every other day; IFN, interferon; Bort, bortezomib; Q2, every two.

maintenance for 2 years, whereas the VAD arm received valuable. In another phase II trial using ixazomib and
thalidomide maintenance. Median PFS was 35 months in the lenalidomide combination maintenance,32 ixazomib dose
bortezomib-treated arm compared with 28 months in the was reduced to 3 mg or less (from the standard 4 mg) in
VAD/thalidomide-treated arm. A landmark analysis sug- some patients because of the development of neuropathy.
gested an OS benefit, although it is not clear if the benefit Overall, the combination was well-tolerated, with the ma-
was derived solely from the bortezomib maintenance or jority of patients still receiving treatment at 30-plus cycles
induction, or from the higher discontinuation rates with with an estimated 2-year PFS of 83%.
thalidomide maintenance. There was particular improve- In the allogeneic setting, the BMT CTN 1302 phase II trial
ment in the high-risk subgroup with 17p deletion that re- for patients with protocol-defined high-risk multiple mye-
ceived bortezomib (PFS of 26 months vs. 12 months in the loma (NCT02440464) randomly assigned patients to receive
thalidomide arm).30 Another three-arm Spanish trial com- ixazomib maintenance or placebo starting at 60- to 120-days
pared bortezomib plus thalidomide, thalidomide, or IFN as after allo-HCT. Because bortezomib has been shown to
maintenance with a planned duration of 3 years. PFS was reduce graft-versus-host disease (GVHD), the effects of
superior with the bortezomib/thalidomide combination, but ixazomib on GVHD and relapse rates are being evaluated.
there was no OS benefit.31
Immunotherapy
Carfilzomib and ixazomib. Other proteasome inhibitors Immunotherapy for multiple myeloma encompasses approved
could offer the potential advantages of minimizing neu- myeloma-directed passive antibody or active approaches that
ropathy with possibly higher potency. Carfilzomib-based enhance tumor-specific immune responses. The post-ASCT
combinations with lenalidomide have been described pre- maintenance setting is the ideal platform for immunother-
viously, whereas ixazomib maintenance is another attractive apy, given low disease burden as well as a favorable immune
option, given its oral administration and lower neuropathy milieu. Multiple immunotherapy options are being evaluated
risk. As shown in Fig. 2, a current trial (NCT02253316) is in trials designed to elicit antitumor immune responses, as well
evaluating ixazomib, lenalidomide, and dexamethasone as augment T-cell function (Table 7).
consolidation followed by random assignment to ixazomib Daratumumab,33 the newly approved anti-CD38directed
or lenalidomide maintenance until disease progression. antibody, has a half-life of 21 days, which makes it an at-
Although not powered to show a benefit with either of the tractive maintenance option. An ongoing IFM phase III trial
maintenance arms, the tolerability and feasibility of ixazo- (NCT02541383) for front-line therapy randomly assigns
mib maintenance and ongoing MRD assessments will be patients to receive daratumumab maintenance for 2 years

TABLE 6. Lenalidomide Maintenance Studies


Outcome
Maintenance Planned Length
Study Comparison of Maintenance PFS OS
McCarthy et al12 Lenalidomide vs. placebo Until progression Median PFS (46 vs. 3-year OS (88%
(CALGB 100104) 27 months; p , .001) vs. 80%; p = .03)
Attal et al75 Lenalidomide vs. placebo Until progression, but Median PFS (41 vs. 4-year OS (73%
(IFM 0502) after 2 months lenalidomide terminated early for SPM 23 months; p , .001) vs. 75%; p = NS)
consolidation
Palumbo et al27 MPR vs. tandem ASCT followed Until progression Median PFS 41.9 vs. 3-year OS (88%
by lenalidomide vs. placebo 21.6 months; (p , .001) vs. 79.2%; p = .14)
Abbreviations: PFS, progression-free survival; OS, overall survival; SPM, secondary primary malignancies; NS, not significant; MPR, melphalan/prednisone/lenalidomide; ASCT,
autologous hematopoietic cell transplantation.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 215


KRISHNAN ET AL

TABLE 7. Comparison of Immunotherapy Maintenance Strategies

Therapy Advantage Disadvantage


Vaccine Ease of administration Patient-specific (manufacturing)
Low toxicity
T-cellbased (Including CAR) Cytolytic trafficking to extramedullary sites Toxicity manufacturing
Antibodies Long half-life Infusional toxicity
Commercially available Cytopenias
Checkpoint Blockade Commercially available Low single-agent response rates in MM
Abbreviations: CAR, chimeric antigen receptor; MM multiple myeloma.

after ASCT. Elotuzumab, a natural killer (NK) celldirected observed in recipients of T-cell replete allografts compared
antibody requires lenalidomide to augment its activity. An with recipients of T-cell depleted or syngeneic grafts.
ongoing phase II trial (NCT 02420860) explores the combi-
nation of elotuzumab with lenalidomide following ASCT.
Modern Randomized Trials of Allo-HCT
After promising phase II data, a series of randomized,
Emerging Paradigms in Maintenance
prospective studies explored the concept of decoupling
Patient-specific maintenance is a concept that utilizes MRD
myeloablation and immune therapy by a tandem approach
as the primary driver of decisions regarding starting or
involving an initial autologous myeloablative transplant
continuing maintenance therapy. Most ongoing phase III and
followed (within 3 to 6 months) by an allogeneic non-
phase II trials are collecting MRD data at various points in
myeloablative transplant from a matched sibling or un-
time, although the optimal method for MRD assessment is
related donor. The results of this strategy have been
debated (multicolor flow cytometry vs. sequencing). To a
discordant in the front-line treatment of multiple myeloma.
smaller extent, decisions regarding choice of maintenance
Two major European studies39,40 reported favorable long-
drug are also patient- and disease-specific (e.g., bortezomib
term OS.41,42 In these studies, compared with tandem au-
for patients with t[4,14] or 17p deletion). Further refinement
tologous transplantation, a second (tandem) allo-HCT from
of this concept with more targeted therapy based on genetic
an HLA-matched donor (after a prior autograft) results in
sequencing is key. Lastly, augmenting immune function
superior PFS and OS despite an increase in early TRM. This is
through a variety of methods including adoptive cell ther-
in contrast to the U.S. study led by the BMT CTN in which no
apy, vaccines, checkpoint blockade, and myeloma-specific
survival or progression benefit was seen in the allo-HCT
antibodies will likely become the backbone of post-ASCT
group.2 Interestingly, in the larger European Group on Blood
interventions.
and Marrow Transplantation (EBMT) study, in addition to
relapse risk being lower in the allograft arm, post-relapse
IMMUNOTHERAPY FOR MULTIPLE MYELOMA: survival was also superior for the allo-HCT cohort compared
ALLOGENEIC TRANSPLANTATION AND BEYOND with tandem autograft recipients.42 This latter phenomenon
Allogeneic transplantation is a well-established immunother- has been attributed to a synergy between novel agents and
apy strategy for multiple myeloma with evidence of the ex- the immune benefits of an allo-HCT.
istence of a potent and often sustained graft-versus-myeloma As discussed in the maintenance section, more recent
effect.4 Conventional myeloablative regimens resulted in pro- studies incorporated maintenance agents (lenalidomide,
hibitive TRM (40% to 60%), but apparent plateaus in transplant bortezomib, and others) in the post allo-HCT setting. These
survival curves indicated long-term disease control. The allo- results were discordant with the HOVON group,43 reporting
geneic arm of U.S. intergroup trial (S93-21) was prematurely substantial GVHD that threatened the feasibility of planned
closed as a result of high early TRM (53%), but showed long lenalidomide maintenance. In contrast, a multicenter U.S.
relapse-free survivorship of 39% at 7 years.34 Development of study,44 in a predominantly high-risk population, indicated
nonmyeloablative and reduced-intensity conditioning regimens promising PFS and OS of 63% and 78%, respectively, at
has decreased TRM,35,36 but the lower TRM was negated by an 18 months. A current multicenter U.S. study (BMT CTN 1302;
increase in relapse risk during later years.37 The risks of chronic NCT02440464) uses the newly approved agent ixazomib in
GVHD and the need for long-term immunosuppression remain maintenance after an allogeneic transplant in patients with
major challenges. The success of allo-HCT as a relapse pre- genetically defined high-risk multiple myeloma, plasma cell
vention strategy resides in the ability of donor-derived allor- leukemia, or early relapse after an ASCT.
eactive T cells to eliminate or suppress multiple myeloma On a practical basis, if allo-HCT is considered part of the
propagating residual cells. Several lines of evidence point to this, therapeutic armamentarium, clinicians are faced with two
including the correlation of chronic GVHD with protection from major questions: (1) identifying the patients who benefit the
relapse,2,4 the ability of donor derived lymphocytes to eliminate most from allo-HCT and (2) identifying the optimal time
residual or relapsing disease,38 and lower relapse rates point for referral for allo-HCT.

216 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


BEYOND AUTOLOGOUS TRANSPLANTATION IN MULTIPLE MYELOMA

Who Should Be Considered for Allo-HCT for Multiple related or unrelated donors with an overall response rate of
Myeloma 90%, including a CR rate of 40%. Cumulative incidence of
In practical terms, this boils down to the identification of 1-year TRM was 25% and was significantly lower in transplants
patients who are at such high clinical or biologic risk that the from fully HLA-matched donors compared with mismatched
TRM risk of allo-HCT is balanced by the intrinsic negative donors (10% vs. 53%, p = .001). After a median follow-up of
prognosis. Biologically, high-risk myeloma is defined by the 43 months, the 5-year PFS and OS were 20% and 26%, re-
presence of chromosomal abnormalities t(4:14), t(14:16), spectively, and were greater in patients who demonstrated
+1q21, 17p by fluorescence in situ hybridization (FISH); 13q post-transplant CR.54 In light of the above data, it is rea-
deletion by karyotyping or high-risk gene expression pro- sonable to consider allo-HCT in high-risk patients who have
filing (GEP).45,46 Based on the recently developed R-ISS relapsed and who demonstrated a deep remission to salvage
staging system that incorporates disease burden and bi- regimens prior to allo-HCT.
ology, the highest risk subgroup (stage III) had a predicted
5-year OS and PFS of 40% and 24%, respectively.47 Similarly, Conditioning Regimen Intensity
relapse within 18 months of ASCT is an extremely negative The optimal intensity of conditioning regimens for allo-HCT is
prognostic marker.48 Effective treatment that establishes still controversial for patients with multiple myeloma. Fully
long-term disease control in these high-risk patient pop- myeloablative regimens have been largely abandoned,
ulations is an unmet need, and allo-HCT becomes a con- whereas nonmyeloablative stem cell transplant regimens
sideration. Both prospective and retrospective studies have (e.g., total body irradiation of 2 Gy) without anti-multiple
explored the role of allo-HCT in patients with high-risk myeloma activity have been associated with lower TRM risk
multiple myeloma with variable outcomes. The EBMT but increased relapse.37 The pendulum of regimen intensity
NMAM 2000 study reported a 21% PFS at the 8-year follow- has now swung back to reduced-intensity conditioning with
up in patients with the higher risk deletion 13 (by FISH) who regimens that incorporate intermediate doses of active anti-
underwent allo-HCT versus 5% in the tandem ASCT group.42 multiple myeloma therapy. The most popular approach in
Knop et al showed that in the highest risk subgroup with the United States is a combination of fludarabine and
del(17p) and del(13q) abnormality,49 median PFS (p = .002) melphalan at a dose of 140 mg/m2 (CIBMTR data). The
and OS (p = .011) were superior compared with tandem addition of proteasome inhibitors to conditioning is being
ASCT. In younger eligible patients with well-defined high-risk explored as an additional strategy to increase the antineo-
features, it is reasonable to consider allo-HCT, especially plastic potential without incurring additional toxicity.
within a clinical trial and early in the clinical course (front-line
or first relapse). The mortality and morbidity in general has IMMUNE-BASED THERAPIES BEYOND ALLO-HCT
steadily declined in recent years, making the short-term risks A variety of novel post-ASCT or allo-HCT immunotherapeutic
lower than in published studies from a decade or more strategies are now being explored for patients with multiple
ago.37 In contrast, after repeated relapses, allo-HCT itself is myeloma (Table 7). These include cellular approaches such
associated with very poor survival, making this modality a as myeloma-specific T cells (via T-cell expansion), marrow
very poor late salvage option.50 In the United States, overall infiltrating lymphocytes and redirected T cells with chimeric
allogeneic transplant activity has declined for patients with antigen receptors (CAR T), and tumor-based vaccines to
multiple myeloma and more transplants, unfortunately, are induce myeloma-specific immunity in the context of en-
now suboptimally performed later in the disease course. hanced antigen presentation. HCT provides an ideal platform
for additional immune-based therapies. The recovery phase
Timing of Allo-HCT for Relapsed Multiple Myeloma from ASCT (or other lymphodepleting therapy) represents
Prospective data that demonstrated a benefit for allo-HCT a favorable platform for adoptive cellular therapy. The
are all derived from the up-front setting after an initial ASCT. homeostatic lymphocyte proliferation following lympho-
Understandably in the modern era, patients and physicians penia is a context in which immune checkpoint blockers may
are still hesitant regarding a treatment with high immediate also be able to reverse multiple myeloma-associated T-cell
TRM risk. In patients whose disease relapsed after an ASCT, exhaustion. Additionally, lymphopenia resulting from ASCT
the benefit of allo-HCT was found to be highest when this eliminates tolerogenic antigen-presenting cells and induces
modality was used earlier in the disease course and when cytokine release that generates a more favorable environ-
used as a strategy for consolidation of remission induced ment for adoptive T-cell therapy. Indirect evidence suggests
by salvage therapy.51,52 A donor versus no donor analysis that the immune system can contribute to the clinical
considered 169 consecutive patients who had relapsed after benefits of ASCT. For example, patients with early lymphoid
an ASCT and underwent HLA-typing immediately after re- recovery after ASCT have superior long-term outcomes.55
lapse. The 2-year PFS was higher in the allo-HCT group (with
donors) at 42% compared with 18% in the no donor group Donor Lymphocyte Infusion
(p = .0001) with similar OS, although TRM was 22% versus Donor lymphocyte infusions have been used upon relapse
1% in the allo-HCT and ASCT groups, respectively.53 In another after allo-HCT and even to preempt relapse, but exacer-
prospective phase II multicenter EBMT trial, 49 patients who bation of GVHD is a major risk. Disease control is gener-
had relapsed after a previous ASCT received allo-HCT from ally superior in patients in whom GVHD develops.38

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 217


KRISHNAN ET AL

Donor lymphocyte infusions have been combined with along with lenalidomide.66 Haploidentical allo-HCT followed
IMIDs or bortezomib.56 Preemptive donor lymphocyte in- by planned NK-cell infusion attempts to use donor-recipient
fusions at a defined time period has been used to enhance KIR ligand mismatch and NK-cell reactivity to facilitate long-
reconstitution of donor T cells and antitumor immunity.57 term remission (NCT02100891).
Emergence of WT1-specific cytotoxic T lymphocytes (WT1-
CTL) has been correlated with better PFS after allo-HCT. Immune Checkpoint Inhibitors
Infusion of donor-derived T cells directed against specific Immune responses against multiple myelomaspecific an-
myeloma antigens such as WT1 or cancer testis antigens58,59 tigens are minimally protective, although detectable in
is another promising area of adoptive T-cell therapy using patients. Inhibitors of PD-1 and PD-L1 interaction are be-
the allo-HCT platform. ing studied as a means of breaking down multiple mye-
loma immune tolerance. In patients with multiple myeloma,
Myeloma Infiltrating T Lymphocytes PD-1 expression was upregulated on T cells concomitant
Marrow lymphocytes in patients with multiple myeloma are with increased PD-L1 expression on plasma cells.67,68 The
enriched for T cells with myeloma-specific antigen specificity. antiPD-1 agent nivolumab as monotherapy was un-
Noonan et al60 described adoptive transfer of such autolo- impressive,69 but promising response rates were observed in
gous marrow-derived, ex vivo activated and expanded T cells combination with lenalidomide, even for patients whose
on day 3 after ASCT.60 They demonstrated measurable disease was refractory.70 The antiPD-1 agent pem-
myeloma-specific activity for the ex vivo expanded product brolizumab is being studied in the lymphocyte recovery
and persistence of myeloma-specific immunity even at 1 year. phase after ASCT (NCT02331368) in combination with
lenalidomide.
CAR T-Cell Therapy
CAR T-cell therapy involves transducing activated T cells with Vaccines
genes encoding T-cell receptors specific to the antigen of The vaccine approach holds great promise for patients with
interest. Although multiple myeloma is not a classic CD19- multiple myeloma. Patients who received a patient-specific
positive malignancy, deep sustained response to anti-CD19 dendritic cell/myeloma fusion vaccine demonstrated the
CAR T cells in conjunction with second salvage ASCT for expansion of multiple myelomaspecific T cells, as well as
patients with relapsed/refractory multiple myeloma was upgrading of response in a subgroup of patients.71 This
recently reported.61 Several promising antigenic targets concept has been expanded to a intergroup randomized
have been identified for the development of anti-multiple phase II trial (BMT CTN 1401) with ASCT followed by lena-
myeloma CARs (40) such as Bcell maturation antigen lidomide maintenance with or without vaccination using the
(BCMA), CD138, kappa light chains, and CS-1. Notably, allogeneic dendritic cell/myeloma fusion vaccine.
CD19-directed CAR T cells (derived from donor lymphocytes) The direct manipulation of T cells by increasing T-cell
have induced remissions without induction of GVHD in pa- number, as well as engineering the T cells for augmented
tients who relapsed after allo-HCT.61,62 Thus, the allo-HCT anti-multiple myeloma affinity was studied in a phase I/II
or ASCT platform could be adapted to subsequent CAR T trial. In this study, autologous T cells were transduced with a
technology. lentiviral vector that encoded the affinity-enhanced NY-ESO
T-cell receptor. Patients received an infusion of these T cells
Natural KillerCell Therapy after ASCT, which was shown to lead to long-term en-
Natural killer cells have innate cytotoxicity against multiple graftment, infiltration of marrow, and trafficking to other
myeloma cells, while multiple myeloma exhibits specific tumor sites.
immune-evasive strategies to circumvent and attenuate NK-
cell function.63 Modulation of NK activity using anti-KIR Ab CONCLUSION
IPH2101 (monoclonal antibody against inhibitory KIR on NK The role of transplant-based approaches as a key step in
cells) is being explored as a means to establish multiple inducing long-term remissions for patients with multiple
myelomaspecific immunity.64 Autologous-, allogeneic-, and myeloma continues to evolve. The next decade of studies will
cord-derived NK cells have been found to be safe and ef- likely establish personalized post-transplant maintenance
ficacious for multiple myeloma.65 In a phase I study, up to strategies designed to achieve the trifecta of MRD negativity,
1 3 108 NK cells/kg freshly expanded cord blood NK cells restoration of multiple myelomaspecific immunity, and
were found to be safe when given before high-dose melphan freedom from multiple myeloma clonal evolution.

References
1. Kumar SK, Rajkumar SV, Dispenzieri A, et al. Improved survival in 2. Krishnan A, Pasquini MC, Logan B, et al; Blood Marrow Transplant
multiple myeloma and the impact of novel therapies. Blood. 2008;111: Clinical Trials Network (BMT CTN). Autologous haemopoietic stem-cell
2516-2520. transplantation followed by allogeneic or autologous haemopoietic

218 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


BEYOND AUTOLOGOUS TRANSPLANTATION IN MULTIPLE MYELOMA

stem-cell transplantation in patients with multiple myeloma (BMT CTN Intergroupe Francophone Du Myelome (IFM/DFCI 2009 Trial). Blood.
0102): a phase 3 biological assignment trial. Lancet Oncol. 2011;12: 2015;126:391-391.
1195-1203. 20. Sonneveld P, Asselbergs E, Zweegman S, et al. Phase 2 study of carfilzomib,
3. Gahrton G, Tura S, Ljungman P, et al; European Group for Bone Marrow thalidomide, and dexamethasone as induction/consolidation therapy for
Transplantation. Allogeneic bone marrow transplantation in multiple newly diagnosed multiple myeloma. Blood. 2015;125:449-456.
myeloma. N Engl J Med. 1991;325:1267-1273. 21. Jakubowiak A, Griffith K, Jasielec J, et al. Post-transplant carfilzomib
4. Tricot G, Vesole DH, Jagannath S, et al. Graft-versus-myeloma effect: (Kyprolis), lenalidomide (Revlimid), and dexamethasone (KRd) consolida-
proof of principle. Blood. 1996;87:1196-1198. tion in newly diagnosed multiple myeloma (NDMM): efficacy and toler-
5. Harousseau JL, Attal M, Avet-Loiseau H. The role of complete response ability of extended treatment. Haematologica. 2015;100:1-800.
in multiple myeloma. Blood. 2009;114:3139-3146. 22. Ladetto M, Pagliano G, Ferrero S, et al. Major tumor shrinking and
6. San-Miguel JF, Mateos MV. Can multiple myeloma become a curable persistent molecular remissions after consolidation with bortezomib,
disease? Haematologica. 2011;1246-1248. thalidomide, and dexamethasone in patients with autografted mye-
7. Zangari M, van Rhee F, Anaissie E, et al. Eight-year median survival in loma. J Clin Oncol. 2010;28:2077-2084.
multiple myeloma after total therapy 2: roles of thalidomide and 23. Bjorkstrand B, Svensson H, Goldschmidt H, et al. Alpha-interferon
consolidation chemotherapy in the context of total therapy 1. Br J maintenance treatment is associated with improved survival after
Haematol. 2008;141:433-444. high-dose treatment and autologous stem cell transplantation in pa-
8. Vesole DH, Barlogie B, Jagannath S, et al. High-dose therapy for re- tients with multiple myeloma: a retrospective registry study from the
fractory multiple myeloma: improved prognosis with better supportive European Group for Blood and Marrow Transplantation (EBMT). Bone
care and double transplants. Blood. 1994;84:950-956. Marrow Transplant. 2001;27:511-515.
9. Barlogie B, Jagannath S, Desikan KR, et al. Total therapy with tandem 24. Attal M, Harousseau JL, Leyvraz S, et al; Inter-Groupe Francophone du
transplants for newly diagnosed multiple myeloma. Blood.1999;93: Myelome (IFM). Maintenance therapy with thalidomide improves
55-65. survival in patients with multiple myeloma. Blood. 2006;108:3289-3294.
10. Barlogie B, Tricot G, Rasmussen E, et al. Total therapy 2 without tha- 25. Morgan GJGW, Gregory WM, Davies FE, et al; National Cancer Research
lidomide in comparison with total therapy 1: role of intensified in- Institute Haematological Oncology Clinical Studies Group. The role of
duction and posttransplantation consolidation therapies. Blood. 2006; maintenance thalidomide therapy in multiple myeloma: MRC Myeloma
107:2633-2638. IX results and meta-analysis. Blood. 2012;119:7-15.
11. Attal M, Lauwers-Cances V, Marit G, et al; IFM Investigators. Lenali- 26. Holstein SA, Owzar K, Richardson PG, et al. Updated analysis of CALGB/
domide maintenance after stem-cell transplantation for multiple my- ECOG/BMT CTN 100104: lenalidomide maintenance therapy after
eloma. N Engl J Med. 2012;366:1782-1791. single autologous stem cell transplant (ASCT) for multiple myeloma.
12. McCarthy PL, Owzar K, Hofmeister CC, et al. Lenalidomide after stem- J Clin Oncol. 2015;33: (suppl; abstr 8523).
cell transplantation for multiple myeloma. N Engl J Med. 2012;366: 27. Palumbo A, Cavallo F, Gay F, et al. Autologous transplantation and main-
1770-1781. tenance therapy in multiple myeloma. N Engl J Med. 2014;371:895-905.
13. Uy GL, Goyal SD, Fisher NM, et al. Bortezomib administered pre-auto- 28. Avet-Loiseau H, Corre J, Lauwers-Cances V, et al. Evaluation of minimal
SCT and as maintenance therapy post transplant for multiple myeloma: residual disease (MRD) By next generation sequencing (NGS) is highly
a single institution phase II study. Bone Marrow Transplant. 2009;43: predictive of progression free survival in the IFM/DFCI 2009 trial. Blood.
793-800. 2015;126:191-191.
14. Mellqvist UH, Gimsing P, Hjertner O, et al; Nordic Myeloma Study 29. Sonneveld P, Schmidt-Wolf IG, van der Holt B, et al. Bortezomib in-
Group. Bortezomib consolidation after autologous stem cell trans- duction and maintenance treatment in patients with newly diagnosed
plantation in multiple myeloma: a Nordic Myeloma Study Group ran- multiple myeloma: results of the randomized phase III HOVON-65/
domized phase 3 trial. Blood. 2013;121:4647-4654. GMMG-HD4 trial. J Clin Oncol. 2012;30:2946-2955.
15. Cavo M, Pantani L, Petrucci MT, et al; GIMEMA (Gruppo Italiano 30. Neben K, Lokhorst HM, Jauch A, et al. Administration of bortezomib
Malattie Ematologiche dellAdulto) Italian Myeloma Network. before and after autologous stem cell transplantation improves out-
Bortezomib-thalidomide-dexamethasone is superior to thalidomide- come in multiple myeloma patients with deletion 17p. Blood. 2012;119:
dexamethasone as consolidation therapy after autologous hemato- 940-948.
poietic stem cell transplantation in patients with newly diagnosed 31. Rosinol L OA, Teruel A. Maintenance therapy after stem cell trans-
multiple myeloma. Blood. 2012;120:9-19. plantation for multiple myeloma with bortezomib/thalidomide vs
16. Roussel M, Lauwers-Cances V, Robillard N, et al. Front-line trans- thalidomide vs alfa 2 b interferon: final results of a phase 3 Pethema/
plantation program with lenalidomide, bortezomib, and dexamethasone GEM randomized trial. Blood. 2012;120: (abstr 334).
combination as induction and consolidation followed by lenalidomide 32. Shah JJ, Feng L, Weber D, et al. Phase II study of the combination of
maintenance in patients with multiple myeloma: a phase II study by ixazomib with lenalidomide as maintenance therapy following autol-
the Intergroupe Francophone du Mye lome. J Clin Oncol. 2014;32: ogous stem cell transplant in patients with multiple myeloma. Blood.
2712-2717. 2015;126:3155-3155.
17. Leleu X, Fouquet G, Hebraud B, et al; Intergroupe Francophone du 33. Lokhorst HM, Plesner T, Laubach JP, et al. Targeting CD38 with dar-
Myelome (IFM). Consolidation with VTd significantly improves the atumumab monotherapy in multiple myeloma. N Engl J Med. 2015;373:
complete remission rate and time to progression following VTd in- 1207-1219.
duction and single autologous stem cell transplantation in multiple 34. Barlogie B, Kyle RA, Anderson KC, et al. Standard chemotherapy
myeloma. Leukemia. 2013;27:2242-2244. compared with high-dose chemoradiotherapy for multiple myeloma:
18. Nooka AK, Kaufman JL, Muppidi S, et al. Consolidation and maintenance final results of phase III US Intergroup Trial S9321. J Clin Oncol. 2006;24:
therapy with lenalidomide, bortezomib and dexamethasone (RVD) in 929-936.
high-risk myeloma patients. Leukemia. 2014;28:690-693. 35. Giralt S, Aleman A, Anagnostopoulos A, et al. Fludarabine/melphalan
19. Attal M, Lauwers-Cances V, Hulin C, et al. Autologous transplantation conditioning for allogeneic transplantation in patients with multiple
for multiple myeloma in the era of new drugs: a phase III study of the myeloma. Bone Marrow Transplant. 2002;30:367-373.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 219


KRISHNAN ET AL

36. Kroger N, Sayer HG, Schwerdtfeger R, et al. Unrelated stem cell study based on donor availability. Biol Blood Marrow Transplant. 2012;18:
transplantation in multiple myeloma after a reduced-intensity condi- 617-626.
tioning with pretransplantation antithymocyte globulin is highly ef- 54. Kroger N, Schwerdtfeger R, Kiehl M, et al. Autologous stem cell
fective with low transplantation-related mortality. Blood. 2002;100: transplantation followed by a dose-reduced allograft induces high
3919-3924. complete remission rate in multiple myeloma. Blood. 2002;100:
37. Kumar S, Zhang MJ, Li P, et al. Trends in allogeneic stem cell trans- 755-760.
plantation for multiple myeloma: a CIBMTR analysis. Blood. 2011;118: 55. Porrata LF, Gertz MA, Inwards DJ, et al. Early lymphocyte recovery
1979-1988. predicts superior survival after autologous hematopoietic stem cell
38. Lokhorst HM, Wu K, Verdonck LF, et al. The occurrence of graft-versus-host transplantation in multiple myeloma or non-Hodgkin lymphoma. Blood.
disease is the major predictive factor for response to donor lymphocyte 2001;98:579-585.
infusions in multiple myeloma. Blood. 2004;103:4362-4364. 56. Kroger N, Badbaran A, Lioznov M, et al. Post-transplant immunotherapy
39. Bruno B, Rotta M, Patriarca F, et al. A comparison of allografting with with donor-lymphocyte infusion and novel agents to upgrade partial
autografting for newly diagnosed myeloma. N Engl J Med. 2007;356: into complete and molecular remission in allografted patients with
1110-1120. multiple myeloma. Exp Hematol. 2009;37:791-798.
40. Bjorkstrand B, Iacobelli S, Hegenbart U, et al. Tandem autologous/ 57. Bellucci R, Alyea EP, Weller E, et al. Immunologic effects of prophylactic
reduced-intensity conditioning allogeneic stem-cell transplantation donor lymphocyte infusion after allogeneic marrow transplantation for
versus autologous transplantation in myeloma: long-term follow-up. multiple myeloma. Blood. 2002;99:4610-4617.
J Clin Oncol. 2011;29:3016-3022. 58. de Carvalho F, Alves VL, Braga WM, et al. MAGE-C1/CT7 and MAGE-C2/
41. Giaccone L, Storer B, Patriarca F, et al. Long-term follow-up of a CT10 are frequently expressed in multiple myeloma and can be ex-
comparison of nonmyeloablative allografting with autografting for plored in combined immunotherapy for this malignancy. Cancer
newly diagnosed myeloma. Blood. 2011;117:6721-6727. Immunol Immunother. 2013;62:191-195.
42. Gahrton G, Iacobelli S, Bjorkstrand B, et al; EBMT Chronic Malignancies 59. Tyler EM, Jungbluth AA, OReilly RJ, et al. WT1-specific T-cell responses
Working Party Plasma Cell Disorders Subcommittee. Autologous/ in high-risk multiple myeloma patients undergoing allogeneic T cell-
reduced-intensity allogeneic stem cell transplantation vs autologous depleted hematopoietic stem cell transplantation and donor lymphocyte
transplantation in multiple myeloma: long-term results of the EBMT- infusions. Blood. 2013;121:308-317.
NMAM2000 study. Blood. 2013;121:5055-5063. 60. Noonan KA, Huff CA, Davis J, et al. Adoptive transfer of activated
43. Kneppers E, van der Holt B, Kersten MJ, et al. Lenalidomide mainte- marrow-infiltrating lymphocytes induces measurable antitumor im-
nance after nonmyeloablative allogeneic stem cell transplantation in munity in the bone marrow in multiple myeloma. Sci Transl Med. 2015;
multiple myeloma is not feasible: results of the HOVON 76 Trial. Blood. 7:288ra78.
2011;118:2413-2419. 61. Garfall AL, Stadtmauer EA, June CH. Chimeric antigen receptor T cells in
44. Alsina M, Becker PS, Zhong X, et al. Lenalidomide maintenance for high- myeloma. N Engl J Med. 2016;374:194.
risk multiple myeloma after allogeneic hematopoietic cell trans- 62. Garfall AL, Fraietta JA, Maus MV. Immunotherapy with chimeric antigen
plantation. Biol Blood Marrow Transplant. 2014;20:1183-1189. receptors for multiple myeloma. Discov Med. 2014;17:37-46.
45. Chng WJ, Dispenzieri A, Chim CS, et al; International Myeloma Working 63. Childs RW, Carlsten M. Therapeutic approaches to enhance natural
Group. IMWG consensus on risk stratification in multiple myeloma. killer cell cytotoxicity against cancer: the force awakens. Nat Rev Drug
Leukemia. 2014;28:269-277. Discov. 2015;14:487-498.
46. Mikhael JR, Dingli D, Roy V, et al; Mayo Clinic. Management of newly 64. Benson DM Jr, Hofmeister CC, Padmanabhan S, et al. A phase 1 trial of
diagnosed symptomatic multiple myeloma: updated Mayo Stratifica- the anti-KIR antibody IPH2101 in patients with relapsed/refractory
tion of Myeloma and Risk-Adapted Therapy (mSMART) consensus multiple myeloma. Blood. 2012;120:4324-4333.
guidelines 2013. Mayo Clin Proc. 2013;88:360-376. 65. Szmania S, Lapteva N, Garg T, et al. Ex vivo-expanded natural killer cells
47. Palumbo A, Avet-Loiseau H, Oliva S, et al. Revised International Staging demonstrate robust proliferation in vivo in high-risk relapsed multiple
System for Multiple Myeloma: a report from International Myeloma myeloma patients. J Immunother. 2015;38:24-36.
Working Group. J Clin Oncol. 2015;33:2863-2869. 66. Shah N, Li L, Kaur I, et al. Infusion of ex vivo expanded allogeneic cord
48. Kumar S, Mahmood ST, Lacy MQ, et al. Impact of early relapse after blood-derived natural killer cells in combination with autologous stem
auto-SCT for multiple myeloma. Bone Marrow Transplant. 2008;42: cell transplantation for multiple myeloma: results of a phase I study.
413-420. Blood. 2015;126:929-929.
49. Knop S LP, Hebart H, et al. Autologous followed by allogeneic versus 67. Benson DM Jr, Bakan CE, Mishra A, et al. The PD-1/PD-L1 axis modulates
tandem-autologous stem cell transplant in newly diagnosed FISH- the natural killer cell versus multiple myeloma effect: a therapeutic
del13q myeloma. Blood. 2014;124:43. target for CT-011, a novel monoclonal anti-PD-1 antibody. Blood. 2010;
50. Freytes CO, Vesole DH, LeRademacher J, et al. Second transplants for 116:2286-2294.
multiple myeloma relapsing after a previous autotransplant-reduced- 68. Rosenblatt J, Glotzbecker B, Mills H, et al. PD-1 blockade by CT-011, anti-
intensity allogeneic vs autologous transplantation. Bone Marrow PD-1 antibody, enhances ex vivo T-cell responses to autologous den-
Transplant. 2014;49:416-421. dritic cell/myeloma fusion vaccine. J Immunother. 2011;34:409-418.
51. de Lavallade H, El-Cheikh J, Faucher C, et al. Reduced-intensity con- 69. Lesokhin AM, Ansell SM, Armand P, et al. Preliminary results of a phase I
ditioning allogeneic SCT as salvage treatment for relapsed multiple study of nivolumab(BMS-936558) in patients with relapsed or re-
myeloma. Bone Marrow Transplant. 2008;41:953-960. fractory lymphoid malignancies. Paper presented at: 56th ASH Annual
52. Michallet M, Sobh M, El-Cheikh J, et al. Evolving strategies with Meeting and Exposition; December 2014; San Francisco, CA.
immunomodulating drugs and tandem autologous/allogeneic hema- 70. San Miguel J, Mateos M-V, Shah JJ, et al. Pembrolizumab in combination
topoietic stem cell transplantation in first line high risk multiple my- with lenalidomide and low-dose dexamethasone for relapsed/refractory
eloma patients. Exp Hematol. 2013;41:1008-1015. multiple myeloma (RRMM): Keynote-023. Blood. 2015;126:505-505.
53. Patriarca F, Einsele H, Spina F, et al. Allogeneic stem cell transplantation 71. Rosenblatt J, Avivi I, Vasir B, et al. Vaccination with dendritic cell/tumor
in multiple myeloma relapsed after autograft: a multicenter retrospective fusions following autologous stem cell transplant induces immunologic

220 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


BEYOND AUTOLOGOUS TRANSPLANTATION IN MULTIPLE MYELOMA

and clinical responses in multiple myeloma patients. Clin Cancer Res. myeloma patients with metaphase cytogenetic abnormalities. Blood.
2013;19:3640-3648. 2008;112:3115-3121.
72. Cavo M, Pantani L, Petrucci MT, et al; GIMEMA (Gruppo Italiano Malattie 75. Spencer A PH, Roberts AW, et al. Consolidation therapy with low dose
Ematologiche dellAdulto) Italian Myeloma Network. Bortezomib- thalidomide and prednisolone prolong survival of multiple myeloma
thalidomide-dexamethasone is superior to thalidomide-dexamethasone patients undergoing a single autologous stem cell transplantation
as consolidation therapy after autologous hematopoietic stem cell procedure. J Clin Oncol. 2009;27:6.
transplantation in patients with newly diagnosed multiple myeloma. 76. Lokhorst HM, van der Holt B, Zweegman S, et al. A randomized phase
Blood. 2012;120:9-19. 3 study on the effect of thalidomide combined with adriamycin,
73. Roussel M, Lauwers-Cances V, Robillard N, et al. Front-line trans- dexamethasone and high dose melphalan, followed by thalidomide
plantation program with lenalidomide, bortezomib, and dexamethasone maintenance in patients with multiple myeloma. Blood. 2010;115:
combination as induction and consolidation followed by lenalidomide 1113-1120.
maintenance in patients with multiple myeloma: a phase II study by 77. Stewart AK, Trudel S, Bahlis NJ, et al. A randomized phase 3 trial of
the Intergroupe Francophone du Myelome. J Clin Oncol. 2014;32: thalidomide and prednisone as maintenance therapy after ASCT in
2712-2717. patients with MM with a quality-of-life assessment: the National Cancer
74. Barlogie B, Pineda-Roman M, van Rhee F, et al. Thalidomide arm of Total Institute of Canada Clinicals Trials Group Myeloma 10 Trial. Blood. 2013;
Therapy 2 improves complete remission duration and survival in 121:1517-1523.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 221


HEMATOLOGIC MALIGNANCIESPLASMA CELL
DYSCRASIA

Defining and Redefining Myeloma

CHAIR
S. Vincent Rajkumar, MD
Mayo Clinic
Rochester, MN

SPEAKERS
Evangelos Terpos, MD, PhD
National and Kapodistrian University of Athens
Athens, Greece

Irene M. Ghobrial, MD
Dana-Farber Cancer Institute
Boston, MA
GHOBRIAL ET AL

Future Directions in the Evaluation and Treatment of


Precursor Plasma Cell Disorders
Salomon Manier, MD, Karma Z. Salem, MD, David Liu, MD, and Irene M. Ghobrial, MD

OVERVIEW

Multiple myeloma (MM) is an incurable disease that progresses from a premalignant stage termed monoclonal gammopathy
of undetermined significance (MGUS) and an intermediate stage of smoldering multiple myeloma (SMM). Recent major
advances in therapy with more effective and less toxic treatments have brought reconsideration of early therapeutic
intervention in management of SMM, with the goal of reducing progression of the disease before the occurrence of end-
organ damage to MM and improving survival. Key to this effort is accurate identification of patients at high risk of pro-
gression who would truly benefit from early intervention. In this review, we discuss the current definitions, risk factors, risk
stratification, prognosis, and management of MGUS and SMM, as well as new emerging therapeutic options under active
investigation.

M ultiple myeloma evolves through a spectrum of dis-


ease from a premalignant stage of MGUS to an inter-
mediate stage of SMM and finally presents with symptoms
as MM based on the updated diagnostic criteria. However,
it should be noted that the revised diagnostic criteria for
MM upstages only a small proportion of patients with
and signs of end-organ damage that lead to the diagnosis of SMM. Thus, SMM as an entity continue to be relevant and
MM.1 Recent studies have indicated that almost all cases of important. Therefore, there is a need to better define the
MM are preceded by the precursor state of MGUS or SMM.2,3 clinical and molecular characteristics of patients with
Over the years, the diagnosis of MM required evidence of MGUS and SMM that predict progression to MM. Here,
end-organ damage attributable to the neoplastic clone of we review the diagnostic criteria and risks of progres-
plasma cells, the so-called CRAB criteria identified by hy- sion for MGUS and SMM and delineate potential therape-
percalcemia, renal failure, anemia, and osteolytic bone utic options that can be used for patients with high-risk
lesions.4 This definition was intended to be conservative to SMM.
avoid unnecessary and toxic administration of chemotherapy
to patients with asymptomatic disease. However, with major
advances in therapy, and identification of potential biomar- DEFINITION, INCIDENCE, AND RISK OF
kers that can distinguish MM from premalignant stages, it PROGRESSION OF MGUS
became necessary to revise the disease definition of MM.5-7 MGUS is a premalignant precursor of MM.9,10 It is defined
In 2014, the International Myeloma Working Group (IMWG) by a serum M-protein concentration lower than 3 g/dL and/or
updated the diagnostic criteria for MM to add three markers an abnormal FLC ratio (, 0.26 or . 1.65), less than 10%
that can identify additional patients not yet meeting CRAB clonal plasma cells in the BM, and absence of MDEs or
criteria who should nevertheless be treated. These were evidence of Waldenstrom macroglobulinemia or amyloid-
termed myeloma-defining events (MDEs) and constituted osis (Table 1).8 When first clinically recognized, MGUS has
the presence of clonal bone marrow (BM) plasma cells likely been present in an undetected state for a median
greater than or equal to 60%, serum free light chain (FLC) duration of more than 10 years.11
ratio greater than or equal to 100, provided the involved MGUS is present in over 3% to 4% of the population older
FLC level is higher than or equal to 100 mg/L, or more than than age 5010 and increases with age to 8.9% in people older
one focal lesion on MRI or CT and PET-CT.8 than age 85.1 Therefore, one of the most important risk
Importantly, the revised definition excluded patients factors for MGUS is age.1,12 In addition to an accumulation of
previously considered to have SMM with ultra-high risk cases, the age-related increase in prevalence of MGUS is
of progression (80% within 2 years) who are now classified related to a true increase in incidence with age.

From the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Irene M. Ghobrial, MD, Dana-Farber Cancer Institute, 450 Brookline Ave., Boston, MA 02115; email: Irene_ghobrial@dfci.harvard.edu.

2016 by American Society of Clinical Oncology.

e400 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


DIAGNOSIS AND MANAGEMENT OF MGUS AND SMOLDERING MYELOMA

Apart from age, there are several factors that are asso- TABLE 1. IMWG Diagnostic Criteria for MGUS, SMM,
ciated with a higher risk of developing MGUS, including and MDEs
African American race and a familial history of myeloma.
Other factors such as the presence of autoimmune disorders IMWG Criteria, 2014 Version8
are less well delineated. Persons of African and African MGUS Serum M protein , 3 g/dL and/or abnormal FLC ratio
American descent have a threefold increased prevalence (, 0.26 or . 1.65) with increased level of appropriate
involved light chain
even after adjusting for socioeconomic and other risk fac-
Clonal BM plasma cells , 10%
tors, suggesting a genetic predisposition among this pop-
ulation.13 An increased prevalence of MGUS in Africans Absence of MDEs or amyloidosis
relative to Caucasians has also been reported in Ghana.14,15 SMM Serum M protein . 3 g/dL
Two studies have shown that the risk of MM and MGUS AND/OR clonal BM plasma cells . 10% and , 60%
is increased threefold in relatives of individuals with AND/OR urinary monoclonal protein . 500 mg per 24
MGUS.16,17 In addition, an increased risk of developing non- hours
Hodgkin lymphoma and chronic lymphoid leukemia was also Absence of MDEs or amyloidosis
observed in this group. Collectively, these data are consis- MDEs Evidence of end-organ damage (CRAB criteria):
tent with an inherited twofold to fourfold genetic suscep- Hypercalcemia: serum Ca2+ . 0.25 mmol/L (. 1 mg/dL)
tibility to MM.18 The genetic basis of inherited MM above upper limit of normal or . 2.75 mmol/L
(. 11 mg/dL)
susceptibility is incompletely understood. However, recent
genome-wide association studies (GWAS) identified seven Renal insufficiency: serum creatinine . 173 mmol/L
(. 2 mg/dL)
loci contributing to inherited genetic susceptibility to
Anemia: hemoglobin value of . 2 g/dL below the lower
MM.18-21 These seven loci were also associated with an limit of normal or a hemoglobin value , 10 g/dL
increased risk of developing MGUS.22 Another GWAS study
Bone lesions: one or more lytic lesion(s) on skeletal
in the Nordic region identified a novel MM risk locus at radiography, CT, or PET-CT
ELL2.23 Clonal BM plasma cell percentage $ 60%
There are three subtypes of MGUS: nonimmunoglobulin Involved/uninvolved serum FLC ratio $ 100
M (IgM) MGUS, IgM MGUS, and light-chain MGUS. Non-IgM
More than one focal lesions on MRI
MGUS carries a risk of progression to MM or solitary
Abbreviations: IMWG, International Myeloma Working Group; MGUS,
plasmacytoma, whereas IgM MGUS is associated with a risk monoclonalgammopathy of undetermined significance; SMM, smoldering multiple
of progression to Waldenstrom macroglobulinemia. Light- myeloma; MM, multiple myeloma; MDEs, myeloma-defining events; FLC, serum free
light chain; BM, bone marrow.
chain MGUS is a newly discovered entity that is associated
with a risk of progression to the light-chain type of MM. All
forms of MGUS can progress to Ig light-chain amyloidosis. ratio, the size of the M protein, and the type of M protein
MGUS is associated with a lifelong risk of transformation (Table 2). Patients with serum M protein , 1.5 g/dL, IgG
to MM or a related malignancy at a rate of 1% per year.9 In class, and normal FLC ratio are considered low-risk MGUS
some patients, however, the risk may be as high as 58% in and have only a 2% lifetime risk of progression. In general,
20 years. The size and subtype of the M protein at diagnosis of patients with MGUS must be reassessed in 6 months, and
MGUS and an abnormal serum FLC ratio are well-established if stable yearly thereafter. A number of lifestyle and envi-
prognostic factors for progression.24 As such, MGUS can be ronmental risk factors have been proposed to increase the
risk stratified using three simple variables: the serum FLC risk of progression from MGUS to MM, including obesity,
immune dysfunction, and agricultural, chemical, and radia-
tion exposure. Of these, obesity is probably the most con-
sistently reported association.25-27 Studies are ongoing to
KEY POINTS determine the mechanism of progression of MGUS and to
explore preventive strategies.
Multiple myeloma is always preceded by precursor In addition to the risk of progression, MGUS can also be
conditions including MGUS and smoldering myeloma. associated with several other clinical problems including
The rate of progression of MGUS is at 1% per year, but sensorimotor peripheral neuropathy (MGUS neuropathy),
the rate of progression of SMM is 10% per year, with a membranoproliferative glomerulonephritis, lichen myx-
high rate of progression of 50% at 2 years for those with edematosus (papular mucinosis, scleromyxedema), pyo-
high-risk features.
derma gangrenosum, or necrobiotic xanthogranuloma.
Patients with high-risk SMM should be offered clinical
trials to prevent progression or carefully observed to
avoid end-organ damage.
Patients with MGUS and SMM have different risk DEFINITION, INCIDENCE, AND RISK OF
stratifications and rates of progression. PROGRESSION OF SMM
Some patients may benefit from early therapeutic SMM is a heterogeneous disease entity that includes pa-
interventions, specifically in high-risk SMM. tients with higher disease burden than in MGUS but remain
asymptomatic.28 The term SMM was first described by Kyle

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e401


GHOBRIAL ET AL

TABLE 2. Risk of Progression of Monoclonal Gammopathy of Undetermined Significance to Myeloma or Related


Disorders

Cumulative Absolute Risk of


Relative Risk of Cumulative Absolute Risk of Progression at 20 Years Accounting
Risk Group Progression Progression at 20 Years (%)* for Death as a Competing Risk (%)**
Low Risk: Serum M Protein 1 5 2
< 1.5 g/dL, IgG Subtype,
Normal Free Light Chain Ratio
(0.261.65)
Low-Intermediate Risk: Any One 5.4 21 10
Factor Abnormal
High-Intermediate Risk: Any Two 10.1 37 18
Factors Abnormal
High Risk: All Three Factors 20.8 58 27
Abnormal
Adapted from Rajkumar et al56 with permission from the publisher.
*Estimates in this column represent the risk of progression assuming that patients do not die of other causes during this period.
**Estimates in this column represent the risk of progression calculated by using a model that accounts for the fact that patients can die of unrelated causes during this time.
Abbreviation: IgG, immunoglobulin G.

and Greipp in 198029 and was followed by many other The update to the disease definition of MM automatically
descriptions terming it indolent MM,30 or Durie Salmon resulted in revised diagnostic criteria for SMM as it excludes
stage I.31 In 2003, the IMWG defined SMM as having a serum patients with ultra-highrisk SMM who are now considered
M-protein level higher than or equal to 3 g/L and/or greater as having overt MM (Table 1).8 Despite that, it remains a
than or equal to 10% monoclonal plasma cells in the BM major clinical dilemma with an overall risk of progression of
(Table 3).1,4 Although the incidence and prevalence of SMM 10% per year for the first 5 years, 3% per year for the next
in the population is not well defined, it has been estimated to 5 years, and 1% per year for the last 10 years. This suggests
represent approximately 8% to 20% of patients within the that the current definition of SMM is highly biologically
MM spectrum.28 and clinically heterogeneous.32 Indeed, SMM represents a

TABLE 3. Risk Stratification of Patients with Smoldering Myeloma Based on Mayo Clinic and Spanish Criteria

Model Risk Factors No. of Risk Factors 5-Year Progression (%) Relative Risk
Mayo Clinic Model M protein $ 3 g/dL 1 25 1
$ 10% BM plasma cells 2 51 2.0
FLC ratio , 0.125 or . 8 3 76 3.0
Total 51 NA
Spanish (PETHEMA) Model $ 95% aPC 0 4 1
Immunoparesis 1 46 11.5
2 72 18
Total 46 NA
Proposed New Criteria for $ 10% BM plasma cells Serum M protein $ 30 g/L
High-Risk SMM
With one of the criteria in the IgA SMM
right column
Immunoparesis with reduction of two uninvolved Ig isotypes
Serum involved/uninvolved FLC ratio $ 8 (but , 100)
Progressive increase in M-protein level (evolving type of SMM)
BM clonal plasma cells 50% to 60%
aPC immunophenotype ($ 95% of BM plasma cells are clonal)
and reduction of one or more uninvolved Ig isotypes
t(4;14) or del 17p or 1q gain
Increased circulating plasma cells
MRI with diffuse abnormalities or one focal lesion
PET-CT with focal lesion with increased uptake without underlying osteolytic bone
destruction
Abbreviations: BM, bone marrow; FLC, serum free light chain; NA, not applicable, aPC, abnormal plasma cells; IgA, immunoglobulin A; SMM, smoldering multiple myeloma.

e402 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


DIAGNOSIS AND MANAGEMENT OF MGUS AND SMOLDERING MYELOMA

clinical entity where a subset of patients have a very indolent genetically aberrant as MM and does not appear to be
course of disease that mimics an MGUS-like state, whereas associated with a particular chromosomal imbalance but
others have a more aggressive course of disease that has rather with an expansion of altered clones that are already
been described as early myeloma or CRAB-negative present in MGUS.43
myeloma. Unfortunately, there are currently no molecular A study of transcriptional profiling using gene expression
factors to differentiate these two clinically and biologically profiling has identified signatures that can identify patients
distinct groups of patients. For this reason, additional studies with MGUS versus SMM versus MM.44 However, this study
are required to identify markers of progression within these has major limitations given that the percentage of plasma
patients. cells present in MGUS cases is low and the samples are
There are three subtypes of SMM: IgA, IgG, and light chain. inherently contaminated with normal nontransformed plasma
The median time to progression (TTP) for IgA and IgG SMM cells.
is 27 and 75 months, respectively.32,33 Light-chain SMM Based on all these factors, a new classification for high-risk
has a cumulative probability of progression to active MM SMM has been proposed by Rajkumar et al to identify pa-
or light-chain amyloidosis of 27.8% at 5 years, 44.6% at tients at high risk of progression to MM (25% per year) as
10 years, and 56.5% at 15 years.34 The current factors as- defined by the criteria listed in Table 3.32,33,35,38,39,45-49
sociated with risk of progression are mainly based on the Although these biomarkers were defined before the re-
level of tumor burden in these patients assessed by the visions to the diagnostic criteria, because the proportion of
degree of tumor involvement in the BM and the quantifi- patients with SMM affected and upstaged as a result of the
cation of monoclonal protein in the peripheral blood. new criteria are small, the effect on the estimates for
The two most widely used risk stratification methods progression would likely be minimal. Identification of high-
are the Mayo Clinic32 and the PETHEMA Spanish group risk SMM is of particular importance because these patients
classifications.35 The Mayo Clinic criteria are primarily based are at considerable risk of end-organ damage and are
on the levels of serum protein markers (serum protein candidates for clinical trials and/or intervention.7 In con-
electrophoresis and FLC assay) and the percentage of plasma trast, patients with SMM without high-risk factors likely
cells in the BM.32,33 The risk stratification of the PETHEMA have a risk of progression of 5% per year or less.
Study Group, on the other hand, focused on the use of
multiparameter flow cytometry of the BM to quantify the
ratio of abnormal, neoplastic plasma cells to normal plasma MANAGEMENT OF MGUS AND SMM
cells and reduction of uninvolved Igs.35 Interestingly, a head- Patients with MGUS are not offered therapeutic options to
to-head comparison between the PETHEMA and the Mayo date and close observation is indicated in these patients. A
Clinic risk models showed notable discordance reflected in recent study demonstrated that prior knowledge of MGUS
many patients being classified as high risk in one model and and close monitoring had significantly better overall survival
low risk in the other model.36 Other risk factors that have (median survival, 2.8 years) than patients with MM without
been examined include the role of IgA (vs. IgG) isotype, the prior knowledge of MGUS (median survival, 2.1 years).50 In
presence of proteinuria, circulating plasma cells, a high fact, low M-protein concentration (, 0.5 g/dL) at MGUS
proliferative rate of BM plasma cells, and abnormal MRI diagnosis was associated with poorer MM survival, pre-
findings.12,28,37 sumably because those patients were not closely followed
SMM can also be subclassified based on underlying cy- for disease progression.50
togenetic abnormalities.38,39 Patients with t(4;14), 1q gain, All patients suspected to have SMM need an MRI of the
and/or del17p are considered high-risk SMM (median TTP of spine and pelvis (or ideally whole-body MRI) or whole-body
approximately 24 months). Patients with hyperdiploidy are CT or PET-CT.1,8,51 BM examination with FISH studies and
considered intermediate-risk (median TTP of approximately multiparametric flow cytometry are also required. The
34 months). Other cytogenetic abnormalities including t(11;14) standard of care for SMM remains observation with re-
are considered standard risk (median TTP, 54 months). evaluations of the patients every 34 months.1,8,49 In low-
Patients with SMM who have no evidence of cytogenetic risk patients, follow-up can be reduced to once every
abnormalities on fluorescence in situ hybridization (FISH) 6 months after the first 5 years. Imaging studies must be
studies are considered low risk (median TTP, 101 months). repeated if changes in clinical features or M protein occur.
An international workshop assembled to review cytoge- For patients at high risk, follow-up should continue in-
netic studies to evaluate whether MGUS and SMM cases definitely, and should include periodic imaging studies to
have the same detectable anomalies that are often found in rule out asymptomatic progression. Patients with SMM can
MM.40 Point mutations in NRAS and KRAS, MYC upregulation,41 be initiated on therapy without waiting for CRAB features to
and gain or loss of chromosome 1q or 1p seem to correlate appear if follow-up testing shows the development of other
with disease progression from MGUS and SMM.42 A pro- MDE or early detection of MM bone disease on the basis of
gressive increase in the incidence of copy number ab- advanced imaging studies. Patients with a baseline MRI
normalities from MGUS to SMM and to MM has also been showing diffuse infiltration, solitary focal lesion, or equivocal
recently observed.43 Although MM has more copy num- lesions, need follow-up examinations in 36 months to rule
ber abnormalities than its precursor states, MGUS is as out progression.8

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e403


GHOBRIAL ET AL

Investigators attempted to assess whether early thera- and a superior 3-year overall survival (94% vs. 80%; p = .03)
peutic intervention can lead to substantial improvement in from the time of registration. However, this study was
survival and response.28 There are two major goals in early criticized by many groups because of how asymptomatic
therapeutic intervention: the first is prevention of pro- biochemical progression was handled in both arms, the short
gression, and the second is definitive therapy to achieve overall survival of the abstention group and the use of
complete remission with the hope that all subclones will be salvage therapy in the abstention group. Because of these
eradicated at this early disease state and cure can be concerns, additional studies are needed before imple-
achieved.37,52 menting therapeutic interventions as a standard of care in
The major barrier to early intervention has been defining patients with high-risk SMM.
the group of patients who would truly benefit from this early
treatment as they would have otherwise progressed to
symptomatic disease. Indeed, if SMM is a heterogeneous Investigational Therapies
mix of patients with early myeloma and MGUS-like mye- More intensive treatment approaches similar to MM are
loma, then identification of those with early MM will allow also being investigated in high-risk SMM. These include
for intervention only in those patients who truly warrant triplet therapy with carfilzomib, lenalidomide, and dexa-
therapy. methasone, as well as combining combination therapy,
autologous stem cell transplantation, and maintenance.55
Immune-based approaches using elotuzumab or darat-
Bisphophonates
umumab or vaccine therapies are also being investigated in
Bisphosphonates have shown promise in reducing the risk of
patients with high-risk SMM.
skeletal-related events (SREs) in SMM, but have not been
On the basis of available data, we recommend that pa-
shown to delay progression to MM or prolong survival. In
tients with high-risk SMM be offered clinical trials testing
a randomized trial of pamidronate (once monthly for
early intervention or observed closely. However, given the
12 months) versus observation, the incidence of SRE was
high risk of progression, select patients with high-risk SMM
39% versus 73%, respectively (p = .009).53 In another trial of
with multiple risk factors or evidence of biologic progression
zoledronic acid (monthly for 12 months) versus observation,
(rising M-protein level) can be considered for MM therapy.49
the incidence of SRE was 56% versus 78%, respectively
There are no specific factors to make this determination, and
(p = .041).54 The reduction in SREs is an important endpoint,
clinical judgment is needed.
and based on these two trials, consideration must be given
to bisphosphonates. We recommend once-yearly bisphospho-
nate in patients at low risk, and once every 34 months in select FUTURE DIRECTIONS
patients with high-risk SMM. Further research is needed to identify factors involved in
progression of MGUS and SMM to MM. We must identify
Lenalidomide additional reliable biomarkers of malignancy. Finally, we
The most critical study of therapy in SMM that has reignited need a portfolio of clinical trials designed to determine
interest in therapeutic intervention in this patient pop- whether early intervention can improve survival or provide a
ulation came from the PETHEMA group using lenalidomide path to prevent or cure MM. The question remains whether
and dexamethasone compared with observation. Mateos these efforts will lead to a cure in myeloma or potentially to
et al7 reported on 119 patients with high-risk SMM who an early screening and intervention modality that will com-
received either observation or lenalidomide and dexa- pletely eradicate the progression to symptomatic disease,
methasone in an open-label randomized trial. Patients making myeloma a preventable disease. Only well-designed
treated with lenalidomide and dexamethasone had a su- clinical trials will be able to determine which interventions
perior 3-year survival without progression to symptomatic are most effective, what populations should be targeted,
disease (progression-free survival; 77% vs. 30%; p , .001) and when interventions should take place.

References
1. Kyle RA, Durie BG, Rajkumar SV, et al; International Myeloma Working 3. Weiss BM, Abadie J, Verma P, et al. A monoclonal gammopathy pre-
Group. Monoclonal gammopathy of undetermined significance (MGUS) cedes multiple myeloma in most patients. Blood. 2009;113:5418-5422.
and smoldering (asymptomatic) multiple myeloma: IMWG consensus 4. International Myeloma Working Group. Criteria for the classification of
perspectives risk factors for progression and guidelines for monitoring monoclonal gammopathies, multiple myeloma and related disorders:
and management. Leukemia. 2010;24:1121-1127. a report of the International Myeloma Working Group. Br J Haematol.
2. Landgren O, Kyle RA, Pfeiffer RM, et al. Monoclonal gammopathy of 2003;121:749-757.
undetermined significance (MGUS) consistently precedes multiple 5. Rajkumar SV, Larson D, Kyle RA. Diagnosis of smoldering multiple
myeloma: a prospective study. Blood. 2009;113:5412-5417. myeloma. N Engl J Med. 2011;365:474-475.

e404 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


DIAGNOSIS AND MANAGEMENT OF MGUS AND SMOLDERING MYELOMA

6. Larsen JT, Kumar SK, Dispenzieri A, et al. Serum free light chain ratio as a 25. Thompson MA, Kyle RA, Melton LJ III, et al. Effect of statins, smoking and
biomarker for high-risk smoldering multiple myeloma. Leukemia. 2013; obesity on progression of monoclonal gammopathy of undetermined
27:941-946. significance: a case-control study. Haematologica. 2004;89:626-628.
7. Mateos M-V, Hernandez MT, Giraldo P, et al. Lenalidomide plus 26. Kyle RA, Rajkumar SV. Epidemiology of the plasma-cell disorders. Best
dexamethasone for high-risk smoldering multiple myeloma. N Engl J Pract Res Clin Haematol. 2007;20:637-664.
Med. 2013;369:438-447. 27. Landgren O, Rajkumar SV, Pfeiffer RM, et al. Obesity is associated with
8. Rajkumar SV, Dimopoulos MA, Palumbo A, et al. International Myeloma an increased risk of monoclonal gammopathy of undetermined sig-
Working Group updated criteria for the diagnosis of multiple myeloma. nificance among black and white women. Blood. 2010;116:1056-1059.
Lancet Oncol. 2014;15:e538-e548. 28. Dispenzieri A, Stewart AK, Chanan-Khan A, et al. Smoldering multiple
9. Kyle RA, Therneau TM, Rajkumar SV, et al. A long-term study of myeloma requiring treatment: time for a new definition? Blood. 2013;
prognosis in monoclonal gammopathy of undetermined significance. 122:4172-4181.
N Engl J Med. 2002;346:564-569. 29. Kyle RA, Greipp PR. Smoldering multiple myeloma. N Engl J Med. 1980;
10. Kyle RA, Therneau TM, Rajkumar SV, et al. Prevalence of monoclonal 302:1347-1349.
gammopathy of undetermined significance. N Engl J Med. 2006;354: 30. Alexanian R. Localized and indolent myeloma. Blood. 1980;56:521-525.
1362-1369. 31. Durie BG, Salmon SE. A clinical staging system for multiple myeloma.
11. Therneau TM, Kyle RA, Melton LJ III, et al. Incidence of monoclonal Correlation of measured myeloma cell mass with presenting clinical
gammopathy of undetermined significance and estimation of duration features, response to treatment, and survival. Cancer. 1975;36:
before first clinical recognition. Mayo Clin Proc. 2012;87:1071-1079. 842-854.
12. Agarwal A, Ghobrial IM. Monoclonal gammopathy of undetermined 32. Kyle RA, Remstein ED, Therneau TM, et al. Clinical course and prognosis
significance and smoldering multiple myeloma: a review of the current of smoldering (asymptomatic) multiple myeloma. N Engl J Med. 2007;
understanding of epidemiology, biology, risk stratification, and man- 356:2582-2590.
agement of myeloma precursor disease. Clin Cancer Res. 2013;19:985- 33. Dispenzieri A, Kyle RA, Katzmann JA, et al. Immunoglobulin free light
994. chain ratio is an independent risk factor for progression of smoldering
13. Greenberg AJ, Vachon CM, Rajkumar SV. Disparities in the prevalence, (asymptomatic) multiple myeloma. Blood. 2008;111:785-789.
pathogenesis and progression of monoclonal gammopathy of un- 34. Kyle RA, Larson DR, Therneau TM, et al. Clinical course of light-chain
determined significance and multiple myeloma between blacks and smouldering multiple myeloma (idiopathic Bence Jones proteinuria):
whites. Leukemia. 2012;26:609-614. a retrospective cohort study. Lancet Haematol. 2014;111:e28-e36.
14. Buadi F, Hsing AW, Katzmann JA, et al. High prevalence of polyclonal 35. Perez-Persona E, Vidriales MB, Mateo G, et al. New criteria to identify
hypergamma-globulinemia in adult males in Ghana, Africa. Am J He- risk of progression in monoclonal gammopathy of uncertain significance
matol. 2011;86:554-558. and smoldering multiple myeloma based on multiparameter flow
15. Landgren O, Katzmann JA, Hsing AW, et al. Prevalence of monoclonal cytometry analysis of bone marrow plasma cells. Blood. 2007;110:
gammopathy of undetermined significance among men in Ghana. Mayo 2586-2592.
Clin Proc. 2007;82:1468-1473. 36. Cherry BM, Korde N, Kwok M, et al. Modeling progression risk for
16. Landgren O, Kristinsson SY, Goldin LR, et al. Risk of plasma cell and smoldering multiple myeloma: results from a prospective clinical study.
lymphoproliferative disorders among 14621 first-degree relatives of Leuk Lymphoma. 2013;54:2215-2218.
4458 patients with monoclonal gammopathy of undetermined signif- 37. Landgren O. Monoclonal gammopathy of undetermined significance
icance in Sweden. Blood. 2009;114:791-795. and smoldering multiple myeloma: biological insights and early
17. Vachon CM, Kyle RA, Therneau TM, et al. Increased risk of monoclonal treatment strategies. Hematology Am Soc Hematol Educ Program.
gammopathy in first-degree relatives of patients with multiple mye- 2013;2013:478-487.
loma or monoclonal gammopathy of undetermined significance. Blood. 38. Rajkumar SV, Gupta V, Fonseca R, et al. Impact of primary molecular
2009;114:785-790. cytogenetic abnormalities and risk of progression in smoldering mul-
18. Morgan GJ, Johnson DC, Weinhold N, et al. Inherited genetic suscep- tiple myeloma. Leukemia. 2013;27:1738-1744.
tibility to multiple myeloma. Leukemia. 2014;28:518-524. 39. Neben K, Jauch A, Hielscher T, et al. Progression in smoldering myeloma
19. Chubb D, Weinhold N, Broderick P, et al. Common variation at 3q26.2, is independently determined by the chromosomal abnormalities del
6p21.33, 17p11.2 and 22q13.1 influences multiple myeloma risk. Nat (17p), t(4;14), gain 1q, hyperdiploidy, and tumor load. J Clin Oncol. 2013;
Genet. 2013;45:1221-1225. 31:4325-4332.
20. Weinhold N, Johnson DC, Chubb D, et al. The CCND1 c.870G.A 40. Fonseca R, Barlogie B, Bataille R, et al. Genetics and cytogenetics of
polymorphism is a risk factor for t(11;14)(q13;q32) multiple myeloma. multiple myeloma: a workshop report. Cancer Res. 2004;64:1546-1558.
Nat Genet. 2013;45:522-525. 41. Chng WJ, Huang GF, Chung TH, et al. Clinical and biological implications
21. Broderick P, Chubb D, Johnson DC, et al. Common variation at 3p22.1 of MYC activation: a common difference between MGUS and newly
and 7p15.3 influences multiple myeloma risk. Nat Genet. 2012;44: diagnosed multiple myeloma. Leukemia. 2011;25:1026-1035.
58-61. 42. Chiecchio L, Dagrada GP, Protheroe RK, et al; UK Myeloma Forum. Loss
22. Weinhold N, Johnson DC, Rawstron AC, et al. Inherited genetic sus- of 1p and rearrangement of MYC are associated with progression of
ceptibility to monoclonal gammopathy of unknown significance. Blood. smouldering myeloma to myeloma: sequential analysis of a single case.
2014;123:2513-2517, quiz 2593. Haematologica. 2009;94:1024-1028.
23. Swaminathan B, Thorleifsson G, Joud M, et al. Variants in ELL2 influ- 43. Lopez-Corral
L, Sarasquete ME, Be`a S, et al. SNP-based mapping arrays
encing immunoglobulin levels associate with multiple myeloma. Nat reveal high genomic complexity in monoclonal gammopathies, from
Commun. 2015;6:7213. MGUS to myeloma status. Leukemia. 2012;26:2521-2529.
24. Rajkumar SV, Kyle RA, Therneau TM, et al. Serum free light chain ratio is 44. Zhan F, Barlogie B, Arzoumanian V, et al. Gene-expression signature of
an independent risk factor for progression in monoclonal gammopathy benign monoclonal gammopathy evident in multiple myeloma is linked
of undetermined significance. Blood. 2005;106:812-817. to good prognosis. Blood. 2007;109:1692-1700.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e405


GHOBRIAL ET AL

45. Rosin~ol L, Blade J, Esteve J, et al. Smoldering multiple myeloma: natural 52. Ghobrial IM, Landgren O. How I treat smoldering multiple myeloma.
history and recognition of an evolving type. Br J Haematol. 2003;123: Blood. 2014;124:3380-3388.
631-636. 53. DArena G, Gobbi PG, Broglia C, et al; Gimema (Gruppo Italiano Malattie
46. Dhodapkar MV, Sexton R, Waheed S, et al. Clinical, genomic, and Ematologiche DellAdulto); Multiple Myeloma Working Party; Gisl
imaging predictors of myeloma progression from asymptomatic (Gruppo Italiano Studio Linfomi) Cooperative Group. Pamidronate
monoclonal gammopathies (SWOG S0120). Blood. 2014;123:78-85. versus observation in asymptomatic myeloma: final results with long-
47. Bianchi G, Kyle RA, Larson DR, et al. High levels of peripheral blood term follow-up of a randomized study. Leuk Lymphoma. 2011;52:
circulating plasma cells as a specific risk factor for progression of 771-775.
smoldering multiple myeloma. Leukemia. 2013;27:680-685. 54. Musto P, Petrucci MT, Bringhen S, et al; GIMEMA (Italian Group for
48. Hillengass J, Fechtner K, Weber MA, et al. Prognostic significance of Adult Hematologic Diseases)/Multiple Myeloma Working Party and the
focal lesions in whole-body magnetic resonance imaging in patients Italian Myeloma Network. A multicenter, randomized clinical trial
with asymptomatic multiple myeloma. J Clin Oncol. 2010;28:1606-1610. comparing zoledronic acid versus observation in patients with
49. Rajkumar SV, Landgren O, Mateos MV. Smoldering multiple myeloma. asymptomatic myeloma. Cancer. 2008;113:1588-1595.
Blood. 2015;125:3069-3075. 55. Zingone A, et al. Phase II clinical and correlative study of carfilzomib,
50. Sigurdardottir EE, Turesson I, Lund SH, et al. The role of diagnosis and lenalidomide, and dexamethasone followed by lenalidomide extended
clinical follow-up of monoclonal gammopathy of undetermined signifi- dosing (CRD-R) induces high rates of MRD negativity in newly diagnosed
cance on survival in multiple myeloma. JAMA Oncol. 2015;1:168-174. multiple myeloma (MM) patients. Blood. 2013;122:538-538.
51. Blade J, Dimopoulos M, Rosin~ol L, et al. Smoldering (asymptomatic) 56. Rajkumar SV, Kyle RA, Therneau TM, et al. Serum free light chain ratio is
multiple myeloma: current diagnostic criteria, new predictors of out- an independent risk factor for progression in monoclonal gammopathy
come, and follow-up recommendations. J Clin Oncol. 2010;28:690-697. of undetermined significance (MGUS). Blood. 2005;106:812-817.

e406 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


IMAGING FOR MULTIPLE MYELOMA

The Role of Imaging in the Treatment of Patients With


Multiple Myeloma in 2016
Evangelos Terpos, MD, PhD, Meletios A. Dimopoulos, MD, and Lia A. Moulopoulos, MD

OVERVIEW

The novel criteria for the diagnosis of symptomatic multiple myeloma have revealed the value of modern imaging for the
management of patients with myeloma. Whole-body low-dose CT (LDCT) has increased sensitivity over conventional ra-
diography for the detection of osteolytic lesions, and several myeloma organizations and institutions have suggested that
whole-body LDCT should replace conventional radiography for the work-up of patients with myeloma. MRI is the best
imaging method for the depiction of marrow infiltration by myeloma cells. Whole-body MRI (or at least MRI of the spine and
pelvis if whole-body MRI is not available) should be performed for all patients with smoldering multiple myeloma with no
lytic lesions to look for occult disease, which may justify treatment. In addition, MRI accurately illustrates the presence of
plasmacytomas, spinal cord, and/or nerve compression for surgical intervention or radiation therapy; it is also recom-
mended for the work-up of solitary bone plasmacytoma, and it may distinguish malignant from benign fractures (which is
very important in cases of patients in biochemical remission with no other signs of progression). Diffusion weighted imaging
(DWI) seems to improve MRI diagnosis in patients with myeloma. PET/CT is a functional imaging technique, more sensitive
than conventional radiography for the detection of lytic lesions, which probably allows better definition of complete
response and minimal residual disease compared with all other imaging methods. PET/CT has shown the best results in the
follow-up of patients with myeloma and has an independent prognostic value both at diagnosis and following treatment.
PET/CT can also be used for the work-up of solitary bone plasmacytoma and nonsecretory myeloma.

L ytic bone disease is a major feature of multiple myeloma:


70% to 80% of patients have osteolytic lesions at diagnosis,
and up to 90% develop lytic lesions during the course of their
during the course of the disease. The skeletal survey (whole
body x-rays) at diagnosis should include plain radiographs of
the whole skeleton, as previously described.3 Osteolyses
disease.1 The novel criteria for the diagnosis of symptomatic have the typical appearance of punched-out lesions with
multiple myeloma have revealed the value of modern imaging the absence of reactive sclerosis (Fig. 1) and are more
for the management of patients with multiple myeloma, as common in the vertebrae, ribs, skull, and pelvis.4 Although
they include (1) the presence at least one lytic lesion detected the whole-body x-ray was the standard of care for many
not only by conventional radiography but also by CT, whole- years, it has several limitations: (1) for a lytic lesion to be-
body LDCT, or PET/CT, and (2) the presence of one or more come apparent, more than 30% loss of trabecular bone must
focal bone marrow lesions on MRI studies.2 Furthermore, novel occur; (2) it is difficult to assess certain areas, such as the
imaging techniques, such as MRI and PET/CT, provide prog- pelvis and the spine; (3) there are limitations to the de-
nostic information and have been recently proven of value, for tection of lytic lesion response to anti-myeloma therapy
the better definition of response to anti-myeloma therapy. because of delayed evidence of healing; (4) specificity is
reduced for the differential diagnosis of myeloma-related
IS CONVENTIONAL RADIOGRAPHY THE GOLD fracture and benign fracture (very important, particularly in
STANDARD FOR THE DIAGNOSIS OF BONE cases of new vertebral compression fractures in the absence
DISEASE IN PATIENTS WITH MYELOMA IN 2016? of other criteria of relapse); (5) it is dependent on the ob-
The Role of Whole-Body LDCT server (there is very low reproducibility among centers;
Conventional radiography has been widely used for the higher number of osteolytic lesions detected in academic
identification of osteolytic lesions both at diagnosis and centers compared with nonacademic centers); and (6)

From the School of Medicine, National and Kapodistrian University of Athens, Alexandra General Hospital, Athens, Greece; School of Medicine, National and Kapodistrian University of
Athens, Areteion Hospital, Athens, Greece.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Evangelos Terpos, MD, PhD, Department of Clinical Therapeutics, University of Athens School of Medicine, Alexandra General Hospital, 80 Vas. Sofias Ave.,
11528, Athens, Greece; email: eterpos@med.uoa.gr.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e407


TERPOS, DIMOPOULOS, AND MOULOPOULOS

studies are long and often not tolerable for patients in severe et al).5-11 Major disadvantages of whole-body LDCT include
pain.3,4 Thus, the development of novel imaging methods increased length of time required for radiologists to report
has led to the substitution of whole-body x-ray by more their findings, lack of availability in several centers,2,5 and lack
advanced techniques, such as the whole-body LDCT in many of specificity for the differential diagnosis between malignant
European centers or by PET/CT in the United States. and osteoporotic fractures, despite improvements during
Whole-body LDCT was introduced to allow the detection of recent years.12 Furthermore, although exposure to radiation
osteolytic lesions in the whole skeleton with high accuracy, no is much lower compared with standard CT, it continues to be
need for contrast agents and low radiation dose compared higher than whole-body x-ray: mean dose of whole-body
with standard CT (two- to threefold lower radiation dose LDCT is approximately 3.6 and 2.8 mSv for females and
versus conventional CT).5,6 In several studies, whole-body LDCT males, respectively, compared with 1.2 mSv for whole-body
was found to be superior to whole-body x-ray for the detection x-ray.13 Nevertheless, the higher diagnostic accuracy of
of osteolytic lesions (Table 1).5,7-11 In one of the largest studies whole-body LDCT and patient comfort, which is particularly
staging patients with myeloma, 61% of patients with normal important for older patients, renders the dose/quality ratio
whole-body x-ray results had more than one osteolytic lesions favorable for whole-body LDCT. For these reasons, the Eu-
on whole-body LDCT.10 According to the new criteria for ropean Myeloma Network has suggested that whole-body
symptomatic myeloma, these patients should receive therapy. LDCT replace conventional radiography as the standard im-
In the same study, the total number of lesions detected by aging technique for evaluation of bone disease in multiple
whole-body LDCT was 968 compared with 248 for whole-body myeloma, where available.14 Figure 2 shows images from a
x-ray (p , .001), with the only limitation of this study being its whole-body LDCT study of one of our patients with myeloma.
retrospective origin.10 In a more recent prospective study,
which included 52 patients with myeloma at diagnosis, whole- The Value of MRI in Myeloma: Advantages and
body LDCT revealed osteolysis in 12 patients (23%) with Limitations
negative whole-body x-ray, and it proved to be more sensitive MRI detection of bone involvement in myeloma: comparison
than whole-body x-ray mainly in the axial skeleton (p , .001). with conventional radiography. MRI has been estab-
Whole-body LDCT was superior in detecting lesions in patients lished as a valuable technique for the diagnosis of bone
with osteopenia and osteoporosis.11 involvement in multiple myeloma. MRI detects bone mar-
In total, whole-body LDCT advantages over whole-body row infiltration by myeloma cells; whole-body x-ray and CT
x-ray include (1) superior diagnostic sensitivity for de- detect osseous destruction. Conventional MRI protocols for
piction of osteolytic lesions, especially in areas where the multiple myeloma include T1-weighted, T2-weighted with
whole-body x-ray detection rate is low (i.e., pelvis and spine); fat suppression, in- and opposed-phase imaging, and
(2) superiority in estimating fracture risk and bone instability; contrast-enhanced T1-weighted sequences. Five MRI pat-
(3) duration of the examination, which is 5 minutes or less, an terns of marrow involvement have been recognized in
important issue for patients in extreme pain; (4) production of multiple myeloma: (1) a focal pattern that consists of lo-
higher-quality 3D high resolution images for planning biopsies calized areas of myeloma cell infiltration 5 mm or greater in
and therapeutic interventions; and (5) demonstration of diameter (present in 30%50% of patients with multiple
unsuspected manifestations of myeloma or other disease, myeloma); (2) a diffuse pattern characterized by almost
especially in the lungs and kidneys (33% in the study by Wolf complete replacement of normal marrow by myeloma cells
(present in 25%40% of patients with multiple myeloma);
(3) a combined diffuse and focal pattern (present in 10% of
patients with multiple myeloma); (4) a normal bone marrow
KEY POINTS pattern (present in 15%25% of patients with multiple
myeloma); and (5) a variegated or salt and pepper pattern
Whole-body LDCT is superior to conventional with innumerable small bone marrow focal lesions (present
radiography for the detection of osteolytic lesions, and it in 1%5% of patients with multiple myeloma; Fig. 3).3,4,15
is suggested to replace it in the work-up of patients with Several studies have shown that MRI is more sensitive than
myeloma. whole-body x-ray for the detection of bone involvement in
MRI is the best imaging method for the depiction of multiple myeloma.16-19 The largest included 611 patients
marrow infiltration by myeloma cells. who were treated with total therapy protocols in Arkansas.
Whole-body MRI (or at least MRI of the spine and pelvis MRI detected focal lesions in 74% of the patients, while
if whole-body MRI is not available) should be performed whole-body x-ray detected osteolytic lesions in 56% of them.
for all patients with smoldering multiple myeloma with
MRI was superior to whole-body x-ray particularly in
no lytic lesions to look for occult disease, which may
justify treatment.
detecting bone involvement.16 A meta-analysis, which
PET/CT allows better definition of complete response compared the detection rate of different imaging tech-
and minimal residual disease. niques, confirmed the superiority of MRI over whole-body
PET/CT has an independent prognostic value both at x-ray, mainly in the axial skeleton.20
diagnosis and after treatment. Because of its high sensitivity in revealing bone marrow
involvement, MRI is now used for the discrimination

e408 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


IMAGING FOR MULTIPLE MYELOMA

FIGURE 1. (A) Characteristic Punched-Out Lesions in the Skull and (B) Large Osteolytic Lesions in the Left
Humerus

between asymptomatic/smoldering and symptomatic mul- In this point, it is important to note the differences between
tiple myeloma. Several studies have shown that approxi- the detection rates of whole-body MRI, which is not avail-
mately 40% to 50% of patients with normal whole-body x-ray able everywhere, and the detection rate of spine and pelvis
had abnormal findings on MRI examinations.17-19 Two MRI. In a study of 100 patients with multiple myeloma and
studies showed that patients with smoldering multiple monoclonal gammopathy of undetermined significance who
myeloma who had more than one focal lesion on MRI had a underwent whole-body MRI, 10% of patients presented with
median time to progression (TTP) to symptomatic disease focal lesions in the peripheral skeleton only; this is the
of 13 to 15 months and a 2-year probability of progression of number of patients who will not be diagnosed with lesions if
approximately 70%.21,22 In both studies, the presence of an MRI of the spine and pelvis is performed, instead of
more than one focal lesion on MRI was an independent whole-body MRI.23
adverse prognostic factor for progression to symptomatic
disease. Based on these studies, the International Myeloma MRI detection of bone involvement in myeloma:
Working Group (IMWG) included the use of whole-body MRI comparison with whole-body LDCT and PET/CT. In two
(or MRI of the spine and pelvis if whole-body MRI is not small studies, with 41 and 46 patients with newly diagnosed
available) in the work-up for smoldering multiple myeloma.2 multiple myeloma, respectively, whole-body MRI was able

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e409


TERPOS, DIMOPOULOS, AND MOULOPOULOS

TABLE 1. Major Studies Comparing Whole-Body LDCT and Conventional Radiography


Study Study Design No. of Patients Reference Test Key Findings
Gleeson et al9 P 39 WBXR, WB-MRI, bone marrow WBLDCT . WBXR, superior detection rate, comparable
biopsy with WB-MRI
Kropil et al8 P 29 WBXR WBLDCT . WBXR, ratio of detection of lytic lesion
WBLDCT/WBXR = 2.06 (4.60 for the spine; 7 for the
skull; 2.54 for the pelvis and 2.07 for the thoracic cage)
Princewill et al10 R 51 WBXR WBLDCT . WBXR; ratio of detection of lytic lesion
WBLDCT/WBXR = 3.9 (74 for the thoracic cage; 6.67 for
the pelvis and the fat bones; 4.92 for the spine; 2.31 for
the long bone and 1.09 for the skull); 61% of the
patients upstaged with WBLDCT
Wolf et al11 P 52 WBXR WBLDCT . WBXR, particularly in the axial skeleton. 63%
patients upstaged with WBLDCT, 23% patients nega-
tive at WBXR and positive at WBLDCT
Abbreviations: P, prospective; WBRX; whole-body x-ray (conventional skeletal survey); WB-MRI, whole-body MRI; WBLDCT, whole-body low-dose CT; R, retrospective.

to detect a higher number of patients with bone marrow disease.24,25 However, in the largest comparison to date of
involvement compared with whole-body LDCT or PET/CT. 303 patients with myeloma, there was no superiority of MRI
The superiority of MRI over PET/CT was mainly because of over PET/CT for the detection of focal lesions; though, a
the higher sensitivity of MRI for the detection of diffuse diffuse MRI pattern of bone marrow involvement was not

FIGURE 2. Whole-Body LDCT Images Showing Lytic Lesions in the Skull, Spine, and Pelvis, Bone Marrow Deposits in
the Femoral Cavity, and Vertebral Body Fractures

e410 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


IMAGING FOR MULTIPLE MYELOMA

FIGURE 3. MRI Patterns of Marrow Involvement in Myeloma

(A) Focal pattern: On T1-weighted images, focal lesions are darker than yellow marrow and slightly hypointense or isointense to intervertebral disc and muscle. (B) Diffuse
pattern: On T1-weighted images, the normal bone marrow signal is completely replaced by the abnormal process, and the intervertebral discs appear brighter or isointense
to the diseased marrow. (C) Variegated pattern: On T1-weighted images, the bone marrow is very inhomogeneous with innumerable small lesions.

included in this comparison.26 Finally, in a retrospective regimens, the combination of diffuse MRI pattern and high-
study of 210 MRI and 210 PET/CT studies of patients with risk cytogenetics and ISS-3, identified a group of patients
multiple myeloma, MRI achieved better results than PET/CT who experienced poor outcome: a median OS of 21 months
for the detection of myeloma at diagnosis and at progres- and a 35% probability for 3-year OS.29 Similarly, in a recent
sion; PET/CT, however, detected findings of response to anti- study of 161 patients with multiple myeloma who un-
myeloma therapy earlier than MRI.27 derwent an autologous transplantation (ASCT), the combi-
nation of a high-risk MRI score (defined as the presence of
Prognostic significance of MRI. MRI patterns have been moderate to severe diffuse infiltration, or the presence of
compared with myeloma features at presentation. More more than 25 focal lesions on whole-body MRI or more than
specifically, a diffuse MRI pattern correlated with high-risk seven focal lesions on axial MRI) and high-risk cytogenetics
cytogenetics, advanced disease stage (ISS-3), and increased identified a group of patients with median progression-free
angiogenesis.28-30 Patients with diffuse MRI pattern expe- survival (PFS) and OS of 23 and 56 months, respectively.33
rienced poorer overall survival (OS) compared with pa- The cut-off value of seven focal lesions on MRI was also used
tients with focal or normal patterns, both in the era of by the Arkansas group to identify patients with inferior OS
conventional chemotherapy or in the era of novel anti- (3-year OS probability: 73% vs. 86% for those with zero to
myeloma agents.29-32 More importantly, in a study that seven focal lesions).34 Despite the prognostic significance of
included 228 symptomatic patients who received first-line MRI, no treatment has been suggested to date for patients
therapy with bortezomib- or immunomodulatory drugbased with high-risk MRI features.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e411


TERPOS, DIMOPOULOS, AND MOULOPOULOS

The role of MRI in the follow-up of patients with of a patient with myeloma in remission with a new vertebral
myeloma. Although there are several studies evaluating the fracture and no other evidence of progression. MRI can offer
role of imaging at diagnosis, there are limited data for the important information and is able to differentiate myeloma
value of imaging during treatment and follow-up of patients from osteoporotic fractures in more than 90% of the cases.41
with myeloma. The IMWG suggests that there is no need to Furthermore, extramedullary plasmacytomas, present in up
repeat the skeletal survey in a patient who is responding to to 20% of patients during the course of their disease, are
treatment unless he develops bone symptoms and that easily seen on whole-body MRI.42 MRI is also recommended
other imaging studies (CT, MRI, and PET/CT) to detect soft for the work-up of patients with a solitary plasmacytoma of
tissue masses arising from bone lesions or extramedullary the bone. MRI may reveal unsuspected bone lesions in
disease may be indicated according to clinical circum- approximately one-third of such patients43; in these pa-
stances.35 We believe that these recommendations must tients, systemic treatment must be applied instead of ra-
be changed, as the physician cannot detect asymptomatic diation therapy, which is the treatment of choice for solitary
increases in the dimensions of pre-existing lytic lesions or the bone plasmacytoma.44 MRI accurately localizes spinal cord
presence of new lytic lesions or new plasmacytomas (all are and/or nerve root compression for surgical intervention or
criteria for defining disease progression36) without appro- radiation therapy.2,4,15 Finally, avascular necrosis of the
priate imaging. femoral head and cardiac amyloidosis may also be evaluated
Is there any role for MRI in this setting? And if yes, how with MRI.45,46
often must we repeat MRI after initial evaluation? Resolu- Limitations of MRI include prolonged acquisition time,
tion of previously detected focal lesions occurred in ap- availability issues, problems with claustrophobic patients
proximately 60% of patients who were treated with total and with patients carrying metal devices in their body, and
therapy protocols in Arkansas. When the disease pro- high cost.15
gressed, after the achievement of complete response (CR),
whole-body MRI revealed new focal lesions or enlargement Novel MRI techniques. DWI MRI, which derives its contrast
of previously detected focal lesions in 26% and 28% of mainly from differences in the diffusivity of water mole-
patients, respectively, while 15% of patients demonstrated cules in the tissue environment, is a novel and promising
both of these features at their MRI evaluation.16 In general, MRI technique. DWI MRI uses the calculation of apparent
MRI findings of response to therapy (CR or partial response diffusion coefficient values to better evaluate myeloma
[PR]) include resolution or decrease of the extent of focal burden and MRI infiltration patterns.47,48 Apparent diffu-
lesions or diffuse patterns in conventional chemotherapy or sion coefficient values are higher in patients with multiple
novel anti-myeloma agent era.37-39 In a small study with 33 myeloma at diagnosis, compared with patients in remission
patients who underwent an ASCT, MRI had 79% accuracy 20 weeks after initiation of treatment.49 DWI MRI was
and 86% specificity but only 64% sensitivity for the detection found superior to whole-body x-ray for the detection of
of remission because of false-positive results of nonviable bone involvement in 20 patients with relapsed/refractory
lesions.39 Thus, for better evaluation of remission, functional
imaging techniques (e.g., PET/CT or possibly DWI MRI) seem
to be more appropriate and will be discussed in the following FIGURE 4. MRI Differentiating a Vertebral Fracture
sections.
Regarding patients with smoldering multiple myeloma
with a negative MRI study at diagnosis, the timing of follow-
up with MRI and the significance of any findings on these
studies have not yet been established. In a single such study,
the Heidelberg group performed a second MRI 3 to 6 months
after the first in 63 patients with smoldering multiple my-
eloma. Approximately one-half of the patients showed
progression on MRI, while 40% developed symptomatic
disease, according to CRAB criteria (i.e., calcium, renal
failure, anemia, and bone disease).40 More data are needed
before recommending time intervals between the first and
subsequent MRIs for patients with smoldering multiple
myeloma.

Advantages and limitations of conventional MRI. The


major advantage of MRI over whole-body LDCT or con-
ventional CT is the discrimination between myelomatous
and normal marrow. This is extremely helpful in the di-
agnosis of the malignant or benign nature of a vertebral Left: From an underlying tumor. Right: From an osteoporotic fracture without
fracture (Fig. 4). Several times, clinicians face the condition signs of myeloma cell infiltration.

e412 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


IMAGING FOR MULTIPLE MYELOMA

multiple myeloma in all areas of the skeleton except the there is only one prospective study, which compared
skull, where both examinations had equal sensitivity.50 In PET/MRI with PET/CT in 30 patients with myeloma with both
another small study with 24 patients with myeloma (both techniques performed sequentially. There was high corre-
treated and untreated), DWI MRI was found more sensitive lation between the two techniques, regarding the number of
than 18F-fluorodeoxyglucose (FDG)-PET in detecting mye- active lesions and average standardized uptake value
loma lesions.51 In a recent study, 17 patients were eval- (SUV).59 Further studies with PET/MRI will reveal if there is
uated using DWI MRI and FDG-PET/CT, and the findings any value of this technique for patients with multiple
were compared with bone marrow biopsy data. In all myeloma.
studied regions, whole-body DWI scores were higher
compared with FDG-PET/CT. DWI MRI was particularly PET/CT and the Treatment of Patients With Myeloma
accurate in diagnosing diffuse disease (diffuse disease was PET/CT detection of bone involvement in myeloma. FDG-
observed in 37% of regions imaged on whole-body DWI PET/CT is a functional imaging method that combines
scans compared with only 7% on FDG-PET/CT); both demonstration of hypermetabolic activity in intramedullary
techniques were equally sensitive in the detection of focal and extramedullary sites (PET) with evidence of osteolysis
lesions.52 Preliminary reports suggest that DWI MRI may be (CT). Several studies have shown that PET/CT is more
used for the better definition of response to therapy, but sensitive compared with whole-body x-ray for the de-
this has to be confirmed in larger studies and in comparison tection of osteolytic lesions in multiple myeloma.25,60-62
with PET/CT results.48,53 This has been confirmed by the largest meta-analysis in
Dynamic contrast-enhanced (DCE) MRI evaluates the the field.20 The higher detection rate of PET/CT over
distribution of an intravenous contrast agent both inside and whole-body x-ray for the presence of osteolytic lesions is
outside the blood vessels over time. DCE MRI provides data especially important for patients with smoldering multi-
for the microcirculation of a specific area. In multiple my- ple myeloma. In one study with 120 patients with smol-
eloma, DCE MRI derives parameters correlated with mi- dering multiple myeloma based on the previous IMWG
crovascular density in the bone marrow both before and criteria,25 16% of patients with normal whole-body x-ray
after therapy,54-56 as well as with adverse disease features had positive PET/CT results. The median TTP for patients
and OS.57 Finally, the combination of DWI and DCE MRI with positive PET/CT results was 1.1 years compared with
produced a score with 76% agreement with the IMWG 4.5 for patients with negative PET/CT results, while the
criteria of response to therapy in 27 patients with myeloma probability of progression at 2 years for patients with
under treatment.58 More data are needed to evaluate such positive PET/CT results was 58%.63 The largest study in the
scores in larger studies. field involved 188 patients with suspected smoldering
PET/MRI represents a novel imaging modality with at- multiple myeloma examined with PET/CT. PET/CT was
tractive potential clinical applications. In multiple myeloma, positive in 39% of patients. The probability of progression

TABLE 2. Summary of the Novel IMPeTUs Criteria for the PET/CT Standardization for Multiple Myeloma

Lesion Type Site Number of Lesions (x) Grading


Diffuse Bone marrow* Deauville five-point scale
Focal Skull x =1 (no lesions) Deauville five-point scale
Spine x = 2 (1-3 lesions)
Extraspinal x = 3 (4-10 lesions)
x = 4 (. 10 lesions)
Lytic x = 1 (no lesions)
x = 2 (1-3 lesions)
x = 3 (4-10 lesions)
x = 4 (. 10 lesions
Fracture At least one
Paramedullary At least one
Extramedullary At least one Nodal**/extranodal Deauville five-point scale
*A if hypermetabolism in limbs and ribs.
**For nodal disease: C, cervical; SC, supraclavicular; M, mediastinal; Ax, axillary; Rp, retroperitoneal; Mes, mesenteric; In, inguinal.
For extranodal disease: Li, liver; Mus, muscle; Spl, spleen; Sk, skin; Oth, other.
Adapted by Nanni et al76 with permission.
Deauville five-point scale:
1 5 No uptake at all
2 # Mediastinal blood pool uptake (SUVmax)
3 . Mediastinal blood pool uptake, # liver uptake
4 . Liver uptake more than 10%
5 . Liver uptake (twice)

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e413


TERPOS, DIMOPOULOS, AND MOULOPOULOS

to symptomatic multiple myeloma within 2 years was 75% Value of PET/CT for better definition of CR to anti-myeloma
for patients with a positive PET/CT under observation therapy. Data obtained from PET/CT of 40 patients with
compared with only 30% for patients with a negative multiple myeloma, including average SUV and FDG kinetic
PET/CT. This probability was higher if hypermetabolic parameters K1, influx and fractal dimension correlated
activity was combined with underlying osteolysis (2-year significantly with percentage of bone marrow infiltration by
progression rate: 87%). The median TTP was 21 months plasma cells on trephine biopsies (PC %).65 Furthermore,
compared with 60 months for patients with a positive PET/CT efficiently detected extramedullary disease in pa-
PET/CT and those with a negative PET/CT, respectively.64 The tients both at diagnosis and at relapse.66 Consequently,
results of these two studies support the integration of PET/CT was tested for better definition of CR in 282 patients
changes in imaging requirements in the new IMWG di- with multiple myeloma. It was performed at diagnosis and
agnostic criteria for multiple myeloma; detection of osteolytic every 12 to 18 months afterward. At diagnosis, 42% of
lesions by PET/CT is a criterion for symptomatic multiple patients with multiple myeloma had more than three focal
myeloma.2 lesions; in 50% of these patients, SUVmax was more than 4.2.
Compared with MRI, as mentioned previously, PET/CT After treatment, PET/CT was negative for 70% of patients,
performs equally well in detecting focal lesions, but MRI while 53% of patients achieved CR according to IMWG
is better at detecting diffuse disease.24,25,62 criteria. Approximately 30% of patients at CR had a positive

TABLE 3. Advantages and Limitations of Different Imaging Techniques


Advantages Limitations
WBXR Cost Poor sensitivity/low detection rate
Availability Findings present only after advanced bone damage
Historical use/validation Patient discomfort with repositioning, multiple films; long image
acquisition time
No evaluation of bone marrow; no differentiation between
malignant and benign fractures
Lack of accurate visualization of specific areas, such as the pelvis
and the spine
Difficulties in the assessment of lytic lesions response to anti-
myeloma therapy
Observer dependency
WBLDCT Increased sensitivity and specificity for the detection of lytic Unclear prognostic significance of lesion number
lesions
3D structural information for CT-guided biopsy, surgery, and Relatively higher radiation exposure and more expensive than
radiotherapy planning WBXR
Can depict EMD, bone marrow involvement, and lytic lesions
Rapid acquisition time
Inexpensive compared with MRI and PET
Comfortable for patients
PET/CT Functional method High cost
Assesses disease activity before and after treatment; better Limited availability
definition of CR and MRD
Depicts extramedullary disease False-positive diagnoses because of infection/inflammation
Prognostic significance pre- and post-treatment Lack of standardization
Novel radioisotopes may offer additional disease-relevant Poor spatial resolution
information
MRI Is able to exclude smoldering/asymptomatic myeloma High cost
No radiation exposure Long acquisition time, claustrophobia
Assesses bone marrow both diffuse infiltration and focal May exclude patients with indwelling metal objects; contrast
bone marrow lesions contraindicated in severe renal insufficiency
Superior for detection of spinal cord compression and soft-
tissue masses
Number of focal lesions has prognostic significance
Detects extramedullary disease
3D structural information for CT-guided biopsy, surgery, and
radiotherapy planning
Abbreviations: WBXR, whole-body x-ray; WBLDCT, whole-body low-dose CT; EMD, extramedullary disease; CR, complete response; MRD, minimal residual disease.

e414 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


IMAGING FOR MULTIPLE MYELOMA

PET/CT. More importantly, PET/CT negativity was an in- normalization of PET/CT before maintenance could predict for
dependent predictor for prolonged PFS and OS in patients OS (30-month OS rate: 70% in patients with a positive PET/CT
with a CR. In addition, for patients with a CR, median PFS was vs. 94.6% in patients with negative PET/CT; p = .01).75
50 months for patients with a positive PET/CT and 90 months At this point, it is crucial to mention that one of the major
for patients with a negative PET/CT.67 PET/CT, therefore, limitations of PET/CT is the lack of standardization and the
provides more accurate definition of CR, and it has been controversies regarding SUV level of positivity. Recently, an
suggested that it should be incorporated to CR criteria.68 Italian panel of experts introduced novel criteria for the in-
terpretation of PET/CT images; these are summarized in
Prognostic significance of PET/CT. Several studies have Table 2.76 Large multicenter studies with prospective evalu-
confirmed the value of PET/CT as an independent factor for ation of these new criteria will reveal their clinical effect.
survival of patients with multiple myeloma both at di-
agnosis and after treatment.69-74 In 192 newly diagnosed Other PET/CT indications and limitations. PET/CT may be
patients who underwent ASCT, the presence of extra- used for the work-up of patients with solitary bone plas-
medullary disease and SUVmax greater than 4.2 on PET/CT macytoma at diagnosis.77 However, it is not clear whether
performed at diagnosis and the persistence of FDG uptake PET/CT or MRI is more suitable in this setting because
post-ASCT were independent variables adversely affecting restaging PET/CT after radiotherapy has a number of false-
PFS.69 In the largest study in the field, 429 patients who positive findings.78 PET/CT also has a role for patients with
were treated with total therapy protocols in Arkansas were nonsecretory or oligosecretory myeloma for the detection of
evaluated with both MRI and PET/CT at diagnosis and seven active lesions in the body.79 Major limitations of PET/CT in-
days post-ASCT. From the imaging variables, in the multivariate clude high cost, lack of availability in many centers and
analysis, only the detection of more than two osteolytic lesions countries, and false-positive results because of inflammation
by whole-body x-ray at diagnosis and the detection of more from other underlying pathology.
than three focal lesions by PET/CT 7 days post-ASCT were
independent prognostic factors for inferior OS. Limitation of
this study was the exclusion of the diffuse MRI pattern from the CONCLUSION
analysis.34 Despite this limitation, studies reported to date Whole-body LDCT seems to be most suitable for the detection
support the role of PET/CT after therapy, deeming it the best of osteolytic bone disease in patients with multiple myeloma
imaging technique for the follow-up of patients with myeloma. at diagnosis, replacing whole-body x-ray. Whole-body MRI (or
Indeed, in a recent study, 134 patients who were eligible for at least MRI of the spine and pelvis if whole-body MRI is not
treatment with ASCT were randomly assigned to eight cycles of available) should be performed for all patients with smol-
bortezomib-lenalidomide-dexamethasone (VRD) followed by dering multiple myeloma with no lytic lesions to look for
1-year maintenance with lenalidomide, or three cycles of VRD occult disease, which may justify treatment. PET/CT seems to
followed by ASCT plus two cycles of VRD consolidation and be inferior to MRI regarding the detection of marrow in-
1-year lenalidomide maintenance. PET/CT and whole-body volvement in multiple myeloma, but it is probably the best
MRI were performed after induction and before mainte- technique for optimal definition of CR and follow-up of pa-
nance. Both techniques were positive at diagnosis in more than tients with myeloma. Table 3 lists the advantages and limi-
90% of patients. After induction therapy and before mainte- tations of the most important imaging modalities. More
nance, more patients continued to have positive MRI than studies with novel imaging MRI techniques and the broader
PET/CT (93% vs. 55% and 83% vs. 21%, respectively), possibly use of PET/CT will define the role of these techniques in
because of earlier reduction of activity of PET/CT lesions. Both myeloma at diagnosis and follow-up, with the particular aim
after induction and before maintenance, normalization of to better define CR or minimal residual disease and to select
PET/CT and not of MRI could predict for PFS, while only the optimal window for their performance.

References

1. Terpos E, Berenson J, Raje N, et al. Management of bone disease in 5. Pianko MJ, Terpos E, Roodman GD, et al. Whole-body low-dose
multiple myeloma. Expert Rev Hematol. 2014;7:113-125. computed tomography and advanced imaging techniques for multi-
2. Rajkumar SV, Dimopoulos MA, Palumbo A, et al. International Myeloma ple myeloma bone disease. Clin Cancer Res. 2014;20:5888-5897.
Working Group updated criteria for the diagnosis of multiple myeloma. 6. Ippolito D, Besostri V, Bonaffini PA, et al. Diagnostic value of whole-
Lancet Oncol. 2014;15:e538-e548. body low-dose computed tomography (WBLDCT) in bone lesions
3. Dimopoulos M, Terpos E, Comenzo RL, et al. International myeloma detection in patients with multiple myeloma (MM). Eur J Radiol. 2013;82:
working group consensus statement and guidelines regarding the 2322-2327.
current role of imaging techniques in the diagnosis and monitoring of 7. Horger M, Claussen CD, Bross-Bach U, et al. Whole-body low-dose
multiple myeloma. Leukemia. 2009;23:1545-1556. multidetector row-CT in the diagnosis of multiple myeloma: an
4. Terpos E, Moulopoulos LA, Dimopoulos MA. Advances in imaging and the alternative to conventional radiography. Eur J Radiol. 2005;54:
management of myeloma bone disease. J Clin Oncol. 2011;29:1907-1915. 289-297.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e415


TERPOS, DIMOPOULOS, AND MOULOPOULOS

8. Kropil P, Fenk R, Fritz LB, et al. Comparison of whole-body 64-slice 26. Waheed S, Mitchell A, Usmani S, et al. Standard and novel imaging
multidetector computed tomography and conventional radiography methods for multiple myeloma: correlates with prognostic laboratory
in staging of multiple myeloma. Eur Radiol. 2008;18:51-58. variables including gene expression profiling data. Haematologica.
9. Gleeson TG, Moriarty J, Shortt CP, et al. Accuracy of whole-body low- 2013;98:71-78.
dose multidetector CT (WBLDCT) versus skeletal survey in the detection 27. Spinnato P, Bazzocchi A, Brioli A, et al. Contrast enhanced MRI and
18
of myelomatous lesions, and correlation of disease distribution with F-FDG PET-CT in the assessment of multiple myeloma: a comparison
whole-body MRI (WBMRI). Skeletal Radiol. 2009;38:225-236. of results in different phases of the disease. Eur J Radiol. 2012;81:
10. Princewill K, Kyere S, Awan O, et al. Multiple myeloma lesion detection 4013-4018.
with whole body CT versus radiographic skeletal survey. Cancer Invest. 28. Moulopoulos LA, Dimopoulos MA, Christoulas D, et al. Diffuse MRI
2013;31:206-211. marrow pattern correlates with increased angiogenesis, advanced
11. Wolf MB, Murray F, Kilk K, et al. Sensitivity of whole-body CT and MRI disease features and poor prognosis in newly diagnosed myeloma
versus projection radiography in the detection of osteolyses in patients treated with novel agents. Leukemia. 2010;24:1206-1212.
with monoclonal plasma cell disease. Eur J Radiol. 2014;83:1222-1230. 29. Moulopoulos LA, Dimopoulos MA, Kastritis E, et al. Diffuse pattern of
12. Cretti F, Perugini G. Patient dose evaluation for the whole-body low- bone marrow involvement on magnetic resonance imaging is associ-
dose multidetector CT (WBLDMDCT) skeleton study in multiple mye- ated with high risk cytogenetics and poor outcome in newly diagnosed,
loma (MM). Radiol Med (Torino). 2016;121:93-105. symptomatic patients with multiple myeloma: a single center experi-
13. Borggrefe J, Giravent S, Campbell G, et al. Association of osteolytic ence on 228 patients. Am J Hematol. 2012;87:861-864.
lesions, bone mineral loss and trabecular sclerosis with prevalent 30. Mai EK, Hielscher T, Kloth JK, et al. Association between magnetic
vertebral fractures in patients with multiple myeloma. Eur J Radiol. resonance imaging patterns and baseline disease features in multiple
2015;84:2269-2274. myeloma: analyzing surrogates of tumour mass and biology. Eur Radiol.
14. Terpos E, Kleber M, Engelhardt M, et al. European Myeloma Network Epub 2016 Jan 15.
guidelines for the management of multiple myeloma-related compli- 31. Moulopoulos LA, Gika D, Anagnostopoulos A, et al. Prognostic significance
cations. Haematologica. 2015;100:1254-1266. of magnetic resonance imaging of bone marrow in previously untreated
15. Dimopoulos MA, Hillengass J, Usmani S, et al. Role of magnetic reso- patients with multiple myeloma. Ann Oncol. 2005;16:1824-1828.
nance imaging in the management of patients with multiple myeloma: 32. Song MK, Chung JS, Lee JJ, et al. Magnetic resonance imaging pattern of
a consensus statement. J Clin Oncol. 2015;33:657-664. bone marrow involvement as a new predictive parameter of disease
16. Walker R, Barlogie B, Haessler J, et al. Magnetic resonance imaging in progression in newly diagnosed patients with multiple myeloma eligible for
multiple myeloma: diagnostic and clinical implications. J Clin Oncol. autologous stem cell transplantation. Br J Haematol. 2014;165:777-785.
2007;25:1121-1128. 33. Mai EK, Hielscher T, Kloth JK, et al. A magnetic resonance imaging-based
17. Moulopoulos LA, Varma DG, Dimopoulos MA, et al. Multiple myeloma: prognostic scoring system to predict outcome in transplant-eligible
spinal MR imaging in patients with untreated newly diagnosed disease. patients with multiple myeloma. Haematologica. 2015;100:818-825.
Radiology. 1992;185:833-840. 34. Usmani SZ, Mitchell A, Waheed S, et al. Prognostic implications of serial
18. Moulopoulos LA, Dimopoulos MA, Smith TL, et al. Prognostic signifi- 18-fluoro-deoxyglucose emission tomography in multiple myeloma
cance of magnetic resonance imaging in patients with asymptomatic treated with total therapy 3. Blood. 2013;121:1819-1823.
multiple myeloma. J Clin Oncol. 1995;13:251-256. 35. Dimopoulos M, Kyle R, Fermand JP, et al. Consensus recommendations
19. Kastritis E, Terpos E, Moulopoulos L, et al. Extensive bone marrow for standard investigative workup: report of the International Myeloma
infiltration and abnormal free light chain ratio identifies patients with Workshop Consensus Panel 3. Blood. 2011;117:4701-4705.
asymptomatic myeloma at high risk for progression to symptomatic 36. Kyle RA, Rajkumar SV. Criteria for diagnosis, staging, risk stratification
disease. Leukemia. 2013;27:947-953. and response assessment of multiple myeloma. Leukemia. 2009;23:3-9.
20. Regelink JC, Minnema MC, Terpos E, et al. Comparison of modern and 37. Moulopoulos LA, Dimopoulos MA, Alexanian R, et al. Multiple myeloma:
conventional imaging techniques in establishing multiple myeloma- MR patterns of response to treatment. Radiology. 1994;193:441-446.
related bone disease: a systematic review. Br J Haematol. 2013;162: 38. Hillengass J, Ayyaz S, Kilk K, et al. Changes in magnetic resonance
50-61 . imaging before and after autologous stem cell transplantation correlate
21. Hillengass J, Fechtner K, Weber MA, et al. Prognostic significance of with response and survival in multiple myeloma. Haematologica. 2012;
focal lesions in whole-body magnetic resonance imaging in patients 97:1757-1760.
with asymptomatic multiple myeloma. J Clin Oncol. 2010;28:1606-1610. 39. Bannas P, Hentschel HB, Bley TA, et al. Diagnostic performance of
22. Kastritis E, Moulopoulos LA, Terpos E, et al. The prognostic importance whole-body MRI for the detection of persistent or relapsing disease in
of the presence of more than one focal lesion in spine MRI of patients multiple myeloma after stem cell transplantation. Eur Radiol. 2012;22:
with asymptomatic (smoldering) multiple myeloma. Leukemia. 2014; 2007-2012.
28:2402-2403. 40. Merz M, Hielscher T, Wagner B, et al. Predictive value of longitudinal
23. Bauerle T, Hillengass J, Fechtner K, et al. Multiple myeloma and whole-body magnetic resonance imaging in patients with smoldering
monoclonal gammopathy of undetermined significance: importance of multiple myeloma. Leukemia. 2014;28:1902-1908.
whole-body versus spinal MR imaging. Radiology. 2009;252:477-485. 41. Baur A, Stabler A, Bruning R, et al. Diffusion-weighted MR imaging of
24. Baur-Melnyk A, Buhmann S, Becker C, et al. Whole-body MRI versus bone marrow: differentiation of benign versus pathologic compression
whole-body MDCT for staging of multiple myeloma. AJR Am J Roent- fractures. Radiology. 1998;207:349-356.
genol. 2008;190:1097-1104. 42. Varettoni M, Corso A, Pica G, et al. Incidence, presenting features and
25. Zamagni E, Nanni C, Patriarca F, et al. A prospective comparison of outcome of extramedullary disease in multiple myeloma: a longitudinal
18
F-fluorodeoxyglucose positron emission tomography-computed tomog- study on 1003 consecutive patients. Ann Oncol. 2010;21:325-330.
raphy, magnetic resonance imaging and whole-body planar radiographs in 43. Moulopoulos LA, Dimopoulos MA, Weber D, et al. Magnetic resonance
the assessment of bone disease in newly diagnosed multiple myeloma. imaging in the staging of solitary plasmacytoma of bone. J Clin Oncol.
Haematologica. 2007;92:50-55. 1993;11:1311-1315.

e416 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


IMAGING FOR MULTIPLE MYELOMA

44. Dimopoulos MA, Moulopoulos LA, Maniatis A, et al. Solitary plasma- 62. Breyer RJ III, Mulligan ME, Smith SE, et al. Comparison of imaging with
cytoma of bone and asymptomatic multiple myeloma. Blood. 2000;96: FDG PET/CT with other imaging modalities in myeloma. Skeletal Radiol.
2037-2044. 2006;35:632-640.
45. Lafforgue P, Dahan E, Chagnaud C, et al. Early-stage avascular necrosis 63. Zamagni E, Nanni C, Gay F, et al. 18F-FDG PET/CT focal, but not
of the femoral head: MR imaging for prognosis in 31 cases with at least 2 osteolytic, lesions predict the progression of smoldering myeloma to
years of follow-up. Radiology. 1993;187:199-204. active disease. Leukemia. 2015;30:417-422.
46. Syed IS, Glockner JF, Feng D, et al. Role of cardiac magnetic resonance 64. Siontis B, Kumar S, Dispenzieri A, et al. Positron emission tomography-
imaging in the detection of cardiac amyloidosis. JACC Cardiovasc Im- computed tomography in the diagnostic evaluation of smoldering
aging. 2010;3:155-164. multiple myeloma: identification of patients needing therapy. Blood
47. Nonomura Y, Yasumoto M, Yoshimura R, et al. Relationship between Cancer J. 2015;5:e364.
bone marrow cellularity and apparent diffusion coefficient. J Magn 65. Sachpekidis C, Mai EK, Goldschmidt H, et al. (18)F-FDG dynamic PET/CT
Reson Imaging. 2001;13:757-760. in patients with multiple myeloma: patterns of tracer uptake and
48. Terpos E, Koutoulidis V, Fontara S, et al. Diffusion-weighted imaging correlation with bone marrow plasma cell infiltration rate. Clin Nucl
improves accuracy in the diagnosis of MRI patterns of marrow in- Med. 2015;40:e300-e307.
volvement in newly diagnosed myeloma: results of a prospective study 66. Tirumani SH, Sakellis C, Jacene H, et al. Role of FDG-PET/CT in extra-
in 99 patients. Blood. 2015;126 (suppl; abstr 4178). medullary multiple myeloma: correlation of FDG-PET/CT findings with
49. Messiou C, Giles S, Collins DJ, et al. Assessing response of myeloma clinical outcome. Clin Nucl Med. 2016;41:e7-e13.
bone disease with diffusion-weighted MRI. Br J Radiol. 2012;85: 67. Zamagni E, Nanni C, Mancuso K, et al. PET/CT improves the definition of
e1198-e1203. complete response and allows to detect otherwise unidentifiable skeletal
50. Giles SL, deSouza NM, Collins DJ, et al. Assessing myeloma bone disease progression in multiple myeloma. Clin Cancer Res. 2015;21:4384-4390.
with whole-body diffusion-weighted imaging: comparison with x-ray 68. Paiva B, van Dongen JJ, Orfao A. New criteria for response assessment:
skeletal survey by region and relationship with laboratory estimates of role of minimal residual disease in multiple myeloma. Blood. 2015;125:
disease burden. Clin Radiol. 2015;70:614-621. 3059-3068.
51. Sachpekidis C, Mosebach J, Freitag MT, et al. Application of (18)F-FDG 69. Zamagni E, Patriarca F, Nanni C, et al. Prognostic relevance of 18-F FDG
PET and diffusion weighted imaging (DWI) in multiple myeloma: PET/CT in newly diagnosed multiple myeloma patients treated with up-
comparison of functional imaging modalities. Am J Nucl Med Mol front autologous transplantation. Blood. 2011;118:5989-5995.
Imaging. 2015;5:479-492. 70. Patriarca F, Carobolante F, Zamagni E, et al. The role of positron
52. Pawlyn C, Fowkes L, Otero S, et al. Whole-body diffusion-weighted MRI: emission tomography with 18F-fluorodeoxyglucose integrated with
a new gold standard for assessing disease burden in patients with computed tomography in the evaluation of patients with multiple
multiple myeloma? Leukemia. Epub 2015 Dec 9. myeloma undergoing allogeneic stem cell transplantation. Biol Blood
53. Horger M, Weisel K, Horger W, et al. Whole-body diffusion-weighted Marrow Transplant. 2015;21:1068-1073.
MRI with apparent diffusion coefficient mapping for early response 71. Fonti R, Pace L, Cerchione C, et al. 18F-FDG PET/CT, 99mTc-MIBI, and MRI
monitoring in multiple myeloma: preliminary results. AJR Am J in the prediction of outcome of patients with multiple myeloma:
Roentgenol. 2011;196:W790-W795. a comparative study. Clin Nucl Med. 2015;40:303-308.
54. Hillengass J, Zechmann C, Bauerle T, et al. Dynamic contrast-enhanced 72. Bartel TB, Haessler J, Brown TL, et al. F18-fluorodeoxyglucose pos-
magnetic resonance imaging identifies a subgroup of patients with itron emission tomography in the context of other imaging tech-
asymptomatic monoclonal plasma cell disease and pathologic micro- niques and prognostic factors in multiple myeloma. Blood. 2009;114:
circulation. Clin Cancer Res. 2009;15:3118-3125. 2068-2076.
55. Huang SY, Chen BB, Lu HY, et al. Correlation among DCE-MRI mea- 73. Lapa C, Luckerath K, Malzahn U, et al. 18 FDG-PET/CT for prognostic
surements of bone marrow angiogenesis, microvessel density, and stratification of patients with multiple myeloma relapse after stem cell
extramedullary disease in patients with multiple myeloma. Am J He- transplantation. Oncotarget. 2014;5:7381-7391.
matol. 2012;87:837-839. 74. Cascini GL, Falcone C, Console D, et al. Whole-body MRI and PET/CT in
56. Merz M, Ritsch J, Kunz C, et al. Dynamic contrast-enhanced magnetic multiple myeloma patients during staging and after treatment: personal
resonance imaging for assessment of antiangiogenic treatment effects experience in a longitudinal study. Radiol Med (Torino). 2013;118:
in multiple myeloma. Clin Cancer Res. 2015;21:106-112. 930-948.
57. Merz M, Moehler TM, Ritsch J, et al. Prognostic significance of 75. Moreau P, Attal M, Karlin L, et al. Prospective evaluation of MRI and PET-
increased bone marrow microcirculation in newly diagnosed mul- CT at diagnosis and before maintenance therapy in symptomatic pa-
tiple myeloma: results of a prospective DCE-MRI study. Eur Radiol. tients with multiple myeloma included in the IFM/DFCI 2009 trial.
Epub 2015 Jul 29. Blood. 2015;126 (suppl; abstr 395).
58. Dutoit JC, Claus E, Offner F, et al. Combined evaluation of conventional 76. Nanni C, Zamagni E, Versari A, et al. Image interpretation criteria for FDG
MRI, dynamic contrast-enhanced MRI and diffusion weighted imaging PET/CT in multiple myeloma: a new proposal from an Italian expert
for response evaluation of patients with multiple myeloma. Eur J Radiol. panel. IMPeTUs (Italian Myeloma criteria for PET USe). Eur J Nucl Med
2016;85:373-382. Mol Imaging. 2015;43:414-421.
59. Sachpekidis C, Hillengass J, Goldschmidt H, et al. Comparison of (18) 77. Fouquet G, Guidez S, Herbaux C, et al. Impact of initial FDG-PET/CT and
F-FDG PET/CT and PET/MRI in patients with multiple myeloma. Am J serum-free light chain on transformation of conventionally defined
Nucl Med Mol Imaging. 2015;5:469-478. solitary plasmacytoma to multiple myeloma. Clin Cancer Res. 2014;20:
60. Bredella MA, Steinbach L, Caputo G, et al. Value of FDG PET in the 3254-3260.
assessment of patients with multiple myeloma. AJR Am J Roentgenol. 78. Alongi P, Zanoni L, Incerti E, et al. 18F-FDG PET/CT for early post-
2005;184:1199-1204. radiotherapy assessment in solitary bone plasmacytomas. Clin Nucl
61. Lutje S, de Rooy JW, Croockewit S, et al. Role of radiography, MRI and Med. 2015;40:e399-e404.
FDG-PET/CT in diagnosing, staging and therapeutical evaluation of 79. Lonial S, Kaufman JL. Non-secretory myeloma: a clinicians guide.
patients with multiple myeloma. Ann Hematol. 2009;88:1161-1168. Oncology (Williston Park). 2013;27:924-928, 930.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e417


S. VINCENT RAJKUMAR

Updated Diagnostic Criteria and Staging System for Multiple


Myeloma
S. Vincent Rajkumar, MD

OVERVIEW

There has been remarkable progress made in the diagnosis and treatment of multiple myeloma (MM). The median survival of
the disease has doubled as a result of several new active drugs. These advances have necessitated a revision of the disease
definition and staging of MM. Until recently, MM was defined by the presence of end-organ damage, specifically hy-
percalcemia, renal failure, anemia, and bone lesions (CRAB features) that can be attributed to the clonal process. In 2014, the
International Myeloma Working Group (IMWG) updated the diagnostic criteria for MM to add three specific biomarkers that
can be used to diagnose the disease in patients who did not have CRAB features: clonal bone marrow plasma cells greater
than or equal to 60%, serum free light chain (FLC) ratio greater than or equal to 100 provided involved FLC level is 100 mg/L or
higher, or more than one focal lesion on MRI. In addition, the definition was revised to allow CT and PET-CT to diagnose MM
bone disease. These changes enable early diagnosis and allow the initiation of effective therapy to prevent the development
of end-organ damage for patients who are at the highest risk. A new staging system has been developed that incorporates
high-risk cytogenetic abnormalities in addition to standard laboratory markers of prognosis.

M ultiple myeloma evolves from a clinically silent pre-


malignant stage termed monoclonal gammopathy of
undetermined significance (MGUS).1,2 MGUS is a classic
REVISED DIAGNOSTIC CRITERIA FOR MULTIPLE
MYELOMA
In 2014, the IMWG revised the diagnostic criteria for MM.10
premalignant condition with a low risk of malignant con- The revised diagnostic criteria for MM allow the use of
version, but the risk of progression persists indefinitely. A specific biomarkers to define the disease in addition to the
small subset of patients has an intermediate clinical phe- established CRAB features. They also allow the use of
notype between MGUS and MM, and they are referred to modern imaging tools to diagnose MM bone disease and
as having smoldering multiple myeloma (SMM).3 MGUS clarify several other diagnostic requirements. Table 1 pro-
is associated with a risk of progression to MM or related vides the revised IMWG criteria for diagnosis of MM and
malignancy at a rate of approximately 1% per year, whereas related plasma cell disorders.10
SMM has a much higher risk of progression of approxi-
mately 10% per year.4,5 MGUS and SMM are typically
asymptomatic and are typically diagnosed incidentally Myeloma-Defining Events
when a monoclonal (M) protein is detected during labo- The diagnosis of MM requires 10% or more clonal plasma
ratory work-up of patients who have a wide spectrum of cells on bone marrow examination or a biopsy-proven
clinical conditions. plasmacytoma plus the presence of one or more
Over the years the diagnosis of MM required evidence myeloma-defining events.10 Myeloma-defining events in-
of end-organ damage attributable to the neoplastic clone clude the presence of one or more CRAB features, or one
of plasma cells: hypercalcemia, renal failure, anemia, and or more biomarkers of malignancy. The three biomarkers
osteolytic bone lesions, commonly referred to as CRAB included in the definition of MM are associated with an
features.6 This definition was conservative and intended to approximately 80% risk of progression to symptomatic end-
prevent patients with MGUS and SMM from receiving un- organ damage in two or more independent studies. They are
necessary and toxic chemotherapy. With major advances in clonal bone marrow plasma cells greater than or equal to
therapy and identification of biomarkers that can distinguish 60%, serum FLC ratio of 100 or higher, provided involved FLC
MM from premalignant phases, it became necessary to level is 100 mg/L or higher, or more than one focal lesion
revise the disease definition of MM.7-9 on MRI.

From the Division of Hematology, Mayo Clinic, Rochester, MN.

Disclosures of potential conflicts of interest provided by the author are available with the online article at asco.org/edbook.

Corresponding author: S. Vincent Rajkumar, MD, Division of Hematology, Mayo Clinic, 200 First St. SW, Rochester, MN 55905; email: rajkumar.vincent@mayo.edu.

2016 by American Society of Clinical Oncology.

e418 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


NEW DIAGNOSTIC AND STAGING CRITERIA IN MYELOMA

Extreme bone marrow clonal plasmacytosis. Clonal bone with SMM with an involved/uninvolved FLC ratio of 100 or
marrow plasma cell involvement of greater than or equal to higher had a 64% risk of progression within 2 years.12 A
60% is extremely unusual in the absence of CRAB features. markedly abnormal FLC ratio is thus an accurate predictor of
In a Mayo Clinic study of SMM, only six of 276 patients (2%) patients who have a high risk of progression to end-organ
had clonal bone marrow plasma cells greater than or equal damage within a short period of time. To reduce the pos-
to 60%. In that study, patients with this level of marrow sibility of error, in addition to the FLC ratio being 100 or
involvement progressed to symptomatic malignancy rapidly, higher, the IMWG also added a requirement for a minimal
with a median progression-free survival of 7.7 months.7 In a involved FLC level of at least 100 mg/L to be considered as
separate cohort of 651 patients with SMM, only 21 (3.2%) a myeloma-defining event.10
had clonal bone marrow plasma cells greater than or equal
to 60%, and 95% of these patients progressed to having MM MRI with more than one focal lesion. MRI imaging study of
within 2 years.7 A study of 96 patients with SMM from the the whole-body spine/pelvis can reveal focal or diffuse
Greek Myeloma Group also found a markedly high risk of changes in SMM. In a study by Hillengass et al, 23 of 149
progression in this subgroup of patients, with a median time (15%) patients with SMM had more than one focal lesion
to progression (TTP) of 15 months.11 Similar results were seen on whole-body MRI.14 The median TTP in these patients
reported by the University of Pennsylvania; six of 121 pa- was 13 months, and the progression rate at 2 years was 70%.
tients (5%) with SMM had greater than or equal to 60% bone These results were confirmed by Kastritis et al, who found
marrow involvement, and all progressed to having MM in more than one focal lesion on spinal MRI in nine of 65
less than 2 years.12 patients (14%) with SMM.15 The median TTP was 15 months
and 69% progressed to MM within 2 years. To increase
Marked elevation of serum involved/uninvolved FLC predictive value, the IMWG added a requirement that focal
ratio. In SMM, an abnormal FLC ratio is associated with a lesions must be at least 5 mm or more in size and recom-
higher risk of progression to MM.13 As the ratio increases, mended follow-up examinations in 3 to 6 months for pa-
so does the risk of progression. In a Mayo Clinic study of tients who had a solitary focal lesion, equivocal findings, or
586 patients with SMM, a markedly abnormal involved/ diffuse infiltration.10
uninvolved FLC ratio ($ 100) was seen in 90 (15%) pa-
tients.8 The risk of progression to symptomatic end-organ Assessment of myeloma bone disease. There have been
damage within the first 2 years with an FLC ratio of 100 or many advances to imaging methods used to detect bone and
higher was 72%; the risk of progression to MM or amyloid extramedullary disease in MM. These include low-dose
light-chain amyloidosis in 2 years was 79%. A similar finding whole-body CT, MRI, and (18)Ffluorodeoxyglucose PET
was reported by Kastritis et al, who studied 96 patients with (FDG-PET) and FDG-PET with PET-CT.16-22 The old IMWG
SMM. In their study, 7% of patients with SMM had an criteria for the diagnosis of MM primarily relied on con-
involved/uninvolved FLC ratio of 100 or higher, and almost ventional radiographs to detect bone disease.6 A systematic
all progressed to symptomatic disease within 18 months.11 review compared MRI, FDG-PET, PET CT, and whole-body CT
In a third study, at the University of Pennsylvania, patients to conventional whole-body skeletal radiography in MM.23
Newer imaging techniques had greater sensitivity compared
with radiographic bone survey for detection of MM bone
lesions, with as high as 80% more lesions detected by the
KEY POINTS newer imaging techniques. CT and MRI performed equally
with respect to sensitivity.
The diagnostic criteria for multiple myeloma and related
disorders have been updated by the International
Based on these studies, the revised IMWG criteria for MM
Myeloma Working Group. permit the use of CT, low-dose whole-body CT, PET-CT, and
Patients with 60% or more clonal plasma cell whole-body CT to diagnose lytic bone disease in MM. One or
involvement of the marrow, serum free light chain ratio more sites of osteolytic bone destruction of at least 5 mm or
of 100 or higher (provided involved free light chain more in size is required. Increased uptake on PET-CT alone is
level 100 mg/L), and/or greater than one focal lesion not adequate; there must be evidence of actual osteolytic
on MRI are defined as MM even in the absence of end- bone destruction on the CT portion of the examination. A
organ damage. biopsy of one of the bone lesions should be considered if
Low-dose whole-body CT, MRI, and FDG-PET and FDG- there is any doubt about the diagnosis. These changes will
PET with PET-CT are more sensitive in detecting enable early and accurate diagnosis of MM. In addition to
myeloma and must be considered in diagnosis and
these changes, the IMWG clarified that the presence of
monitoring.
In terms of renal involvement, only light chain cast
osteoporosis, vertebral compression fractures, or bone
nephropathy is considered a myeloma-defining event. densitometric changes in the absence of lytic lesions is not
The Revised International Staging System has been sufficient evidence of myeloma bone disease.
developed for myeloma that incorportates high-risk Because the accurate diagnosis of MM is critical, at least
cytogenetic abnormalities. one advanced imaging examination (PET-CT, low-dose
whole-body CT, or MRI of the whole body or spine) is

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e419


S. VINCENT RAJKUMAR

TABLE 1. International Myeloma Working Group Diagnostic Criteria for Multiple Myeloma and Related Plasma
Cell Disorders

Disorder Disease Definition


Non-IgM MGUS All three criteria must be met:
Serum monoclonal protein (non-IgM type) , 3 g/dL
Clonal bone marrow plasma cells , 10%*
Absence of end-organ damage such as CRAB features that can be attributed to the plasma cell proliferative disorder
Smoldering MM Both criteria must be met:
Serum monoclonal protein (IgG or IgA) $ 3 gm/dL, or urinary monoclonal protein $ 500 mg per 24 h and/or
clonal bone marrow plasma cells 10%60%
Absence of myeloma-defining events or amyloidosis
MM Both criteria must be met:
Clonal bone marrow plasma cells $ 10% or biopsy-proven bony or extramedullary plasmacytoma
Any one or more of the following myeloma defining events:
Evidence of end organ damage that can be attributed to the underlying plasma cell proliferative disorder,
specifically:
Hypercalcemia: serum calcium . 0.25 mmol/L (. 1 mg/dL) higher than the upper limit of normal or
. 2.75 mmol/L (. 11 mg/dL)
Renal insufficiency: creatinine clearance , 40 mL/min or serum creatinine . 177 mmol/L (. 2 mg/dL)
Anemia: hemoglobin value of . 2 g/dL below the lower limit of normal, or a hemoglobin value
, 10 g/dL
Bone lesions: one or more osteolytic lesions on skeletal radiography, CT, or PET-CT
Clonal bone marrow plasma cell percentage $ 60%
Involved: uninvolved serum FLC ratio $ 100 (involved FLC level must be $ 100 mg/L)
. 1 focal lesion on MRI studies (at least 5 mm in size)
IgM MGUS All three criteria must be met:
Serum IgM monoclonal protein , 3 gm/dL
Bone marrow lymphoplasmacytic infiltration , 10%
No evidence of anemia, constitutional symptoms, hyperviscosity, lymphadenopathy, or hepatospleno-
megaly that can be attributed to the underlying lymphoproliferative disorder.
Light-Chain MGUS All criteria must be met:
Abnormal FLC ratio (, 0.26 or . 1.65)
Increased level of the appropriate involved light chain (increased kappa FLC in patients with ratio . 1.65 and
increased lambda FLC in patients with ratio , 0.26)
No immunoglobulin heavy-chain expression on immunofixation
Absence of end-organ damage that can be attributed to the plasma cell proliferative disorder
Clonal bone marrow plasma cells , 10%
Urinary monoclonal protein , 500 mg/24 h
Solitary Plasmacytoma All four criteria must be met:
Biopsy-proven solitary lesion of bone or soft tissue with evidence of clonal plasma cells
Normal bone marrow with no evidence of clonal plasma cells
Normal skeletal survey and MRI (or CT) of spine and pelvis (except for the primary solitary lesion)
Absence of end-organ damage such as CRAB features that can be attributed to a lympho-plasma cell
proliferative disorder
Solitary Plasmacytoma With Minimal All four criteria must be met:
Marrow Involvement**
Biopsy-proven solitary lesion of bone or soft tissue with evidence of clonal plasma cells
Clonal bone marrow plasma cells , 10%
Normal skeletal survey and MRI (or CT) of spine and pelvis (except for the primary solitary lesion)
Absence of end-organ damage such as CRAB features that can be attributed to a lympho-plasma cell
proliferative disorder
Abbreviations: MGUS, monoclonal gammopathy of undertermined significance; CRAB features, hypercalcemia, renal insufficiency, anemia, and bone lesions; MM, multiple myeloma; FLC, free light chain;
SMM, smoldering multiple myeloma.
*A bone marrow examination can be deferred for patients with low-risk MGUS (IgG type, M protein , 15 gm/L, normal FLC ratio) in whom there are no clinical features concerning for myeloma.
**
Solitary plasmacytoma with 10% or more clonal plasma cells is considered as MM.
Reproduced from Rajkumar et al.10

e420 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


NEW DIAGNOSTIC AND STAGING CRITERIA IN MYELOMA

recommended for patients before concluding that a patient marrow examination (Table 1). Treatment consists of radi-
has SMM or solitary plasmacytoma.10,24 The choice between ation therapy at 40 Gy to 50 Gy to the involved site.
various imaging methods can vary depending on the clinical Patients with an apparent solitary plasmacytoma who
situation and availability. have limited (, 10%) clonal marrow involvement are con-
sidered to have solitary plasmacytoma with minimal marrow
Other clarifications to MM diagnostic criteria. Several involvement. These patients are also treated similar to
clarifications to the diagnostic criteria for MM were made. patients with solitary plasmacytoma. The risk of recurrence
Importantly, the IMWG clarified that in terms of renal disease, or progression to myeloma within 3 years is approximately
only suspected or proven light-chain cast nephropathy is 10% for patients with solitary plasmacytoma versus 20% to
considered a myeloma-defining event. Other renal disorders 60% for patients with solitary plasmacytoma and minimal
associated with M proteins, such as light-chain deposition marrow involvement.
disease, membranoproliferative glomerulonephritis, and
amyloid light-chain amyloidosis, are considered unique dis- REVISED INTERNATIONAL STAGING SYSTEM FOR
eases and not MM. A renal biopsy to clarify the underlying MYELOMA
cause of renal failure is recommended for patients with There is major variation in survival of MM depending on
suspected cast nephropathy, especially if the serum involved host factors, tumor burden (stage), biology (cytogenetic
FLC levels are less than 500 mg/L.25 An estimated glomerular abnormalities), and response to therapy.26 Tumor burden
filtration rate less than 40 mL/min is preferred to the serum in MM has traditionally been assessed using the Durie-
creatinine concentration for purposes of fulfilling the CRAB Salmon Staging (DSS) 27 and the International Staging
criteria. System (ISS).28,29 Both these staging systems have some
limitations. The DSS primarily classified patients based on
Solitary plasmacytoma. Solitary plasmacytoma is an early- tumor burden and had problems with reproducibility be-
stage plasma cell malignancy that is in between MGUS/SMM cause some judgment is needed in interpretation of MM
and MM along the spectrum of plasma cell disorders. It is bone disease. The ISS is more reproducible but includes a
defined by the presence of a single biopsy-proven plas- host factor determinant, namely serum albumin. Thus,
macytoma (bony or extramedullary) and a normal bone outcome using ISS can be disproportionately affected by

TABLE 2. Cytogenetic Abnormalities on Clinical Course and Prognosis in Multiple Myeloma

Clinical Setting in Which Abnormality Is Detected


Cytogenetic Abnormality Smoldering Multiple Myeloma Multiple Myeloma
Trisomies Intermediate-risk of progression, median TTP of 3 years Good prognosis, standard-risk MM, median
OS 7-10 years
Most have myeloma bone disease at diagnosis
Excellent response to lenalidomide-based therapy
t(11;14) (q13;q32) Standard-risk of progression, median TTP of 5 years Good prognosis, standard-risk MM, median
OS 7-10 years
t(6;14) (p21;q32) Standard-risk of progression, median TTP of 5 years Good prognosis, standard-risk MM, median
OS 7-10 years
t(4;14) (p16;q32) High-risk of progression, median TTP of 2 years Intermediate-risk MM, median OS 5 years
Needs bortezomib-based initial therapy, early
ASCT (if eligible), followed by bortezomib-
based consolidation/maintenance
t(14;16) (q32;q23) Standard-risk of progression, median TTP of 5 years High-risk MM, median OS 3 years
Associated with high levels of FLC and 25% present
with acute renal failure as initial MDE
t(14;20) (q32;q11) Standard-risk of progression, median TTP of 5 years High-risk MM, median OS 3 years
Gain(1q21) High-risk of progression, median TTP of 2 years Intermediate-risk MM, median OS 5 years
Del(17p) High-risk of progression, median TTP of 2 years High-risk MM, median OS 3 years
Trisomies Plus Any One of the IgH Standard-risk of progression, median TTP of 5 years May ameliorate adverse prognosis conferred by
Translocations high risk IgH translocations, and del 17p
Isolated Monosomy 13, or Isolated Standard-risk of progression, median TTP of 5 years Effect on prognosis is not clear
Monosomy 14
Normal Low-risk of progression, median TTP of 7-10 years Good prognosis, probably reflecting low tumor
burden, median OS . 7-10 years
Abbreviations: TTP, time to progression; MM, multiple myeloma; OS, overall survival; ASCT, autologous stem cell transplantation; MDE, myeloma-defining event.
Reproduced from Rajan and Rajkumar.32

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e421


S. VINCENT RAJKUMAR

TABLE 3. Revised International Staging System for Myeloma


Stage Frequency (% of Patients) 5-Year Survival Rate (%)
Stage I 28 82
ISS stage I (serum albumin > 3.5, serum beta-2-microglobulin < 3.5) and
No high-risk cytogenetics
Normal LDH
Stage II 62 62
Neither stage I or III
Stage III 10 40
ISS stage III (serum beta-2-microglobulin > 5.5) and
High-risk cytogenetics [t(4;14), t(14;16), or del(17p)] or elevated LDH
Abbreviations: LDH, lactate dehydrogenase; ISS, International Staging System.
Derived from Palumbo et al.33

factors that are not disease-specific. Neither staging system the clinic in terms of counseling patients regarding prog-
considers disease biologya key determinant of overall nosis, as well as in clinical trials to compare outcomes across
survival in the disease. clinical trials.
Disease biology in MM is best reflected based on the
molecular subtype of the disease and the presence or ab-
sence of specific cytogenetic abnormalities.30,31 For exam- FUTURE DIRECTIONS
ple, abnormalities such as t(4;14), t(14;16), t(14;20), gain The updated diagnostic criteria for MM represent a para-
(1q), del(1p), and del(17p) influence disease course, re- digm shift in our approach to the disease. These changes will
sponse to therapy, and prognosis in MM (Table 2).32 The allow us to intervene before end-organ damage in selected
Revised International Staging System (RISS) combines ele- patients who are at imminent risk of symptomatic pro-
ments of tumor burden (ISS) and disease biology (presence gression. The revised staging system for MM allows us to
of high-risk cytogenetic abnormalities or elevated lactate better predict outcome and tailor treatments accordingly.
dehydrogenase level) to create a unified prognostic index Advances in imaging have not only enabled early and ac-
that helps in clinical care as well as in comparison of clinical curate diagnosis, but also provide ways of monitoring re-
trial data (Table 3).33 The RISS was developed based on a sponse to therapy. We must identify additional reliable
study of 4,445 patients with newly diagnosed MM from biomarkers of malignancy. We must also develop a portfolio
11 international trials. This study showed that the 5-year of clinical trials designed to determine whether early in-
survival rate of patients with stage I, II, and III RISS was 82%, tervention can improve survival or provide a path to
62%, and 40%, respectively. The RISS will be of importance in cure MM.

References
1. Landgren O, Kyle RA, Pfeiffer RM, et al. Monoclonal gammopathy of 8. Larsen JT, Kumar SK, Dispenzieri A, et al. Serum free light chain ratio as a
undetermined significance (MGUS) consistently precedes multiple biomarker for high-risk smoldering multiple myeloma. Leukemia. 2013;
myeloma: a prospective study. Blood. 2009;113:5412-5417. 27:941-946.
2. Weiss BM, Abadie J, Verma P, et al. A monoclonal gammopathy pre- 9. Mateos M-V, Hernandez M-T, Giraldo P, et al. Lenalidomide plus
cedes multiple myeloma in most patients. Blood. 2009;113:5418-5422. dexamethasone for high-risk smoldering multiple myeloma. N Engl J
3. Rajkumar SV, Merlini G, San Miguel JF. Haematological cancer: Rede- Med. 2013;369:438-447.
fining myeloma. Nat Rev Clin Oncol. 2012;9:494-496. 10. Rajkumar SV, Dimopoulos MA, Palumbo A, et al. International Myeloma
4. Kyle RA, Remstein ED, Therneau TM, et al. Clinical course and prognosis Working Group updated criteria for the diagnosis of multiple myeloma.
of smoldering (asymptomatic) multiple myeloma. N Engl J Med. 2007; Lancet Oncol. 2014;15:e538-e548.
356:2582-2590. 11. Kastritis E, Terpos E, Moulopoulos L, et al. Extensive bone marrow
5. Kyle RA, Therneau TM, Rajkumar SV, et al. A long-term study of infiltration and abnormal free light chain ratio identifies patients with
prognosis in monoclonal gammopathy of undetermined significance. asymptomatic myeloma at high risk for progression to symptomatic
N Engl J Med. 2002;346:564-569. disease. Leukemia. 2013;27:947-953.
6. International Myeloma Working Group. Criteria for the classification of 12. Waxman AJ, Mick R, Garfall AL, et al. Modeling the risk of progression in
monoclonal gammopathies, multiple myeloma and related disorders: smoldering multiple myeloma. J Clin Oncol. 2014;32:A8607.
a report of the International Myeloma Working Group. Br J Haematol. 13. Dispenzieri A, Kyle RA, Katzmann JA, et al. Immunoglobulin free
2003;121:749-757. light chain ratio is an independent risk factor for progression of
7. Rajkumar SV, Larson D, Kyle RA. Diagnosis of smoldering multiple smoldering (asymptomatic) multiple myeloma. Blood. 2008;111:
myeloma. N Engl J Med. 2011;365:474-475. 785-789.

e422 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


NEW DIAGNOSTIC AND STAGING CRITERIA IN MYELOMA

14. Hillengass J, Fechtner K, Weber MA, et al. Prognostic significance of related bone disease: a systematic review. Br J Haematol. 2013;162:
focal lesions in whole-body magnetic resonance imaging in patients 50-61.
with asymptomatic multiple myeloma. J Clin Oncol. 2010;28:1606-1610. 24. Dimopoulos MA, Hillengass J, Usmani S, et al. Role of magnetic reso-
15. Kastritis E, Moulopoulos LA, Terpos E, et al. The prognostic importance nance imaging in the management of patients with multiple myeloma:
of the presence of more than one focal lesion in spine MRI of patients a consensus statement. J Clin Oncol. 2015;33:657-664.
with asymptomatic (smoldering) multiple myeloma. Leukemia. 2014; 25. Hutchison CA, Batuman V, Behrens J, et al; International Kidney and
28:2402-2403. Monoclonal Gammopathy Research Group. The pathogenesis and di-
16. Dimopoulos M, Terpos E, Comenzo RL, et al; IMWG. International agnosis of acute kidney injury in multiple myeloma. Nat Rev Nephrol.
myeloma working group consensus statement and guidelines regarding 2012;8:43-51.
the current role of imaging techniques in the diagnosis and monitoring 26. Russell SJ, Rajkumar SV. Multiple myeloma and the road to personalised
of multiple Myeloma. Leukemia. 2009;23:1545-1556. medicine. Lancet Oncol. 2011;12:617-619.
17. Hillengass J, Landgren O. Challenges and opportunities of novel imaging 27. Durie BG, Salmon SE. A clinical staging system for multiple myeloma.
techniques in monoclonal plasma cell disorders: imaging early mye- Correlation of measured myeloma cell mass with presenting clinical
loma. Leuk Lymphoma. 2013;54:1355-1363. features, response to treatment, and survival. Cancer. 1975;36:
18. Zamagni E, Cavo M. The role of imaging techniques in the management 842-854.
of multiple myeloma. Br J Haematol. 2012;159:499-513. 28. Greipp PR, San Miguel J, Durie BG, et al. International staging system for
19. Zamagni E, Nanni C, Patriarca F, et al. A prospective comparison of 18F- multiple myeloma. J Clin Oncol. 2005;23:3412-3420.
fluorodeoxyglucose positron emission tomography-computed tomog- 29. Hari PN, Zhang MJ, Roy V, et al. Is the International Staging System
raphy, magnetic resonance imaging and whole-body planar radiographs superior to the Durie-Salmon staging system? A comparison in multiple
in the assessment of bone disease in newly diagnosed multiple mye- myeloma patients undergoing autologous transplant. Leukemia. 2009;
loma. Haematologica. 2007;92:50-55. 23:1528-1534.
20. Walker R, Barlogie B, Haessler J, et al. Magnetic resonance imaging in 30. Kumar SK, Mikhael JR, Buadi FK, et al. Management of newly diagnosed
multiple myeloma: diagnostic and clinical implications. J Clin Oncol. symptomatic multiple myeloma: updated Mayo Stratification of My-
2007;25:1121-1128. eloma and Risk-Adapted Therapy (mSMART) consensus guidelines.
21. Bartel TB, Haessler J, Brown TL, et al. F18-fluorodeoxyglucose positron Mayo Clin Proc. 2009;84:1095-1110.
emission tomography in the context of other imaging techniques and 31. Kumar S, Fonseca R, Ketterling RP, et al. Trisomies in multiple myeloma:
prognostic factors in multiple myeloma. Blood. 2009;114:2068-2076. impact on survival in patients with high-risk cytogenetics. Blood. 2012;
22. Waheed S, Mitchell A, Usmani S, et al. Standard and novel imaging 119:2100-2105.
methods for multiple myeloma: correlates with prognostic laboratory 32. Rajan AM, Rajkumar SV. Interpretation of cytogenetic results in multiple
variables including gene expression profiling data. Haematologica. myeloma for clinical practice. Blood Cancer J. 2015;5:e365.
2013;98:71-78. 33. Palumbo A, Avet-Loiseau H, Oliva S, et al. Revised International Staging
23. Regelink JC, Minnema MC, Terpos E, et al. Comparison of modern and System for multiple myeloma: a report from International Myeloma
conventional imaging techniques in establishing multiple myeloma- Working Group. J Clin Oncol. 2015;33:2863-2869.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e423


HEMATOLOGIC MALIGNANCIESPLASMA CELL
DYSCRASIA

Multiple Myeloma: Are We Ready


for Personalized Therapy?

CHAIR
Nikhil C. Munshi, MD
Dana-Farber Cancer Institute
Boston, MA

SPEAKERS
Herve Avet-Loiseau, MD, PhD
University Hospital
Nantes, France

Sagar Lonial, MD, FACP


Winship Cancer Institute of Emory University
Atlanta, GA
MINIMAL RESIDUAL DISEASE BY NEXT-GENERATION SEQUENCING

Minimal Residual Disease by Next-Generation Sequencing:


Pros and Cons
Herve Avet-Loiseau, MD, PhD

OVERVIEW

The wealth of data recently generated highlights that minimal residual disease (MRD)negative status can be achieved in a
large proportion of patients. These studies, in addition to a meta-analysis, clearly suggest significant improvement in both
event-free survival (EFS) and overall survival (OS) among those patients achieving MRDnegative status, especially with
sensitivity of one cell in 1 million bone marrow cells. There is an evolving consensus that achieving MRDnegative status
should become the ultimate goal of therapeutic intervention. Further future efforts should now be directed at determining
how MRD status can be used to guide and personalize further therapy including type of consolidation and maintenance
therapy.

T he treatment landscape for multiple myeloma (MM) has


been radically transformed during the past decade by the
introduction of several new drugs with different mecha-
post-transplant consolidation, and prolonged maintenance
therapy, nearly all patients achieve a treatment response,
with over 50% of these patients reaching complete re-
nisms of action, which in turn has led to the improved mission (CR) in some of the more recently reported studies
survival of patients with MM.1 Diagnostic and response and up to 80% in recent phase II trials.4-9 Frustratingly, the
criteria in MM have used a combination of tumor burden vast majority of patients experience relapse despite achiev-
and functional consequences of tumor expansion (such as ing such deep responses, reflecting persistent disease that
CRAB features), with recent revisions incorporating thresh- cannot be detected using the currently recommended disease
olds for the serum-free light chain (sFLC) ratio and bone evaluation techniques. Consequently, new methods are ur-
marrow plasma cell percentage as well as using bone lesions gently required to detect and quantitate MRD beyond the
on sensitive imaging as predictors of incipient progression to level of detection of the current clinical response criteria, and
MM and, therefore, as acceptable triggers for initiation of there is a need to revise the current definition of disease
therapy.2 Response evaluation in MM has traditionally been response for it to evolve in parallel to the changing treatment
based on assessing serum and/or urine monoclonal protein paradigm.
concentrations via protein electrophoresis as a surrogate for
tumor burden with immunofixation, allowing for detection
of trace amounts of paraprotein. Thus, the most recent it- DEPTH OF RESPONSE AND LONG-TERM
eration of the response criteria was developed a decade ago OUTCOME
by the International Myeloma Working Group (IMWG; IMWG The relationship between depth of response and long-term
Consensus Criteria for Response Assessment) and was based outcomes has been one of the most debated topics in MM.
on serum and urine protein electrophoresis, serum and urine The relationship between CR and progression-free survival
immunofixation, sFLC, and bone marrow plasma cell quan- (PFS) and/or time to progression (TTP) has been more
titation plus clonality assessment.3 consistent than the relationship between CR and OS. This
The consensus criteria were uniformly incorporated into putative incongruity is a frequent phenomenon in cancer
clinical trials, allowing better comparison of different drugs, therapy and probably due to multiple factors, including
drug combinations, and treatment strategies, and the re- interactions between (1) disease biology, (2) different treat-
visions over the years have allowed it to stay ahead of the ment strategies after CR has been reached, and (3) the true
advances in treatment. With the recent introduction of more depth of response beyond the conventional (and low-sensitive)
effective multidrug combinations, especially when used in approaches defining CR after different therapies. In this
the context of allogeneic stem cell transplantation (ASCT), context, several studies using newer and more sensitive

From the Laboratory for Genomics in Myeloma, Institut Universitaire du Cancer and University Hospital, Centre de Recherche en Cancerologie de Toulouse, Toulouse, France.

Disclosures of potential conflicts of interest provided by the author are available with the online article at asco.org/edbook.

Corresponding author: Herve Avet-Loiseau, MD, PhD, Unite de Genomique du Myelome, IUC-T Oncopole, 1 Avenue Irene Joliot-Curie, 31059, Toulouse, France;
email: avet-loiseau.h@chu-toulouse.fr.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e425


HERVE AVET-LOISEAU

techniques have been able to demonstrate the persistence for discriminating and categorizing normal and MM plasma
of MRD not detected by the current CR evaluation meth- cells include CD138, CD38, CD45, CD56, CD19, and cyto-
odologies in a significant fraction of patients. The level of plasmic kappa and lambda immunoglobulin light chains.
persistent MRD, undetected by conventional methods, is Additional markers, many of which are aberrantly expressed
likely to be one of the most important features contributing on MM plasma cells, have also been found to be of value and
to the link between the depth of response and long-term include CD27, CD28, CD81, and CD117. However, given the
outcomes. Independent of the method used to define MRD heterogeneity of expression of these markers and differ-
(cell-based, molecular-based, or imaging-based), prior studies ences in both the number of events studied and in the
consistently show that among patients who have achieved analytical strategies used, there has been significant con-
CR, MRDpositive cases consistently have an inferior PFS fusion in the field and inconsistent clinical interpretation of
than patients with MRDnegative disease.10-16 Given the results from different studies.20 Recently, attempts have
substantial proportion of patients achieving CR with the cur- been made to develop consensus guidelines to standardize
rent therapies, it is time to go beyond the CR criteria and the flow-based assessment of disease in MM and other
define MRD assessment in MM.16 related plasma cell disorders.17,18
Several studies have demonstrated the utility of MFC in
the detection of MRD in the bone marrow. The Spanish
DETECTION OF MINIMAL RESIDUAL DISEASE IN Myeloma Group (PETHEMA/GEM) used four-color flow
BONE MARROW cytometry to study MRD among 295 patients with newly
Bone marrow examination has been the cornerstone diagnosed MM receiving uniform treatment including ASCT,
of disease assessment in the absence of a measurable and they demonstrated that MRD was one of the most
monoclonal protein in serum or urine, whether this repre- important predictors of outcome.10 Minimal residual dis-
sents nonsecretory disease or complete response to ther- ease negativity at day 100 after ASCT correlated with im-
apy. In recent years, increasingly sensitive assays have proved PFS and OS; furthermore, the impact of MRD
been adopted for the evaluation of bone marrow aspi- negativity was equally relevant among patients who had
rates, including multiparameter flow cytometry (MFC), achieved a conventional CR. Similarly, Rawstron et al eval-
allele-specific oligonucleotide polymerase chain reaction uated the role of six-color MFC in the assessment of MRD at
(ASO-PCR), and next-generation sequencing (NGS) of im- various stages of therapy among patients with newly di-
munoglobulin gene sequences, in an effort to increase the agnosed MM who enrolled on the MRC IX clinical trial.13
sensitivity of detection of MM cells. Such methods allow Among patients undergoing ASCT, absence of MRD at day
examination of several hundred thousand to millions of 100 was associated with substantially improved PFS, irre-
bone marrow cells (or their corresponding amount of DNA) spective of cytogenetics or achievement of CR. Of note, in all
per assay and can provide a quantitative assessment of any these studies, three- to six-color MFC approaches with a
residual tumor cells in the bone marrow in a relatively short sensitivity of one in 10,000 myeloma cells were used.
timeframe. Thus, the recent advances in the flow technology allowing
interrogation of several million cells have significantly
improved the sensitivity of the assay, particularly when
Multiparameter Flow Cytometric Methods for
combined with simultaneous usage of eight or more colors/
Minimal Residual Disease Detection
markers for an increased specificity. Recent consensus in-
First-generation MFC methods. Although MFC-based as-
dicates that such approaches are optimally suited for MRD
sessment of bone marrow has been conducted in MM for a
testing in the settings of multiple myeloma.20
number of years, it is only recently that the technology has
gained wide acceptance in the routine work-up of patients
Next-generation flow. Recent attempts at standardization
with MM. The most commonly used surface markers used
and automated read outs makes MFC an attractive test for
sensitive, routine detection of MRD in the bone marrow
compartment.21,22 However, to have uniform MFC-based
KEY POINTS MRD response criteria, it is mandatory to have consensus in
the way MRD is evaluated; accordingly, concerted effort has
Traditional complete response is not adequate for been put into effect to standardize the flow-based ap-
predicting long-term outcome or acting as an endpoint proaches and remove the subjectivity introduced by indi-
for clinical studies. vidual interpretations by defining the reagent characteristics,
Newer technologies allow detection of myeloma cells in
defining the acquisition and plasma cell identification pa-
bone marrow with great sensitivity.
All studies confirm that patients with MRDnegative
rameters, and introducing novel common data analysis tools.
status have improved EFS and OS. The current EuroFlow next-generation flow (NGF) method
MRD status should become a standard in clinical studies for MRD detection in MM relies on two 8-color combinations
and be considered for intervention decision making in that combine surface antigens for the identification of
the future. phenotypically aberrant clonal plasma cells, as well as
intracytoplasmic kappa and lambda light chain expression to

e426 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


MINIMAL RESIDUAL DISEASE BY NEXT-GENERATION SEQUENCING

confirm their clonality. This two-tube NGF approach has now marrow samples from baseline and from the time of very
been extensively validated (over 1,000 MRD samples). It is good partial response or CR from patients enrolled in the
very robust and improves reliability, consistency, and sensi- GEM trials were studied by NGS to identify a tumor clo-
tivity because of the acquisition of a greater number of cells. notype at baseline and then re-evaluated for the presence of
One of the most attractive features of the eight-color the same clonotype in the subsequent sample. A dominant
method is its balance between effectiveness (sensitivity plus MM clone could be identified at baseline in 91% of the
specificity) and wide availability because eight-color in- patients. Of the 121 patients with an identifiable clonotype
struments are commonly used in many hospitals. To improve at baseline, 110 had follow-up samples, of which 73%
efficiency in the laboratory and to reduce costs, it is antici- remained positive by sequencing at MRD levels of one in
pated that alternate single-tube 10- and 14-color methods will 100,000 MM cells. Among patients who achieved a very
be suggested by some centers. The single-tube approach will good partial response, an MRD-negative status (less than
undergo detailed cross-validation with the reference NGF one tumor cell in 100,000 cells) bestowed a better PFS and
method for standardization. To fulfill criteria for NGF, doc- OS. Among the group of patients with a CR, a higher pro-
umentation of cross-validation with reference NGF, ongoing portion of cases had MRD negativity that also associated
quality control assessment, routine assessment of more than with improved PFS. Korde et al also used NGS in 43 patients
5 million cells to estimate MRD, and a sensitivity of one in with MM treated with carfilzomib, lenalidomide, dexa-
100,000 or higher is necessary. methasone, and they observed a 12-month PFS for MRD-
negative versus MRD-positive patients of 100% and 79%,
respectively (p , .001).24 Results from the IFM-DFCI trial
Molecular Methods for Minimal Residual Disease that randomly selected 700 patients to receive either eight
Detection cycles of VRD (arm A) or three cycles of VRD, ASCT, and
Allele-specific oligonucleotide polymerase chain reaction. The then two consolidation VRD cycles (arm B).25 All patients
other methodology that has been studied extensively in the then received lenalidomide maintenance for 12 months.
past and for which a wealth of data exist is ASO-qPCR, in- A total of 246 patients were evaluated by NGS before
cluding a few reports in which it has been compared head-to- maintenance and 178 after maintenance. Among the
head with MFC assays. Despite MRD evaluation by ASO-qPCR patients in CR, the 4-year PFS was 83% and 30% for patients
being a sensitive and specific approach, it is applicable in a who were MRD negative and MRD positive, respectively,
lower proportion of patients with MM and is more time- postmaintenance.
consuming when compared with MFC. This technique is now
supplanted by NGS methods.
Comparison of Techniques for Minimal Residual
Next-generation sequencing. Recently, there has been sig- Disease Detection in Bone Marrow
nificant interest in NGS for detection of MM MRD in the As described above, various techniques have been studied
bone marrow, and data supporting its use continue to for detection of MRD. Each of these techniques (based on
emerge. Most of the published data have been generated the plasma cell phenotype and/or plasma cell genotype) has
using the LymphoSIGHT platform (Sequenta/Adaptive Inc.), advantages and disadvantages that must be taken into
which utilizes consensus primers for the amplification and consideration (Table 1). The ideal MRD test should fulfill
sequencing of immunoglobulin gene segments. Specifically, several relevant characteristics: (1) high applicability (use-
genomic DNA is amplified using locus-specific primers ful among most patients), (2) highly sensitive and specific,
designed for IGH-VDJH or IGH-DJH or IGK. Once amplified, the (3) excellent feasibility (results can be obtained for most
immunoglobulin gene DNA is sequenced and the frequencies patients), (4) easily accessible, (5) requirement for a limited
of the different clonotypes in the sample are determined. sample that can be transported with ease, (6) reproduc-
Patients with detectable MM clones (. 5%) at baseline can ibility, and (7) proven clinical value.
then be studied at subsequent time points to determine the While none of these tests fully satisfies all these ideal
presence and quantity of that particular clone using se- characteristics at the current time, NGS and NGF fulfill most
quencing approaches. Ladetto et al compared IGH gene of them and can be translated into an advanced platform
based MRD detection by ASO-qPCR and NGS to assess that can be uniformly applied across institutions and coun-
whether NGS could overcome some of the limitations of ASO- tries. In terms of sensitivity, the different methodologies
qPCR and further increase its sensitivity and specificity.23 have been reported to have variable levels of sensitivity.
Clonotypes identified by NGS and ASO-qPCR were identical or Both NGF and NGS have been reported to have the ability to
greater than 97% homologous in 96% of cases, and both tools detect one in 100,0001,000,000 MM cells in more recent
appeared to have a sensitivity of approximately one in studies. We strongly encourage the inclusion of both meth-
100,000, but NGS has the added advantage of not requiring odologies in prospective trials, to the extent possible, to
patient-specific primers. Recent studies show that NGS can collect data that would allow us to better understand the
achieve a sensitivity of one in 1,000,000. advantages and disadvantages of the individual approaches
Next-generation sequencing has been compared with as well as the sensitivity of detection required in various
first-generation four-color MFC in one Spanish study.24 Bone clinical settings.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e427


HERVE AVET-LOISEAU

Table 1. Comparison of Different Bone Marrow Minimal Residual Disease Techniques


Characteristic ASO-qPCR MFC VDJ Sequencing
Applicability 60%70% Nearly 100% $ 90%
Need for Baseline Yes, requires production of Not required, abnormal plasma Baseline samples required for
Sample patient-specific probes cells can be identified in any sample identification of the dominant
by their distinct immunophenotypic clonotype; alternatively, a stored
pattern vs. normal plasma cells sample from time-point with
detectable disease can be used
to define baseline status
Sample Requirements , 1 million cells sufficient Recent, sensitive methodology , 1 million cells sufficient
requires over 5 million cells
to be analyzed
Sample Processing Can be delayed Needs assessment within 2448 Can be delayed; can use both fresh
hours; requires a fresh sample and stored samples
Sample Quality Not possible. Additional Immediate with global bone Not possible. Additional studies
Control studies required marrow cell analysis required.
Sensitivity $ 1 in 100,000 $ 1 in 100,000 $ 1 in 100,000
In frequent clones may not
be evaluable
Turnaround and Labor intensive, requires Can be done in few hours, automated Typically days for turnaround,
Complexity development of patient-specific software available requires intense bioinformatics
primers/probes, may take several support. Use of local laboratories
days may speed up turnaround.
Clonal Consideration Detects only single clone or Considers all clones with similar Can take into account all minor
some of the related clones phenotype but evolving clone with clones with infrequent occurrence
change in phenotype may not be
evaluable
Abbreviations: ASO-qPCR, allele-specific oligonucleotide polymerase chain reaction; MFC, multiparameter flow cytometry.

DETECTION OF BONE AND EXTRAMEDULLARY double ASCT among 192 patients with newly diagnosed MM.
DISEASE Persistence of a standard uptake value (SUV) of greater than
The current approaches for the detection and measurement 4.2 after induction therapy predicted for early relapse, and
of tumor burden after therapy relies on bone marrow as- the 4-year PFS and OS were superior for those patients
sessment. However, bone marrow involvement in MM can with a negative PET-CT at day 100 post-ASCT. Recently, the
be heterogeneous, thus increasing the likelihood of a false- Italian group presented updated results from their study of
negative assessment. More importantly, it does not allow 282 patients with newly diagnosed MM who had PET imaging
detection of the disease outside the bone marrow. The rec- at baseline. After treatment, PET negativity was achieved in
ognition of extramedullary disease is increasingly encoun- 70% of patients, whereas conventionally defined CR was
tered in the clinic as a result of more sensitive imaging studies achieved in 53%. Among the patients in CR, 29% still had
and extended survival of patients with MM. positive PET, translating to an inferior PFS (44 vs. 84 months)
18 and OS (5-year estimate of 70% vs. 90%). In this study,
F-fluorodeoxyglucose PET (FDG-PET) is a powerful tool
used to assess tumor metabolic activity and the effect of persistence of SUVmax greater than 4.2 was the single factor
therapy on the tumor cell metabolism. Multiple studies have independently associated with skeletal progression in the
demonstrated a prognostic capacity related to the detection absence of conventional measures of disease progression.
of PET-positive lesions in patients with MM at diagnosis and Similar data have been recently reported by the French group.
at time of relapse.26-31 In addition to the metabolic as- In the IFM-DFCI trial, which randomized high-dose melphalan,
sessment, the low-dose CT that is typically done for local- PET-CT positivity premaintenance was predictive of a shorter
ization along with FDG-PET allows for a sensitive screen for PFS and OS.
the detection of MM-associated bone disease. In an initial
study from the University of Arkansas, complete FDG sup- INCORPORATION IN CLINICAL PRACTICE
pression in the focal lesions before first transplantation was Having established that achieving MRD-negative status
associated with substantially improved survival outcomes. provides significantly superior survival outcome and that we
In a subsequent study in the context of Total Therapy (TT) 3, can reliably measure MRD-negative status with a sensitivity
it was shown that persistent FDG avidity 7 days after initi- of one cell in 1 million, the next important question in
ation of therapy was associated with poorer survival out- myeloma is how MRD status will inform our therapeutic
comes and was independent of other prognostic factors. In decision algorithm, to both provide the most effective
an Italian study, PET-CT was performed at diagnosis, after therapy and develop strategies to individualize therapy. The
thalidomide-dexamethasone induction therapy, and after questions to be answered are whether outcome is similar

e428 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


MINIMAL RESIDUAL DISEASE BY NEXT-GENERATION SEQUENCING

between patients achieving early MRD negativity versus begging to play an important role in predicting superior
those who achieve late MRD-negative status following outcome. Minimal residual disease may serve as a biomarker
multiple interventions. This may help determine the need to inform therapy and as a surrogate for OS. Specifically,
for consolidation therapy for a given patient. The next achieving MRD-negative status may become a goal of future
question is whether patients with MRD-negative status can studies using induction, transplant, consolidation, and/or
get similar benefit with less intense (one drug vs. two drugs) maintenance therapies. With available technologies and
or shorter duration of maintenance. The last questions will ease of MRD measurement, it is now time to carry out
be whether the same principles apply to older individuals additional large prospective studies to define the clinical
and whether MRD negativity should also be an endpoint for significance of MRD and its impact on patient outcome in
patients with relapsed disease. myeloma. In our quest toward personalizing therapy for
patients, as in chronic myeloid leukemia, it may now be
CONCLUSION possible to both assess and monitor MRD using standardized
In the era of novel agents and combination therapies assays and decide both intensity and length of therapy for
achieving very high conventional CR rates, MRD status is individual patients to improve patient outcome.

References
1. Kumar SK, Dispenzieri A, Lacy MQ, et al. Continued improvement in disease by multiparameter flow cytometry predict unsustained com-
survival in multiple myeloma: changes in early mortality and outcomes plete response after autologous stem cell transplantation in multiple
in older patients. Leukemia. 2013;28:122-128. myeloma. Blood. 2012;119:687-691.
2. Rajkumar SV, Dimopoulos MA, Palumbo A, et al. International Myeloma 13. Rawstron AC, Child JA, de Tute RM, et al. Minimal residual disease
Working Group updated criteria for the diagnosis of multiple myeloma. assessed by multiparameter flow cytometry in multiple myeloma:
Lancet Oncol. 2014;15:e538-e548. impact on outcome in the Medical Research Council Myeloma IX Study.
3. Durie BG, Harousseau JL, Miguel JS, et al; International Myeloma J Clin Oncol. 2013;31:2540-2547.
Working Group. International uniform response criteria for multiple 14. Puig N, Sarasquete ME, Balanzategui A, et al. Critical evaluation of ASO
myeloma. Leukemia. 2006;20:1467-1473. RQ-PCR for minimal residual disease evaluation in multiple myeloma.
4. Attal M, Lauwers-Cances V, Marit G, et al; IFM Investigators. Lenali- A comparative analysis with flow cytometry. Leukemia. 2013;28:391-397.
domide maintenance after stem-cell transplantation for multiple my- 15. Martinez-Lopez J, Lahuerta JJ, Pepin F, et al. Prognostic value of deep
eloma. N Engl J Med. 2012;366:1782-1791. sequencing method for minimal residual disease detection in multiple
5. McCarthy PL, Owzar K, Hofmeister CC, et al. Lenalidomide after stem-cell myeloma. Blood. 2014;123:3073-3079.
transplantation for multiple myeloma. N Engl J Med. 2012;366:1770-1781. 16. Paiva B, Puig N, Garcia-Sanz R, et al. Grupo Espanol de Mieloma
6. Jakubowiak AJ, Dytfeld D, Griffith KA, et al. A phase 1/2 study of /Programa para el Estudio de la Terapeutica en Hemopatias Malignas
carfilzomib in combination with lenalidomide and low-dose dexa- cooperative study g. Is this the time to introduce minimal residual
methasone as a frontline treatment for multiple myeloma. Blood. disease in multiple myeloma clinical practice? Clin Cancer Res. 2015;21:
2012;120:1801-1809. 2001-2008.
7. Kumar S, Flinn I, Richardson PG, et al. Randomized, multicenter, phase 2 17. van Dongen JJ, Lhermitte L, Bottcher S, et al; EuroFlow Consortium
study (EVOLUTION) of combinations of bortezomib, dexamethasone, (EU-FP6, LSHB-CT-2006-018708). EuroFlow antibody panels for stan-
cyclophosphamide, and lenalidomide in previously untreated multiple dardized n-dimensional flow cytometric immunophenotyping of normal,
myeloma. Blood. 2012;119:4375-4382. reactive and malignant leukocytes. Leukemia. 2012;26:1908-1975.
8. Cavo M, Tacchetti P, Patriarca F, et al; GIMEMA Italian Myeloma 18. Kalina T, Flores-Montero J, van der Velden VH, et al; EuroFlow Con-
Network. Bortezomib with thalidomide plus dexamethasone compared sortium (EU-FP6, LSHB-CT-2006-018708). EuroFlow standardization of
with thalidomide plus dexamethasone as induction therapy before, and flow cytometer instrument settings and immunophenotyping pro-
consolidation therapy after, double autologous stem-cell transplan- tocols. Leukemia. 2012;26:1986-2010.
tation in newly diagnosed multiple myeloma: a randomised phase 3 19. Flanders A, Stetler-Stevenson M, Landgren O. Minimal residual disease
study. Lancet. 2010;376:2075-2085. testing in multiple myeloma by flow cytometry: major heterogeneity.
9. Harousseau JL, Attal M, Avet-Loiseau H. The role of complete response Blood. 2013;122:1088-1089.
in multiple myeloma. Blood. 2009;114:3139-3146. 20. Stetler-Stevenson M, Paiva B, Stoolman L, et al. Consensus guidelines
10. Paiva B, Vidriales MB, Cervero J, et al; GEM (Grupo Espan ~ol de MM)/ for myeloma minimal residual disease sample staining and data ac-
PETHEMA Cooperative Study Groups. Multiparameter flow cytometric quisition. Cytometry B Clin Cytom. Epub 2015 Jul 6.
remission is the most relevant prognostic factor for multiple myeloma 21. van Dongen JJ, van der Velden VH, Bruggemann M, et al. Minimal residual
patients who undergo autologous stem cell transplantation. Blood. disease diagnostics in acute lymphoblastic leukemia: need for sensitive,
2008;112:4017-4023. fast, and standardized technologies. Blood. 2015;125:3996-4009.
11. Paiva B, Martinez-Lopez J, Vidriales MB, et al. Comparison of immu- 22. Paiva B, van Dongen JJ, Orfao A. New criteria for response assessment:
nofixation, serum free light chain, and immunophenotyping for re- role of minimal residual disease in multiple myeloma. Blood. 2015;125:
sponse evaluation and prognostication in multiple myeloma. J Clin 3059-3068.
Oncol. 2011;29:1627-1633. 23. Ladetto M, Bruggemann M, Monitillo L, et al. Next-generation se-
12. Paiva B, Gutierrez NC, Rosin ~ol L, et al; PETHEMA/GEM Cooperative quencing and real-time quantitative PCR for minimal residual disease
Study Groups. High-risk cytogenetics and persistent minimal residual detection in B-cell disorders. Leukemia. 2013;28:1299-1307.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e429


HERVE AVET-LOISEAU

24. Korde N, Roschewski M, Zingone A, et al. Treatment with carfilzomib- 28. Bartel TB, Haessler J, Brown TL, et al. F18-fluorodeoxyglucose positron
lenalidomide-dexamethasone with lenalidomide extension in patients emission tomography in the context of other imaging techniques and
with smoldering or newly diagnosed multiple myeloma. JAMA Oncol. prognostic factors in multiple myeloma. Blood. 2009;114:2068-2076.
2015;1:746-754. 29. van Lammeren-Venema D, Regelink JC, Riphagen II, et al. F-fluoro-
25. Avet-Loiseau H, Corre L, Lauwers-Cances V, et al. Evaluation of minimal deoxyglucose positron emission tomography in assessment of myeloma-
residual disease (MRD) by next generation sequencing (NGS) is highly related bone disease: a systematic review. Cancer. 2012;118:1971-1981.
predictive of PFS in the IFM/DFCI 2009 trial. Blood. 2015;126:191. 30. Elliott BM, Peti S, Osman K, et al. Combining FDG-PET/CT with labo-
26. Zamagni E, Patriarca F, Nanni C, et al. Prognostic relevance of 18-F FDG ratory data yields superior results for prediction of relapse in multiple
PET/CT in newly diagnosed multiple myeloma patients treated with up- myeloma. Eur J Haematol. 2011;86:289-298.
front autologous transplantation. Blood. 2011;118:5989-5995. 31. Zamagni E, Nanni C, Mancuso K, et al. PET/CT improves the definition of
27. Usmani SZ, Mitchell A, Waheed S, et al. Prognostic implications of serial complete response and allows to detect otherwise unidentifiable
18-fluoro-deoxyglucose emission tomography in multiple myeloma skeletal progression in multiple myeloma. Clin Cancer Res. 2015;21:
treated with total therapy 3. Blood. 2013;121:1819-1823. 4384-4390.

e430 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


NEW TARGETS AND AGENTS IN HIGH-RISK MULTIPLE MYELOMA

New Targets and New Agents in High-Risk Multiple Myeloma


Ajay K. Nooka, MD, FACP, and Sagar Lonial, MD

OVERVIEW

Advances in the treatment of multiple myeloma have resulted in dramatic improvements in outcomes for patients. The
newly emerging profiling of mutations emerging as a consequence of large prospective sequencing studies such as the
CoMMpass Study or other efforts from European investigators are not further helping to define the place and role for
personalized medicine in myeloma. While mutations such as NRAS, KRAS, and BRAF do occur in myeloma, it is not clear that
targeting them as a single drug strategy will result in meaningful responses or durations of response. Personalized medicine
in multiple myeloma at this time likely entails the use of risk-based approaches for maintenance therapy, the use of current
biology-based treatments such as proteasome inhibitors, and immunomodulatory agents, with an eye towards the use of
mutation-specific treatments in the setting of minimal residual disease or in concert with biology-based treatments overall.

T he improvement in outcomes of patients with high-risk


myeloma in recent years can be attributed to a con-
fluence of numerous factors. First, improved stratification of
(CR) more important than in the high-risk cohort. Attaining
CR using agents with novel mechanisms of action is critical to
success. Second, the treatment approach used must be well
patients with myeloma by their risk status has allowed us to tolerated and not induce further genomic changes leading to
better define the high-risk myeloma subset. Improved drug resistance. This is important because long-term du-
stratification enabled us to specifically focus on developing ration of therapy is a critical part of this strategic approach
treatment algorithms aimed at delivering uninterrupted for high-risk patients. Although the high-risk myeloma
intensive antimyeloma therapies, with resultant improve- spectrum has been defined mostly by cytogenetics, a few
ment in survival outcomes. Second, the availability of novel groups have identified high-risk patients based on the dis-
therapies with proven clinical efficacy among patients with ease presentation as extramedullary myelomathat is,
myeloma, especially among high-risk patients, led to un- plasma cell leukemiaand also by gene expression pro-
precedented survival improvements. More importantly, vast filing. We have incorporated these high-risk attributes in the
clinical experience with these newer agents has not only review of individual drugs
enabled us to better manage the toxicities of new agents,
but has also enabled us to consciously withdraw from the Immunomodulatory Drugs
excessive use of cytotoxic agents, such as alkylating agents, Thalidomide. The use of thalidomide is currently limited
among these patients with a highly genomically unstable because of the availability of next-generation agents with
disease. This has further led to minimizing regimen-related greater efficacy and safety. In the context of high-risk pat-
toxicity and decreased the incidence of alkylator-related ents, thalidomide usage has not been shown to improve
secondary myelodysplastic syndromes. In this article, we prognosis. The MRC-IX trial evaluated its role among pa-
describe the clinical experience of effective antimyeloma tients with adverse immunofluorescence with fluorescence
agents among high-risk patients and provide the rationale in situ hybridization (iFISH; defined as gain(1q), t(4;14),
for incorporating these most effective combination regi- t(14;16), t(14;20), del(17p), and del(1p32)) demonstrated no
mens in the treatment of patients with high-risk myeloma. progression-free survival (PFS) benefit and, in fact, worse
overall survival (OS).1 The poor survival among the high-risk
CURRENTLY APPROVED AGENTS AND THEIR patients receiving thalidomide maintenance was attributed
ACTIVITY IN HIGH-RISK MYELOMA to the clonal advantage of the high-risk patients, the se-
Successfully treating high-risk patients requires two im- lective pressure of thalidomide accounting for acquired drug
portant steps. First, among all patients with myeloma, no- resistance, and the subsequent poor survival after relapse.
where is the importance of achieving a complete response Conflicting data were seen from the other groups that have

From the Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Sagar Lonial, MD, Emory University, 1365 Clifton Rd., Building C, Room 3003, Atlanta, GA 30322; email: sloni01@emory.edu.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e431


NOOKA AND LONIAL

used metaphase cytogenetic abnormalities to define high poor prognosis conferred by t(4;14). Although additional
risk. The OS of the thalidomide arm of the Total Therapy 2 studies have confirmed that lenalidomide/dexamethasone
(TT2) regimen was superior among patients exhibiting cy- is a suboptimal regimen among patients with del(17)p, the
togenetic abnormalities, but when examined in the context positive impact of this regimen on patients with t(4;14)
of gene expression profiling data, the benefit of thalidomide has never been recapitulated in any other trial to date.8,9
was limited to those patients with low-risk disease defined In the maintenance setting, Attal at al presented their
by gene expression profiling.2 Several other studies have findings comparing fixed-duration lenalidomide mainte-
repeatedly demonstrated the inability to induce responses nance placebo post-transplant.10 They reported that the
with the use of thalidomide among patients with high-risk presence of t(4;14) or del(17)p are independent com-
myeloma presenting with extramedullary myeloma.3,4 In pared with risk factors for poor survival on the multi-
current practice, thalidomide has a nominal role in the variate analysis, but they did not comment on the impact
treatment of patients with high-risk myeloma. of maintenance therapy on these high-risk patients.10

Lenalidomide. The use of lenalidomide and dexametha- Pomalidomide. The influence of cytogenetics in patients
sone as induction therapy among high-risk patients did not with relapsed or refractory myeloma treated with pomali-
yield superior results. Among 40 patients (16%) exhibiting domide and low-dose dexamethasone have been evaluated
t(4;14), t(14;16), t(14;20), or del(17)p on FISH analysis, the by Dimopoulos and colleagues as a subgroup analysis from
median PFS was 1.4 years (compared with standard-risk the registration phase III MM003 trial. The results dem-
median PFS of 3.4 years; p = .0002). Not surprisingly, the onstrate quite a variance between the outcomes of pa-
5-year OS was 47% versus 77% (p = .0001), favoring the tients with del(17p) and t(4;14). The median PFS rates for
standard-risk patients.5 Clearly, lenalidomide/dexamethasone standard risk versus del(17p) versus t(4;14) were 4.2, 4.6,
was deemed as a suboptimal induction regimen, and a shift and 2.8 months, respectively; the median OS rates were
toward triplet induction regimen has been made by all 14.0, 12.6, and 7.5 months, respectively, suggesting that
parties, at least for the high-risk subgroup. The results from the pomalidomide/low-dose dexamethasone regimen may
the FIRST trial also did not demonstrate any survival benefit overcome the poor prognosis of patients with del(17p) but
for transplant-ineligible patients exhibiting high-risk fea- not t(4;14).11 Similar results were demonstrated in a dedi-
tures treated with lenalidomide/dexamethasone continu- cated high-risk trial from the Intergroupe Francophone du
ously (Table 1).6 These results suggest that a modified Myelome; patients with del(17)p and t(4:14) treated with
combination regimen such as lenalidomide, bortezomib, and pomalidomide/low-dose dexamethasone achieved median
dexamethasone (RVD)-lite might be an appropriate in- time to progression (TTP) for del(17p) and t(4;14) of 7.3
duction regimen for high-risk transplant-ineligible pa- versus 2.8 months, and the 8-month OS was 41% versus
tients, though this has yet to be demonstrated. Among 12.4%, respectively.12 This regimen could serve as an in-
patients with relapsed or refractory myeloma, Reece et al teresting backbone upon which to further build combina-
evaluated the impact of cytogenetics, using lenalidomide/ tions that would benefit patients with del(17)p and t(4;14).
dexamethasone in relapsed myeloma.7 Consistent with Based on this concept, IFM-2014-01 is evaluating the regi-
other experiences, patients with del(17)p experienced men of pomalidomide/low-dose dexamethasone with the
a worse outcome, with a median OS of 4.7 months. In addition of ixazomib exclusively among patients with re-
contrast, patients with t(4;14) experienced a median OS lapsed or refractory high-risk myeloma who have received
similar to patients without any cytogenetic abnormalities, more than two lines of therapy with an enrollment goal of
suggesting lenalidomide/dexamethasone may abrogate the 50 patients.

Proteasome Inhibitors
KEY POINTS Bortezomib. The UAMS group as part of the Total Therapy 3
(TT3) experience incorporated bortezomib in the induction
Identification of risk status should be performed on all treatment of myeloma. Compared with TT2, a meaningful
patients at diagnosis. benefit was apparent among younger patients with high-risk
Immune-based treatments may provide unique ways by myeloma defined by GEP70. The 2-year event-free survival
which to target high-risk myeloma. rates (TT3 vs. TT2: 68% vs. 30%) and OS rates (TT3 vs. TT2:
Combination therapy with currently available novel 75% vs. 50%) strongly favored inclusion of bortezomib in
agents may help mitigate the outcomes of high-risk
induction therapy for patients with high-risk myeloma.13
disease.
Cellular immune-based treatments are currently in
Additionally, in the earlier relapsed trials with bortezomib,
evolution with rapidly expanding targets. Jagannath et al evaluated the utility of bortezomib among
Specific mutation-driven treatments are in high-risk cohorts (defined as del13 by FISH and/or meta-
development and may offer benefit as part of phase cytogenetics). This analysis suggested that bortezo-
a combination approach with biology-based treatments. mib may be able to deliver responses uniformly, irrespective
of the risk status.14 Among patients with newly diagnosed

e432 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


TABLE 1. Outcomes Among Patients With High-Risk Myeloma From Newly Diagnosed Trials
Std Risk, High Risk,
No. No. t(4;14), del(17)p, Median
Study, Year/ Regimen (No. Patients Patients No. Patients No. Patients Std Risk t(4;14) del(17)p Follow-
Phase Patients) (%) (%) (%) (%) VGPR VGPR VGPR Std Risk PFS t(4;14) PFS del(17)p PFS Std Risk OS t(4;14) OS del(17)p OS up
Cavo VTD SCT 3 2 NR 53 (24) NR 15 (7) NR NR NR 74 69 69 NR NR NR NR
et al,16 VTD (236) vs.
NR 57 (26) 17 (8) 63 (3 y PFS) 37 (3 y PFS) 37 (3 y PFS)
2010/III TD SCT 3
2 TD (238)
Avet-Loiseau VDSCT (507) 106 (21) 54 (11) NR NR NR 36 mo 28 mo 14 mo 79% (4 y OS) 63% (4 y OS) 50% (4 y OS) 24
et al,15
2010/III
Sonneveld PAD SCT 3 NR NR 50/295 (17%) 39/312 (13%) 76% NR NR 27% 16% 22% 72% 52% 65% 94
et al,18 2 V (413) vs.
NR 56% 24% (5 y PFS) 8% (5 y PFS) 5% (5 y PFS) 64% (5 y OS) 33% (5 y OS) 18% (5 y OS)
2015/III VADSCT 32T
(4141)
Mateos VMP Rd 66 19 NR 63% 68% NR 32 mo 30 mo 30 mo 65% 45% 45% 37
et al,59 (sequential)
76 13 66% 46% 36 mo 24 mo 24 mo 72% (4 y OS) 27% (4 y OS) 27% (4 y OS)
2015/III (118) vs. VMPRd
alternating (115)
Avet-Loiseau Rd continuous 620 (81.3) 142 (18.7) NR 49.3% 30.2% 30.2% 31.1% 8.4% 8.4% 77.1% 40.7% 40.7% NR
et al,22 (248) vs. Rd18
47.3% 34.7% 34.7% 21.2% 17.5% 17.5% 71% 39.6% 39.6%
2015/III* (261) vs. MPT
(253) 38.8% 10.6% 10.6% 24.9% (3 y PFS) 14.6% (3 y PFS) 14.6% (3 y PFS) 64.8% (3 y OS) 46.8% (3 y OS) 46.8% (3 y
OS)
Nooka RVD SCT RVD N/A 45 (100) 2 (5) 19 (42) N/A 96% 96% N/A 32 mo 28 mo N/A 93% (3 y OS) 94% (3 y OS) 26
et al,19 (45)
2013
Abbreviations: VGPR, very good partial response; PFS, progression-free survival; OS, overall survival; mo, months; VTD, bortezomib, thalidomide, and dexamethasone; SCT, stem cell transplant; NR, not reported; TD, thalidomide and
dexamethasone; VD, bortezomib and dexamethasone; y, years; V, bortezomib; T, thalidomide; VMP, bortezomib/melphalan/prednisone; Rd, lenalidomide and dexamethasone; MPT, melphalan/thalidomide/prednisone; RVD, lenalidomide,
bortezomib, and dexamethasone.
*Cytogenetics available for 762 of 1,623 patients in the FIRST trial.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK


e433
NEW TARGETS AND AGENTS IN HIGH-RISK MULTIPLE MYELOMA
NOOKA AND LONIAL

myeloma, several trials evaluated the impact of bortezomib lenalidomide/dexamethasone was 59.6% versus 73.5%,
on high-risk patients. From the IFM trial that compared respectively. The median PFS among patients who received
bortezomib and dexamethasone to the chemotherapy KRD with standard risk versus t(4;14) versus del(17)p was
combination regimen vincristine, doxorubicin, and dexa- 29.6, 23.1, and 24.5 months, and for Rd was 19.5, 16.7, and
methasone as induction therapy before high-dose therapy 11.1 months, respectively (Table 2). Though there are
and autologous transplant, differential outcomes were several limitations to the data set including unavailability of
observed for t(4;14) and del(17)p. Patients with t(4;14) cytogenetics for more than half the patients, these results
appeared to have significantly improved outcomes with suggest the proteasome inhibitor/immunomodulatory drug
bortezomib/dexamethasone as induction therapy compared combination can overcome the poor prognosis conferred
with those with del(17)p (Table 1).15 Another induction trial by these high-risk cytogenetics.22 In another recent trial
evaluated the impact of bortezomib, thalidomide, and evaluating carfilzomib in combination with pomalidomide/
dexamethasone (VTD) versus thalidomide and dexametha- low-dose dexamethasone (KPD) for relapsed/refractory my-
sone on outcomes of high-risk cohorts including patients eloma, high ORRs were seen with this combination, even
with newly diagnosed myeloma. Cavo and colleagues among patients with high-risk genetics (Table 2).23
demonstrated that VTD with bortezomib as part of con-
solidation could eliminate the negative impact of t(4;14), Ixazomib. Ixazomib is the most recently approved oral
although this was not the case with the thalidomide/ proteasome inhibitor that has been evaluated in the
dexamethasone arm.16 A subsequent trial, HOVON/GMMG, TOURMALINE-MM1 trial. This phase III study randomly se-
also evaluated the impact of bortezomib as part of induction lected 722 patients to receive lenalidomide/dexamethasone
and as maintenance.17 As illustrated in Table 1, the use of or to receive ixazomib in combination with lenalidomide/
bortezomib as maintenance therapy was able to reduce the dexamethasone (IRD). The combination of IRD was shown
negative impact of high-risk cytogenetics on outcomes. to be superior in delivering high or at least very good
Prolonged bortezomib treatment mostly abrogated the partial response (VGPR) rates among both high-risk and
del(17p) effect on PFS and OS but had limited benefit among standard-risk patients. At least VGPR with IRD for standard
patients with t(4;14).18 Our group evaluated the use of the risk versus t(4;14) versus del(17)p was 51%, 53%, and 39%,
combination of RVD at modified doses as a post-transplant and for lenalidomide/dexamethasone was 44%, 28%, and
maintenance strategy among patients with high-risk mye- 21%, respectively. The median PFS with IRD for standard
loma.19 The use of RVD maintenance after transplant sig- risk versus t(4;14) versus del(17)p was 20.6, 18.5, and
nificantly improved PFS and OS for high-risk patients, greater 21.4 months, and for lenalidomide/dexamethasone was
than what has been reported with single-agent maintenance 15.6, 12, and 9 months, respectively (Table 2).24 These re-
with either lenalidomide or bortezomib. sults also suggest the same observations seen with KRD,
a proteasome inhibitor/immunomodulatory drug combi-
Carfilzomib. The efficacy of carfilzomib as a single agent was nation is the most optimal regimen to deliver the best results
evaluated among patients with high-risk myeloma in the PX- for patients with high-risk cytogenetics.
171-003-A1 study. Patients with t(4;14) had the highest
overall response rate (ORR) of 38.9%. Those with del(17)p
had the lowest ORR of 16.7%. Among high-risk patients, the
IMMUNE-BASED APPROACHES
Immunotherapeutic approaches as antimyeloma therapies
median PFS and OS trended higher among the patients with
aimed at targeting the malignant plasma cell have been
t(4;14). Among patients with t(4;14) only, the OS was
explored for some time with limited success. Though iden-
15.8 months, and, among all high-risk patients,20 the OS was
tifying the ideal target was a major challenge, the current
shorter compared with the standard-risk patients (9 vs.
clinical activity using the monoclonal antibodies targeting
23 months; Table 2). In the ENDEAVOR trial, a phase III
SLAMF7 and CD38 demonstrate much promise. Acceptable
study that compared two proteasome inhibitors,21 carfil-
safety in combination with other antimyeloma agents allow
zomib with dexamethasone was superior to bortezomib/
for their utility in treating myeloma, even among the pa-
dexamethasone among all patients with myeloma irrespective
tients heavily pretreated for myeloma. Furthermore, drug
of baseline cytogenetic risk status. However, the median
development using novel immune-based antimyeloma ap-
PFS for high-risk patients was lower compared with the
proaches such as checkpoint inhibitors and cellular therapies
standard-risk patients. Carfilzomib/dexamethasone im-
are highly promising.
proved the outcome of high-risk patients but did not
overcome the adverse impact of high-risk cytogenetics. A
similar experience from the ASPIRE trial that compared Monoclonal Antibodies
carfilzomib, lenalidomide, and dexamethasone (KRD) with Elotuzumab. Elotuzumab is a recently approved monoclo-
lenalidomide/dexamethasone in a phase III trial dem- nal antibody targeting the surface protein SLAMF7. Its U.S.
onstrated KRD exerting higher-quality responses both Food and Drug Administration approval in combination with
among the high-risk and standard-risk patients. The lenalidomide and dexamethasone for the treatment of
overall responses among the high-risk versus standard- patients with multiple myeloma who have received one to
risk patients with KRD was 79.2% versus 91.2% and for three prior lines of therapies was based on the results

e434 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


TABLE 2. Outcomes Among Patients With High-Risk Myeloma From Relapsed/Refractory Trials
Std Risk, High Risk, del(17)p,
Regimen No. No. t(4;14), No. Median
Study, Year/ (No. Patients Patients No. Patients Patients Std Risk t(4;14) del(17)p PFS Std Risk t(4;14) del(17)p OS Std Risk t(4;14) del(17)p Follow-up
Phase Patients) (%) (%) (%) (%) VGPR VGPR VGPR (mo) PFS PFS PFS (mo) OS OS OS (mo)
Lonial ERd (321) 30 (9) 102 (32) NR NR NR 19.4 20.34 18.46 NR NR NR NR 24.5
et al,25 2015 vs.
31 (10) 104 (32) 14.9 15.74 14.85
Rd (325)
Avet-Loiseau KRd (396) 147 (37) 48 (12) 33 (69) 18 (38) 75.5 60.5% 60.5% 26.3 29.6 23.1 24.1 NR NR NR NR 32
et al,22 vs.
170 (43) 52 (13) 31 (60) 21 (40) 45.3 27% 27% 17.6 19.5 16.7 11.1
2015/III Rd (396)
Moreau IRd (360) vs. 199 (55) 75 (21) NR NR 51% 53% 39% 20.6 20.6 18.5 21.4 NR NR NR NR 23
et al,24 Rd (362)
216 (60) 62 (17) NR NR 44% 28% 15% 14.7 15.6 12.0 9.7
2015/III
Chng Kd (464) vs. 284 (61) 97 (21) NR NR 56.7% 44.3% 44.3% 18.7 NR 8.8 8.8 NR NR NR NR N/A
et al,21 Vd (465)
291 (63) 113 (24) NR NR 27.4% 27.5% 27.5% 9.4 10.2 6.0 6.0
2015/III
Reece Rd (130) 28 (21.5) 12 (9.2) NR NR NR 7.1 8 (TTP) 2.2 (TTP) 22.7 NR NR 4.7 19.7
et al,7 (TTP)
2009/II
Avet-Loiseau Rd (207) 7 (6) 14 (5) NR NR NR 9.6 5.5 15.1 9.4
et al15, 2010
Leleu Pd (50) N/A 50 (100) 22 (46) 32 (64) N/A 3% 9% 7.3 N/A 41% (8 mo TTP) 12.4% (8 mo TTP) 12 N/A NR NR 10
et al,12 (TTP)
2015/III
Dimopoulos Pd (302) vs. 126 (56) 99 (44) 44 (15) 44 (15) 6.8 2.3 6.8 4 4.2 2.8 4.6 12.6 14 7.5 12.6 15.4
et al,11 HiDex
66 (62) 41 (38) 15 (10) 23 (15) 1.4 0 0 1.9 2.3 1.9 1.1 7.7 9 4.9 7.7
2015/III (153)
Jakubowiak K (266) 167 (72.9) 62 (27.1) 18 (8) 30 (13) 8.4 0 0 4.6 4.5 3.5 19 11.8 7.0 N/A
et al,20
2013/II
Shah et al,60 KPd (32) 10 (31) 3 (10) 5 (15) 16% NR NR 7.2 NR NR 60% (12 mo) 20.6 NR NR 80% (12 mo) 26
2015/I
Abbreviations: VGPR, very good partial response; PFS, progression-free survival; OS, overall survival; mo, months; ERd, elotuzumab in combination with lenalidomide and dexamethasone; Rd, lenalidomide and dexamethasone; NR, not reported;
KRd, carfilzomib, lenalidomide, and dexamethasone; IRd, ixazomib in combination with lenalidomide and dexamethasone; Kd, carfilzomib and dexamethasone; Vd, bortezomib and dexamethasone; TTP, time to progression; Pd, pomalidomide and
low-dose dexamethasone; HiDex, high-dose dexamethasone; K, carfilzomib; KPd, carfilzomib in combination with pomalidomide and low-dose dexamethasone.
Note: unknown cytogenetic status: KRD, 201 (51%) vs. Rd, 174 (44%) from ASPIRE trial; unknown cytogenetic status: IRd, 86 (24%) vs. Rd, 84 (23%) from TOURMALINE-MM1 trial; unknown cytogenetic status: KD, (18%) vs. Vd, (13%) from
TOURMALINE-MM1 trial; unknown cytogenetic status for 47 patients in PX-171-003-A1 study. Available for 229 of 266 patients; MM003 cytogenetics available for 225 (75%) in Pd and 107 (70%) patients.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK


e435
NEW TARGETS AND AGENTS IN HIGH-RISK MULTIPLE MYELOMA
NOOKA AND LONIAL

meeting the primary endpoint of PFS and ORR from the with elotuzumab consolidation to receive lenalidomide
ELOQUENT-2 trial. In this trial, 646 patients were ran- versus lenalidomide with elotuzumab maintenance for the
domly selected to receive elotuzumab in combination with primary endpoint of CR post-induction and 3-year PFS
lenalidomide and dexamethasone (ERD) or lenalidomide/ post = first randomization. In the relapsed setting, trials of
dexamethasone among patients sensitive to lenalidomide. elotuzumab in combination with pomalidomide and low-
The ORRs were 79% and median PFS of 19.4 versus dose dexamethasone (EPD; accrual goal: 60 patients) and
14.9 months (p = .0014) favored the ERD arm.25 The 3-year combination nivolumab to EPD (accrual goal: 40 patients)
extended PFS was recently presented at the 2015 American are underway. More importantly, the safety and tolerability
Society of Hematology (ASH) Annual Meeting and demonstrated of elotuzumab in combination with other monoclonal an-
that more than one-quarter of the patients receiving ERD tibodies such as lirilumab or urelumab are also ongoing.
continue to be free from progression, suggesting long-term
benefits with immune-based approaches. The prespecified Daratumumab. Daratumumab, a humanized IgG1 anti-
interim analysis for OS indicated a strong trend favoring ERD CD38 antibody, is the most active monoclonal antibody in
(43.7 vs. 39.6 months; p = .0257). myeloma to date. It was recently approved in the United
Data regarding the efficacy of elotuzumab among patients at States at the active dose of 16 mg/kg as a single agent
high risk are very encouraging. Approximately 32% of patients resulting in response rates of 29%.28 The combined results of
had the cytogenetic abnormality of del(17)p and 9% of patients GEN501 part 2 and the SIRIUS trials comprising 148 patients
had t(4;14), and the outcomes for these patients have been were presented at ASH 2015. At a median follow-up of
surprisingly comparable with those of the patients at standard 14.8 months, 50% of the responders were progression free
risk. The PFS benefit of the combination of ERD relative to at 1 year. Among most patients, responses deepened with
lenalidomide/dexamethasone was evident among both of continued treatment and were tolerated well. The 1-year OS
these subsets; hazard ratios (HR) for del(17)p and t(4;14) were rate among this heavily pretreated patient population was
0.65 (95% CI, 0.450.94) and 0.53 (95% CI, 0.290.95), re- 69% (95% CI, 60.4%75.6%) and the median OS was
spectively. At a median follow-up of 24 months, the PFS for 19.9 months, demonstrating its efficacy as a single agent.29
standard risk versus del(17)p versus t(4;14) patients were More importantly, the minimal adverse event profile and the
similar at 19.4, 18.46, and 20.34 months, respectively. established safety and tolerability of this antibody (48% of
Based on a phase I trial showing the tolerability of elo- patients had infusion reactions, mostly # grade 2) led to the
tuzumab in combination with bortezomib and dexametha- design of multiple combination trials.
sone (EVD) among heavily pretreated patients,26 another Daratumumab in combination with lenalidomide/
randomized phase II trial was designed that randomly se- dexamethasone, among patients with early-relapse multiple
lected 152 bortezomib-sensitive patients with myeloma who myeloma who have received a median of two lines of therapy,
had received one to three prior lines of therapy to receive demonstrated response rates of 81%, and close to two-thirds
EVD or bortezomib/dexamethasone. At a median follow- of patients who enrolled in the trial appeared to have
up of 27.3 months, a PFS benefit of close to 3 months achieved at least a VGPR (63%).30 In this trial, the toxicity
was observed for the EVD arm (EVD vs. bortezomib/ profile was acceptable and more likely related to the lena-
dexamethasone: 9.7 vs. 6.9 months, respectively; p = .018). lidomide than the daratumumab. Although greater than or
Similarly, the OS trended in favor of EVD (2-year OS, EVD vs. equal to grade 3 or worse toxicities were seen in 88% of
bortezomib/dexamethasone: 73% vs. 66%).27 patients, most of these were at least grade 3 neutropenia
Multiple combination trials of elotuzumab with other (78%) that have resolved with supportive growth factor
antimyeloma agents are ongoing. Study of the safety and therapies. In another recent study presented by Chari et al,
efficacy of elotuzumab in combination with RVD (ERVD) the combination of daratumumab with pomalidomide/low-
among transplant-eligible patients is currently ongoing. dose dexamethasone among patients who have received four
Preliminary data suggest that this as a safe combination. lines of therapy resulted in response rates of 71% and at least
Another trial (S1211) of RVD with or without elotuzumab VGPR rates seen among 43% of patients. More than half (53%)
specifically focused on accruing newly diagnosed patients of patients were progression free at 1-year among this heavily
with high-risk multiple myeloma is underway with an ex- pretreated population.31 Daratumumab certainly has a good
pected accrual of 122 patients. GMMG-HD6 in a randomized safety and efficacy profile and can be combined with a
phase III trial is evaluating the same combination (ERVD) number of antimyeloma therapies. The question of whether
versus RVD among newly diagnosed transplant-eligible we can rechallenge with a subsequent daratumumab-based
patients. The study also has a second set of patients ran- combination therapy among those patients who are deemed
domly selected to receive RVD after receiving stem cell refractory to daratumumab single-agent therapy remains to
transplant (SCT) versus ERVD consolidation and lenalido- be answered. Data of responses among the high-risk patients
mide versus lenalidomide with elotuzumab maintenance, are encouraging. At least a 20% ORR among patients with
with the primary endpoint of PFS. Another similarly designed heavily pretreated, relapsed, refractory, high-risk multiple
trial from the DSMM XVII evaluating the combination (KRD myeloma and those with extramedullary myeloma as a single
vs. KRD with elotuzumab) also has a second trial of randomly agent is of great interest. More than one quarter of the
selected patients after receiving SCT and KRD versus KRD patients in the SIRIUS trial had either del(17)p (17%) or t(4;14)

e436 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


NEW TARGETS AND AGENTS IN HIGH-RISK MULTIPLE MYELOMA

(10%), and the responses were not limited to standard-risk and, at the same time, limiting the toxicities seen with an al-
patients alone.28 logeneic transplant. The main goal of cellular therapies is to
Several daratumumab trials are in progress. The HOVON/ induce a robust antimyeloma immune response that can have a
IFM trial is evaluating the combination of VTD with dar- potential effect on disease control in the long term. The utility of
atumumab in the induction, consolidation, and daratumumab chimeric antigen receptor T cells (CAR-T cells), natural killer cells,
maintenance for the primary endpoint of stringent complete and regulatory T cells is encouraging but preliminary (Table 3).
remission, PFS, and OS. From the Alliance groups, the
concept of evaluating lenalidomide/dexamethasone versus CAR-T cells. Engineering autologous T cells to express antigen-
daratumumab/lenalidomide/dexamethasone in transplant- specific CAR-T cells can be a potential immune-based
ineligible patients and RVD versus daratumumab with RVD therapeutic approach for B-cell malignancies. The utility of
are in the concept phase. In the maintenance setting, CAR-T cells has also been recently investigated in myeloma
daratumumab/lenalidomide versus lenalidomide post- management. One of the means by which myeloma cells evade
transplant until progression or unacceptable toxicity is immunosurveillance is by enabling mutations in the major his-
under evaluation (communication with Dr. McCarthy). tocompatibility complex (MHC). T cells bind to the T-cell receptor
Several other phase III trials of daratumumab in combination with the MHC on the tumor cell. However, in the presence of
with lenalidomide/dexamethasone versus lenalidomide/ MHC mutations, normal T cells cannot bind to MHC T-cell re-
dexamethasone or in combination with bortezomib/ ceptors. CAR-T cells have been genetically modified to have a
dexamethasone versus bortezomib/dexamethasone have different extracellular antigen-binding site that allows CAR-T
completed accrual and are awaiting results. Several other cells to identify malignant cells without the use of the MHC.33 A
phase III trials in transplant-ineligible patients (daratumumab case of sustained CR to infusion of cytotoxic T lymphocyte (CTL)
with or without bortezomib/melphalan/prednisone) are 019 cells in conjunction with autologous transplantation in a
ongoing. Given the longer infusion times and the higher patient with refractory myeloma was recently reported by
rate of infusion reactions seen (although majority of these Stadtmauer and colleagues.34 They were able to demonstrate
are # grade 2), daratumumab is currently being evaluated as a that the clinically relevant target of CTL019 in this case may have
subcutaneous formulation with the addition of recombinant been non-neoplastic CD19+ cells, which have been implicated in
human hyaluronidase (rHuPH20) among patients with multiple immune evasion and resistance to therapy in solid tumors.
myeloma. However, one can argue the observed response could be at-
A second CD38 monoclonal antibody, SAR650984 (isatux- tributed to be a response from alkylating therapies alone based
imab), in combination with lenalidomide/dexamethasone on the normal reconstitution of CD19+ B cells and loss of de-
demonstrated an ORR of 65% and VGPR rates of 32% among tectable CTL019. Nevertheless, the fact that six of the 10 patients
patients with multiple myeloma who were heavily pretreated (six treated remained progression free is hypothesis-generating in
prior lines of therapy).32 Patients experienced a median PFS of that this is not just an effect of melphalan alone. Additional
6.2 months reported at a median follow-up of 6 months. Another CAR-T cell studies are in progress, and early clinical results are
phase IB study of isatuximab in combination with pomalidomide promising, though other targets such as B-cell maturation an-
and dexamethasone is currently enrolling patients. tigen may prove to be more suitable for myeloma.35

Cellular Therapies Therapeutic Vaccinations


The idea behind using cellular immunotherapies is to attain Dendritic cell vaccines. The utility of dendritic cell vaccines
similar benefits to those observed after allogeneic transplant is currently being explored in myeloma. Vaccines aimed at

TABLE 3. Other Cellular Therapies

Cellular Therapy Identification Rationale Clinical Activity


NK Cells NK cells are members of the cellular immune Given the depression of NK activity in
system that play a key role in defense refractory myeloma, and the idea of
against viruses and tumors. In humans, enhancing NK efficacy to improve
NK cells are identified as CD32CD56 antimyeloma activity has led to ongoing
lymphocytes but may differ in their trials exploring the utility of NK cellbased
immunoregulatory role based on their therapy in myeloma.
further expression.61
Tregs Tregs are CD4 + T-cell population They are believed to strongly inhibit Among 10 patients undergoing ASCT, Treg
(CD4+CD25+FoxP3+ T cells), comprise antitumor immune responses among depletion for patients with myeloma was
5%10% of peripheral CD4 T cells, and patients with myeloma. It is unclear if deemed to be safe, and further studies to
are responsible for the control of Tregs play a role in protection from determine the role of Treg depletion are
autoimmune phenomena. malignancy, but Treg depletion leads to ongoing.62
induction of tumor rejection in murine
models. Treg depletion may enhance the
function of tumor antigenspecific T cells.
Abbreviations: NK, natural killer; Tregs, regulatory T cells; ASCT, autologous stem cell transplant.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e437


NOOKA AND LONIAL

developing myeloma-specific immunity have become a TARGETED THERAPIES


major area of focus. Currently, several groups are working on Boise and colleagues have published an interesting model for
either whole-cell or antigen-specific dendritic cell vaccines the two sides of myeloma therapy, the Tao of myeloma,
to enhance innate immunity to obtain the antitumor effect. where Yin and Yang represented the normal plasma cell and
Currently, these are early approaches and in phase I. A malignant plasma cell biology, respectively.42 Our current
dendritic cellmyeloma cell fusion vaccine study among agents have focused on the biology side of the Tao, but among
patients with multiple myeloma supported by the Bone high-risk patients, focusing on the mutational profile and the
Marrow Transplant Clinical Trials Network as a post- abnormal expression may be equally important.43
transplant maneuver to gain disease control is underway.

Peptide vaccines. PVX-410 is composed of a tetrapeptide


MDM2 Inhibitors Targeting p53 Deletion or Mutation
p53, a tumor suppressor gene located on the short arm of
from three unique regions of myeloma-associated antigens
chromosome 17, is implicated in normal cellular pro-
(XBP1, CD138, and CS1). The goal is to induce immunity
liferation, differentiation, and apoptosis. Loss of p53 is as-
against myeloma cells by selectively stimulating tumor-
sociated with poor survival among patients with myeloma
associated antigen-specific CTLs. The safety and tolerabil-
(del(17)p).44 Another protein, the murine double-minute 2
ity of PVX-410, alone and in combination with lenalidomide,
(MDM2), is a negative regulator of p53 and plays a role in
show considerable promise in treatment of patients with
proliferation and survival of myeloma cells by suppressing
smoldering myeloma.36 Future studies in combination with
the p53 function. MDM2 inhibitors to block the MDM2-p53
MEDI4736, a PD-L1 antibody and lenalidomide PVX-410, will
protein-protein interaction have potential as antimyeloma
be initiated shortly. Immucin, an anti-MUC1 signal peptide
therapeutic strategies for patients with myeloma with
vaccine, induced a robust CD8+- and CD4+-specific T-cell
del(17)p; however, this hypothesis presumes there is a
response in all 15 patients following autologous SCT and
normal and functional copy of p53 in the cell.45 There are
demonstrated a marked antitumor humoral response.37
emerging data suggesting an increased frequency of p53
mutations among with del(17)p upon greater exposure
Checkpoint Blockade to treatment. This finding may limit the utility of MDM2
PD-1, or its ligand, PD-L1, interactions may indirectly
inhibitors. Currently, DS-3032b, an oral MDM2 in-
modulate the response to tumor antigens through T-cell/
hibitor, is in initial trials for relapsed refractory myeloma
antigen-presenting cell interactions. PD-1 engagement may
(NCT02579824).
represent one way by which tumors evade immuno-
surveillance.38 The PD-1/PD-L1 pathway is shown by mul-
tiple groups to promote progression of myeloma indirectly BRAF Inhibitors
by undermining immune control of the malignancy.39 PD-1 Prevalence of patients with myeloma with actionable BRAF
blockade has been pursued as a promising therapeutic mutations (V600E) is much higher than what was originally
strategy to reverse immune tolerance and enhance T-cell thought. Approximately 5% of patients harbor either a clone
effector function in several tumor types. The broad ex- or a subclone of BRAF V600E.46 It has been postulated that
pression of PD-1 and its ligands in the microenvironment of myeloma clones harboring BRAF V600E might have a survival
myeloma and the preclinical data reveal an important role of advantage and likely present as aggressive, extramedullary
the PD-1 pathway in immune evasion by myeloma cells, and disease.47,48 Though recent analysis evaluating BRAF V600E
there may be immunotherapeutic potential of PD-1/PD-L mutations in early-stage myeloma suggests no relation to
inhibition in myeloma. Several PD-1 antibodies such as prognosis, this could represent the differential prognosis of
nivolumab, pidiluzumab (CT-011), and PD-L1 antibodies BRAF subclones at various phases of myeloma progression.46
(MEDI4736) are under investigation. Pembrolizumab, a There have been several reports of patients with BRAF-
humanized IgG4 antiPD-1 monoclonal antibody designed mutated, relapsed, refractory myeloma who were treated
to block interaction of PD-1 with PD-L1 and PD-L2, was successfully with the BRAF inhibitor vemurafenib.47
evaluated in combination with immunomodulatory drugs
(lenalidomide/dexamethasone40 and pomalidomide/low- MEK Inhibitors
dose dexamethasone41) with an intent to enhance tumor The deregulation of the RAS/RAF/MEK/extracellular signal
suppression. The ORR with lenalidomide/dexamethasone regulated kinase (ERK; RAS/mitogen-activated protein
was 76% ($ VGPR 24%)40 and the ORR with pomalidomide/ kinases) signal transduction pathway leads to abnormal
low-dose dexamethasone was 60% ($ VGPR 19%), 41 cellular proliferation, impaired apoptosis, enhanced an-
showing impressive activity with a tolerable safety profile. giogenesis, metastasis, and the development of drug re-
Forty-two percent of the patients in the pembrolizumab and sistance.49 Activating RAS mutations, which have a reported
pomalidomide/low-dose dexamethasone studies exhibited incidence varying between 32% and 50% in multiple mye-
high-risk features (del 17p, t(4:14) and/or t(14:16)). Among loma, are also thought to deregulate this pathway.50 The
these high-risk patients, a 50% ORR was observed.41 Several presence of RAS mutations have previously been shown to
other combination trials with PD-1 antibodies and PD-L1 confer clinical prognosis.50,51 Mutation-specific inhibitor use
antibodies are under investigation in myeloma so far is in the early phases. Several factors that

e438 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


NEW TARGETS AND AGENTS IN HIGH-RISK MULTIPLE MYELOMA

are not understood fully limit the therapeutic application of processing and the regulation of gene transcription. BRD as a
the KRAS/NRAS inhibitors in myeloma. Variability of the potential therapeutic target has resulted in the development
dominant clone relative to size of the clonal population of BRD and BRD and extraterminal protein inhibitors.56 The
carrying the KRAS/NRAS mutations point to the presence of BRD inhibitor CPI203 exerted much preclinical activity in
intraclonal heterogeneity and also the potential difficulties both the bortezomib- and melphalan-resistant lines and
in the use of targeted treatment strategies.43 patient samples.57 Specific BRD4 degraders show promising
activity against preclinical models of multiple myeloma.58
FGFR3 Inhibitors
Simultaneous overexpression of two oncogenes, FGFR3 CONCLUSION
(fibroblast growth factor receptor 3) and MMSET (multiple It is an exciting time in myeloma research, where future
myeloma SET domain), represent a subset of patients with progress is being built on the current success. The suc-
myeloma with t(4;14) translocation, often indicating poor cessful incorporation of immunomodulatory agents and
prognosis.52 Though preclinical experience suggested anti- proteasome inhibitor combinations in high-risk multiple
myeloma activity in FGFR3-expressing cell lines,53 experi- myeloma management has improved the survival among
ence with small-molecule inhibitors has not made much this group of patients. With monoclonal antibodies targeting
progress. It could represent that 30% of t(4;14) patients do CD38, SLAMF7, and PD-1/PD-L1 pathways available in
not express FGFR3.52 Further drug development with FGFR3 clinical practice and, more importantly, the preliminary
monoclonal antibodies54 and MMSET inhibitors55 are cur- results showing superior efficacy among high-risk patients,
rently under evaluation. future strategies of treating high-risk patients should in-
corporate these agents in the absence of target-specific
BRD4 Inhibitors antimyeloma therapy. Identifying more relevant biologic
Bromodomain (BRD) and extraterminal proteins play an targets/actionable mutations and developing drugs for
important role in cellular proliferation, cell cycle progres- these targets will be the next step to enhance deeper re-
sion, and chromatin compaction. They use BRD modules sponses, resulting in durable remissions among the high-risk
for lysine acetylation, a pivotal mechanism in chromatin patients with multiple myeloma.

References

1. Morgan GJ, Gregory WM, Davies FE, et al; National Cancer Research 9. Kapoor P, Kumar S, Fonseca R, et al. Impact of risk stratification on
Institute Haematological Oncology Clinical Studies Group. The role of outcome among patients with multiple myeloma receiving initial
maintenance thalidomide therapy in multiple myeloma: MRC Myeloma therapy with lenalidomide and dexamethasone. Blood. 2009;114:
IX results and meta-analysis. Blood. 2012;119:7-15. 518-521.
2. Barlogie B, Pineda-Roman M, van Rhee F, et al. Thalidomide arm of total 10. Attal M, Lauwers-Cances V, Marit G, et al. Lenalidomide maintenance
therapy 2 improves complete remission duration and survival in my- after stem-cell transplantation for multiple myeloma: follow-up anal-
eloma patients with metaphase cytogenetic abnormalities. Blood. ysis of the IFM 2005-02 trial. Blood. 2013;122:406.
2008;112:3115-3121. 11. Dimopoulos MA, Weisel KC, Song KW, et al. Cytogenetics and long-term
3. Wu P, Davies FE, Boyd K, et al. The impact of extramedullary disease at survival of patients with refractory or relapsed and refractory multiple
presentation on the outcome of myeloma. Leuk Lymphoma. 2009;50: myeloma treated with pomalidomide and low-dose dexamethasone.
230-235. Haematologica. 2015;100:1327-1333.
4. Rosin~ol L, Cibeira MT, Blade J, et al. Extramedullary multiple myeloma 12. Leleu X, Karlin L, Macro M, et al; Intergroupe Francophone du Mye lome
escapes the effect of thalidomide. Haematologica. 2004;89:832-836. (IFM). Pomalidomide plus low-dose dexamethasone in multiple mye-
5. Srivastava G, Rana V, Lacy MQ, et al. Long-term outcome with lena- loma with deletion 17p and/or translocation (4;14): IFM 2010-02 trial
lidomide and dexamethasone therapy for newly diagnosed multiple results. Blood. 2015;125:1411-1417.
myeloma. Leukemia. 2013;27:2062-2066. 13. Barlogie B, Anaissie E, van Rhee F, et al. Incorporating bortezomib into
6. Avet-Loiseau H, Hulin C, Benboubker L, et al. Impact of cytogenetics on upfront treatment for multiple myeloma: early results of total therapy
outcomes of transplant-ineligible patients with newly diagnosed 3. Br J Haematol. 2007;138:176-185.
multiple myeloma treated with continuous lenalidomide plus low-dose 14. Jagannath S, Richardson PG, Sonneveld P, et al. Bortezomib appears to
dexamethasone in the first (MM-020) trial. Blood. 2015;126:730. overcome the poor prognosis conferred by chromosome 13 deletion in
7. Reece D, Song KW, Fu T, et al. Influence of cytogenetics in patients with phase 2 and 3 trials. Leukemia. 2007;21:151-157.
relapsed or refractory multiple myeloma treated with lenalidomide plus 15. Avet-Loiseau H, Leleu X, Roussel M, et al. Bortezomib plus dexa-
dexamethasone: adverse effect of deletion 17p13. Blood. 2009;114: methasone induction improves outcome of patients with t(4;14) my-
522-525. eloma but not outcome of patients with del(17p). J Clin Oncol. 2010;28:
8. Avet-Loiseau H, Soulier J, Fermand JP, et al; IFM and MAG groups. 4630-4634.
Impact of high-risk cytogenetics and prior therapy on outcomes in 16. Cavo M, Tacchetti P, Patriarca F, et al; GIMEMA Italian Myeloma
patients with advanced relapsed or refractory multiple myeloma Network. Bortezomib with thalidomide plus dexamethasone compared
treated with lenalidomide plus dexamethasone. Leukemia. 2010;24: with thalidomide plus dexamethasone as induction therapy before, and
623-628. consolidation therapy after, double autologous stem-cell transplantation

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e439


NOOKA AND LONIAL

in newly diagnosed multiple myeloma: a randomised phase 3 study. relapsed or relapsed and refractory multiple myeloma. Blood. 2015;
Lancet. 2010;376:2075-2085. 126:508.
17. Sonneveld P, Schmidt-Wolf IG, van der Holt B, et al. Bortezomib in- 32. Martin TG, Baz R, Benson DM, et al. A phase Ib dose escalation trial of
duction and maintenance treatment in patients with newly diagnosed SAR650984 (Anti-CD-38 mAb) in combination with lenalidomide and
multiple myeloma: results of the randomized phase III HOVON-65/ dexamethasone in relapsed/refractory multiple myeloma. Blood. 2014;
GMMG-HD4 trial. J Clin Oncol. 2012;30:2946-2955. 124:83.
18. Sonneveld P, Salwender H-J, Van Der Holt B, et al. Bortezomib induction 33. Ayed AO, Chang LJ, Moreb JS. Immunotherapy for multiple myeloma:
and maintenance in patients with newly diagnosed multiple myeloma: current status and future directions. Crit Rev Oncol Hematol. 2015;96:
long-term follow-up of the HOVON-65/GMMG-HD4 trial. Blood. 2015; 399-412.
126:27. 34. Garfall AL, Maus MV, Hwang W-T, et al. Chimeric antigen receptor
19. Nooka AK, Kaufman JL, Muppidi S, et al. Consolidation and maintenance T cells against CD19 for multiple myeloma. N Engl J Med. 2015;373:
therapy with lenalidomide, bortezomib and dexamethasone (RVD) in 1040-1047.
high-risk myeloma patients. Leukemia. 2014;28:690-693. 35. Chekmasova AA, Horton HM, Garrett TE, et al. A novel and highly potent
20. Jakubowiak AJ, Siegel DS, Martin T, et al. Treatment outcomes in pa- CAR T cell drug product for treatment of BCMA-expressing hemato-
tients with relapsed and refractory multiple myeloma and high-risk logical malignances. Blood. 2015;126:3094.
cytogenetics receiving single-agent carfilzomib in the PX-171-003-A1 36. Wang M, Nooka AK, Yee AJ, et al. Initial results of a phase 1/2a, dose
study. Leukemia. 2013;27:2351-2356. escalation study of PVX-410 multi-peptide cancer vaccine in patients
21. Chng W-J, Goldschmidt H, Dimopoulos MA, et al. Efficacy and safety of with smoldering multiple myeloma (SMM). Blood. 2014;124:4737.
carfilzomib and dexamethasone vs bortezomib and dexamethasone in 37. Carmon L, Avivi I, Riva K, et al. Phase I/II trial of immucin a pan HLA, anti-
patients with relapsed multiple myeloma based on cytogenetic risk MUC1 signal peptide vaccine, in multiple myeloma patients with
status: subgroup analysis from the phase 3 study endeavor residual disease or progression following autologous stem cell trans-
(NCT01568866). Blood. 2015;126:30. plantation. Blood. 2013;122:1943.
22. Avet-Loiseau H, Fonseca R, Siegel D, et al. Efficacy and safety of car- 38. Brahmer JR, Tykodi SS, Chow LQM, et al. Safety and activity of anti-PD-
filzomib, lenalidomide, and dexamethasone vs lenalidomide and L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366:
dexamethasone in patients with relapsed multiple myeloma based on 2455-2465.
cytogenetic risk status: subgroup analysis from the phase 3 study aspire 39. Iwai Y, Ishida M, Tanaka Y, et al. Involvement of PD-L1 on tumor cells in
(NCT01080391). Blood. 2015;126:731. the escape from host immune system and tumor immunotherapy by
23. Shah J, Stadtmauer E, Abonour R, et al. Phase I/II dose expansion of a PD-L1 blockade. Proc Natl Acad Sci USA. 2002;99:12293-12297.
multi-center trial of carfilzomib and pomalidomide with dexametha- 40. San Miguel J, Mateos M-V, Shah JJ, et al. Pembrolizumab in combi-
sone (Car-Pom-d) in patients with relapsed/refractory multiple mye- nation with lenalidomide and low-dose dexamethasone for relapsed/
loma. Paper presented at: 2013 American Society of Hematology refractory multiple myeloma (RRMM): keynote-023. Blood. 2015;
Annual Meeting; December 2013; New Orleans, LA. 126:505.
24. Moreau P, Masszi T, Grzasko N, et al. Ixazomib, an investigational oral 41. Badros AZ, Kocoglu MH, Ma N, et al. A phase II Study of anti PD-1
proteasome inhibitor (PI), in combination with lenalidomide and antibody pembrolizumab, pomalidomide and dexamethasone in pa-
dexamethasone (IRd), significantly extends progression-free survival tients with relapsed/refractory multiple myeloma (RRMM). Blood.
(PFS) for patients (Pts) with relapsed and/or refractory multiple my- 2015;126:506.
eloma (RRMM): the phase 3 tourmaline-MM1 study (NCT01564537). 42. Boise LH, Kaufman JL, Bahlis NJ, et al. The tao of myeloma. Blood. 2014;
Blood. 2015;126:727. 124:1873-1879.
25. Lonial S, Dimopoulos M, Palumbo A, et al; ELOQUENT-2 Investigators. 43. Lohr JG, Stojanov P, Carter SL, et al; Multiple Myeloma Research
Elotuzumab therapy for relapsed or refractory multiple myeloma. Consortium. Widespread genetic heterogeneity in multiple myeloma:
N Engl J Med. 2015;373:621-631. implications for targeted therapy. Cancer Cell. 2014;25:91-101.
26. Jakubowiak AJ, Benson DM, Bensinger W, et al. Phase I trial of anti-CS1 44. Chng WJ, Dispenzieri A, Chim CS, et al; International Myeloma Working
monoclonal antibody elotuzumab in combination with bortezomib in Group. IMWG consensus on risk stratification in multiple myeloma.
the treatment of relapsed/refractory multiple myeloma. J Clin Oncol. Leukemia. 2014;28:269-277.
2012;30:1960-1965. 45. Zhang B, Golding BT, Hardcastle IR. Small-molecule MDM2-p53 in-
27. Palumbo A, Offidani M, Pegourie B, et al. Elotuzumab plus bortezomib hibitors: recent advances. Future Med Chem. 2015;7:631-645.
and dexamethasone versus bortezomib and dexamethasone in patients 46. Rustad EH, Dai HY, Hov H, et al. BRAF V600E mutation in early-stage
with relapsed/refractory multiple myeloma: 2-year follow-up. Blood. multiple myeloma: good response to broad acting drugs and no relation
2015;126:510. to prognosis. Blood Cancer J. 2015;5:e299.
28. Lonial S, Weiss BM, Usmani SZ, et al. Daratumumab monotherapy in 47. Andrulis M, Lehners N, Capper D, et al. Targeting the BRAF V600E
patients with treatment-refractory multiple myeloma (SIRIUS): an mutation in multiple myeloma. Cancer Discov. 2013;3:862-869.
open-label, randomised, phase 2 trial. The Lancet. 48. Bohn OL, Hsu K, Hyman DM, et al. BRAF V600E mutation and clonal
29. Usmani S, Weiss B, Bahlis NJ, et al. Clinical efficacy of daratumumab evolution in a patient with relapsed refractory myeloma with plas-
monotherapy in patients with heavily pretreated relapsed or refractory mablastic differentiation. Clin Lymphoma Myeloma Leuk. 2014;14:
multiple myeloma. Blood. 2015;126:29. e65-e68.
30. Plesner T, Arkenau H-T, Gimsing P, et al. Daratumumab in combination 49. McCubrey JA, Steelman LS, Chappell WH, et al. Roles of the Raf/MEK/
with lenalidomide and dexamethasone in patients with relapsed or ERK pathway in cell growth, malignant transformation and drug re-
relapsed and refractory multiple myeloma: updated results of a phase sistance. Biochim Biophys Acta. 2007;1773:1263-1284.
1/2 Study (GEN503). Blood. 2015;126:507. 50. Liu P, Leong T, Quam L, et al. Activating mutations of N- and K-RAS in
31. Chari A, Lonial S, Suvannasankha A, et al. Open-label, multicenter, phase multiple myeloma show different clinical associations: analysis of the
1b study of daratumumab in combination with pomalidomide and Eastern Cooperative Oncology Group phase III trial. Blood. 1996;88:
dexamethasone in patients with at least 2 lines of prior therapy and 2699-2706.

e440 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


NEW TARGETS AND AGENTS IN HIGH-RISK MULTIPLE MYELOMA

51. Mulligan G, Lichter DI, Di Bacco A, et al. Mutation of NRAS but not KRAS 57. Siegel MB, Liu SQ, Davare MA, et al. Small molecule inhibitor screen
significantly reduces myeloma sensitivity to single-agent bortezomib identifies synergistic activity of the bromodomain inhibitor CPI203 and
therapy. Blood. 2014;123:632-639. bortezomib in drug resistant myeloma. Oncotarget. 2015;6:18921-18932.
52. Xie Z, Chng WJ. MMSET: role and therapeutic opportunities in multiple 58. Zhang X, Lu J, Qian Y, et al. Proteolytic targeting chimeric molecules
myeloma. Biomed Res Intl. 2014;2014:636514. (PROTACs) specific for bromodomain-containing protein (BRD) 4 are
53. Trudel S, Li ZH, Wei E, et al. CHIR-258, a novel, multitargeted tyrosine active against pre-clinical models of multiple myeloma. Blood. 2015;
kinase inhibitor for the potential treatment of t(4;14) multiple mye- 126:917.
loma. Blood. 2005;105:2941-2948. 59. Mateos M-V, Martnez-Lopez J, Hernandez M-T, et al. Sequential versus
54. Trudel S, Stewart AK, Rom E, et al. The inhibitory anti-FGFR3 antibody, alternating administration of VMP and Rd in elderly patients with newly
PRO-001, is cytotoxic to t(4;14) multiple myeloma cells. Blood. 2006; diagnosed MM. Blood. 2016;127(4):420-425.
107:4039-4046. 60. Shah JJ, Stadtmauer EA, Abonour R, et al. Carfilzomib, pomalidomide,
55. Martinez-Garcia E, Popovic R, Min DJ, et al. The MMSET histone and dexamethasone for relapsed or refractory myeloma. Blood. 2015;
methyl transferase switches global histone methylation and alters 126:2284-2290.
gene expression in t(4;14) multiple myeloma cells. Blood. 2011;117: 61. Dosani T, Carlsten M, Maric I, et al. The cellular immune system in
211-220. myelomagenesis: NK cells and T cells in the development of myeloma
56. Chaidos A, Caputo V, Karadimitris A. Inhibition of bromodomain and and their uses in immunotherapies. Blood Cancer J. 2015;5:e306.
extra-terminal proteins (BET) as a potential therapeutic approach in 62. Keudell Gv, Rosenbaum CA, Zimmerman TM, et al. Pilot study of
haematological malignancies: emerging preclinical and clinical evi- regulatory T cell depletion in the setting of autologous stem cell
dence. Ther Adv Hematol. 2015;6:128-141. transplantation for multiple myeloma. Blood. 2013;122:4607.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e441


SZALAT AND MUNSHI

Next-Generation Sequencing Informing Therapeutic Decisions


and Personalized Approaches
Raphael Szalat, MD, and Nikhil C. Munshi, MD

OVERVIEW

Multiple myeloma is a heterogeneous disease featured by different molecular subtypes. In the last decade, new therapeutics
including second- and third-generation proteasome inhibitors and immunomodulatory agents, monoclonal antibodies,
and other novel targeted agents have completely transformed the outcome of the disease. The task ahead is to develop
strategies to identify effective combinations and sequences of agents that can exploit the genetic make-up of myeloma cells
to improve efficacy. Moreover, a subgroup of high-risk patients who experience early disease relapse and shorter survival
also requires early identification and specific intervention. Next-generation sequencing (NGS) technologies now allow us to
accomplish some of these goals. As described here, besides improving our understanding of the disease, it is beginning to
influence our clinical decisions and therapeutic choices. In this article, we describe the current state-of-the-art role of NGS in
myeloma from identifying high-risk disease, to drug selection, and, ultimately, to guide personalized therapy.

M ultiple myeloma is a clonal plasma cell disease that


evolves as a multistep process classically characterized
by a premalignant phase (monoclonal gammopathy of un-
who experience shorter survival at diagnosis. A classification
combining these characteristics has also been proposed.10
To date, there are no validated criteria that take into account
determined significance), a nonsymptomatic phase (smol- the chemosensitivity of the disease or status and changes at
dering multiple myeloma), and, eventually, a symptomatic relapse. Furthermore, these criteria do not take into ac-
disease that will be subjected to remissions and relapses count the heterogeneity existing within patients to define
over time. Multiple myeloma is biologically highly hetero- high or low risk. As an example, based on cytogenetic
geneous, defined by two groupshyperdiploid and non- classification, a combination of high-risk features such as
hyperdiploid multiple myelomaaccording to karyotype 17p13 deletion may coexist with trisomies of some of the
studies, and featured by 710 molecular subgroups based on chromosomes, a good-risk feature. A recent study by the
gene expression studies.1-3 Intergroupe Francophone du Myelome has now reported
The therapeutic landscape of multiple myeloma has been how the combination of these features change the outcome.
transformed by the advent of new and very efficient drugs If patients have trisomies of chromosome 3 and 5 along with
including second and third generations of proteasome in- other poor risk cytogenetics, the overall outcome may im-
hibitors as well as immunomodulatory agents and mono- prove; however, the presence of chromosome 21 trisomy
clonal antibodies. Such therapies have transformed patient may further adversely affect the outcome.12,13 Conversely,
outcomes and quality of life.4-9 However, the majority of some patients not identified by the current stratification as
patients experience relapse, and, for a subgroup of patients, high-risk patients have an unpredicted short survival. With
the prognosis remains poor with early relapse and short the availability of additional novel therapeutic options, new
survival. Risk stratification is crucial to identify these patients markers are now more important than ever in predicting not
and to develop and adapt treatment regimens accordingly. only patient risk but also treatment selection and outcome
The actual recommendations to assess multiple myeloma to provide the best available therapy in multiple myeloma.
risk at diagnosis are based on the combination of two bi- Over last 15 years, various high-throughput technologies
ologic criteria determined by the International Staging have become available. DNA-based studies include whole-
System (ISS; beta-2 microglobulin and albumin levels) and genome and whole-exome sequencing, array comparative
three cytogenetic abnormalities (17p13 deletion, t(4;14), genomic hybridization, and high-density single nucleotide
and t(14;16)), usually identified by fluorescence in situ hy- polymorphism arrays. These techniques allow identification
bridization.10,11 These criteria allow identification of patients of recurrent mutations and affected pathways, mutational

From the Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; VA Boston Healthcare System, Boston, MA.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Nikhil C. Munshi, MD, Dana-Farber Cancer Institute, 450 Brookline Ave., Boston, MA 02215; email: nikhil_munshi@dfci.harvard.edu.

2016 by American Society of Clinical Oncology.

e442 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


NEXT-GENERATION SEQUENCING

signatures, features of clonality and clonal evolution, and mutations in the MAPK and NF-kappa B pathway are most
copy number alterations. At the RNA level, gene expression probably drivers of myelomagenesis.19
profiling of the malignant plasma cells has been widely Although most of the recurrent mutations do not appear
evaluated and reported as a promising method for prog- to correlate with survival, a global increased number of
nostication in multiple myeloma.14-16 RNA sequencing is mutations correlate with a higher risk of relapse and death.17
now progressively replacing microarray by providing wider In one study, some less-recurrent mutations involving DNA
information related to protein-coding RNA (gene expres- repair and apoptotic pathways have been reported to be
sion, splicing and isoforms expression, fusions, mutations) associated with poor survival. Indeed TP53, ATM, ATR, and
and to noncoding RNA (micro-RNA, long noncoding RNA, CCND1 mutations are significantly associated with poor
small nucleolar RNA, small nuclear RNA, transfer RNA, ri- outcome.19 As described below, mutational information can
bosomal RNA, and extracellular RNA). The clinical signifi- be used for various aspects of patient care in myeloma.
cance of noncoding RNA, which constitutes a large volume Besides prognostication, mutational profiling is beginning
of RNA, is still being investigated. These and other tech- to guide therapy and aid in development of personalized
nological advances including chromatin studies have been medicine. The increasing number of targeted or nontargeted
explored and validated for their ability to risk stratify and therapies that are now available make this strategy par-
develop personalized effective therapy. In this article, we ticularly attractive. Thus, in a cohort of 133 patients with
describe the most recent and important findings generated relapsing myeloma, the presence of NRAS mutations eval-
by NGS (Table 1) and how it may contribute to better uated by targeted sequencing was significantly associated
identify high-risk patients and to guide therapy in multiple with poor response to bortezomib.21 Conversely, IRF4
myeloma. mutations, if present, are associated with better outcome
with immunomodulatory agent therapy.19 MEK inhibitors
WHOLE-GENOME AND WHOLE-EXOME for MAPK pathway, ATR/ATM inhibitors, and CCND1 in-
SEQUENCING hibitors are some examples of adapted strategies that
Three independent large studies (total of 733 patients) have can now be considered for patients with these specific
evaluated the mutational profile of multiple myeloma by mutations.
whole-exome and/or whole-genome sequencing.17-19 These Study of NGS has confirmed that, at the time of diagnosis,
studies have led to the discovery of the recurrent mutational patients have a number of coexistent clones. Some muta-
landscape in multiple myeloma. Although no universal driver tions, especially those occurring early in the tumor devel-
mutation has been discovered, KRAS, NRAS, FAM46C, DIS3, opment, are present in all cells and are called clonal
BRAF, and TP53 have been identified as the most recur- mutations. However, some mutations occur later and are
rent mutations. NRAS, KRAS, and BRAF, which are part of the present in subset of cells; these are known as subclonal
mitogen-activated protein kinase (MAPK) pathway, and mutations. The clonal and subclonal content changes and
TP53 are also recurrently mutated in other malignancies.20 evolves over time. In multiple myeloma, no clear data are
Along with the MAPK pathway, genes involved in the nuclear currently available to precisely understand the impact of
factor (NF)-kappa B pathway are recurrently mutated in clonal composition on survival.17 When two or more samples
up to 17% of multiple myeloma cases, suggesting that from the same patients are studied in myeloma, four distinct
patterns of clonal evolution are observed (linear, branching,
no change, and differential clonal response).17 This ability to
monitor the clonal evolution over time is also an important
source of information that can be exploited in clinic. More-
KEY POINTS
over, the study of types of mutations have now allowed
Whole-genome and whole-exome sequencing have us to identify two signatures ascribed to mechanisms of
identified recurrent mutations in multiple myeloma mutational processes.22 One of the two signatures belong to
including mutations associated with poor outcome the APOBEC family and is observed more often in relapsed
(ATM, ATR, TP53, and CCND1) and druggable target samples.17 These signatures should lead to new therapeutic
mutations (BRAF, MAPK pathway, FGFR3). strategies that may reduce the genomic instability.
Specific mutations, mutational load, copy number Targeted sequencing strategies focusing on a relatively
abnormalities, and gene expression signatures are small number of genes are under evaluation.23,24 This has
reliable markers for prognostication in multiple included studies of frequently mutated groups of genes or in
myeloma. one specific instance using deep sequencing of a single gene,
NGS data can be used to accurately evaluate response to
for example, IgH, to study minimal residual disease.25
treatment characterizing minimal residual disease.
The molecular and genomic characteristics of a patient
continue to evolve and thus may require repeated RNA PROFILE
reassessment over disease course Gene expression profiling using microarrays has identified
New markers generated by NGS can be incorporated in different multiple myeloma subtypes.2,3,26-28 It has allowed
the clinical practice to elaborate a personalized therapy. improved understanding of the myeloma biology, however,
it has not yet helped to select specific therapeutic agents

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e443


SZALAT AND MUNSHI

TABLE 1. Genomic Methodologies and Their Practical Clinical Uses


Possible Results Clinical Application Comments
DNA-Based Studies
Whole-Genome or Mutations; clonality and clonal Identify mutations for prognostication This method provides extensive useful data but
Whole-Exome evolution; chromosome and druggable targets; identify clonal requires expertise to interpret data
Sequencing rearrangements content; Clonal evolution
Targeted Sequencing Identifies specific mutations; Identify specific mutations impacting Most practical clinically applicable method; MRD
minimal residual disease evalua- outcome and with potential for evaluation is becoming an important endpoint in
tion (IgH deep sequencing) therapeutic targeting; response therapy and also for prognostication; useful for
assessment for MRD personalized therapy in future
aCGH Evaluation of copy number abnor- Correlate copy number changes to Limited utilization as same information can be
SNP Array malities; identification of ampli- prognosis; correlate specific SNPs to obtained by WGS/WES
fications and deletions; identify disease occurrence by GWAS studies
specific SNPs
Methylation Array Regulation of gene expression; May inform utilization of methylation Currently not used to inform therapy; useful in
evaluation of promoter targeting agents research; future potential for personalized
methylation therapy as more agents targeting
methylation are developed
ChIP Sequencing Histone and other epigenomic Select epigenome targeting agents Will allow selection of agents in myeloma with
DNA-binding modifications specific epigenomic changes, however,
impacting gene expression currently remains investigational; useful in
research to understand biology
RNA-Based Studies
Microarray-Based Gene expression profile; micro-RNA GEP-based signatures allow risk Widely and commercially available; ease of utili-
Studies expression profile stratification zation of the data; being phased-out by RNA
sequencing
RNA Sequencing Gene expression profile; micro- Identify high-risk disease; may inform This method provides broader information and is
RNA; long noncoding RNA; alter- selection of agents in future more reliable for clinical trials; requires
nate splicing isoforms; mutations expertise to interpret data
Abbreviations: aCGH, array comparative genomic hybridization; ChIP, chromatin immunoprecipitation; GEP, gene-expression profiling; GWAS, genome-wide association study; MRD,
minimal residual disease; SNP, single nucleotide polymorphism; WGS, whole-genome sequencing; WES, whole-exome sequencing

that could be predicted to be effective. Using gene ex- biology and that any new gene expression studies will re-
pression profiling, various gene expression signatures have quire integration of these data in the future.31,32
been developed that could identify patients with high- and
low-risk disease. Three different signatures have been
described; a 70-gene signature by the Arkansas group, a
NONCODING RNA
RNA sequencing data also provide information on long
15-gene signature by the Intergroupe Francophone du
noncoding RNA, which can significantly affect cellular bi-
Myelome, and a 92-gene signature by the HOVON
ology. Large RNA sequencing data have identified a unique
group.14-16 All three signatures identify around 20% to 25%
long noncoding RNA pattern in multiple myeloma and an
of patients who are high risk with less than 2-year survival.
independent, significant correlation with poor survival.33
However, each of these signatures includes distinct genes
Similarly, micro-RNAs are noncoding RNAs that affect
with almost no overlap between the signatures, making its
transcriptome function. It has been reported as a marker
practical and universal use complicated. A recent study has
for prognosis in multiple myeloma in several studies.34-37
proposed a combined unique signature that may be used in
In addition to prognostication, micro-RNA are being in-
clinical practice in the future.29
vestigated as therapeutic targets with potential for de-
Importantly, RNA sequencing has now replaced micro-
velopment of small molecules that affect micro-RNA
array to study gene expression. This method allows study of
function.38-42
coding and noncoding RNA, identifying not only gene ex-
pression levels, but also differential splicing and isoform
expression, mutational profiling, and gene fusions. RNA se- EPIGENOMIC MODIFICATIONS
quencing data from a large cohort of uniformly treated DNA methylation and histone modifications that include
patients have also identified that alternative splicing and acetylation and methylation lead to modification of gene
small noncoding RNA expression patterns are specific in expression and function. Histone methylation has been
multiple myeloma compared with normal plasma cells. In- studied, mostly in the t(4;14) of multiple myeloma cells that
terestingly, within the same level of expression of a given overexpress histone methyl transferase and multiple mye-
gene, only some isoforms are associated with poor out- loma SET domain (MMSET, NSD2, or WHSC1)regulating
come,30 suggesting that post-translational regulation, such gene expression43-45 and influence multiple myeloma cell
as splicing, plays an important role in multiple myeloma behavior.46,47 Although the impact of epigenomic changes

e444 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


NEXT-GENERATION SEQUENCING

on outcome has not been studied in detail, it has already The feasibility of using a single or limited number of
provided various therapeutic targets including inhibitors of techniques to get the most from this information is under
histone deacetylase (HDACs) or enhancer of zeste homolog 2 development. As an example, DNA and RNA sequencing can
(EZH2) inhibitors.48 Several studies have also demonstrated provide most of the information.52 Although currently used
that DNA methylation is differentially regulated in multi- in a limited number of institutions for research purposes
ple myeloma cells compared with normal plasma cells, only, routine DNA sequencingand in some cases, targeted
with a pattern of global hypomethylation and focal sequencingis performed along with either array-based
hypermethylation.49-51 However, therapies targeting DNA gene expression profiling or RNA sequencing. As an in-
methylation have not been effective in multiple myeloma. tegrated approach of analysis is developed and validated, a
more robust prognostic model will be available for clinical
CLINICAL APPLICATION OF GENOMIC DATA application. Moreover, the NGS data have revealed that a
Prognostic Marker substantial number of new mutations evolve at the time of
Next-generation sequencing data enlighten the biology of relapse and suggest the need to re-examine genomic profiles
myeloma and provide information on disease behavior to on more than one occasion during the course of the disease.
inform prognosis and, particularly, to define low- and
high-risk disease. To date, several validated markers can be
integrated and combined with ISS and cytogenetic features Inform Therapy
to accurately identify low- and high-risk multiple myeloma. The ability to characterize the prognosis of patients with
Array comparative genomic hybridization and high-density multiple myeloma by NGS can be used to rationally design a
single nucleotide polymorphism arrays have been partic- personalized therapeutic plan at diagnosis and at relapse.
ularly useful to study large cohorts of patients. Major First, risk stratification allows for identification of high-risk
findings have been amp(1q23.3), amp(5q31.3), and patients who may benefit from more intense therapeutic
del(12p13.31), which carry poor prognosis. Now, the newer interventions. Currently, the main focus of intensification for
gene and micro-RNA expression signatures, and specific these patients is in consolidation and maintenance thera-
mutations and clonal shifts, constitute the basis of new pies. We suggest that patients identified as high risk at di-
markers of prognosis. An important challenge is to agnosis should be treated with the most efficient and
generate a unique tool that would combine all of these available therapy as an induction regimen, including a
validated data. Ideally, such a tool would use gene and combination of immunomodulatory agents, proteasome
micro-RNA expression signatures, copy number alter- inhibitors, dexamethasone, and possibly a monoclonal an-
ations, chromosome rearrangements, and relevant muta- tibody. The impact of autotransplant among high-risk pa-
tions for their prognostic value and possibly to be tients may not be of the same extent as among patients
potentially targeted by specific therapies. Additional data with low-risk disease; however, it remains an important con-
concerning noncoding RNA, the roles of clonality and clonal solidation therapy. This phase must be followed for high-risk
evolution, the roles of specific mutations, and epigenomic patients by consolidation with a more intense regimen and a
and post-translational regulation are also under evaluation two-drug maintenance regimen.53
and should provide even more reliable markers in the near Attempts at identifying effective therapy using gene ex-
future (Table 2). pression profiling have not been very successful. 54 The

TABLE 2. New Markers Generated by Next-Generation Sequencing Under Evaluation to Predict High-Risk Multiple
Myeloma
Marker Method Potential Impact
Mutational Signature WES/WGS Distinct mutational processes may contribute differentially to tumor progression
May provide future therapeutic targets
Clonal and Subclonal WES/WGS Clonal content may have prognostic significance
Evolution
May predict treatment sensitivity
May require special consideration to judge response to therapy
Lnc-RNA Signature RNA-seq Lnc-RNA are involved in MM biology and may provide potential therapeutic targets
Lnc-RNA signature may have prognostic significance
RNA Splicing RNA-seq Differentially expressed isoforms constitute a marker of prognosis
Spliced isoforms have differential biologic activity making them attractive therapeutic targets with increased
specificity
Epigenomic Profile Microarray-ChIP Highly regulated region may correlate with patient outcome
Some epigenomic modifications have already been targeted in myeloma (panobinostat, HDAC inhibitor)
Abbreviations: ChIP, chromatin immunoprecipitation; lnc-RNA, long noncoding RNA; MM, multiple myeloma; RNA-seq, RNA sequencing; WGS, whole-genome sequencing; WES,
whole-exome sequencing.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e445


SZALAT AND MUNSHI

results suggest that a simple gene expression level is unable useful information to select a specific therapy that would
to identify effective agents that can achieve complete target DNA methylation or histone methylation or acety-
remission. The use of DNA sequencing seems particularly lation (MMSET expression abnormality, HDAC expression
promising to identify specific mutations that correlate changes). The development of new histone deacetylase
with a clinical response to therapy or those that are inhibitors and new histone methylase inhibitors is particu-
druggable targets. A strategy that takes into account larly promising.9 However, some mutations may not have
mutations can be integrated into the patient assessment any biologic impact because of low or no expression of the
plan. For example, the presence of NRAS mutations at mutated gene. We have observed that only a little over a
relapse is associated with low response to bortezomib,21 quarter of mutated genes are expressed, and the global
whereas the presence of IRF4 mutations confers higher expression of mutated genes is lower than the expression of
sensitivity to immunomodulatory agents.18 Currently, no nonmutated genes, with approximately only 25% expres-
single agent that specifically targets a mutation is approved sion. This low level of expression may be due to the level of
for routine clinical use in multiple myeloma. However, expression of the nonmutated allele or by the presence of
smaller studies have evaluated the efficacy of such targeted only a subclonal population featured by the mutation. Thus,
agents. The presence of an activating BRAF mutation can be theoretically, the presence of mutation does not necessarily
specifically targeted using a BRAF inhibitor, and efficacy can mean that it has an important role and that we should
also be monitored using assays devised to detect the consider it as therapeutic target. This precision is important
presence of BRAF-mutated myeloma cells.55 In a single case for the interpretation of data in ongoing clinical studies.59
report, vemurafenib, which exclusively inhibits the V600E- Mutational profiling ideally should be coupled with RNA
mutated BRAF, was effective for a patient with the BRAF sequencing studies.
V600E mutation.56 Similarly, in a RAS-mutated, MAPK Important obstacles to these therapies must be consid-
pathwayactivated, relapsed, refractory myeloma, the ered as we develop personalized approaches (Table 3). The
MEK inhibitor trametinib has been shown to achieve deep major limitations may include a dynamic disease process
response. A larger study of trametinib has also shown very with a constantly evolving genome, the coexistence of
encouraging efficacy.57 Numbers of such druggable mu- subclonal populations with only a subset of subclones that
tations are being recognized, each in a small subset of harbor a mutation, and lack of expressed mutant alleles
patients, with the potential to add a targeted agent for making the presence of a mutation not a clinically important
these patients. The large NCI MATCH trial, which is eval- target and rendering a specific targeted treatment in-
uating targeted strategies for a number of malignancies, is efficient. Moreover, other downstream activating mutations
expanding to include myeloma. In this distinctive trial in the same pathway may coexist and would make a targeted
concept, patients tumor cells are initially sequenced to therapy ineffective as well. In one study, a number of pa-
identify unique mutations and for those druggable muta- tients had coexistent KRAS and BRAF mutations.17 For these
tions, a specific inhibitor will be used to evaluate efficacy patients, BRAF-directed therapy will not be effective. De-
across various malignancies. NCI MATCH and other similar spite these potential limitations, NGS is a promising tool
studies will usher in an era where therapy will be tailored to for targeted therapy or to identify a specific drug that
molecular and genomic characteristics rather than just could be combined with current therapies including im-
phenotypic or pathologic diagnosis. munomodulatory agents, proteasome inhibitors, and mono-
Mutations in SF3B1, FGFR3, ATM/ATR, IDH1/2, and CCND1 clonal antibodies.
as well as RAS/RAF, NF-kappa B, and mTOR pathwayrelated
genes have been reported in myeloma. These mutations can Response Assessment
be targeted by appropriate inhibitors. To identify these Deep IgH sequencing has been used to evaluate minimal
mutations, a targeted sequencing approach is being pur- residual disease in multiple myeloma. Next-generation se-
sued.52,58 The study of the epigenome has also provided quencing has been recently reported to be especially useful

TABLE 3. Obstacles to Successful Therapeutic Application of Next-Generation Sequencing Data

Genomic Change Clinical Impact Possible Interventions to Overcome Obstacles


Subclonal Nature of Mutation Limited clinical response despite effective Evaluate subclonal response
targeted therapy
Target multiple subclones, combination therapy
Lack or Limited Expression of the Minimal to no effect of targeted agent Must evaluate both mutation and expression
Mutant Allele
Select alternate target/s
Presence of Mutation Downstream Transient or no response Prefer targeting downstream mutation from the beginning
of Targeted Mutation
Resequence genome if response plateau or progression
Evolution of New Driver Mutation Transient or no response Resequence genome if response plateau or progression
Direct therapy at new target or use broader multitarget agent

e446 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


NEXT-GENERATION SEQUENCING

to detect one cell among 1,000,000 in bone marrow, and the mutated cells, it will only remove 30% of the cells with
absence of myeloma with such precision predicts superior overall response, well below partial response criteria.
survival.25 This aspect of minimal residual disease is dis- However, if we measure BRAF-mutated cells, we can
cussed in accompanying article. In regards to the evaluation conclude a complete clonal response. Sensitive laboratory
of agents targeting specific mutations, it is necessary to methods are being developed to detect such responses to
consider clonal content; what proportion of myeloma cells consider their practical applications.
carry such a mutation? It is well described that some of the
driver mutations are subclonal and only present in small CONCLUSION
number of cells. As we use specific inhibitors, this therapy Next-generation sequencing data identify new markers
will potentially only impact the proportion of cells that carry that accurately characterize prognosis, patient outcome,
such mutations. Thus, the ability to detect a response at the and response to treatment allowing elaboration of per-
clonal level will be an important response criterion in the sonalized therapy. Incorporation of these markers into the
future. For example, if 30% of cells contain a BRAF mutation clinical practice should be evaluated and be made widely
and BRAF-directed therapy is able to eliminate all of these available.

References

1. Morgan GJ, Walker BA, Davies FE. The genetic architecture of multiple patients: a study of the Intergroupe Francophone du Myelome. J Clin
myeloma. Nat Rev Cancer. 2012;12:335-348. Oncol. 2008;26:4798-4805.
2. Zhan F, Huang Y, Colla S, et al. The molecular classification of multiple 15. Kuiper R, Broyl A, de Knegt Y, et al. A gene expression signature for high-
myeloma. Blood. 2006;108:2020-2028. risk multiple myeloma. Leukemia. 2012;26:2406-2413.
3. Broyl A, Hose D, Lokhorst H, et al. Gene expression profiling for mo- 16. Shaughnessy JD Jr, Zhan F, Burington BE, et al. A validated gene expression
lecular classification of multiple myeloma in newly diagnosed patients. model of high-risk multiple myeloma is defined by deregulated expression
Blood. 2010;116:2543-2553. of genes mapping to chromosome 1. Blood. 2007;109:2276-2284.
4. Lokhorst HM, Plesner T, Laubach JP, et al. Targeting CD38 with dar- 17. Bolli N, Avet-Loiseau H, Wedge DC, et al. Heterogeneity of genomic
atumumab monotherapy in multiple myeloma. N Engl J Med. 2015;373: evolution and mutational profiles in multiple myeloma. Nat Commun.
1207-1219. 2014;5:2997.
5. Stewart AK, Rajkumar SV, Dimopoulos MA, et al; ASPIRE Investigators. 18. Lohr JG, Stojanov P, Carter SL, et al; Multiple Myeloma Research
Carfilzomib, lenalidomide, and dexamethasone for relapsed multiple Consortium. Widespread genetic heterogeneity in multiple myeloma:
myeloma. N Engl J Med. 2015;372:142-152. implications for targeted therapy. Cancer Cell. 2014;25:91-101.
6. Attal M, Lauwers-Cances V, Marit G, et al; IFM Investigators. Lenali- 19. Walker BA, Boyle EM, Wardell CP, et al. Mutational spectrum, copy
domide maintenance after stem-cell transplantation for multiple my- number changes, and outcome: results of a sequencing study of
eloma. N Engl J Med. 2012;366:1782-1791. patients with newly diagnosed myeloma. J Clin Oncol. 2015;33:
7. Benboubker L, Dimopoulos MA, Dispenzieri A, et al; FIRST Trial Team. 3911-3920.
Lenalidomide and dexamethasone in transplant-ineligible patients with 20. Kandoth C, McLellan MD, Vandin F, et al. Mutational landscape and
myeloma. N Engl J Med. 2014;371:906-917. significance across 12 major cancer types. Nature. 2013;502:333-339.
8. Lonial S, Dimopoulos M, Palumbo A, et al; ELOQUENT-2 Investigators. 21. Mulligan G, Lichter DI, Di Bacco A, et al. Mutation of NRAS but not KRAS
Elotuzumab therapy for relapsed or refractory multiple myeloma. significantly reduces myeloma sensitivity to single-agent bortezomib
N Engl J Med. 2015;373:621-631. therapy. Blood. 2014;123:632-639.
9. Mimura N, Hideshima T, Anderson KC. Novel therapeutic strategies for 22. Alexandrov LB, Nik-Zainal S, Wedge DC, et al; Australian Pancreatic
multiple myeloma. Exp Hematol. 2015;43:732-741. Cancer Genome Initiative; ICGC Breast Cancer Consortium; ICGC
10. Chng WJ, Dispenzieri A, Chim CS, et al; International Myeloma Working MMML-Seq Consortium; ICGC PedBrain. Signatures of mutational
Group. IMWG consensus on risk stratification in multiple myeloma. processes in human cancer. Nature. 2013;500:415-421.
Leukemia. 2014;28:269-277. 23. Kortum KM, Langer C, Monge J, et al. Longitudinal analysis of 25
11. Palumbo A, Avet-Loiseau H, Oliva S, et al. Revised International Staging sequential sample-pairs using a custom multiple myeloma mutation
System for multiple myeloma: a report from International Myeloma sequencing panel (M(3)P). Ann Hematol. 2015;94:1205-1211.
Working Group. J Clin Oncol. 2015;33:2863-2869. 24. Kortum KM, Langer C, Monge J, et al. Targeted sequencing using a 47
12. Moreau P, Attal M, Garban F, et al; SAKK; IFM Group. Heterogeneity gene multiple myeloma mutation panel (M(3) P) in -17p high risk
of t(4;14) in multiple myeloma. Long-term follow-up of 100 cases disease. Br J Haematol. 2015;168:507-510.
treated with tandem transplantation in IFM99 trials. Leukemia. 2007; 25. Avet-Loiseau H, Corre J, Lauwers-Cances V, et al. Evaluation of minimal
21:2020-2024. residual disease (MRD) by next generation sequencing (ngs) is highly
13. Kumar S, Fonseca R, Ketterling RP, et al. Trisomies in multiple myeloma: predictive of progression free survival in the IFM/DFCI 2009 trial. Blood.
impact on survival in patients with high-risk cytogenetics. Blood. 2012; 2015;126:191.
119:2100-2105. 26. Davies FE, Dring AM, Li C, et al. Insights into the multistep trans-
14. Decaux O, Lode L, Magrangeas F, et al; Intergroupe Francophone du formation of MGUS to myeloma using microarray expression analysis.
Myelome. Prediction of survival in multiple myeloma based on gene Blood. 2003;102:4504-4511.
expression profiles reveals cell cycle and chromosomal instability sig- 27. Zhan F, Hardin J, Kordsmeier B, et al. Global gene expression profiling
natures in high-risk patients and hyperdiploid signatures in low-risk of multiple myeloma, monoclonal gammopathy of undetermined

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e447


SZALAT AND MUNSHI

significance, and normal bone marrow plasma cells. Blood. 2002;99: 43. Pei H, Zhang L, Luo K, et al. MMSET regulates histone H4K20 methylation
1745-1757. and 53BP1 accumulation at DNA damage sites. Nature. 2011;470:
28. Mulligan G, Mitsiades C, Bryant B, et al. Gene expression profiling and 124-128.
correlation with outcome in clinical trials of the proteasome inhibitor 44. Martinez-Garcia E, Popovic R, Min DJ, et al. The MMSET histone methyl
bortezomib. Blood. 2007;109:3177-3188. transferase switches global histone methylation and alters gene ex-
29. Chng WJ, Chung TH, Kumar S, et al. Gene signature combinations pression in t(4;14) multiple myeloma cells. Blood. 2011;117:211-220.
improve prognostic stratification of multiple myeloma patients. Leu- 45. Kuo AJ, Cheung P, Chen K, et al. NSD2 links dimethylation of histone H3
kemia. Epub 2015 Dec 16. at lysine 36 to oncogenic programming. Mol Cell. 2011;44:609-620.
30. Rashid N, Minvielle S, Magrangeas F, et al. Alternative splicing is a 46. Popovic R, Martinez-Garcia E, Giannopoulou EG, et al. Histone methyl-
frequent event and impacts clinical outcome in myeloma: a large RNA- transferase MMSET/NSD2 alters EZH2 binding and reprograms the
seq data analysis of newly-diagnosed myeloma patients. Presented at: myeloma epigenome through global and focal changes in H3K36 and
56th ASH Annual Meeting and Exposition; December 2014; San Fran- H3K27 methylation. PLoS Genet. 2014;10:e1004566.
cisco, CA; abstract 638. 47. Marango J, Shimoyama M, Nishio H, et al. The MMSET protein is a
31. Fulciniti M, Samur MK, Rashid NU, et al. Functional and clinical impact of histone methyltransferase with characteristics of a transcriptional
splicing factor dysregulation in multiple myeloma. Blood. 2015;126:726. corepressor. Blood. 2008;111:3145-3154.
32. Cleynen A, Szalat R, Samur MK, et al. The fusion gene landscape in 48. Afifi S, Michael A, Azimi M, et al. Role of histone deacetylase inhibitors
multiple myeloma, with clinical impact. Blood. 2015;126:835. in relapsed refractory multiple myeloma: a focus on vorinostat and
33. Samur MK, Gulla A, Cleynen A, et al. Differentially expressed and panobinostat. Pharmacotherapy. 2015;35:1173-1188.
prognostically significant lincrnas may impact immune system and 49. Kaiser MF, Johnson DC, Wu P, et al. Global methylation analysis
tumor progression in multiple myeloma (MM). Blood. 2015;126:2989. identifies prognostically important epigenetically inactivated tumor
34. Seckinger A, Meiner T, Moreaux J, et al. miRNAs in multiple myeloma suppressor genes in multiple myeloma. Blood. 2013;122:219-226.
a survival relevant complex regulator of gene expression. Oncotarget. 50. Walker BA, Wardell CP, Chiecchio L, et al. Aberrant global methylation
2015;6:39165-39183. patterns affect the molecular pathogenesis and prognosis of multiple
35. Wu P, Agnelli L, Walker BA, et al. Improved risk stratification in myeloma myeloma. Blood. 2011;117:553-562.
using a microRNA-based classifier. Br J Haematol. 2013;162:348-359. 51. Agirre X, Castellano G, Pascual M, et al. Whole-epigenome analysis in
36. Zhou Y, Chen L, Barlogie B, et al. High-risk myeloma is associated with multiple myeloma reveals DNA hypermethylation of B cell-specific
global elevation of miRNAs and overexpression of EIF2C2/AGO2. Proc enhancers. Genome Res. 2015;25:478-487.
Natl Acad Sci USA. 2010;107:7904-7909. 52. Bolli N, Avet-Loiseau H, Gimondi S, et al. Genomic landscape and its
37. Lionetti M, Biasiolo M, Agnelli L, et al. Identification of microRNA ex- prognostic implications in multiple myeloma using a targeted se-
pression patterns and definition of a microRNA/mRNA regulatory quencing approach. Blood. 2015;126:370.
network in distinct molecular groups of multiple myeloma. Blood. 2009; 53. Lonial S, Boise LH, Kaufman J. How I treat high-risk myeloma. Blood.
114:e20-e26. 2015;126:1536-1543.
38. Gatt ME, Zhao JJ, Ebert MS, et al. MicroRNAs 15a/16-1 function as tumor 54. Amin SB, Yip WK, Minvielle S, et al. Gene expression profile alone is
suppressor genes in multiple myeloma. Blood. Epub 2010 Oct 20. inadequate in predicting complete response in multiple myeloma.
39. Roccaro AM, Sacco A, Thompson B, et al. MicroRNAs 15a and 16 Leukemia. 2014;28:2229-2234.
regulate tumor proliferation in multiple myeloma. Blood. 2009;113: 55. Raab MS, Lehners N, Xu J, et al. Spatially divergent clonal evolution in
6669-6680. multiple myeloma: overcoming resistance to BRAF inhibition. Blood.
40. Fulciniti M, Amodio N, Bandi RL, et al. miR-23b/SP1/c-myc forms a feed- Epub 2016 Feb 16.
forward loop supporting multiple myeloma cell growth. Blood Cancer J. 56. Andrulis M, Lehners N, Capper D, et al. Targeting the BRAF V600E
2016;6:e380. mutation in multiple myeloma. Cancer Discov. 2013;3:862-869.
41. Min DJ, Ezponda T, Kim MK, et al. MMSET stimulates myeloma cell 57. Heuck C, Jethava Y, Khan RZ, et al. Targeted MEK inhibition in patients
growth through microRNA-mediated modulation of c-MYC. Leukemia. with previously treated multiple myeloma. Blood. 2014;124:4775.
2013;27:686-694. 58. Heuck C, Chavan SS, Stein CK, et al. Comprehensive genomic profiling of
42. Rio-Machin A, Ferreira BI, Henry T, et al. Downregulation of specific multiple myeloma in the course of clinical care identifies targetable and
miRNAs in hyperdiploid multiple myeloma mimics the oncogenic effect prognostically significant genomic alterations. Blood. 2015;126:369.
of IgH translocations occurring in the non-hyperdiploid subtype. Leu- 59. Rashid NU, Sperling AS, Bolli N, et al. Differential and limited expression
kemia. 2013;27:925-931. of mutant alleles in multiple myeloma. Blood. 2014;124:3110-3117.

e448 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


LUNG CANCER

Immunotherapy: Beyond
AntiPD-1 and AntiPD-L1
Therapies

CHAIR
Scott J. Antonia, MD, PhD
Moffitt Cancer Center
Tampa, FL

SPEAKERS
Edmund Moon, MD
Hospital of the University of Pennsylvania
Philadelphia, PA

Johan F. Vansteenkiste, MD, PhD


University Hospital KU Leuven
Leuven, Belgium
ANTONIA, VANSTEENKISTE, AND MOON

Immunotherapy: Beyond AntiPD-1 and AntiPD-L1


Therapies
Scott J. Antonia, MD, PhD, Johan F. Vansteenkiste, MD, PhD, and Edmund Moon, MD

OVERVIEW

Advanced-stage nonsmall cell lung cancer (NSCLC) and small cell lung cancer are cancers in which chemotherapy produces a
survival benefit, although it is small. We now know that antiPD-1/PD-L1 has substantial clinical activity in both of these
diseases, with an overall response rate (ORR) of 15%20%. These responses are frequently rapid and durable, increase
median overall survival (OS) compared with chemotherapy, and produce long-term survivors. Despite these very significant
results, many patients do not benefit from antiPD-1/PD-L1. This is because of the potential for malignancies to co-opt
myriad immunosuppressive mechanisms other than aberrant expression of PD-L1. Conceptually, these can be divided into
three categories. First, for some patients there is likely a failure to generate sufficient functional tumor antigen-specific
T cells. Second, for others, tumor antigenspecific T cells may be generated but fail to enter into the tumor parenchyma.
Finally, there are a large number of immunosuppressive mechanisms that have the potential to be operational within the
tumor microenvironment: surface membrane immune checkpoint proteins PD-1, CTLA-4, LAG3, TIM3, BTLA, and adenosine
A2AR; soluble factors and metabolic alterations interleukin (IL)-10, transforming growth factor (TGF)-b, adenosine, IDO, and
arginase; and inhibitory cells, cancer-associated fibroblasts (CAFs), regulatory T cells, myeloid-derived suppressor cells
(MDSCs), and tumor-associated macrophages. In this article, we discuss three strategies to generate more tumor-reactive
T cells for patients: antiCTLA-4, therapeutic tumor vaccination, and adoptive cellular therapy, with T cells redirected to
tumor antigens using T-cell receptor (TCR) or chimeric antigen receptor (CAR) gene modification. We also review some of the
various strategies in development to thwart tumor microenvironment immunosuppressive mechanisms. Strategies to drive
more T cells into tumors remain a significant challenge.

STRATEGIES TO INCREASE THE NUMBER OF the CTLA-4expanded T cells are prevented from being
TUMOR-REACTIVE T CELLS functionally inhibited when they enter into the tumor mi-
AntiCTLA-4 croenvironment. We recently reported the results of two
CTLA-4 is an immune checkpoint protein. Unlike PD-1, separate trials with these monoclonal antibodies, one with
CTLA-4 for patients with cancer suppresses T cells within nivolumab (antiPD-1) and ipilimumab (antiCTLA-4) and the
the lymphoid compartment rather than within the tumor other with MEDI4736 (antiPD-L1) and tremelimumab
microenvironment by limiting T-cell proliferation, and thus (antiCTLA-4). The initial doses and schedules of the nivolumab/
preventing expansion of antitumor T-cell responses. Evidence ipilimumab combination were toxic and produced a response
for this comes from a study demonstrating that among pa- rate of 15%; however, more tolerable doses have now been
tients with melanoma who were treated with ipilimumab, discovered.4 The durvalumab/tremelimumab combination
there was the appearance of tumor antigenspecific T cells was associated with manageable toxicity and produced re-
with specificities in the blood of clinically responsive patients sponses for patients who were PD-L1 biomarkernegative to a
not present in pretreatment and not appearing in the blood similar degree as antiPD-1 monotherapy for patients who
of clinically nonresponsive patients.1 were PD-L1 positive.5

AntiPD-1/PD-L1 in combination with antiCTLA-4. Anti Lung Cancer Vaccines


CTLA-4 (ipilimumab) has not been tested as a single agent Melanoma-associated antigen 3 cancer vaccination and
for patients with NSCLC, but has some activity in combination MAGRIT. We start this section with the MAGRIT trial, as this
with chemotherapy.2,3 In combination with antiPD-1/PD-L1, is the largest vaccination study, in a population that has long

From the Department of Thoracic Oncology Moffitt Cancer Center, Tampa, FL; Respiratory Oncology Unit, University Hospital KU Leuven, Leuven, Belgium; Hospital of the University of
Pennsylvania, Philadelphia, PA.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Scott J. Antonia, MD, PhD, Moffitt Cancer Center, 12902 Magnolia Dr., MRC 3-East, Tampa, FL 33612; email: scott.antonia@moffitt.org.

2016 by American Society of Clinical Oncology.

e450 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


BEYOND ANTIPD-1 AND ANTIPD-L1 THERAPIES

been considered to be optimal candidates: patients with TABLE 1. Steps in the Clinical Development of Cancer
minimal residual disease after definitive therapy. This is also Vaccines
the largest therapeutic study overall in NSCLC, and the
program nicely shows how vaccines were developed in Phase Objective Questions
NSCLC (Table 1). Preclinical Expression Broadly expressed?
The melanoma-associated antigen 3 (MAGE-A3) protein is Conserved in metastatic cells?
totally tumor-specific and present in approximately 35% of Specificity Tumor specific or not?
early-stage NSCLC.6 In a mechanism-of-action study, it was Mechanism of Immunogenic Effective humoral and cellular
shown that MAGE-3 protein with adjuvant AS02B successfully Action response?
induced specific antibodies, CD8+, and CD4+ T cells (whereas Phase I-II Clinical effects Activity?
MAGE-3 protein alone did not).7 In the hypothesis-generating Tolerability?
phase II, double-blind, randomized, placebo-controlled trial, Predictive biomarker?
182 patients with completely resected MAGE-A3positive stage Phase III Patient benefit Outcome (survival)?
IB-II NSCLC received the MAGE-A3 cancer vaccine (recombinant TolerabilityQuality of life effects?
MAGE-A3 protein combined with an immunostimulant) or Predictive biomarker confirmed?
placebo.8 No significant toxicity was observed, and a
potential predictive biomarker was suggested.9
The ensuing large phase III study, MAGRIT (MAGE-A3 as Other recent phase III trials. The mucin MUC1 expression is
Adjuvant NonSmall Cell Lung Cancer Immunotherapy) was altered, mainly by aberrant glycosylation, in many cancer
reported at the 2014 European Society for Medical Oncology types, including NSCLC. The tandem repeat MUC1-peptide
Congress (Table 2). Patients who were MAGE-A3 positive with in a liposomal formulation is the BLP-25, or tecemotide,
completely resected stage IB-II-IIIA NSCLC and adjuvant che- vaccine. START (Stimulating Targeted Antigenic Responses
motherapy, as clinically indicated, were randomly assigned 2:1 to NSCLC Trial) was a phase III, double-blind, random-
to receive MAGE-A3 vaccine or placebo. Almost 14,000 sur- ized, placebo-controlled trial comparing maintenance
gical patients were screened; 4,210 patients were MAGE-A3 therapy with tecemotide (829 patients) or placebo (410
positive (33%) and 2,312 patients were randomly assigned. patients) for patients with unresectable stage III NSCLC
The median disease-free survival (primary endpoint) was slightly who did not progress after sequential or concurrent
better with the MAGE-A3 cancer vaccine (60.5 vs. 57.9 months), chemoradiotherapy.10 The primary endpointoverall
but the difference was unfortunately not significant (hazard survivalwas not significantly different between the vac-
ratio [HR] 1.02; 95% CI, 0.891.18; p = .74). No subgroups with cine and placebo group (25.6 vs. 22.3 months). However,
potential benefit could be identified. The safety of this approach preplanned subgroup analysis showed that the patients
as adjuvant therapy in NSCLC was confirmed, as there were treated with concurrent chemoradiotherapy (829 patients)
higher rates of mild injection-site reactions and flu-like symp- had a 10.2-month improvement in OS (30.8 vs. 20.6 months;
toms than with placebo, but grade 3 and 4 treatment-related adjusted HR 0.78; p = .016). The consequential trial was
symptoms and treatment-related signigicant adverse events START 2, a similar large randomized, placebo-controlled trial
were infrequent. On the basis of this disappointing result, for patients who completed concurrent chemoradiotherapy
further development of the MAGE-A3 vaccine in NSCLC has for unresectable stage III NSCLC (NCT02049151). However,
been abandoned. this trial and further development of tecemotide was
abandoned after disappointing results of a smaller trial
among Japanese patients with stage III NSCLC and concur-
rent chemoradiotherapy.
TG4010 is a vaccine based on a recombinant viral vector
KEY POINTS (attenuated strain of vaccinia virus) expressing both the
tumor-associated antigen MUC1 and IL-2. This vaccine is
AntiPD-1/PD-L1 have clinical activity in lung cancer. explored in the phase IIB/III TIME trial (NCT01383148). This
The majority of patients treated with single agent double-blind, placebo-controlled trial evaluates standard
antiPD-1/PD-L1 do not benefit. first-line chemotherapy with or without TG4010 for patients
Combination therapy with multiple immunotherapeutics with MUC1-positive stage IV NSCLC. In the phase IIB part,
will be necessary to improve clinical efficacy. the predictive value of activated natural killer cells (triple-
Strategies are being developed to increase the number positive activated lymphocytes [TrPAL], CD16+ CD56+ CD69+)
of tumor antigen reactive T cells, to drive more of these
was evaluated on the basis of a progression-free survival (PFS)
into the tumor parenchyma, and to block a variety of
immunosuppressive mechanisms in the tumor
endpoint.11 If patients with TrPAL lower than normal values
microenvironment. had a Bayesian probability of greater than 95% so that the
Given the heterogeneity regarding the specific ways that HR and PFS were less than 1, and if those with TrPAL values
individual tumors evade immune-mediated rejection, greater than normal had a probability of greater than 80%
relevant biomarkers will be necessary to guide therapy. so that the HR for PFS was greater than 1, then the TrPAL
biomarker would be considered validated. The former was

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e451


ANTONIA, VANSTEENKISTE, AND MOON

TABLE 2. Recent Randomized Trials of Cancer Vaccines


No.
Compound Trial Design Patients Study Population Treatment Arms Primary Endpoint Other Endpoints
MAGE-A3 Phase III RCT 2,270 Completely resected MAGE-A3 vaccine vs. Disease-free Prospective validation
(MAGRIT)5 MAGE-A3positive placebo survival: HR 1.02; 95% of predictive gene
stage IB-IIIA NSCLC CI, 0.891.18; p = .738 signature not
feasible
Tecemotide Phase III RCT 1,513 Stage III NSCLC after LBLP-25 vaccine vs. OS: HR 0.88; 95% CI, Subgroup analysis;
(START)6 chemoradiation placebo 0.751.03; p = .123 concurrent
therapy chemoradiation:
OS: HR 0.78; 95% CI,
0.640.95; p = .016
TG4010 Phase II B RCT 222 Untreated patients with Chemotherapy with TrPAL biomarker PFS: HR 0.74; 95% CI,
(TIME; stage IV MUC1- TG4010 vaccine vs. validation: 0.550.98
NCT01383148) positive NSCLC chemotherapy achieved in part
plus placebo
Phase III RCT 800 Untreated patients with Chemotherapy plus OS; phase III part Safety, response rate,
(TIME; stage IV nonsquamous TG4010 vaccine vs. ongoing duration of response
NCT01383148) MUC1-positive NSCLC chemotherapy
plus placebo
Belagen-pumatucel-L Phase III RCT 700 Advanced NSCLC in Belagen pumatucel-L OS: HR 0.94; 95% CI, Subgroup analysis;
(STOP)8 disease control after vs. placebo 0.731.20; p = .594 OS for patients
standard therapy with radiotherapy:
HR 0.61; 95% CI,
0.380.96; p = .032
Abbreviations: MAGE-A3, melanoma-associated antigen 3; HR, hazard ratio; NSCLC, nonsmall cell lung cancer; OS, overall survival; LBLP, stimuvax; RCT, randomized, placebo-
controlled trial; PFS, progression-free survival.

the case; the latter was not. Overall, median PFS was agent with almost no grade 3/4 toxicity,8 and a predictive
5.9 months in the TG4010 group and 5.1 months in the gene signature from metastatic melanoma reproducible in
placebo group (HR 0.74; 95% CI, 0.550.98; p = .019). On early-stage NSCLC.9 Differences in population (stage IB-II-IIIA
the basis of these findings, the phase III part of the trial vs. IB-II), diagnostic testing (MAGE-A3 reverse transcriptase
continues. polymerase chain reaction on formalin-fixed, paraffin-
Belagenpumatucel-L is a vaccine based on a mixture of embedded vs. fresh, frozen tissue), and drug composition
allogeneic tumor cells with TGF-b2 antisense blockade as (adjuvants AS15 vs. AS02B) could be ruled out as causes of
adjuvant. It was studied in the phase III STOP (Survival: the negative MAGRIT results in comparison with the initial
Tumor-free, Overall, and Progression-free) trial for patients phase II randomized trial. Overinterpretation of the existing
with stage III-IV NSCLC achieving a response or dis- data, as is the case for many compounds, may have been in
ease control after first-line therapy.12 Median OS was place: factors for the preclinical data were that most of the
20.3 months with belagenpumatucel-L versus 17.8 months immune responses were antibodies or CD4+ T cells, whereas
with placebo (HR 0.94; p = .594). In the (small) subgroup of it was more difficult to demonstrate CD8+ T cells. For the
patients treated with previous radiotherapy, the OS HR was phase II randomized data, the sample size was limited, with
0.61 (p = .032). possible hidden imbalances across treatment arms. Another
factor was raising the bar in outcome compared with his-
Why were early positive findings not translated into pa- torical data in the placebo arm, thus making it more difficult
tients benefits in phase III? Looking at MAGRIT, it was most to improve. Indeed, in the adjuvant landmark study IALT,13
disappointing that the very well tolerated MAGE-A3 vaccine the 5-year OS was 40.4% with surgery alone and 44.5% with
could not enrich our adjuvant therapies in NSCLC, espe- surgery followed by adjuvant chemotherapy. In MAGRIT, the
cially as no real progress in this setting has been seen for 5-year OS in the placebo arm was 58.7%.
more than a decade. Indeed, the IALT trial established
cisplatin-based chemotherapy as a standard for resected Where should we go with cancer vaccination in
stage II and IIIA tumors13 and was initially presented at the NSCLC? Several recently reported phase III trials with vac-
2003 American Society of Clinical Oncology (ASCO) Annual cination approaches in NSCLC did not reach their primary
Meeting. Since then, we have witnessed failures of gefitinib,14 endpoint (MAGRIT, START, STOP). This essentially means
erlotinib,15 bevacizumab, and pharmacogenetically tailored that immunologic response vaccines are not translated into
chemotherapy.16-18 clinical benefits. Immunologic responses are measured in
MAGRIT was started on the basis of previously docu- the circulatory compartment, typically with antibody re-
mented clear responses to the MAGE-A3 cancer vaccine in sponses and less easily with specific lymphocyte responses,
metastatic melanoma,19 a phase II randomized study with an and these may not be translated sufficiently in the peri-
HR for OS similar to modern adjuvant chemotherapy with an tumoral stromal immune system because of the strong

e452 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


BEYOND ANTIPD-1 AND ANTIPD-L1 THERAPIES

immunosuppressive environment on NSCLC. Indeed, find- TCRs. This could translate into increased toxicity. Another
ings from recent years gave deeper understanding of the limitation is that each TCR is specific for a given peptide-MHC
numerous mechanisms of immune suppression induced complex, thus making it applicable to patients who shared
by cancer cells: inhibitory cells such as regulatory T-lymphocytes both MHC alleles and TAAs.
and MDSCs, inhibitory molecules such as TGF-b and IDO, Chimeric antigen receptor engineering artificially com-
and immune checkpointinhibiting pathways such as those bines a single-chain Fv fragment fused from the variable light
related to CTLA-4 and the PD-1 receptor and its ligand and heavy chains of a TAA-specific antibody with the CD3z-
(PD-L1). 20 signaling chain of the TCR.31 This so-called first-generation
Consequently, combination therapies of agents that re- CAR, which confers antigen-specific tumor lytic capability,
verse this immunosuppressive environment of NSCLC (such can be further modified to express one or more additional
as the monoclonal antibodies that inhibit the PD-1/PD-L1 costimulatory domains to increase T-cell persistence and
pathway) with vaccination may be the best way forward. induce proliferation upon antigen-binding. CD28, 41BB,
Both complement each other in the loop of immune response, and ICOS are a few examples of such domains that can be
vaccination with delivery and enhanced presentation of included in these second- and third-generation CARs
antigens, and checkpoint inhibitors with alleviation of the (Fig. 1B).32 The advantages of CARs include highly specific
immune-suppressive tumor microenvironment. It would be and avid recognition of the targeted antigen and MHC in-
particularly interesting to deliver neo-antigens of neo- dependent signaling initiated by antigen binding. However,
epitopes, as the latter have recently been correlated with in general, CARs are only able to recognize antigens
the response to antiPD-1/PD-L1 therapies.21 Moreover, expressed or presented on the cell membrane of target cells,
this mutational load of neo-antigens and neo-epitopes is limiting the number of candidate TAAs that can be targeted
particularly high in NSCLC found in smokers, especially using CAR technology.
squamous cell lung cancer. Such mutational epitopes are
immunogenic when presented by major histocompatibility Tumor antigens as targets for genetically redirected
complex (MHC)receptors and have recently been associated T cells. Both TCR and CAR engineering strategies to redirect
with increased patient survival.22 adoptively transferred T cells require choosing an optimal
TAA for targeting. Ideally, the targeted TAA is highly
expressed by tumor cells but not by vital normal tissues (or at
Engineering T Cells for Lung Cancer and Thoracic least expressed at low levels by normal tissues). Otherwise,
Malignancies adoptively transferred T cells would be able to induce what
Efforts to redirect T cells to tumor. The more traditional has been coined off-tumor, on-target toxicity. Tumor-
strategy of adoptive T-cell transfer involved polyclonal associated antigens that have been targeted via engineered
expansion of tumor-infiltrating lymphocytes (TILs) from T cells in lung cancer and/or mesothelioma include (1)
tumor resection specimens and reinfusion of expanded NYESO1 (NCT01697527, NCT01967823), a cancer testis an-
populations of naturally occurring tumor-reactive TILs back tigen found in up to 30% of lung cancers;33 (2) VEGF receptor 2
into the patient. Although this strategy induced responses (NCT01218867), expressed in the majority of lung cancer
for patients with metastatic melanoma,23-25 identification samples;34 (3) MAGE-A3 (NCT02111850), found in up to
and ex vivo culturing of TILs from most other solid tumor 40% of lung cancers;6,35 (4) mesothelin (NCT02159716,
types is difficult. Over the last 2 decades, this challenge has NCT01583686, NCT02414269),36,37 a molecule involved in
been overcome by genetically modifying otherwise tumor- cell-cell adhesion but otherwise unclear in function,
nonreactive T cells to bear tumor reactivity. This involves expressed in virtually all mesothelioma samples of the epi-
taking bulk CD8+ and CD4+ T cells via leukapharesis, thelioid subtype and up to 80% of lung cancer samples;38,39
expanding them via coculture with artificial antigen- and (5) Wilm tumor protein 1 (NCT02408016), Wilm tumor
presenting cells or microbeads coated with anti-CD3/CD28 suppressor gene 1, which encodes a transcription factor
protein, and transducing them to express either a TCR clone essential in the normal development of the urogenital
or a CAR, conferring specificity to a tumor-associated anti- system. Refer to the publications by Cheever et al40 and
gen (TAA). Transduction can be performed using viral sys- Hinrichs and Restifo 41 for a more comprehensive review
tems (e.g., retrovirus and lentivirus) 26,27 and nonviral of TAA targets for T-cell immunotherapy in thoracic
systems (e.g., Sleeping Beauty transposon28 and mRNA malignancies.
electroporation29).
T-cell receptor engineering involves the modification of Challenges to genetically redirecting T cells to thoracic
T cells to express high-affinity alpha/beta TCRs bearing malignancies. Engineered T-cell therapy has demonstrated
known specificity and avidity for TAAs (Fig. 1A).30 One major dramatic and durable responses in patients with melanoma
advantage of TCR engineering is the ability to confer T-cell (TCR engineering)42 and leukemia.43 However, applying this
reactivity against both surface and intracellular antigens. immunotherapeutic strategy to many other solid tumors,
One potential drawback is the potential generation of ad- such as lung cancer and mesothelioma,44,45 has been met
ditional, unintended receptor specificities via rearranged with many challenges, limiting the objective responses
pairing of the a and b chains with those of the endogenous among patients. There have been no published reports of

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e453


ANTONIA, VANSTEENKISTE, AND MOON

FIGURE 1. Chimeric Antigen Receptors

Abbreviation: TAA, tumor-associated antigen.

clinical responses to TCR-engineered T cells among patients proportion of patients.36 The challenges inherent to solid
with lung cancer, and the limited data on the clinical ex- tumors have been described repeatedly for many years in
perience of CAR-engineered T cells for patients with me- naturally occurring TILs.46-48 T-cell receptorengineered
sothelioma describe partial responses in only a small T cells,49 and even CAR-engineered T cells, despite their

e454 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


BEYOND ANTIPD-1 AND ANTIPD-L1 THERAPIES

built-in costimulatory signaling motifs,50 succumb to similar is associated with hyaluronan accumulation in stroma, there
changes. These challenges include, but are not limited to, are efforts to combine hyaluronidase administration with
heterogeneous TAA expression and TAA shedding, short engineered T cells to overcome the stromal challenge.63
T-cell survival/persistence, suboptimal T-cell trafficking, the
barrier of tumor-associated stroma, the presence of sup- The challenge of inhibitory checkpoints. Checkpoint
pressive immune cells, upregulation of inhibitory check- blockade has recently received a lot of attention as a prom-
points, the expression of regulatory genes, lack of oxygen ising immunotherapeutic strategy in lung cancer.64 The most
and cellular nutrients, and immunosuppressive soluble studied checkpoint at the present time is PD-1, a negative
factors. Moon et al50,51 recently demonstrated that both regulator of the T-cell immune response that signals upon
TCR- and CAR-engineered T cells undergo profound sup- binding its ligands, PD-L1 and PD-L2, which are often highly
pression of their tumor-lytic and cytokine secretion capa- expressed in solid tumors and their tumor microenviron-
bilities, leading to an inability to cure human lung cancer ments. However, the majority of patients treated with PD-1
and human mesothelioma xenograft tumors. When these checkpoint blockade demonstrate no objective clinical re-
TCR/CAR TILs were isolated and allowed to rest away from sponse. One likely reason for this is that many patients tu-
the tumor microenvironment, they recovered a significant mors lack sufficient infiltration of tumor-reactive TILs to be
degree of their antitumor effector function. Preclinical unmasked by checkpoint blockade. Thus, combining adoptive
studies such as these are being conducted in parallel with the transfer of engineered tumor-reactive T cells with checkpoint
early-phase trials of adoptive T-cell therapy for thoracic blockade may be the key to successfully applying immuno-
malignancies, testing ways to overcome the much antici- therapy to cancers such as lung cancer and mesothelioma.
pated challenges in the solid tumor microenvironment. We Preclinical studies have already demonstrated the ability to
describe a few salient examples. augment TCR and CAR human T-cell control of lung cancer
and mesothelioma tumors by combining them with PD-1
The challenge of T-cell survival/persistence. There is a blockade.51
positive correlation between survival/persistence of geneti-
cally engineered T cells and clinical response.52 There are IMMUNOSUPPRESSION IN THE TUMOR
a few potential strategies of optimizing T-cell survival/ MICROENVIRONMENT
persistence in the context of solid tumors. Immune Checkpoint Proteins
Host preparative conditioning regimens (e.g., fludarabine There are several T-cell surface receptors called immune
and/or cyclophosphamide)53,54 can be administered prior to checkpoint proteins that deliver negative signals to T cells
adoptive transfer of genetically engineered T cells to reduce when they engage their ligands expressed on tissue- or
the number of circulating T cells (i.e., lymphodepletion). This antigen-presenting cells.65 This is a feedback mechanism
will promote the in vivo expansion of transferred T cells by whereby immune responses are dampened when no longer
limiting the competition for cytokines necessary for T-cell needed. This mechanism of control of T cells can be co-opted
survival/persistence (e.g., IL-7, IL-15).55 The majority of by tumors to escape rejection by the immune system.66,67 A
ongoing trials in thoracic malignancies (see above) have hallmark of malignancy is an inflamed tumor microenvi-
conditioning regimens incorporated in their protocols. ronment,68 and inflammatory cytokines such as g-interferon
Another strategy is further genetically modifying the can induce immune checkpoint ligand expression on tumor
TCR/CAR-bearing T cells to secrete the cytokines necessary cells.69 Therefore tumor antigenspecific T cells activated in
for their survival and persistence.56 In some cases, the same tumor-draining lymph nodes are shut down when they enter
cytokines can promote the secondary influx of other host into the tumor microenvironment.
immune cells, leading to additional antitumor/stroma ef-
fects.57 Optimizing the combination of costimulatory do- AntiPD-1/PD-L1 antibodies have significant antitumor
mains in CARs will also augment the cytokine secretion activity in lung cancer. The discovery of aberrant expres-
profiles of transferred T cells, and the costimulatory motifs sion of PD-L1 in tumors leads to the development of the
being tested in CARs are constantly evolving.58,59 therapeutic strategy of monoclonal antibodies designed to
prevent PD-1 binding to PD-L1. There are a number of
The challenge of tumor stroma. Stroma in both lung cancer different monoclonal antibodies specific to PD-1 or PD-L1
and mesothelioma consists of a multicellular matrix at the that are in clinical development for lung cancer, all having
invasion front of tumor and has a very crucial role in tumor similar tumor response rates of 15%20%.70 This demon-
progression.45,60 Mesothelioma tumors have especially strates that lung cancer is an immunotherapeutically re-
pronounced stromal components compared with other solid sponsive disease, and this strategy can have a major impact
tumors.61 There is a complex network of cross-talk between on OS and, more importantly, can influence the tail of the
stromal cells and tumor cells via both cell-cell interactions survival curve.
and soluble factors. Wang et al62 demonstrated the ability to
augment mesothelioma tumor control by combining CAR Other immune checkpoint proteins that may be opera-
T cells targeting CAFs and mesothelin-directed CAR T cells. tional within the tumor microenvironment. It is known
With the well-described observation that tumor progression that the extracellular tumor microenvironment contains

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e455


ANTONIA, VANSTEENKISTE, AND MOON

high levels of adenosine as a consequence of anaerobic Recently, two drugs have been approved for the treatment
glycolysis in hypoxic regions and preferential utilization of of idiopathic pulmonary fibrosis, pirfenidone and nintedanib.
aerobic glycolysis for energy metabolism in nonhypoxic The fact that the fibroblasts in this disease resemble
regions (the Warburg effect), producing a relative excess of CAFse.g., both express FAPled us to develop clinical
AMP; and tumor cell expression of the ectonucleotidase trials where these drugs will be repurposed as cancer im-
CD73 that catabolyzes AMP to produce adenosine.71,72 It munotherapeutics. Because CAFs are also known to support
had been known that adenosine produced within the the malignant phenotype, these drugs may have anticancer
hypoxic microenvironment of inflamed tissue functions to effects beyond relief of immunosuppression.
limit the exuberance of inflammatory responses to reduce
collateral damage of normal tissue by inflammatory cells and Myeloid-derived suppressor cells. Inflammation associ-
cytokines.73,74 This is because of a direct inhibitory effect of ated with the tumor microenvironment plays an important
adenosine on T cells that express adenosine A2ARs, a T-cell role in the development and progression of lung cancer. In
surface immune checkpoint protein,75 and leads to the the context of an inflammatory response, myeloid cells are
discovery that adenosine in the tumor microenvironment the primary recruited effectors. In lung cancer, these cells
interferes with antitumor immunity,76 suggesting that an- are activated macrophages, granulocytes, and MDSC.79
tagonism of the A2AR could be an effective cancer immu- Production of reactive oxygen and nitrogen species is one of
notherapeutic.77 Other immune checkpoint proteins that the major characteristics of all activated myeloid cells. The
can be expressed on T cells include BTLA, LAG3, and TIM3. formation of the free radical peroxynitrite (PNT) is the main
Antibodies that block the binding of these proteins to their result of interaction between superoxide and nitric oxide.
ligands are being developed and tested in clinical trials. Peroxynitrite can react directly with cysteine, methionine,
and tryptophan. A substantial number of studies have
Immune Suppressor Cells demonstrated high levels of nitrotyrosine in lung cancer.
Cancer-associated fibroblasts. Tumors secrete a variety of Gabrilovich et al79 demonstrated that MDSC can induce
growth factors and cytokines, including TGF-b, platelet- T-cell tolerance via production of PNT and nitration/
derived growth factor, basic fibroblast growth factor, IL-6, nitrosylation of TCR and CD8 molecules on the surface of
and IL-1, to recruit and activate CAFs.4-8 Fearons group T cells. T-cell receptors lose their ability to bind specific
first demonstrated that CAFs contribute significantly to peptide/MHC complexes and kill tumor cells.80
immunosuppression within the tumor microenvironment.78 Synthetic triterpenoids work through activation of the
Subsequently, it has been discovered that they do so transcription factor Nrf2 (nuclear factor [erythroid-derived]
through a variety of mechanisms. Recent studies dem- like 2), which induces the up-regulation of an array of anti-
onstrated that CAFs suppressed T-cell proliferation more oxidant molecules, such as glutathione, thioredoxin, catalase,
efficiently than normal fibroblasts and collaborate with and superoxide dismutase.81 We are testing one such triter-
tumor cells to create an immunosuppressive tumor micro- prenoid as an anticancer immunotherapeutic.
environment in head and neck squamous cell carcinoma.10
Cancer-associated fibroblasts have been shown to protect CONCLUSION
lung cancer cells from T-cellmediated destruction. Fibro- The discovery that blocking signaling through the T-cell
blast activation protein (FAP)-apositive CAFs were able to surface immune checkpoint protein PD-1 can produce a
protect tumor cells from tumor necrosis factormediated very significant impact on clinical outcomes of patients with
and interferon gmediated necrosis of tumor cells by T cells lung cancer not only gives us effective drugs that work as
and were also able to sequester T cells away from neoplastic single agents, but perhaps more importantly has given
cells by expressing C-X-C motif chemokine ligand 12. When us the proof-of-principle that lung cancer is certainly an
FAP-apositive CAFs were depleted, immunologic detection immunotherapeutically responsive disease. With the dis-
and destruction of tumor cells was restored, and interferon-g covery of the many ways that tumors can evade rejection by
or tumor necrosis factor were able to cause rapid hypoxic the immune system and the development of strategies to
necrosis.4,11,12 Inhibition of CXC receptor type 4 interacting thwart these comes real hope that we will be able to sig-
with C-X-C motif chemokine ligand 12 of CAFs was shown to nificantly improve on results obtained with antiPD-1/PD-L1
promote T-cell accumulation in the pancreatic tumor site blocking antibody monotherapy. It is clear that there is
and synergize with antiPD-L1.13 Cancer-associated fibro- considerable heterogeneity among patients with lung cancer
blasts are also known to induce immune suppressor cell with respect to which of the numerous potential immu-
types such as regulatory T cells and MDSCs, and immune noevasive mechanisms is operational. This means that in all
suppressor cells were reduced when CAFs were inhibited.14 likelihood we must interfere with several immunoevasive
Cancer-associated fibroblasts have also been shown to re- mechanisms for individual patients and that we must de-
cruit macrophages into tumors and activate M2 phenotypes velop good biomarkers to identify which of these mecha-
through secretion of IL-4, IL-6, and IL-8.9 All of these varied nisms is operational among individual patients to choose
mechanisms help to support an immunosuppressive, tumor- the proper combination of immunotherapeutics for each
promoting microenvironment. individual patient.

e456 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


BEYOND ANTIPD-1 AND ANTIPD-L1 THERAPIES

References

1. Kvistborg P, Philips D, Kelderman S, et al. Anti-CTLA-4 therapy broadens Cancer Group trial (Eudract:2007-000067-15; NCTgov:00478699). J Clin
the melanoma-reactive CD8+ T cell response. Sci Transl Med. 2014;6: Oncol. 2015;33 (suppl; abstr 7507).
254ra128. 19. Kruit WH, van Ojik HH, Brichard VG, et al. Phase 1/2 study of sub-
2. Lynch TJ, Bondarenko I, Luft A, et al. Ipilimumab in combination with cutaneous and intradermal immunization with a recombinant MAGE-3
paclitaxel and carboplatin as first-line treatment in stage IIIB/IV non- protein in patients with detectable metastatic melanoma. Int J Cancer.
small-cell lung cancer: results from a randomized, double-blind, 2005;117:596-604.
multicenter phase II study. J Clin Oncol. 2012;30:2046-2054. 20. Berraondo P, Umansky V, Melero I. Changing the tumor microenvi-
3. Reck M, Bondarenko I, Luft A, et al. Ipilimumab in combination with ronment: new strategies for immunotherapy. Cancer Res. 2012;72:
paclitaxel and carboplatin as first-line therapy in extensive-disease- 5159-5164.
small-cell lung cancer: results from a randomized, double-blind, 21. Rizvi N, Shepherd FA, Antonia SJ, et al. First-line monotherapy with
multicenter phase 2 trial. Ann Oncol. 2013;24:75-83. nivolumab (anti-PD-1; BMS-936558, ONO-4538) in advanced non-small
4. Antonia SJ, Gettinger SN, Chow LQ, et al. Nivolumab (anti-PD-1; BMS- cell lung cancer (NSCLC): safety, efficacy, and correlation of outcomes
936558, ONO-4538) and ipilimumab in first-line NSCLC: interim phase I with PD-L1 status. Int J Radiat Oncol Biol Phys. 2014;90S31 (suppl 5).
results. J Clin Oncol. 2014;32 (suppl; abstr 8023). 22. Brown SD, Warren RL, Gibb EA, et al. Neo-antigens predicted by tumor
5. Antonia S, Goldberg SB, Balmanoukian A, et al. Safety and antitumour genome meta-analysis correlate with increased patient survival.
activity of durvalumab plus tremelimumab in non-small cell lung Genome Res. 2014;24:743-750.
cancer: a multicentre, phase 1b study. Lancet Oncol. Epub 2016 Feb 5. 23. Dudley ME, Wunderlich JR, Yang JC, et al. Adoptive cell transfer therapy
6. Sienel W, Varwerk C, Linder A, et al. Melanoma associated antigen following non-myeloablative but lymphodepleting chemotherapy for
(MAGE)-A3 expression in stages I and II non-small cell lung cancer: results the treatment of patients with refractory metastatic melanoma. J Clin
of a multi-center study. Eur J Cardiothorac Surg. 2004;25:131-134. Oncol. 2005;23:2346-2357.
7. Atanackovic D, Altorki NK, Stockert E, et al. Vaccine-induced CD4+ T cell 24. Dudley ME, Wunderlich JR, Robbins PF, et al. Cancer regression and
responses to MAGE-3 protein in lung cancer patients. J Immunol. 2004; autoimmunity in patients after clonal repopulation with antitumor
172:3289-3296. lymphocytes. Science. 2002;298:850-854.
8. Vansteenkiste J, Zielinski M, Linder A, et al. Adjuvant MAGE-A3 im- 25. Rosenberg SA, Spiess P, Lafreniere R. A new approach to the adoptive
munotherapy in resected non-small-cell lung cancer: phase II ran- immunotherapy of cancer with tumor-infiltrating lymphocytes. Science.
domized study results. J Clin Oncol. 2013;31:2396-2403. 1986;233:1318-1321.
9. Ulloa-Montoya F, Louahed J, Dizier B, et al. Predictive gene signature in 26. Naldini L, Blomer U, Gallay P, et al. In vivo gene delivery and stable
MAGE-A3 antigen-specific cancer immunotherapy. J Clin Oncol. 2013; transduction of nondividing cells by a lentiviral vector. Science. 1996;
31:2388-2395. 272:263-267.
10. Butts C, Socinski MA, Mitchell PL, et al; START trial team. Tecemotide 27. Sharma S, Cantwell M, Kipps TJ, et al. Efficient infection of a human
(L-BLP25) versus placebo after chemoradiotherapy for stage III non- T-cell line and of human primary peripheral blood leukocytes with a
small-cell lung cancer (START): a randomised, double-blind, phase 3 pseudotyped retrovirus vector. Proc Natl Acad Sci USA. 1996;93:11842-
trial. Lancet Oncol 2014;15:59-68. 11847.
11. Quoix EA, Forget F, Papai-Szekely Z, et al. Results of the phase IIb part of 28. Singh H, Huls H, Kebriaei P, et al. A new approach to gene therapy using
TIME study evaluating TG4010 immunotherapy in stage IV non-small Sleeping Beauty to genetically modify clinical-grade T cells to target
cell lung cancer (NSCLC) patients receiving first line chemotherapy. J Clin CD19. Immunol Rev. 2014;257:181-190.
Oncol. 2015;33 (suppl; abstr 3034). 29. Zhao Y, Zheng Z, Cohen CJ, et al. High-efficiency transfection of primary
12. Giaccone G, Bazhenova LA, Nemunaitis J, et al. A phase III study of human and mouse T lymphocytes using RNA electroporation. Mol Ther.
belagenpumatucel-L, an allogeneic tumour cell vaccine, as maintenance 2006;13:151-159.
therapy for non-small cell lung cancer. Eur J Cancer. 2015;51:2321-2329. 30. Schumacher TN. T-cell-receptor gene therapy. Nat Rev Immunol. 2002;
13. Arriagada R, Bergman B, Dunant A, et al. Cisplatin-based adjuvant 2:512-519.
chemotherapy in patients with completely resected non-small-cell lung 31. Gross G, Waks T, Eshhar Z. Expression of immunoglobulin-T-cell re-
cancer. N Engl J Med. 2004;350:351-360. ceptor chimeric molecules as functional receptors with antibody-type
14. Goss GD, Lorimer I, Tsao MS, et al. A phase III randomized, double-blind, specificity. Proc Natl Acad Sci USA. 1989;86:10024-10028.
placebo-controlled trial of the epidermal growth factor receptor inhibitor 32. Eshhar Z, Waks T, Bendavid A, et al. Functional expression of chimeric
gefitinb in completely resected stage IB-IIIA non-small cell lung cancer receptor genes in human T cells. J Immunol Methods. 2001;248:67-76.
(NSCLC): NCIC CTG BR.19. J Clin Oncol. 2010;28:18s(suppl; abstr LBA7005). 33. Nicholaou T, Ebert L, Davis ID, et al. Directions in the immune targeting
15. Kelly K, Altorki NK, Eberhardt WE, et al. Adjuvant erlotinib versus placebo in of cancer: lessons learned from the cancer-testis Ag NY-ESO-1. Immunol
patients with stage IB-IIIA non-small-cell lung cancer (RADIANT): a ran- Cell Biol. 2006;84:303-317.
domized, double-blind, phase III trial. J Clin Oncol. 2015;33:4007-4014. 34. Bonnesen B, Pappot H, Holmstav J, et al. Vascular endothelial growth
16. Friboulet L, Olaussen KA, Pignon JP, et al. ERCC1 isoform expression and DNA factor A and vascular endothelial growth factor receptor 2 expression in
repair in non-small-cell lung cancer. N Engl J Med. 2013;368:1101-1110. non-small cell lung cancer patients: relation to prognosis. Lung Cancer.
17. Bepler G, Williams CC, Schell MJ, et al. Molecular analysis-directed, 2009;66:314-318.
international, phase III trial in patients with advanced non-small-cell 35. Gure AO, Chua R, Williamson B, et al. Cancer-testis genes are coor-
lung cancer. J Clin Oncol. 2013;31:(suppl; abstr 8001). dinately expressed and are markers of poor outcome in non-small cell
18. Massuti B, Cobo M, Rodriguez-Paniagua JM, et al. SLCG SLCGG. Ran- lung cancer. Clin Cancer Res. 2005;11:8055-8062.
domized phase III trial of customized adjuvant chemotherapy (CT) 36. Beatty GL, Haas AR, Maus MV, et al. Mesothelin-specific chimeric
according BRCA-1 expression levels in patients with node positive antigen receptor mRNA-engineered T cells induce anti-tumor activity in
resected non-small cell lung cancer (NSCLS) SCAT: a Spanish Lung solid malignancies. Cancer Immunol Res. 2014 2:112-120.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e457


ANTONIA, VANSTEENKISTE, AND MOON

37. Maus MV, Haas AR, Beatty GL, et al. T cells expressing chimeric antigen 58. Guedan S, Chen X, Madar A, et al. ICOS-based chimeric antigen re-
receptors can cause anaphylaxis in humans. Cancer Immunol Res. 2013; ceptors program bipolar TH17/TH1 cells. Blood. 2014;124:1070-1080.
1:26-31. 59. Hombach AA, Abken H. Of chimeric antigen receptors and antibodies:
38. Ho M, Bera TK, Willingham MC, et al. Mesothelin expression in human OX40 and 41BB costimulation sharpen up T cell-based immunotherapy
lung cancer. Clin Cancer Res. 2007;13:1571-1575. of cancer. Immunotherapy 2013;5:677-681.
~ez NG. Application of mesothelin immunostaining in tumor di-
39. Ordo n 60. Bremnes RM, Donnem T, Al-Saad S, et al. The role of tumor stroma in
agnosis. Am J Surg Pathol. 2003;27:1418-1428. cancer progression and prognosis: emphasis on carcinoma-associated fi-
40. Cheever MA, Allison JP, Ferris AS, et al. The prioritization of cancer broblasts and non-small cell lung cancer. J Thorac Oncol. 2011;6:209-217.
antigens: a national cancer institute pilot project for the acceleration of 61. Shia J, Qin J, Erlandson RA, et al. Malignant mesothelioma with a
translational research. Clin Cancer Res. 2009;15:5323-5337. pronounced myxoid stroma: a clinical and pathological evaluation of 19
41. Hinrichs CS, Restifo NP. Reassessing target antigens for adoptive T-cell cases. Virchows Arch. 2005;447:828-834.
therapy. Nat Biotechnol. 2013;31:999-1008. 62. Wang LC, Lo A, Scholler J, et al. Targeting fibroblast activation protein in
42. Robbins PF, Kassim SH, Tran TL, et al. A pilot trial using lymphocytes tumor stroma with chimeric antigen receptor T cells can inhibit tumor
genetically engineered with an NY-ESO-1-reactive T-cell receptor: long- growth and augment host immunity without severe toxicity. Cancer
term follow-up and correlates with response. Clin Cancer Res. 2015;21: Immunol Res. 2014;2:154-166.
1019-1027. 63. Shepard HM. Breaching the castle walls: hyaluronan depletion as a
43. Maude SL, Frey N, Shaw PA, et al. Chimeric antigen receptor T cells for therapeutic approach to cancer therapy. Front Oncol. 2015;5:192.
sustained remissions in leukemia. N Engl J Med. 2014;371:1507-1517. 64. Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune cor-
44. Chen Z, Fillmore CM, Hammerman PS, et al. Non-small-cell lung relates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366:2443-2454.
cancers: a heterogeneous set of diseases. Nat Rev Cancer. 2014;14: 65. Yao S, Zhu Y, Chen L. Advances in targeting cell surface signalling mol-
535-546. ecules for immune modulation. Nat Rev Drug Discov. 2013;12:130-146.
45. Ujiie H, Kadota K, Nitadori JI, et al. The tumoral and stromal immune 66. Nguyen LT, Ohashi PS. Clinical blockade of PD1 and LAG3potential
microenvironment in malignant pleural mesothelioma: a comprehen- mechanisms of action. Nat Rev Immunol. 2015;15:45-56.
sive analysis reveals prognostic immune markers. OncoImmunology. 67. Pardoll DM. The blockade of immune checkpoints in cancer immu-
2015;4:e1009285. notherapy. Nat Rev Cancer. 2012;12:252-264.
46. Frey AB. Suppression of T cell responses in the tumor microenviron- 68. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation.
ment. Vaccine. 2015;33:7393-7400. Cell. 2011;144:646-674.
47. Monu N, Frey AB. Suppression of proximal T cell receptor signaling and 69. Lee SJ, Jang BC, Lee SW, et al. Interferon regulatory factor-1 is pre-
lytic function in CD8+ tumor-infiltrating T cells. Cancer Res. 2007;67: requisite to the constitutive expression and IFN-gamma-induced
11447-11454. upregulation of B7-H1 (CD274). FEBS Lett. 2006;580:755-762.
48. Baitsch L, Fuertes-Marraco SA, Legat A, et al. The three main stumbling 70. Philips GK, Atkins M. Therapeutic uses of anti-PD-1 and anti-PD-L1
blocks for anticancer T cells. Trends Immunol. 2012;33:364-372. antibodies. Int Immunol. 2015;27:39-46.
49. Kunert A, Straetemans T, Govers C, et al. TCR-engineered T cells meet 71. Jin D, Fan J, Wang L, et al. CD73 on tumor cells impairs antitumor T-cell
new challenges to treat solid tumors: choice of antigen, T cell fitness, responses: a novel mechanism of tumor-induced immune suppression.
and sensitization of tumor milieu. Front Immunol. 2013;4:363. Cancer Res. 2010;70:2245-2255.
50. Moon EK, Wang LC, Dolfi DV, et al. Multifactorial T-cell hypofunction 72. Stagg J, Smyth MJ. Extracellular adenosine triphosphate and adenosine
that is reversible can limit the efficacy of chimeric antigen receptor- in cancer. Oncogene 2010;29:5346-5358.
transduced human T cells in solid tumors. Clin Cancer Res. 2014;20: 73. Di Paola R, Melani A, Esposito E, et al. Adenosine A2A receptor-selective
4262-4273. stimulation reduces signaling pathways involved in the development of
51. Moon EK, Ranganathan R, Eruslanov E, et al. Blockade of programmed intestine ischemia and reperfusion injury. Shock. 2010;33:541-551.
death 1 augments the ability of human T cells engineered to target 74. Ohta A, Sitkovsky M. Role of G-protein-coupled adenosine receptors in
NY-ESO-1 to control tumor growth after adoptive transfer. Clin Cancer downregulation of inflammation and protection from tissue damage.
Res. 2016;22:436-447. Nature. 2001;414:916-920.
52. Rosenberg SA, Yang JC, Sherry RM, et al. Durable complete responses in 75. Zarek PE, Huang CT, Lutz ER, et al. A2A receptor signaling promotes
heavily pretreated patients with metastatic melanoma using T-cell peripheral tolerance by inducing T-cell anergy and the generation of
transfer immunotherapy. Clin Cancer Res. 2011;17:4550-4557. adaptive regulatory T cells. Blood. 2008;111:251-259.
53. Klebanoff CA, Khong HT, Antony PA, et al. Sinks, suppressors and an- 76. Ohta A, Gorelik E, Prasad SJ, et al. A2A adenosine receptor protects
tigen presenters: how lymphodepletion enhances T cell-mediated tumors from antitumor T cells. Proc Natl Acad Sci USA. 2006;103:
tumor immunotherapy. Trends Immunol. 2005;26:111-117. 13132-13137.
54. Wrzesinski C, Paulos CM, Kaiser A, et al. Increased intensity 77. Young A, Mittal D, Stagg J, et al. Targeting cancer-derived adenosine:
lymphodepletion enhances tumor treatment efficacy of adoptively new therapeutic approaches. Cancer Discov. 2014;4:879-888.
transferred tumor-specific T cells. J Immunother. 2010;33:1-7. 78. Kraman M, Bambrough PJ, Arnold JN, et al. Suppression of antitumor
55. Gattinoni L, Finkelstein SE, Klebanoff CA, et al. Removal of homeo- immunity by stromal cells expressing fibroblast activation protein-
static cytokine sinks by lymphodepletion enhances the efficacy of alpha. Science. 2010;330:827-830.
adoptively transferred tumor-specific CD8+ T cells. J Exp Med. 2005; 79. Gabrilovich DI, Ostrand-Rosenberg S, Bronte V. Coordinated regulation
202:907-912. of myeloid cells by tumours. Nat Rev Immunol. 2012;12:253-268.
56. Pegram HJ, Lee JC, Hayman EG, et al. Tumor-targeted T cells modified to 80. Nagaraj S, Gupta K, Pisarev V, et al. Altered recognition of antigen is a
secrete IL-12 eradicate systemic tumors without need for prior con- mechanism of CD8+ T cell tolerance in cancer. Nat Med. 2007;13:
ditioning. Blood. 2012;119:4133-4141. 828-835.
57. Chmielewski M, Hombach AA, Abken H. Of CARs and TRUCKs: chimeric 81. Liby KT, Yore MM, Sporn MB. Triterpenoids and rexinoids as multi-
antigen receptor (CAR) T cells engineered with an inducible cytokine to functional agents for the prevention and treatment of cancer. Nat Rev
modulate the tumor stroma. Immunol Rev. 2014;257:83-90. Cancer. 2007;7:357-369.

e458 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


LUNG CANCER

Local Therapies in the


Management of Oligometastatic
and Metastatic NonSmall Cell
Lung Cancer

CHAIR
Puneeth Iyengar, MD, PhD
The University of Texas Southwestern Medical Center
Dallas, TX

SPEAKERS
Jessica S. Donington, MD
New York University School of Medicine
New York, NY

Robert D. Suh, MD
David Geffen School of Medicine at UCLA
Los Angeles, CA
IYENGAR ET AL

Local Therapy for Limited Metastatic NonSmall Cell Lung


Cancer: What Are the Options and Is There a Benefit?
Puneeth Iyengar, MD, PhD, Steven Lau, MD, PhD, Jessica S. Donington, MD, MSCR, and
Robert D. Suh, MD

OVERVIEW

Distant metastasis is common in nonsmall cell lung cancer (NSCLC) and typically associated with poor prognosis. Aggressive
local therapy including surgery and/or radiation for limited metastatic disease from colorectal cancer and sarcoma is
associated with survival benefit and has become part of the standard of care. In this article, we review the literature and
ongoing studies concerning surgery, radiation, and radiofrequency ablation for oligometastatic NSCLC.

N onsmall cell lung cancer continues to represent dis-


proportionately the number of patients with and the
number of patient deaths from cancer. Over 224,000 new
and cure for both medically inoperable but even medical
operable lung cancer patients.
Unfortunately, most patients with NSCLC are still di-
cases are expected in 2016,1 and the overall survival (OS) for agnosed with advanced, metastatic disease. With the de-
all these patients is anticipated to approach only 15%20%. velopment of targeted therapies and immunotherapies,
Hence, a need to offer better strategies to combat the survival is improving. Yet, for a significant proportion of
disease should be a priority. For early-stage disease, surgery patients with stage IV NSCLC, systemic agents offer limited
continues to represent the gold standard for treatment. In potential for durable control of disease. Historically, with
medically inoperable patients, stereotactic ablative radio- limited metastatic colorectal cancer and sarcomas, the use
therapy (SABR), also referred to as stereotactic body radi- of local therapies including surgery and/or radiation have
ation therapy (SBRT), has become a more integral part of our become part of the standard of care as a means of drastically
treatment armamentarium. For those patients at high risk improving OS along with local control. It therefore begs the
for SABR in this medically inoperable setting, potentially question, as we try to identify more and more subsets of
because of previous irradiation, radiofrequency ablation patients with advanced NSCLC with favorable biology, if
(RFA) has begun to take on a greater role. Despite relentless there is a group of patients with limited metastatic NSCLC
advances in surveillance, imaging, and knowledge, the who could benefit from local treatments in addition to their
number of patients at initial presentation with surgically systemic therapy regimens. If so, what would be the optimal
resectable local disease remains soberly small at under 30%, treatments, when should they be delivered, and what type
compounded by the realization that many with resectable of benefit is realistic? Finally, it may be that local therapy in
disease are rendered medically inoperable by comorbid limited metastatic NSCLC is not beneficial independent of
disease. the favorable biology of this cohort. This information would
Over recent decades, most importantly the last, evolu- also be of vital importance to generate.
tionary inertia has refined local control measures on all This article offers the current evidence for the use of local
fronts, surgical and nonsurgical, through improved tech- therapiessurgery, radiation in the form of SABR or
nology and its application and patient selection. Although hypofractionated regimens, and RFAas part of the overall
standard surgical lobectomy has been and is currently the management for patients with limited metastatic NSCLC. For
gold standard for early-stage NSCLC,2 noninvasive and SABR and RFA, there is additional information regarding
minimally invasive nonsurgical measures for local control their general acceptance into more mainstream treatment,
SABR and image-guided tumor ablation (IGTA), respectively, starting with evidence of their role in early-stage disease.
have been increasingly accepted and used for local control As a note, unless considering the yet to be validated abscopal

From the Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX; NYU School of Medicine, New York, NY; Department of Radiological
Sciences, Thoracic Imaging and Intervention, and Diagnostic Radiology Education, David Geffen School of Medicine at UCLA, Los Angeles, CA; Department of Radiation Oncology, Harold
C. Simmons Comprehensive Cancer Center, The University of Texas Southwestern Medical Center, Dallas, TX.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Puneeth Iyengar, MD, PhD, UT Southwestern Medical Center, 5801 Forest Park Rd., MC 9183, Dallas, TX 75235-9183; email: puneeth.iyengar@utsouthwestern.
edu.

2016 by American Society of Clinical Oncology.

e460 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


LOCAL THERAPY FOR LIMITED METASTATIC NSCLC

response associated with immunotherapy, there is no ac- good performance status should be considered for curative
cepted role for the use of local therapies in the treatment of intent resection. Similarly, patients who present de novo with a
widely metastatic NSCLC unless deemed necessary for single metastasis after full staging should be evaluated for
palliation or other improvement in quality of life. curative resection of both sites. An ongoing question in those
who present with synchronous disease is which treatment
should be undertaken first, the systemic or localized therapies?
SURGERY FOR METASTATIC NSCLC
Historically, surgery for patients with stage IV cancer was
limited to palliation, but in the 1980s, series began to appear Isolated Brain Metastasis
that reported prolonged survival following complete re- Some of the oldest series on surgical treatment of stage IV
section of primary tumors and oligometastatic disease in NSCLC relate to treatment of isolated brain metastasis.4,5 Up
selected patients. It is now recognized that there is a subset to one-quarter of all patients with stage IV NSCLC harbor
of patients in whom an isolated metastasis represents the brain metastasis. Adenocarcinomas are associated with
entire disease burden, and removal can confer considerable higher rates of brain metastasis,6-8 but in 10% of patients
survival prolongation. with metastatic adenocarcinoma, the brain is the only site of
Surgery for metastases from numerous primary locations, involvement.9 Aggressive curative intent treatment of the
including the lung, has increased over the past decade. primary and metastatic site is encouraged in those with good
Metastasectomy for primary NSCLC has been found second performance status and in whom both sites are amenable to
in incidence only to colon cancers.3 An analysis from the complete resection or ablation. Curative intent local treat-
National Inpatient Sample uncovered a 5.8% average annual ments can only be considered after a thorough search for
percent increase in resections of NSCLC metastases between disease at other sites. Mediastinal lymph node involvement
2000 and 2011.3 This increase is attributed to several factors, portends poor prognosis,6-8,10,11 and therefore, invasive me-
including more efficacious and better tolerated chemo- diastinal staging is recommended prior to starting treatment.
therapies and the introduction of targeted agents, which Brain MRI is recommended in addition to PET/CT because
have slowed the progression of metastatic spread and al- of increased sensitivity12 Multiple brain metastasis are not
tered patterns of resistance. Simultaneously, there have an absolute contraindication to this aggressive treatment
been improvements in surgical techniques, with increased approach, but most recommend three or fewer lesions.9
use of minimally invasive approaches, making resection Treatment of the brain lesion can be by resection or radio-
better tolerated and negating long interruption from sys- surgery ablation. Radiosurgery has the advantage of being
temic treatments. able to be performed in almost any location, including the
The majority of patients considered for resection of brain stem.13-15
metastatic NSCLC fall into three categories: those with an Five-year survival following definitive treatment of iso-
isolated metastasis to the brain, adrenal glands, or con- lated brain metastasis and primary NSCLC is 15% and not
tralateral lung. Occasional patients with an isolated me- significantly impacted by synchronous or metachronous
tastasis to other sites are considered, but evidence for presentation. Results from several series with more than 20
prolonged survival following local therapy is sparse. Patients patients are outlined in Table 1.4-8,10,16,17 Prognosis is im-
who have previously undergone complete resection of early- proved in patients who are younger, female, have a lower t-
stage NSCLC and subsequently present with isolated oli- stage, and good performance status.6-8 Administration of
gometastasis after thorough metastatic survey and have adjuvant whole-brain radiation therapy (WBRT) following
resection or radiosurgery is recommended. Randomized data

KEY POINTS Table 1. Survival Outcomes for Patients With Isolated


Brain Metastasis Following Local Therapy of Primary
Many patients with NSCLC will be diagnosed with distant Tumor and Metastasis
metastasis, but a subset of patients with limited
metastatic disease may benefit from aggressive local Synchronous Metachronous
therapies. Survival (%) Survival (%)
Evidence supporting an aggressive approach to Author No. 2 years 5 years No. 2 years 5 years
oligometastatic NSCLC is limited but suggestive of 7
Bonnette et al 103 28 11 - - -
reasonable safety and efficacy.
The potential risks and benefits of aggressive local Girard et al8 51 42 N/R - - -
therapies for an individual patient should be openly Granone et al6 30 47 14 - - -
discussed prior to initiating treatment. Wronski
et al 4
86 14 8 145 29 17
Synchronous versus metachronous oligometastasis may
Nakagawa et al5 60 10 N/R 28 11 N/R
carry disparate prognosis to guide management.
Ongoing studies will clarify the roles of various local Flannery et al17 42 34 21 33 59 13
10
therapies in oligometastatic NSCLC. Furak et al - - - 45 N/R 16
Abbreviation: N/R, not recorded.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e461


IYENGAR ET AL

on WBRT in this setting are limited, but the sole trial There are no prospective studies comparing surgical re-
demonstrated a notable decrease in brain recurrence.18 section to radiation therapy for the treatment of oligome-
There are also no randomized data specifically addressing tastatic disease. Indeed, nonoperative approaches may be
the use of adjuvant chemotherapy following complete preferred for some patients with oligometastasis because of
resection of stage IV disease, but with strong evidence sup- risks of surgical morbidity and mortality as well as comorbid
porting adjuvant chemotherapy for completely resected stage conditions common in patients with NSCLC, which may increase
II and III,19 it is recommended for patients with completely such surgical risks. SABR adapts the techniques of stereotactic
resected primary and brain metastasis.9 radiosurgery for intracranial disease to the delivery of highly
conformal radiation to extracranial targets, and SABR is in-
creasingly used to treat oligometastasis.30
Isolated Adrenal Metastasis
In well-selected patients with isolated adrenal metastasis
from NSCLC, survival following complete resection is 25%.20-23 SABR Is Associated With Excellent Local Control and
Similar to those with isolated brain metastasis, mediastinal Acceptable Toxicity
lymph node involvement portends a poor prognosis, so in- Phase I/II studies have described the use of SABR for oli-
vasive staging is recommended.24 Histology and laterality gometastatic disease in the lung, liver, spine, and multiple
appear to have no impact on survival, and adjuvant chemo- sites and reported local control rates of 70%90% with grade
therapy is recommended. Synchronous and metachronous 3 or greater toxicity rates of less than 10%.31-36 For example,
tumors have similar long-term survival rates, despite younger Rusthoven et al31 enrolled 38 patients with 63 lung me-
age distribution in patients with synchronous presentation.25 tastases on a phase I/II protocol using SABR to 4860 Gy in
Operative mortality is extremely low in reported series, and the three fractions. Twenty-seven (71%) patients had been
majority of patients die of disease progression. previously treated with at least one systemic regimen. At a
median follow-up of 15.4 months, only one local failure was
observed. Importantly, only three (8%) patients experienced at
M1a Disease least 3 toxicity. Separately, Salama et al35 enrolled 61 patients
The appearance of bilateral NSCLC lesions with the same with 113 extracranial metastases on a dose-escalation protocol
histology is a staging challenge. In the absence of other using SABR to 2460 Gy in three fractions. Twenty-three (38%)
disease, it is difficult to distinguish bilateral primary tumors patients had received metastasis-directed therapy prior to
from stage IVa disease. Analysis of mutational status and enrollment. The maximum tolerated dose was not reached.
genetic clonality difference are being investigated, but is not At a median imaging follow-up of 15.0 months, 72 (64%)
clinically reliable at this time.26 The clinical judgment of an lesions were controlled despite initially low radiation doses
experienced multimodality team is essential,9 and the cri- on this protocol. At least grade 3 acute toxicity was observed in
teria described by Martini and Melamed in 1975 remains only two (3%) patients, and at least 3 late toxicity was observed
relevant.27 As with isolated brain and adrenal metastasis, an in six (10%) patients. Furthermore, when patients with oligo-
exhaustive search for additional metastatic disease and metastatic disease treated with ablative local therapies ex-
invasive mediastinal staging are recommended prior to perience distant progression, these failures tend to be limited
considering resection. Parenchymal sparing resections are rather than widespread and more amenable to further ablative
typically recommended when possible in this setting. therapies.37

Summary Patient Selection in Oligometastatic NSCLC


Overall, with more data being generated regarding local Outcomes of NSCLC are worse than other cancers, and
treatment of oligometastatic NSCLC, the role of surgery whether an oligometastatic disease state exists in NSCLC is
will be more properly delineated with the goal of im- debated. Ashworth et al38 performed a systematic review of
proving progression-free survival (PFS) at the least and 49 studies including 2,176 patients with one to five me-
ultimately OS. tastases from NSCLC who underwent surgery or radiation.
Most (82%) patients had controlled primary disease, and
60% of studies were limited to intracranial metastasis.
RADIATION FOR METASTATIC NSCLC Median survival was 14.8 months, median time to pro-
Patients with metastatic cancer are historically considered gression was 12 months, and median 5-year OS was 23.3%.
incurable, and their treatment is palliative in nature. However, Control of primary disease, N stage, and disease-free interval
reports of patients long-term survival after surgical resection of at least 6 to 12 months prior to diagnosis of oligome-
of metastasis began to surface in the 1930s.28 The state of tastasis were found to be prognostic on multivariable
limited metastatic disease without widespread progression analysis.
was ultimately termed oligometastasis in 1995.29 Oli- Ashworth et al39 subsequently performed an individual
gometastasis is now recognized as a unique clinical entity patient data meta-analysis on 757 patients with 1 to 5
in which aggressive, ablative therapies can result in long- synchronous or metachronous metastases from NSCLC
term cure. who underwent surgery or radiation. Median survival was

e462 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


LOCAL THERAPY FOR LIMITED METASTATIC NSCLC

26 months, median PFS was 11 months, and 5-year OS was after local or even systemic therapy or with synchronous
29.4%. Recursive partitioning analysis stratified patients or metachronous lung cancer, those with limited or low
into low-risk (metachronous metastasis; 5-year survival volume metastases particularly after a prolonged disease-
47.8%), intermediate-risk (synchronous metastasis, N0; free interval (oligometastatic), and those in need of palliative
5-year survival 36.2%), and high-risk disease (synchro- and now even salvage therapy. Although multiple methods
nous metastasis, N+; 5-year survival 12.1%). for safe and effective local therapy are necessary, IGTA for
De Ruysscher et al prospectively evaluated aggressive NSCLC is particularly poised for success, given it provides
treatment with curative intent in 39 patients diagnosed with the best balance of comparable survival, superior cost,
NSCLC and one to three synchronous metastases.40 Primary and experiential use patients with high-risk disease and
disease was treated with radiation, and metastatic disease other emerging patient populations, much of which was
was treated with either surgery (24%) or radiation (76%). unfathomable a decade prior.
Twenty-seven (69.2%) patients had thoracic nodal in-
volvement, 34 (87.2%) patients had a single metastasis, and
17 (43.9%) patients had brain metastasis. Median survival IGTA Efficacy
was 13.5 months, median PFS was 12.1 months, and 2-year In 2007, Simon et al44 first published 5-year long-term safety
survival was 23.3%. No grade 3 toxicity or higher related to and efficacy data of RFA in 71 patients with stage I primary
treatment of oligometastatic disease was observed. lung cancer. Overall survival was reported at 78% at 1 year,
Thus, appropriate patient selection is warranted when plan- 57% at 2 years, 36% at 3 years, and 27% at 4 and 5 years. The
ning for an aggressive approach using ablative local therapies. authors re-emphasized that patients with treated cancers
3 cm and smaller versus cancers larger than 3 cm demon-
Combined Therapy for Oligometastatic NSCLC strated better OS at every annual time point: 83% versus
Although ablative local therapies may control known sites 45% at 1 year, 64% versus 25% at 2 years, 57% versus 25% at
of disease, most patients will ultimately progress. Novel 3 years, and 47% versus 25% at 4 and 5 years. In addition,
therapeutic approaches are therefore needed. One prom- those patients with 3 cm and smaller tumors showed sig-
ising approach is the combination of SABR and systemic nificantly longer median time to progression: 45 versus
therapies. Iyengar et al41 prospectively tested the use of 12 months. Although 27% 5-year OS appears dismal at first
SABR and concurrent erlotinib in 24 patients with 52 ex- glance, the study was quite remarkable in achieving this
tracranial metastases from NSCLC who had progressed after survival in the high-risk population, estimated up to 30%
at least one systemic regimen. SABR was delivered to all according to the Surveillance, Epidemiology, and End Results
active sites of disease. Median survival and PFS were 20.4 (SEER) database in 2005,45 most therapeutically neglected
and 14.7 months, respectively. Only three local failures of 47 because of their medical ineligibility before this time. Since
evaluable lesions were observed, and 10 patients progressed 2007, at least three additional series in patients with high-
at distant sites. Others have reported an abscopal effect on risk disease have shown comparable or better 5-year OS:
unirradiated metastases from the combination of SABR and 24%, 25%, and 61%,46-48 the last being quite formidable
immunotherapies.42,43 and a testament to long-term operator experience and the
benefits of lessons learned regarding patient and lesion
selection. In addition to single-center experience, the medical
Summary literature includes the results of two multicenter trials. In
SABR for the treatment of oligometastatic NSCLC is effective Lancet Oncology, the RAPTURE study demonstrated 48%
and well tolerated, and combined modality therapy is a OS and 73% cancer-specific survival at 2 years for primary
promising approach to improve outcomes. Two studies, one out lung cancer population of patients with high-risk disease.49
of MD Anderson Cancer Center (NCT01725165) and the second Moreover, pulmonary function tests did not show any signif-
out of UT Southwestern Medical Center (NCT02045446), are icant decline in forced expiratory volume (FEV), FEV percentage
using SABR and hypofractionated radiation in randomized predicted, forced vital capacity (FVC), or FVC percentage
studies with systemic therapy to determine if local treatment predicted, in any follow-up visits through 12 months,
can improve PFS in patients with up to three and six sites of compared with baseline values. The slight decrease in these
limited metastatic disease, respectively. Other larger studies values in treated patients with lung cancer was not clini-
with OS as an endpoint will hopefully best answer how ben- cally significant and was consistent with the progression of
eficial radiation may be in the stage IV NSCLC setting. underlying chronic pulmonary disease in this subgroup.
More recently, the results from the American College of
RADIOFREQUENCY ABLATION FOR METASTATIC Surgeons Oncology Group (ACOSOG) Z4033 Trial were
NSCLC published.50 The OS rate was 86.3% at 1 year and 69.8% at
For those with NSCLC undergoing RFA, to date the most 2 years. The local tumor recurrencefree rate was 68.9% at
studied thermal ablation technique, the medical literature 1 year and 59.8% at 2 years and worse for tumors at least 2 cm.
has focused on the high-risk, or medically inoperable, Not surprisingly, patients with tumors smaller than 2 cm and a
population. RFA, or IGTA, can be considered for those performance status of 0 or 1 achieved a statistically significant
patients with locally recurrent cancer (oligorecurrent) improved survival of 83 and 78%, respectively, at 2 years.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e463


IYENGAR ET AL

Although not as mature as a technological application for however, despite enhanced local control, the rate of distant
lung malignancies compared with RFA, microwave ablation disease remains highly variable between studies whether
(MWA) and cryoablation, or percutaneous cryotherapy comparing like or unlike therapies. Simply and most plau-
(PCT), have more recently published 5-year results in the sibly, this variability is the direct result of already micro-
treatment of early lung cancer in the medically inoperable scopic disease at the time of treatment, in some surgical
patient population, both having demonstrated comparable, series reported as high as 16% in 1-cm apical cancers.59 In
if not improved, OS and cancer-specific survival and local other words, even complete, or 100%, local control will have
control rates to RFA at 5 years with favorable complication already microscopic locoregional and distant disease. This is
profiles.51,52 Yang et al51 reported OS rates at 1, 2, 3, and 5 further exacerbated in the high-risk population, which may
years after MWA were 89%, 63%, 43%, and 16%, respectively. be ineligible for formal nodal staging through mediastino-
Tumors that were 3.5 cm or smaller were associated with scopy, left to complete reliance on medical imaging, spe-
better survival than were tumors that were larger than 3.5 cm. cifically CT-PET.
The local control rates at 1, 3, and 5 years after MWA were Ultimately, the rate of local and locoregional progression
96%, 64%, and 48%, respectively. In patients undergoing PCT may not negatively impact OS. Specifically in 2009, Lanuti
for primary lung cancer, Moore et al52 in 2015 reported the 5- et al60 looked at locoregional recurrence in patients with
year OS rate at 67.8%, the cancer-specific survival rate at 5 stage I lung cancer initially treated with RFA, and despite
years at 56.6%, and the 5-year PFS rate at 87.9%. retreatment with RFA, radiotherapy, chemoradiotherapy,
and chemotherapy, the authors found no notable difference
in 5-year overall and disease-free survival in patients re-
Argued Comparability
ceiving RFA without recurrence versus those patients with
Over the years, RFA has been unfavorably compared with
treated recurrences.
sublobar resection and SABR. However, upon closer scrutiny,
one major overlooked factor is that those patients treated
with RFA were always the sickest, the patients receiving RFA Other Patient Populations
in most, if not all, studies demonstrating statistically significant Helping to define a new patient population afforded lung
poorer FEV in 1 second53 and older age54 compared with those ablation, Kodama et al61 in 2012 showed 55.7% 5-year OS in
undergoing sublobar resection. Moreover, when comparing those patients with recurrent and second primary lung
patient demographics from the ACOSOG z4033 and z4032 and cancers after surgical resection. Ridge et al62 in 2014 found
RTOG 0236 trials, those patients enrolled in the z4033 were improved local progression after thermal ablation with
significantly older and held significantly lower diffusion synchronous and metachronous lung cancers compared
capacity.55 And finally, Kwan et al56 recently argued com- with ablation of first primaries.
parable overall and lung-cancer specific survival between RFA has been studied in the advanced disease patient
RFA and sublobar resection in patients in the SEER database population. Yu et al63 found RFA a useful tool to bridge from
when propensity scores were used to match patient subgroups. failure of one therapy to the initiation of another therapy in
In many circles, stereotactic body radiotherapy has been patients receiving EGFR. In some patients with advanced
largely hailed as the silver bullet against early-stage lung disease, Gu et al64 found that patients receiving cryoablation
cancer, setting new lofty standards for 5-year OS and local and gefitinib demonstrated significantly better survival at 1
recurrence ranging between 54% and 65% and 4% and 14%, year than those patients receiving gefitinib alone. Similarly,
respectively.57,58 Although certainly impressive, the results Lee et al54 observed that even stage III and IV lung cancers
emerging from these and other similar series have not been treated with RFA and chemotherapy demonstrated signifi-
devoid of criticism. Deeper analyses show that not all treated cantly longer median OS than those treated with only
lesions were pathologically confirmed, with many radiograph- chemotherapy. In patients with localized recurrence after
ically diagnosed, prevalence of more indolent tumors specifically initial local or systemic treatment failure, RFA, when added
adenocarcinoma in situ, and the treated population in- to a predetermined treatment regimen, lengthened overall
cluded both patients with low-risk disease and those with and PFS.65,66
high-risk disease. In fact, when parsing out the high-risk
subgroup from the Onishi et al study in 2007,57 5-year OS
Cost Benefit
drops from the overall reported 65% to 35%, similar to
Although surgical resection showed OS superiority at 1, 2,
published 5-year RFA results. Unlike RFA, moreover, SABRs
and 3 years, Alexander et al67 argued that this superiority
higher potential for collateral injury to healthy peritumoral
came at a significantly increased median cost per months
lung may be detrimental in many patients with high-risk
lived of 1.93 times from reimbursement rates from the study
disease with tenuous lung reserve.
state. When compared with other treatment modalities in
the setting of oligometastatic NSCLC, cryoablation appeared
Will Less Equivalent Local Control Be the Death of the most cost-effective, even when added to the cost of best
Radiofrequency Ablation? supportive care or systemic regimens with an adjunctive
In ascending order, local control improves when comparing cost-effectiveness ratio of $49,008$87,074. In their study,
RFA, MWA, cryoablation, SABR, and surgical lobectomy; cryoablation was associated with very low morbidity and

e464 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


LOCAL THERAPY FOR LIMITED METASTATIC NSCLC

local tumor recurrence rates for all anatomic sites and CONCLUSION
possibly increased OS.68 Most would agree that favorable biology is the primary driver of
Although prospective cost-efficacy studies comparing prognosis in the setting of oligometastasis, and the true impact
IGTA and SBRT are nonexistent, with the exception of a of local interventions on prognosis is unclear. But in an era when
single study using Markov modeling and older RFA data,69 local treatments carry minimal morbidity and mortality, the lack
global Medicare reimbursement in Rhode Island for SABR of clarity should not translate into a denial of intervention in
was 4.25 times that of RFA at $17,000 versus $4,000.70 If well-selected patients with oligometastatic disease.
the higher cost of SABR for better local control and arguably Evidence in favor of aggressive local therapies in oligo-
better OS is justified remains to be seen in the high-risk metastatic NSCLC is limited to retrospective series and
population, particularly given the microscopic locoregional several single-arm prospective series reporting reasonable
and distant disease rates and RFAs repeatability at lower safety and efficacy. In the absence of more robust data and
cost. comparative effectiveness studies, clinical judgment and
provider experience must guide management of patients
Summary with limited metastatic NSCLC. Optimal management of
In summary, advances in thermal energy devices and de- oligometastatic NSCLC is likely to be influenced by patient
livery have led to improved control. As local control im- selection. Patients with a single metastasis and good per-
proves, however, gains in survival will be limited for NSCLC, formance status might best be served by surgical resection. In
given the limitations of radiographic staging and microscopic contrast, patients with limited multiple metastases and those
lymph nodal and distant disease at the time of therapy with inoperable disease may be treated with SABR. Similarly,
including ablation, rendering the need for adjuvant control patients with accessible, small, and inoperable lesions may be
in some situations. Additive local control even in those with candidates for RFA. Multidisciplinary discussion may be helpful
advanced cancer appears to offer survival benefits in some for guiding management on a case-by-case basis. Ideally, pa-
patient subsets with recurrent or advanced disease. When tients would be enrolled and treated on clinical trials including
critically compared with surgical resection and SABR, IGTA is ongoing protocols using hypofractionated radiation and sys-
an attractive option with an acceptable threshold for local temic therapy. Ultimately, rigorous studies in well-selected
control balanced with risk and cost without detriment to patients will be necessary to determine the optimal man-
survival. agement of oligometastatic NSCLC.

References
1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin. 10. Furak J, Trojan I, Szoke T, et al. Lung cancer and its operable brain
2016;66:7-30. metastasis: survival rate and staging problems. Ann Thorac Surg. 2005;
2. Churchill ED, Sweet RH, Soutter L, et al. The surgical management of 79:241-247; discussion 247.
carcinoma of the lung; a study of the cases treated at the Massa- 11. Modi A, Vohra HA, Weeden DF. Does surgery for primary non-small cell
chusetts General Hospital from 1930 to 1950. J Thorac Surg. 1950;20: lung cancer and cerebral metastasis have any impact on survival? In-
349-365. teract Cardiovasc Thorac Surg. 2009;8:467-473.
3. Bartlett EK, Simmons KD, Wachtel H, et al. The rise in metastasectomy 12. Billing PS, Miller DL, Allen MS, et al. Surgical treatment of primary lung
across cancer types over the past decade. Cancer. 2015;121:747-757. cancer with synchronous brain metastases. J Thorac Cardiovasc Surg.
4. Wronski
M, Arbit E, Burt M, et al. Survival after surgical treatment of 2001;122:548-553.
brain metastases from lung cancer: a follow-up study of 231 patients 13. Flannery TW, Suntharalingam M, Regine WF, et al. Long-term survival in
treated between 1976 and 1991. J Neurosurg. 1995;83:605-616. patients with synchronous, solitary brain metastasis from non-small-cell lung
5. Nakagawa H, Miyawaki Y, Fujita T, et al. Surgical treatment of brain cancer treated with radiosurgery. Int J Radiat Oncol Biol Phys. 2008;72:19-23.
metastases of lung cancer: retrospective analysis of 89 cases. J Neurol 14. Fuentes R, Bonfill X, Exposito J. Surgery versus radiosurgery for patients
Neurosurg Psychiatry. 1994;57:950-956. with a solitary brain metastasis from non-small cell lung cancer.
6. Granone P, Margaritora S, DAndrilli A, et al. Non-small cell lung cancer Cochrane Database Syst Rev. 2006;1:CD004840.
with single brain metastasis: the role of surgical treatment. Eur J 15. Mariya Y, Sekizawa G, Matsuoka Y, et al. Outcome of stereotactic
Cardiothorac Surg. 2001;20:361-366. radiosurgery for patients with non-small cell lung cancer metastatic to
7. Bonnette P, Puyo P, Gabriel C, et al; Groupe Thorax. Surgical man- the brain. J Radiat Res (Tokyo). 2010;51:333-342.
agement of non-small cell lung cancer with synchronous brain me- 16. Ambrogi MC, Lucchi M, Dini P, et al. Percutaneous radiofrequency
tastases. Chest. 2001;119:1469-1475. ablation of lung tumours: results in the mid-term. Eur J Cardiothorac
8. Girard N, Cottin V, Tronc F, et al. Chemotherapy is the cornerstone of Surg. 2006;30:177-183.
the combined surgical treatment of lung cancer with synchronous brain 17. Flannery TW, Suntharalingam M, Kwok Y, et al. Gamma knife stereotactic
metastases. Lung Cancer. 2006;53:51-58. radiosurgery for synchronous versus metachronous solitary brain metas-
9. Kozower BD, Larner JM, Detterbeck FC, et al. Special treatment issues in tases from non-small cell lung cancer. Lung Cancer. 2003;42:327-333.
non-small cell lung cancer: Diagnosis and management of lung cancer, 18. Patchell RA, Tibbs PA, Regine WF, et al. Postoperative radiotherapy in
3rd ed: American College of Chest Physicians evidence-based clinical the treatment of single metastases to the brain: a randomized trial.
practice guidelines. Chest 2013;143:e369S-e399S. JAMA. 1998;280:1485-1489.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e465


IYENGAR ET AL

19. Pignon JP, Tribodet H, Scagliotti GV, et al; LACE Collaborative Group. oligometastatic non-small-cell lung cancer. Clin Lung Cancer. 2014;15:
Lung adjuvant cisplatin evaluation: a pooled analysis by the LACE 346-355.
Collaborative Group. J Clin Oncol. 2008;26:3552-3559. 40. De Ruysscher D, Wanders R, van Baardwijk A, et al. Radical treatment of
20. Raz DJ, Lanuti M, Gaissert HC, Wright CD, Mathisen DJ, Wain JC. non-small-cell lung cancer patients with synchronous oligometastases:
Outcomes of patients with isolated adrenal metastasis from non-small long-term results of a prospective phase II trial. J Thorac Oncol. 2012;7:
cell lung carcinoma. Ann Thorac Surg. 2011;92:1788-1792; discussion 1547-1555.
1793. 41. Iyengar P, Kavanagh BD, Wardak Z, et al. Phase II trial of stereotactic
21. Lucchi M, Dini P, Ambrogi MC, et al. Metachronous adrenal masses in body radiation therapy combined with erlotinib for patients with
resected non-small cell lung cancer patients: therapeutic implications of limited but progressive metastatic non-small-cell lung cancer. J Clin
laparoscopic adrenalectomy. Eur J Cardiothorac Surg. 2005;27:753-756. Oncol. 2014;32:3824-3830.
22. Strong VE, DAngelica M, Tang L, et al. Laparoscopic adrenalectomy for 42. Postow MA, Callahan MK, Barker CA, et al. Immunologic correlates of
isolated adrenal metastasis. Ann Surg Oncol. 2007;14:3392-3400. the abscopal effect in a patient with melanoma. N Engl J Med. 2012;366:
23. Mercier O, Fadel E, de Perrot M, et al. Surgical treatment of solitary 925-931.
adrenal metastasis from non-small cell lung cancer. J Thorac Cardiovasc 43. Golden EB, Demaria S, Schiff PB, et al. An abscopal response to radiation
Surg. 2005;130:136-140. and ipilimumab in a patient with metastatic non-small cell lung cancer.
24. Porte H, Siat J, Guibert B, et al. Resection of adrenal metastases from Cancer Immunol Res. 2013;1:365-372.
non-small cell lung cancer: a multicenter study. Ann Thorac Surg. 2001; 44. Simon CJ, Dupuy DE, DiPetrillo TA, et al. Pulmonary radiofrequency
71:981-985. ablation: long-term safety and efficacy in 153 patients. Radiology. 2007;
25. Tanvetyanon T, Robinson LA, Schell MJ, et al. Outcomes of adrenal- 243:268-275.
ectomy for isolated synchronous versus metachronous adrenal me- 45. Mery CM, Pappas AN, Bueno R, et al. Similar long-term survival of
tastases in non-small-cell lung cancer: a systematic review and pooled elderly patients with non-small cell lung cancer treated with lobectomy
analysis. J Clin Oncol. 2008;26:1142-1147. or wedge resection within the surveillance, epidemiology, and end
26. Wu CT, Lin MW, Hsieh MS, et al. New aspects of the clinicopathology results database. Chest. 2005;128:237-245.
and genetic profile of metachronous multiple lung cancers. Ann Surg. 46. Huang L, Han Y, Zhao J, et al. Is radiofrequency thermal ablation a safe
2014;259:1018-1024. and effective procedure in the treatment of pulmonary malignancies?
27. Martini N, Melamed MR. Multiple primary lung cancers. J Thorac Eur J Cardiothoracic Surg. 2011;39:348-351.
Cardiovasc Surg. 1975;70:606-612. 47. Ambrogi MC, Fanucchi O, Cioni R, et al. Long-term results of radio-
28. Barney J, Churchill E. Adenocarcinoma of the kidney with metastasis to frequency ablation treatment of stage I non-small cell lung cancer:
the lung cured by nephrectomy and lobectomy. J Urol. 1939;42: a prospective intention-to-treat study. J Thorac Oncol. 2011;6:
269-276. 2044-2051.
29. Hellman S, Weichselbaum RR. Oligometastases. J Clin Oncol. 1995;13: 48. Hiraki T, Gobara H, Mimura H, et al. Percutaneous radiofrequency
8-10. ablation of clinical stage I non-small cell lung cancer. J Thorac Cardiovasc
30. Pan H, Simpson DR, Mell LK, et al. A survey of stereotactic body ra- Surg. 2011;142:24-30.
diotherapy use in the United States. Cancer. 2011;117:4566-4572. 49. Lencioni R, Crocetti L, Cioni R, et al. Response to radiofrequency ablation
31. Rusthoven KE, Kavanagh BD, Burri SH, et al. Multi-institutional phase I/II of pulmonary tumours: a prospective, intention-to-treat, multicentre
trial of stereotactic body radiation therapy for lung metastases. J Clin clinical trial (the RAPTURE study). Lancet Oncol. 2008;9:621-628.
Oncol. 2009;27:1579-1584. 50. Dupuy DE, Fernando HC, Hillman S, et al. Radiofrequency ablation of
32. Rusthoven KE, Kavanagh BD, Cardenes H, et al. Multi-institutional phase stage IA non-small cell lung cancer in medically inoperable patients:
I/II trial of stereotactic body radiation therapy for liver metastases. J Clin Results from the American College of Surgeons Oncology Group Z4033
Oncol. 2009;27:1572-1578. (Alliance) trial. Cancer. 2015;121:3491-3498.
33. Lee MT, Kim JJ, Dinniwell R, et al. Phase I study of individualized ste- 51. Yang X, Ye X, Zheng A, et al. Percutaneous microwave ablation of stage I
reotactic body radiotherapy of liver metastases. J Clin Oncol. 2009;27: medically inoperable non-small cell lung cancer: clinical evaluation of 47
1585-1591. cases. J Surg Oncol. 2014;110:758-763.
34. Wang XS, Rhines LD, Shiu AS, et al. Stereotactic body radiation therapy 52. Moore W, Talati R, Bhattacharji P, et al. Five-year survival after cry-
for management of spinal metastases in patients without spinal cord oablation of stage I non-small cell lung cancer in medically inoperable
compression: a phase 1-2 trial. Lancet Oncol. 2012;13:395-402. patients. J Vasc Interv Radiol. 2015;26:312-319.
35. Salama JK, Hasselle MD, Chmura SJ, et al. Stereotactic body radio- 53. Kim SR, Han HJ, Park SJ, et al. Comparison between surgery and
therapy for multisite extracranial oligometastases: final report of a dose radiofrequency ablation for stage I non-small cell lung cancer. Eur J
escalation trial in patients with 1 to 5 sites of metastatic disease. Cancer. Radiol. 2012;81:395-399.
2012;118:2962-2970. 54. Lee H, Jin GY, Han YM, et al. Comparison of survival rate in primary non-
36. Milano MT, Katz AW, Zhang H, et al. Oligometastases treated with small-cell lung cancer among elderly patients treated with radio-
stereotactic body radiotherapy: long-term follow-up of prospective frequency ablation, surgery, or chemotherapy. Cardiovasc Intervent
study. Int J Radiat Oncol Biol Phys. 2012;83:878-886. Radiol. 2012;35:343-350.
37. Milano MT, Philip A, Okunieff P. Analysis of patients with oligometa- 55. Crabtree T, Puri V, Timmerman R, et al. Treatment of stage I lung cancer
stases undergoing two or more curative-intent stereotactic radio- in high-risk and inoperable patients: comparison of prospective clinical
therapy courses. Int J Radiat Oncol Biol Phys. 2009;73:832-837. trials using stereotactic body radiotherapy (RTOG 0236), sublobar re-
38. Ashworth A, Rodrigues G, Boldt G, et al. Is there an oligometastatic state section (ACOSOG Z4032), and radiofrequency ablation (ACOSOG
in non-small cell lung cancer? A systematic review of the literature. Lung Z4033). J Thorac Cardiovasc Surg. 2013;145:692-699.
Cancer. 2013;82:197-203. 56. Kwan SW, Mortell KE, Talenfeld AD, Brunner MC. Thermal ablation
39. Ashworth AB, Senan S, Palma DA, et al. An individual patient data matches sublobar resection outcomes in older patients with early-stage
metaanalysis of outcomes and prognostic factors after treatment of non-small cell lung cancer. J Vasc Interv Radiol. 2014;25:1-9.e1.

e466 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


LOCAL THERAPY FOR LIMITED METASTATIC NSCLC

57. Onishi H, Shirato H, Nagata Y, et al. Hypofractionated stereotactic 64. Gu XY, Jiang Z, Fang W. Cryoablation combined with molecular target
radiotherapy (HypoFXSRT) for stage I non-small cell lung cancer: therapy improves the curative effect in patients with advanced non-
updated results of 257 patients in a Japanese multi-institutional study. small cell lung cancer. J Int Med Res. 2011;39:1736-1743.
J Thorac Oncol. 2007;2:S94-S100. 65. Schoellnast H, Deodhar A, Hsu M, et al. Recurrent non-small cell lung
58. Uematsu M, Fukui T, Tahara K, et al. Long-term results of computed cancer: evaluation of CT-guided radiofrequency ablation as salvage
tomography guided hypofractionated stereotactic radiotherapy for therapy. Acta Radiol. 2012;53:893-899.
stage I non-small cell lung cancers. Int J Radiat Oncol Biol Phys. 2008;72: 66. Li X, Zhao M, Wang J, et al. Percutaneous CT-guided radiofrequency
S37. ablation as supplemental therapy after systemic chemotherapy for
59. Ketchedjian A, Daly BD, Fernando HC, et al. Location as an important selected advanced non-small cell lung cancers. AJR Am J Roentgenol.
predictor of lymph node involvement for pulmonary adenocarcinoma. 2013;201:1362-1367.
J Thorac Cardiovasc Surg. 2006;132:544-548. 67. Alexander ES, Machan JT, Ng T, et al. Cost and effectiveness of radi-
60. Lanuti M, Sharma A, Digumarthy SR, et al. Radiofrequency ablation for ofrequency ablation versus limited surgical resection for stage I non-
treatment of medically inoperable stage I non-small cell lung cancer. small-cell lung cancer in elderly patients: is less more? J Vasc Interv
J Thorac Cardiovasc Surg. 2009;137:160-166. Radiol. 2013;24:476-482.
61. Kodama H, Yamakado K, Takaki H, et al. Lung radiofrequency ablation 68. Bang HJ, Littrup PJ, Currier BP, et al. Percutaneous cryoablation of
for the treatment of unresectable recurrent non-small-cell lung cancer metastatic lesions from non-small-cell lung carcinoma: initial survival,
after surgical intervention. Cardiovasc Intervent Radiol. 2012;35: local control, and cost observations. J Vasc Interv Radiol. 2012;23:
563-569. 761-769.
62. Ridge CA, Silk M, Petre EN, et al. Radiofrequency ablation of T1 lung 69. Sher DJ, Wee JO, Punglia RS. Cost-effectiveness analysis of stereotactic
carcinoma: comparison of outcomes for first primary, metachronous, body radiotherapy and radiofrequency ablation for medically in-
and synchronous lung tumors. J Vasc Interv Radiol. 2014;25:989-996. operable, early-stage non-small cell lung cancer. Int J Radiat Oncol Biol
63. Yu HA, Sima CS, Huang J, et al. Local therapy with continued EGFR Phys. 2011;81:e767-e774.
tyrosine kinase inhibitor therapy as a treatment strategy in EGFR- 70. Dupuy DE. Treatment of medically inoperable non-small-cell lung
mutant advanced lung cancers that have developed acquired re- cancer with stereotactic body radiation therapy versus image-guided
sistance to EGFR tyrosine kinase inhibitors. J Thorac Oncol. 2013;8: tumor ablation: can interventional radiology compete? J Vasc Interv
346-351. Radiol. 2013;24:1139-1145.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e467


LUNG CANCER

Lung Cancer Screening and


Prevention

CHAIR
Denise R. Aberle, MD
David Geffen School of Medicine at UCLA
Los Angeles, CA

SPEAKERS
Abbie Begnaud, MD
University of Minnesota
Minneapolis, MN

Graham W. Warren, MD, PhD


Medical University of South Carolina
Charleston, SC
BEGNAUD, HALL, AND ALLEN

Lung Cancer Screening With Low-Dose CT: Implementation


Amid Changing Public Policy at One Health Care System
Abbie Begnaud, MD, Thomas Hall, MBA, and Tadashi Allen, MD

OVERVIEW

Screening for lung cancer with low-dose CT has evolved rapidly in recent years since the National Lung Screening Trial (NLST)
results. Subsequent professional and governmental organization guidelines have shaped policy and reimbursement for the
service. Increasingly available guidance describes eligible patients and components necessary for a high-quality lung cancer
screening program; however, practical instruction and implementation experience is not widely reported. We launched a
lung cancer screening program in the face of reimbursement and guideline uncertainties at a large academic health center.
We report our experience with implementation, including challenges and proposed solutions. Initially, we saw less referrals
than expected for screening, and many patients referred for screening did not clearly meet eligibility guidelines. We
educated primary care providers and implemented system tools to encourage referral of eligible patients. Moreover, in
response to the Centers for Medicare & Medicaid Services (CMS) final coverage determination, we report our programmatic
adaptation to meet these requirements. In addition to the components common to all quality programs, individual health
delivery systems will face unique barriers related to patient population, available resources, and referral patterns.

S ince the publication of the landmark NLST in 2011, many


health care systems prepared to implement a lung cancer
screening program. The NLST was the first randomized
radiology, thoracic surgery, and pulmonology, to design the
program. Initial research included an assessment of available
clinical resources, a market analysis, and a cost analysis to
controlled trial to show a mortality benefit when screening start the program. According to our estimates, approxi-
high-risk populations for lung cancer with low-dose CT of the mately 94,634 people at high risk for lung cancer, based on
chest.1 At the time of NLST publication, the most updated smoking history, live in the metropolitan area. Only one
guideline from the U.S. Preventive Services Task Force other screening program was open in the area at the time of
(USPSTF) cited insufficient evidence to recommend for or our program planning and launch.
against screening for lung cancer.2 Similarly, the American A team of clinicians with lung nodule management ex-
College of Chest Physicians recommended screening only pertise is an integral component of a lung cancer screening
within the confines of a clinical trial.3 program.4 Our multidisciplinary thoracic oncology group
The University of Minnesota Health is an academic medical was established in 2011 and includes board-certified tho-
practice in the center of a large metropolitan area. Our health racic surgeons, radiation oncologists, thoracic and inter-
system affiliate, Fairview Health Systems, is one of several that ventional radiologists, interventional pulmonologists, and
provide care throughout the state. One advantage of this medical oncologistsall specializing in the care of patients
system is that any patient treated in the hospitals or clinics has a with lung cancer. An experienced, specialized team con-
universal electronic health record (EHR, Epic). However, dif- tributes to optimizing outcomes of lung cancer screening by
ferent physician practice groups provide services within the
avoiding unnecessary diagnostic procedures and achieving
system. Imaging services are contracted to both University of
the best possible outcomes for lung resection surgery.5 The
Minnesota physicians and a large community radiology practice.
multidisciplinary team reviews indeterminate lung nodules
in a weekly lung nodule conference with diagnostic thoracic
MATERIALS AND METHODS radiology to recommend a patient-specific plan for diagnosis
Determination of Need and Assessment of Resources and/or disease management. Minimally invasive diagnostic
In early 2013, we gathered a team, consisting of repre- testing is available at our institution, including percutaneous
sentatives from business development, thoracic oncology, lung biopsy, video-assisted thoracic surgery, and linear

From the Department of Medicine, University of Minnesota, Minneapolis, MN; Fairview Health System, University of Minnesota Cancer Care, Minneapolis, MN; Department of Radiology,
University of Minnesota, Minneapolis, MN.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Abbie Begnaud, MD, 420 East Delaware St., MMC 27, Minneapolis, MN 55455; email: abegnaud@umn.edu.

2016 by American Society of Clinical Oncology.

e468 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


LUNG CANCER SCREENING WITH LOW-DOSE CT

probe endobronchial ultrasound and guided bronchoscopy Despite the limited screening interval and upper age limit
techniques (such as electromagnetic navigational bron- of 75 reported in the NLST, we predicted benefit from
choscopy and radial endobronchial ultrasound). The team continued annual screening. Hence, we added lung cancer
uses advanced diagnostic technologies with individualized screening to the health maintenance activity where other
attention to lesion characteristics, patient preferences, and preventive health measures are tracked, including cancer
medical comorbidities. screenings and vaccinations. The American Association for
Thoracic Surgery also recommended annual screening for
people age 5579.7 Ultimately, the USPFTF and CMS re-
Program Planning leased recommendations for both a broader age range and
Establishing a workflow for ordering examinations, inter- screening extending beyond three annual examinations. We
preting images, relaying results, and ensuring follow-up educated primary care provider groups about screening
was a crucial phase of planning. The follow-up is driven eligibility and the process we developed.
by the guidelines by the National Comprehensive Cancer
Network, 6 American Association of Thoracic Surgery, 7
and American College of Radiology (ACR).8 Implementation Early Implementation
of the program required partnership with EHR builders, After creating a new chest CT lung cancer screening order in
billing and patient financial services, and communications the EHR, establishing a result workflow with radiology, and
specialists. educating providers, our program launched on December
Although self-referral for screening was initially consid- 12, 2013. Because most insurance companies were not yet
ered, ultimately we required an order from a credentialed covering lung cancer screening examinations, we offered the
provider for screening. Subsequently released CMS re- service at $150, which was a fair market price at the time.
quirements mandate an order and ordering provider, but Several insurance companies began covering screening
health systems might elect to offer screening to patients throughout 2014, well before the updated USPSTF guideline.
with private insurance or those who would pay for services At the time of screening, patients submit detailed in-
out of pocket. The anticipation of need for result follow-up formation about their risk factors for lung cancer via a
influenced our decision to require all patients to have an questionnaire (developed with collaboration from academic
ordering provider for lung cancer screening. epidemiologists and clinical psychologists, subsequently
A unique EHR order was created specifically for lung cancer adapted to match CMS registry data requirements). Imaging
screening. The name of the order and associated search key technologists enter the information into the EHR and share
words included CT, lung cancer screening, and low- with the interpreting radiologist. The questionnaire assesses
dose CT. Initially the order had few mandatory responses each patients risk profile including smoking history, radon
related to eligibility criteria: patients symptoms suggestive and particulate exposures, family history of lung cancer, and
of lung cancer, age, and smoking history. When this order is personal medical history.
signed, providers are also prompted to include in the pa- We implemented a structured reporting system for results
tients printed after-visit summary the Lung Cancer called U-Lung-RADS, which was modeled after Bi-RADS and
Screening, Frequently Asked Questions and Resources to was very similar to the system ultimately released by ACR in
Help You Quit Smoking. Subsequent CMS requirements April 2014 (Table 1).9 In addition to a numeric score based on
changed the order to include the required elements for all the likelihood of cancer, additional modifiers indicate the
patients. presence of incidental or suspicious findings. We imple-
mented the ACR Lung-RADS 1.0 immediately after it was
released.
Our results follow-up team is comprised of a call center,
KEY POINTS certified nurse specialists, and a physician. The team
monitors semiweekly and monthly reports for screening
Executing a new preventive health care service requires examination results. All patients who were screened
education of and acceptance from providers and receive a letter describing the results of the screening CT and
patients. the next steps for follow-up in plain language. The follow-up
Implementing program for screening for a lung cancer team also helps facilitate orders for follow-up scans and
requires multidisciplinary collaboration. appointments and verifies that they are performed. The
Ensuring regulatory compliance and best practice in lung team also notifies the primary care physician of any in-
cancer screening requires system tools and ongoing
cidental findings. Incidental findings unrelated to lung
oversight.
Despite education, providers may inappropriately refer
cancer were found in 7.5% of patients in the NLST,10
patients for lung cancer screening. therefore a mechanism for management is imperative.
Electronic health record tools can be used to Patients who have Lung-RADs 1 or 2 (no suspicious findings)
supplement provider and patient education about and and meet eligibility for screening have the lung cancer
compliance with lung cancer screening. screening topic added to their health maintenance modifier.
The health maintenance activity is visible to all providers,

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e469


BEGNAUD, HALL, AND ALLEN

TABLE 1. Lung-RADS Version 1.0 Assessment Categories


Primary
Category Descriptor Category Descriptor Category Management
Incomplete 0 Additional lung cancer screening CT images and/or
comparison with prior chest CT examinations is
needed
Negative No nodules and definitely benign nodules 1 Continue annual screening with LDCT in 12 months
Benign Appearance or Nodules with a very low likelihood of becoming a 2
Behavior clinically active cancer due to size or lack of growth
Probably Benign Probably benign finding(s): short term follow-up 3 6-month LDCT
suggested, includes nodules with a low likelihood
of becoming a clinically active cancer
Suspicious Findings for which additional diagnostic testing and/or 4A 3-month LDCT; PET/CT may be used when there is a
tissue sampling is recommended $ 8-mm solid component
4B Chest CT with or without contrast, PET/CT and/or tissue
sampling depending on the probability of malignancy
and comorbidities; PET/CT may be used when there is
a $ 8-mm solid component
Significant, Other S
Prior Lung Cancer C
Release date: April 28, 2014.
Abbreviation: LDCT, low-dose CT.

and, when overdue, an alert appears in the patients chart. However, implementing alerts like these must balance pa-
For any positive findings (Lung-RADS 3 or higher), the cer- tient care and health care provider information overload and
tified nurse specialist or physician is notified immediately so alert desensitization.12 Moreover, in order for a BPA to be
that the patient can be discussed in multidisciplinary team useful, patients must have a very detailed smoking history
conference to establish a plan for follow-up or diagnostic recorded in the EHR.
procedure. A unified EHR allows multidisciplinary chart and
image review as requested by ordering providers for ex- Improving examination completion rates. Even as exami-
ternally performed screening examinations with abnormal nation orders increased, many went unscheduled and were
findings. not performed. We believe this low completion rate to be
primarily due to reimbursement uncertainty and out-of-
Overcoming Barriers pocket payment for the examination. Because several na-
Improving examination ordering rates. The number of tional private insurance companies added lung cancer
orders for screening has been less than expected relative to screening coverage prior to the USPSTF recommendation,
the estimated eligible population. Several identifiable bar- we eliminated the self-pay approach and began reviewing
riers are likely responsible. First, adoption of new recom- orders for insurance eligibility. If an eligible patients in-
mendations usually grows over time.11 Adoption is almost surance did not cover screening, they could receive the
certainly limited by provider awareness of screening avail- screening examination under a local grant for this purpose.
ability and eligibility, as well as the time required to in- Thus, we were able to remove a cash price barrier and offer
corporate yet another preventive care service into patient the procedure to all eligible patients as an insurance-neutral
interactions. Secondly, uncertainty regarding insurance screening examination. We notified providers about the
coverage may discourage health care providers from grant and elimination of the self-pay option using an internal
ordering a screening. As we expand to new regions in the newsletter. We also sent letters that explained the benefits
state, we offer educational sessions to primary care practices of screening and offered examination with insurance or
served by those centers. Furthermore, systemwide educa- grant coverage to all patients with orders that had not been
tional events have been held in the form of grand rounds at completed. Finally, our imaging centers do not proactively
multiple sites and lunch webinars for primary care providers. schedule ordered examinations; rather, patients are asked
An effective method of improving adoption of guidelines to initiate the scheduling process. Proactive scheduling of
for lung cancer screening may be to use the EHRs best ordered examinations by radiology would likely improve
practice alert (BPA) functionality. A pop-up alert prompts a completion rates as well.
health care provider about a clinical topic, including eligi-
bility for preventive care activities. Activating an alert for Improving result reporting. Streamlining result reporting
lung cancer screening CT orders for all of our patients also was an initial challenge. We believe it is important to
who meet the criteria would probably improve ordering deliver results to patients in a way that minimizes anxiety
rates and awareness among our ordering physicians. about abnormal findings. We envisioned an experienced

e470 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


LUNG CANCER SCREENING WITH LOW-DOSE CT

certified nurse specialist to deliver abnormal results along TABLE 2. Recommended Components of High-Quality
with a treatment recommendation to the patient. However, Lung Cancer Screening Program4
early examination results follow-up by the ordering provider
was generally not in accordance with national guidelines. Policy in place to offer lung cancer screening only to patients who meet
criteria established by the U.S. Preventive Services Task Force. At
When this happened, we contacted the providers to notify least 90% of patients screening should match this policy.
them of the service we intended to provide and revised
Annual screening until the age of 80, a person has not smoked for
any misinformed recommendations. As providers learned to 15 years, or becomes unwilling or unable to undergo curative lung
trust that we would handle results follow-up, the process resection surgery.
became more streamlined. Primary care physicians are in- Technical specifications of CT conform to ACR-STR standards, and data
formed of all patient communication and recommendations. are collected to ensure mean radiation dose is in compliance with
those.
As a service to patients and providers who scheduled at a
non-university-affiliated imaging center in the health sys- Threshold of nodule size that is considered positive, and data are
collected describing the number and size of nodules detected.
tem, we added an opt-in question to the imaging order in
Structured reporting system, such as Lung-RADS, is used, and data are
September 2015. Ordering providers who opt-in transfer collected on compliance with structured reporting.
nodule follow-up to the centralized results team.
Designated clinicians with expertise in lung nodule management
(diagnostic radiology, interventional radiology, pulmonary
medicine, thoracic surgery, medical oncology, radiation oncology)
Smoking Cessation Program use accepted lung nodule management strategy.
The effect of screening for lung cancer on smoking rates is Integrated smoking cessation program and data collection related to
not fully known13 but thought to be most favorable when smoking cessation interventions offered.
smoking cessation assistance is offered with screening.14
eligibility and discussion of risks and benefits of screening,
Moreover, smoking cessation is an important part of pre-
and offer smoking cessation if applicable. The screening
ventive health and cost-effective lung cancer prevention.15
order itself should also include eligibility criteria.
We use a variety of tools to help patients quit smoking. A
To render an interpretation that will be reimbursed by
pharmacist provides pharmacologic assistance in medica-
CMS, radiologists must be board certified, experienced in
tion therapy management for smoking cessation, and
chest CT, and participate in continuing medical educa-
smoking cessation tools are incorporated in the lung cancer
tion. Imaging centers eligible for reimbursement of lung
screening ordering process in the EHR. Furthermore, our
cancer screening must have experience with low-dose CT
institution is a site for a National Institutes of Healthfunded
lung cancer screening and be accredited as an advanced
smoking cessation trial, with participation offered to
diagnostic imaging center. In addition, effective radiation
smokers who are being screened. Therefore, we consoli-
dose of reimbursable examinations must be less than 1.5
dated available institution resources within a website to
mSv, and data must be collected and submitted to a CMS-
provide centralized access to smoking cessation assistance.
approved registry for each screening examination. Al-
We incorporated smoking cessation resources in our lung
though an extensive list of data elements was initially
cancer screening FAQs, which was given to patients at the
proposed, the final Decision Memo included 10 data
time of order placement. Although many barriers to ideal
elements pertaining to the ordering provider, inter-
provision of smoking cessation assistance exist,16,17 it is
preting radiologist, patient smoking history, and CT
unequivocally recommended to accompany lung cancer
scanner.
screening.4
Adapting EHR tools. To facilitate compliance with CMS
Adapting to New Guidelines and CMS Requirements requirements, we modified the EHR tools. In addition to
Professional society guidelines. In late 2014, the American making modifications to the lung cancer screening imaging
College of Chest Physicians and the American Thoracic So- order, we created a SmartSet, which is a bundle of orders
ciety jointly published a statement that outlined the key and documentation related to a topic. The SmartSet guides
components of a high-quality lung cancer screening program clinicians on patient eligibility, required billing codes, sug-
(Table 2).4 Guidelines are also available for lung cancer gested wording for the shared decision-making visit, orders
screening from the ACR, Society of Thoracic Radiology,8 and for smoking cessation tools, and the imaging order. When
the American Association of Thoracic Surgeons.7 Our pro- signed, the SmartSet outputs patient instructions with
gram had already been built with these best practices in screening FAQs and scheduling information. A BPA was
mind and required no modifications to meet the guidelines. created to direct the ordering provider from the stand-alone
order to the SmartSet, as many providers had become ac-
CMS Decision Memo. The CMS Decision Memo was re- customed to using the order laone.
leased in November 201418 and incorporated many of the
elements of a high-quality screening program previously RESULTS
recommended (Table 3). In addition, a credentialed provider Examination Ordering and Completion Rates
must conduct a shared decision-making visit with the patient Systemwide, 902 unique patients received orders for
when ordering the initial screening examination, document screening using low-dose CT. However, only 63% of these

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e471


BEGNAUD, HALL, AND ALLEN

TABLE 3. CMS Lung Cancer Screening Eligibility Criteria19


Type Criteria
Beneficiary Eligibility Criteria 1. Age 5577
2. Asymptomatic (no signs or symptoms of lung cancer)
3. Tobacco smoking history of at least 30 pack-years (one pack-year 5 smoking one pack per day for one year; 1 pack 5
20 cigarettes)
4. Current smoker or one who has quit smoking within the last 15 years
5. Receives a written order for LDCT lung cancer screening that meets the following criteria:
a. For the initial LDCT lung cancer screening service: a beneficiary must receive a written order for LDCT lung cancer
screening during a lung cancer screening counseling and shared decision-making visit, furnished by a physician (as
defined in Section 1861(r)(1) of the Social Security Act) or qualified nonphysician practitioner (meaning a physician
assistant, nurse practitioner, or clinical nurse specialist as defined in 1861(aa)(5) of the Social Security Act). A lung
cancer screening counseling and shared decision-making visit includes the following elements (and is appropriately
documented in the beneficiarys medical records):
i. Determination of beneficiary eligibility including age, absence of signs or symptoms of lung cancer, a specific
calculation of cigarette smoking pack-years; and if a former smoker, the number of years since quitting;
ii. Shared decision-making, including the use of one or more decision aids, to include benefits and harms of
screening, follow-up diagnostic testing, overdiagnosis, false-positive rate, and total radiation exposure;
iii. Counseling on the importance of adherence to annual lung cancer LDCT screening, impact of comorbidities, and
ability or willingness to undergo diagnosis and treatment;
iv. Counseling on the importance of maintaining cigarette smoking abstinence if former smoker; or the importance
of smoking cessation if current smoker and, if appropriate, furnishing of information about tobacco cessation
interventions; and
v. If appropriate, the furnishing of a written order for lung cancer screening with LDCT.
b. For subsequent LDCT lung cancer screenings: the beneficiary must receive a written order for LDCT lung cancer
screening, which may be furnished during any appropriate visit with a physician (as defined in Section 1861(r)(1) of the
Social Security Act) or qualified nonphysician practitioner (meaning a physician assistant, nurse practitioner, or clinical
nurse specialist as defined in Section 1861(aa)(5) of the Social Security Act). If a physician or qualified nonphysician
practitioner elects to provide a lung cancer screening counseling and shared decision-making visit for subsequent lung
cancer screenings with LDCT, the visit must meet the criteria described above for a counseling and shared decision-
making visit.
c. Written orders for both initial and subsequent LDCT lung cancer screenings must contain the following information,
which must also be appropriately documented in the beneficiarys medical records:
i. Beneficiary date of birth;
ii. Actual pack-year smoking history (number);
iii. Current smoking status, and for former smokers, the number of years since quitting smoking;
iv. Statement that the beneficiary is asymptomatic (no signs or symptoms of lung cancer); and
National Provider Identifier (NPI) of the ordering practitioner.
Reading Radiologist Eligibility 1. Board certification or board eligibility with the American Board of Radiology or equivalent organization
Criteria 2. Documented training in diagnostic radiology and radiation safety
3. Involvement in the supervision and interpretation of at least 300 chest CT acquisitions in the past 3 years
4. Documented participation in continuing medical education in accordance with current American College of Radiology
standards
5. Furnish lung cancer screening with LDCT in a radiology imaging facility that meets the radiology imaging facility
eligibility criteria below
Radiology Imaging Facility 1. Performs LDCT with volumetric CT dose index (CTDIvol) of # 3.0 mGy (milligray) for standard size patients (defined to
Eligibility Criteria be 57 and approximately 155 pounds) with appropriate reductions in CTDIvol for smaller patients and appropriate
increases in CTDIvol for larger patients
2. Utilizes a standardized lung nodule identification, classification, and reporting system
3. Makes available smoking cessation interventions for current smokers and collects and submits data to a CMS-
approved registry for each LDCT lung cancer screening performed.

examinations have been completed (Fig. 1). Over time, both (Table 4). About one-third of patients had inadequate
examinations orders and completion rates increased. Since smoking history as recorded in the EHR history tool to
the function has been offered, 81% of examinations com- determine eligibility. Many of these patients did report
pleted opted in for centralized result management. Using smoking for at least 30 pack-years on the patient
the Lung-RADS interpretation strategy, 70% of patients had a questionnaire administered prior to low-dose CT. An-
positive finding (Lung-RADS 2 or higher). However, only 17% other nearly one-third of patients were not eligible for
of patients had a finding that required follow-up sooner than screening based on age or smoking history, as recorded.
annually (Fig. 2). Among patients who had smoking intensity (packs per
day and smoking years) recorded, implementation of a
BPA clarifying eligibility criteria had no impact on the
Patient Eligibility proportion of patients referred for screening who met
EHR record of eligibility was measured by completed eligibility criteria (30 or more pack years). Similarly, the
smoking history tool. Of the 572 screening examinations proportion of screening referrals for patients whose
completed, less than 20% were eligible by smoking history recorded quit smoking date was more than 15 years ago

e472 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


LUNG CANCER SCREENING WITH LOW-DOSE CT

FIGURE 1. Lung Cancer Screening Order Completion TABLE 4. Patient Characteristics Contributing to
by Quarter Eligibility for Lung Cancer Screening as Recorded in
Electronic Health Records

Patient Characteristics Number of Patients (572, %)


Current Smoker 268 (47%)
Former Smoker 282 (49%)
No Recorded Quit Date 48 (282 patients, 17%)
Quit > 15 years 31 (5%)
Pack-year < 30 117 (20%)
Inadequate Entry to Calculate 206 (36%)
Age-inappropriate 21 (4%)
Apparently Eligible 123 (22%)

did not substantially change after implementation of a screening program before current regulatory requirements
the BPA. were released.
Approximately one-third of the orders were for patients
Counseling and Shared Decision-Making Visit whose eligibility for screening could not be confirmed.
Nearly all examinations (95%) were ordered during an office Eligibility was unconfirmed due to insufficient documen-
visit. Yet many of those did not have documentation of tation of smoking history, documented smoking intensity
counseling and a shared decision-making visit. Shared- less than 30 pack-years, or quit more than 15 years prior to
decision making documentation has improved over time examination. Our health system has had some success but
(Fig. 3), but, in January 2016, it still only occurred in half of continues to optimize EHR tools to facilitate compliance
patients with completed screening examinations. Prior to with recommended eligibility. Most patients did report
implementation of a BPA indicating CMS requirements, 87% smoking history on prescreening questionnaires that was
of patients did not have documentation of a shared decision- adequate for lung cancer screening eligibility. We believe
making visit. Since the BPA function, compliance with shared that infrequent and inaccurate recording of smoking his-
decisions-making documentation in patients with Medicare tory by clinicians to be responsible. Furthermore, the EHR
or Medicaid has improved to 46%. tool for recording smoking history is inadequate to fully
capture the variability in smoking intensity over time that
describes many smokers habits. A tool that more specif-
DISCUSSION ically captures light smoking, heavy smoking, and quit
Lung cancer screening is a relatively new preventive health periods can help record an accurate account of smoke
care service (secondary prevention). As such, developing and exposure. In addition to improving EHR tools and utiliza-
implementing new processes can be guided by national tion, confirmation of eligibility prior to screening using low-
leaders but must also be customized to meet the needs of dose CT will likely be needed to reach perfect compliance
local and regional health care systems. We began implementing with eligibility requirements.
Early orders had a low completion rate, which we believe
to be due to the absence of insurance coverage and the self-
FIGURE 2. Lung-RADS Results for Completed Orders pay cost of the examination ($150). After the CMS final
Decision Memo, examination orders quadrupled, supporting
our notion that uncertainty about reimbursement drove
initial low uptake. Feedback from providers suggests that
anxiety prevents patients from scheduling examinations
despite having the information to do so. Furthermore, in
most clinics, patients are asked to schedule examinations
themselves rather than having examinations scheduled
for them. Completion rates continue to improve, despite
having a patient-initiated scheduling system.
Our findings suggest that reimbursement-driven en-
forcement of patient eligibility as required by Medicare is
necessary to apply screening to the high-risk groups who are
most likely to benefit. We also concur with the need for
continued monitoring of screening outcomes through
national registries to further refine screening eligibility and
utility.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e473


BEGNAUD, HALL, AND ALLEN

FIGURE 3. Lung Cancer Screening Orders Completed in 2015 and Accompanied by Shared Decision Making
Documentation

Abbreviation: SDM, shared decision making.

This article demonstrates our experience implementing reimbursement for eligible patients. As diagnostic radiology
a lung cancer screening program outside of a federally technology improves and adjunctive methods of early de-
supported clinical trial. Future trends will be greatly shaped tection of lung cancer become available, the riskbenefit
by outcomes reported from national registries and ratio for screening for lung cancer will continue to evolve
by experience with pervasive screening in a climate of and, ideally, improve.

References
1. Aberle DR, Adams AM, Berg CD, et al. Reduced lung-cancer mortality computed tomography (CT): 2014 (Resolution 4). J Thorac Imaging.
with low-dose computed tomographic screening. N Engl J Med. 2011; 2014;29:310-316.
365:395-409. 9. American College of Radiology. Lung-RADS Version 1.0 Assess-
2. U.S. Preventive Services Task Force. Lung cancer screening: recom- ment Categories Release date: April 28, 2014. https://www.acr.org/
mendation statement. Ann Intern Med. 2004;140:738-739. ~/media/ACR/Documents/PDF/QualitySafety/Resources/LungRADS/
3. Bach PB, Silvestri GA, Hanger M, et al. Screening for lung cancer: ACCP AssessmentCategories.pdf. Accessed February 13, 2016.
evidence-based clinical practice guidelines (2nd edition). Chest. 2007; 10. Church TR, Black WC, Aberle DR, et al. Results of initial low-dose
132:69S-77S. computed tomographic screening for lung cancer. N Engl J Med.
4. Mazzone P, Powell CA, Arenberg D, et al. Components necessary for high 2013;368:1980-1991.
quality lung cancer screening: American College of Chest Physicians and 11. Francke AL, Smit MC, de Veer AJ, et al. Factors influencing the
American Thoracic Society Policy Statement. Chest. 2015;147:295-303. implementation of clinical guidelines for health care professionals:
5. Farjah F, Flum DR, Varghese TK Jr, et al. Surgeon specialty and long-term a systematic meta-review. BMC Med Inform Decis Mak. 2008;8:38.
survival after pulmonary resection for lung cancer. Ann Thorac Surg. 12. Ash JS, Berg M, Coiera E. Some unintended consequences of information
2009;87:995-1004, discussion 1005-1006. technology in health care: the nature of patient care information system-
6. National Comprehensive Cancer Network. Lung Cancer Screening. https:// related errors. J Am Med Inform Assoc. 2004;11:104-112.
www.nccn.org/store/login/login.aspx?ReturnURL=http://www.nccn.org/ 13. Tanoue LT, Tanner NT, Gould MK, et al. Lung cancer screening. Am J
professionals/physician_gls/pdf/lung_screening.pdf. Accessed February Respir Crit Care Med. 2015;191:19-33.
13, 2016. 14. Park ER, Gareen IF, Japuntich S, et al. Primary care provider-delivered
7. Jaklitsch MT, Jacobson FL, Austin JH, et al. The American Association for smoking cessation interventions and smoking cessation among par-
Thoracic Surgery guidelines for lung cancer screening using low-dose ticipants in the National Lung Screening Trial. JAMA Intern Med. 2015;
computed tomography scans for lung cancer survivors and other high- 175:1509-1516.
risk groups. J Thorac Cardiovasc Surg. 2012;144:33-38. 15. Villanti AC, Jiang Y, Abrams DB, et al. A cost-utility analysis of lung cancer
8. Kazerooni EA, Austin JH, Black WC, et al. ACR-STR practice parameter for screening and the additional benefits of incorporating smoking ces-
the performance and reporting of lung cancer screening thoracic sation interventions. PLoS One. 2013;8:e71379.

e474 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


LUNG CANCER SCREENING WITH LOW-DOSE CT

16. Manley MW, Griffin T, Foldes SS, et al. The role of health plans in details/nca-proposed-decision-memo.aspx?NCAId=274&NcaName=
tobacco control. Annu Rev Public Health. 2003;24:247-266. Screening+for+Lung+Cancer+with+Low+Dose+Computed+Tomography+
17. Panaitescu C, Moffat MA, Williams S, et al. Barriers to the provision (LDCT)&MEDCACId=68&IsPopup=y&bc=AAAAAAAAAgAAAA%3d%3d&.
of smoking cessation assistance: a qualitative study among Ro- Accessed February 13, 2016.
manian family physicians. NPJ Prim Care Respir Med. 2014;24: 19. Centers for Medicare & Medicaid Services. Decision Memo for
14022. Screening for Lung Cancer with Low Dose Computed Tomography
18. Centers for Medicare & Medicaid Services. Proposed Decision Memo (LDCT) (CAG-00439N). https://www.cms.gov/medicare-coverage-
for Screening for Lung Cancer with Low Dose Computed Tomography database/details/nca-decision-memo.aspx?NCAId=274. Accessed Feb-
(LDCT) (CAG-00439N). https://www.cms.gov/medicare-coverage-database/ ruary 13, 2016.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e475


ADDRESSING TOBACCO WITH CANCER SCREENING AND TREATMENT

Lung Cancer Screening, Cancer Treatment, and Addressing the


Continuum of Health Risks Caused by Tobacco
Graham W. Warren, MD, PhD, Jamie S. Ostroff, PhD, and John R. Goffin, MD

OVERVIEW

Tobacco use is the largest preventable risk factor for the development of several cancers, and continued tobacco use by
patients with cancer and survivors of cancer causes adverse outcomes. Worldwide tobacco control efforts have reduced
tobacco use and improved health outcomes in many countries, but several countries continue to suffer from increased
tobacco use and associated adverse health effects. Continued tobacco use by patients undergoing cancer screening or
treatment results in continued risk for cancer-related and noncancer-related health conditions. Although integrating to-
bacco assessment and cessation support into lung cancer screening and cancer care is well justified and feasible, most
patients with cancer unfortunately do not receive evidence-based tobacco cessation support. Combining evidence-based
methods of treating tobacco addiction, such as behavioral counseling and pharmacotherapy, with practical clinical con-
siderations in the setting of lung cancer screening and cancer treatment should result in substantial improvements in access
to evidence-based care and resultant improvements in health risks and cancer treatment outcomes.

T obacco use, particularly combustible tobacco, remains


the leading preventable cause of cancer worldwide.
Since the original U.S. Surgeon Generals report (SGR) on
Although smoking cessation can reverse many of the
adverse effects of smoking among the general population
and among patients with cancer, most oncologists un-
tobacco in 1964 concluding that smoking was causally linked fortunately do not address tobacco use by providing
to lung and laryngeal cancer, there have been numerous evidence-based tobacco cessation support.3,4 There are
SGRs confirming the adverse effects of tobacco on a spec- several potential reasons why oncologists and other pro-
trum of diseases, including increased risk for developing a viders may not provide cessation support,5 but the funda-
wide range of cancers.1 The 2014 SGR extended findings of mental consequence is that many people continue to be
prior reports by concluding that continued smoking by exposed to cancer risk and adverse cancer treatment out-
patients with cancer and survivors of cancer causes in- comes because they do not have access to evidence-based
creased overall mortality, cancer-specific mortality, risk for tobacco cessation support. The overall purpose of this ed-
second primary cancer, and associations with increased ucational article is to convey the continuum of risk associ-
cancer treatment toxicity.2 As updated in the 2014 SGR, ated with tobacco use cancer risk and cancer treatment
smoking and tobacco addiction usually starts in youth, outcomes, discuss recent data as related to lung cancer
continues into adulthood, and results in chronic exposure to screening and cancer treatment in the context of tobacco
chemicals that lead to the development of cancer, heart use, and provide practical considerations that can be helpful
disease, pulmonary disease, and several other adverse to increase access to tobacco cessation support. There are
health conditions. Many of the effects of tobacco are partly limited studies evaluating methods to implement smoking
or entirely reversible, with reduced risks of adverse health cessation into lung cancer screening and cancer care.
conditions associated with longer periods of smoking ab- However, this article will further elaborate on practical
stinence. As it pertains to patients with cancer and the risk of clinical considerations that could be helpful in the design and
developing cancer, smoking cessation can reduce the risks of implementation of tobacco cessation approaches across the
developing cancer, can improve cancer treatment out- continuum of cancer risk and treatment.
comes, and can reduce or prevent the development of other There are thousands of references describing the adverse
tobacco-related diseases.2 health risks associated with tobacco use and hundreds of

From the Departments of Radiation Oncology and Cell and Molecular Pharmacology, Medical University of South Carolina, Charleston, SC; Department of Psychiatry and Behavioral
Sciences, Memorial Sloan Kettering Cancer Center, New York, NY; Department of Oncology, McMaster University, Hamilton, Ontario, Canada.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Graham W. Warren, MD, PhD, Department of Radiation Oncology, Medical University of South Carolina, 169 Ashley Ave., MSC 318, Charleston, SC 29425;
email: warrengw@musc.edu.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 223


WARREN, OSTROFF, AND GOFFIN

large reviews describing specific areas of risk and risk mit- mortality associated with tobacco-related diseases.
igation as related to tobacco use. For the purposes of this However, in most low- and middle-income countries,
educational article, we will highlight substantial review work tobacco use (particularly combustible tobacco use
presented over the past 50 years by the U.S. Public Health [cigarette smoking]) is increasing. Growing tobacco
Service (PHS) through updated SGRs that have focused on consumption in China has offset decreased tobacco
tobacco-associated health risks, addiction, and mitigation,1 consumption in other developed countries, and one-
with particular emphasis on the 2014 SGR2 that summarizes third of all male individuals who smoke are Chinese. In
many current and former conclusions associated with to-
many countries, male tobacco consumption is de-
bacco use. Whenever possible, references will be provided
creasing, whereas female tobacco use has remained
to online resources that keep information up to date, which
unchanged or is increasing.
is important as related to ongoing product, use, and health
information related to tobacco. Lung cancer, chronic obstructive pulmonary disease,
and heart disease are leading causes of death from
tobacco. More than 80% of lung cancer and lung cancer
WORLDWIDE TOBACCO USE AND THE deaths are attributed to smoking, and approximately
CONTINUUM OF RISK one-third of all cancer deaths are attributed to tobacco
Cigarette smoke is the predominant form of worldwide use. However, tobacco use causes disease in almost
tobacco consumption, although in different countries dif- every organ in the body. Thus, continued tobacco use
ferent patterns exist that are highly influenced by culture, after a cancer diagnosis results in continued risk to other
gender, governmental regulation, advertising, and
organs and decreases the effectiveness of cancer
manufacturing. Readers are encouraged to review the fifth
treatment.
edition of the Tobacco Atlas, updated in 2015,6 available
online and in print. Produced by the American Cancer So- Exposure to secondhand smoke can confer similar dis-
ease risks in a dose- and time-dependent manner.
ciety and the World Lung Foundation, the Tobacco Atlas
highlights patterns of worldwide tobacco use. Highlights Higher risk is associated with higher and more prolonged
from the Tobacco Atlas and 2014 SGR include the following: exposure.

Approximately 100 million people died of tobacco-


related causes in the 20th century and an estimated
Addiction has both behavioral and biologic underpin-
nings. Most people start using tobacco during childhood
1 billion people will die in the 21st century from tobacco- or adolescence, primarily influenced by social pressure,
related causes if current tobacco use patterns persist. An advertising, and curiosity. Chronic tobacco use and
estimated 50% of people who use tobacco will die of a addiction is reflective of nicotine dependence, which
tobacco-related disease. can affect constitutively expressed nicotinic acetylcho-
An estimated 5.8 trillion cigarettes were smoked world-
wide in 2014. In many high-income countries, tobacco
line receptors in the brain and throughout the body.
Tobacco use has strong social associations, in that many
use has plateaued and in many cases decreased over the people who smoke often have friends and relatives who
past 30 to 40 years with concomitant decreases in smoke.
Tobacco use is higher among people with lower educa-
tion, among individuals with lower income, and among
people who are diagnosed with a psychiatric disease.
KEY POINTS
However, many financially disadvantaged people will
Tobacco use confers a continuum of risk across the continue to expend resources to continue tobacco use.
general population as well as among patients diagnosed
with cancer, justifying the need to address tobacco use
Tobacco addiction usually starts early in life. Most
people have tried to stop smoking several times and
before, during, and after a cancer diagnosis. most continue to want to stop smoking. Tobacco ces-
Worldwide tobacco consumption patterns continue to sation is often very difficult and requires multiple at-
place a substantial burden on the economy and health of
tempts to achieve success. However, people who have
people in every nation.
Addressing tobacco use is a critical component of lung quit smoking in the past remain at risk for relapse,
cancer screening and cancer treatment. particularly if they are exposed to secondhand smoke.
Although strong evidence exists for methods to help
patients stop tobacco use, clinical implementation is
It is estimated that worldwide tobacco use causes $1
trillion annually in economic damage, with $289 billion
limited with little evidence-based support. spent in treating the health consequences of smoking in
Considering the clinical setting, available resources, and
the United States alone. Only a very small portion of
method of tobacco assessment, cessation support and
developing a sustainable clinical approach are practical tobacco product revenues (typically much less than 1%)
considerations when implementing change to improve are used for tobacco control initiatives.
access to tobacco cessation support. Effective strategies to reduce tobacco use include in-
creased product taxation, smoke-free policies and laws,

224 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


ADDRESSING TOBACCO WITH CANCER SCREENING AND TREATMENT

legislative regulation, education such as graphic product survival. The large, randomized National Lung Screening Trial
labels, limiting or eliminating marketing, increased (NLST) provided the first strong evidence of benefit from
pricing, and providing smoking cessation resources. screening with low-dose computed tomography (LDCT)
International treaties and agreements, such as the compared with chest x-ray.12 Investigators screened an el-
World Health Organization Framework Convention on igible population (age 5574) who had at least a 30 pack-year
Tobacco Control, that coordinate these activities can be smoking history and reported smoking within the past 15
highly effective in influencing national and international years, in which participants had a baseline LDCT scan that
change. However, litigation is a widely used method by was followed by two annual screening LDCT scans. With a
the tobacco industry to limit tobacco control initiatives. follow-up of 6.5 years, LDCT screening decreased lung
Although it was discussed in both the 2014 SGR and the cancerspecific mortality by 20%, overall survival was im-
Tobacco Atlas, an important topic to discuss individually is proved by 6.7%, and CT LDCT screening diagnosed more
continuously evolving tobacco product design. Changes stage I to stage II disease (57% vs. 39%). Based on NLST data,
in cigarette design over the past 50 years have resulted in several agencies have recommended LDCT screening for an
cigarettes that are easier to inhale, leading to changes in eligible population provided that screening is undertaken in
disease risk. For instance, lung cancer was once a rare the setting of appropriate expertise.
disease that significantly increased with the advent of mass With the advent of LDCT screening as evidence-based
cigarette production and changes in cigarette design that medicine, it has been increasingly important to consider
allowed deeper inhalation resulted in the shift of lung cancer the utility of smoking cessation at the time of CT screening.
patterns from centrally to peripherally located cancers.7 Several advocates have promoted screening for tobacco use
Unfortunately, changes in tobacco design did not result in and providing tobacco cessation support to people who use
improved health: recent data demonstrate that people who tobacco. In general, as supported by current National
smoke are now more heavily addicted, with continued in- Comprehensive Cancer Network (NCCN) guidelines, smoking
creased health risks.8 However, the benefits of cessation cessation is recommended for any individuals who are
remain intact with reduced health risks associated with undergoing LDCT screening and reporting current tobacco
longer periods of cessation. use.13 Newly released NCCN Smoking Cessation Guidelines
As new tobacco products are developed, it will take years provide additional guidance and details for providing to-
or decades to determine the risk. Electronic cigarettes (e- bacco cessation support to patients with cancer as well as
cigarettes) or electronic nicotine delivery systems (ENDS) patients undergoing LDCT screening.14
are relatively new products with largely unknown health Perhaps the best evidence for promoting smoking ces-
effects and patterns of use. Although many online resources sation in the context of LDCT screening is the recent report
are promoting the use of ENDS to facilitate tobacco ces- by Tanner et al,15 who evaluated the effect of smoking and
sation, the effects of ENDS to assist in tobacco cessation are smoking cessation on survival in the NLST. Current smoking
not yet known and ENDS cannot yet be promoted as a to- increased overall and cancer-specific mortality among in-
bacco cessation device.9 It is important to note that the dividuals who participated in LDCT screening, but 7 years of
health effects of ENDS may be dependent on the population smoking cessation reduced mortality at a level comparable
in question: ENDS use by youth who have never used to- to the benefit of LDCT, and 15 years of cessation reduced
bacco are likely to result in different health effects compared mortality by 38%. Given the large population of current
with adults who smoke and transition to ENDS use.9 Cur- smokers who are eligible for lung cancer screening, cost-
rently, physicians and clinical providers are limited in pro- effectiveness is an important consideration for policy
viding evidence-based guidance regarding ENDS use makers considering implementation. Studies based on NLST
because most of the evidence is simply lacking at this time. data suggest that screening for lung cancer may have an
incremental cost-effectiveness ratio (ICER) per quality of
life-year saved in the $50,000 to $80,000 range.16 It is al-
TOBACCO USE IN THE CONTEXT OF LUNG ready established that smoking cessation programs are
CANCER SCREENING AND CANCER TREATMENT highly cost-effective with an ICER less than $9,000 per
When considering integration of tobacco cessation into early life-year saved.17 Simulation work suggests that the addition
detection of lung cancer, it is important to note that al- of a smoking cessation program to an LDCT screening
though diagnosis with resectable nonsmall cell lung cancer program can substantially improve cost-effectiveness.18 As
can result in 5-year survival exceeding 70%, most patients such, combining LDCT and smoking cessation programs may
with lung cancer are diagnosed with symptoms, are not add incremental treatment cost but also enhance cost ef-
operable, and have a dismal long-term prognosis.10 Cap- ficiency and provide the proven health benefits associated
turing patients at an early curative stage is the optimal with each.
strategy for managing lung cancer. Early efforts to screen for When considering tobacco use by patients with cancer, the
lung cancer with chest x-rays or sputum cytology may have 2014 SGR concluded that smoking by patients with cancer
resulted in a modest shift in staging, but with no effect on and survivors of cancer caused adverse outcomes and that
survival.11 Subsequent screening of large cohorts with CT smoking cessation improves cancer treatment outcomes.2
scans suggested improved stage shifting and improved Smoking and tobacco products alter cancer biology, promoting

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 225


WARREN, OSTROFF, AND GOFFIN

increased proliferation, angiogenesis, migration, invasion, to quit correlated with actual cessation.26 Consistently
metastasis, and resistance to cytotoxic cancer therapy.19 negative results from the Early Lung Cancer Action Program
Examination of the exhaustive evidence in the 2014 SGR demonstrated that baseline negative LDCT scan results were
demonstrates that one or more adverse effects of smoking associated with lower point abstinence rates at follow-up
extend to virtually all cancer disease sites and cancer compared with positive LDCT scan results, but they had no
treatments. As a result, the adverse effects of smoking are significant effect on prolonged abstinence.27 However, NLST
generally ubiquitous across cancer as a whole and ad- participants with physicians who assisted with cessation and
dressing tobacco use is pertinent to the treatment of all arranged follow-up had a 40% to 46% higher rate of ab-
patients with cancer. The authors are unaware of detailed stinence at 1 year, although physician rates of assisting and
cost-benefit analyses for implementing smoking cessation arranging were low.28
into the care of patients with cancer; however, given the There have been several thoughtful recent reviews on
extremely high costs of treating cancer and the relatively low addressing tobacco use in the context of cancer care.29-32
cost of providing smoking cessation support, it is very likely The principles of addressing tobacco use mimic evidence-based
that implementing smoking cessation practices into cancer PHS guidelines in the general population33 recommending the
care may be one of the most cost-effective methods to use of behavioral counseling and pharmacotherapy to help
improve cancer treatment outcomes. patients quit smoking. The 5As model (Ask, Assess, Advise,
Assist, Arrange) is a widely accepted method of addressing
tobacco use. The 2As and R (or 2As and C) model refers to Ask,
LUNG CANCER SCREENING, CANCER Advise, Refer or to Ask, Advise, Connect, respectively, in which
TREATMENT, AND TOBACCO CESSATION patients are referred or connected to evidence-based smoking
The question arises as to whether a LDCT screening pop- cessation support through a dedicated tobacco cessation
ulation is well suited to promoting smoking cessation. program. As reviewed in these references, patients with
Presumably, individuals participating in LDCT screening may cancer are often receptive to smoking cessation support
represent a population motivated to improve their health. but suffer from similar patterns of tobacco use and risk of
Compared with the general population, participants in the relapse compared with the general population. What is
NLST were more commonly married, better educated, more particularly unique to patients with cancer is that many
frequently ex-smokers, and younger. Persistent smokers in newly diagnosed patients will receive cancer treatment
the NLST were associated with having lower education, requiring multiple visits to an oncology provider over a
being unmarried, being younger, and having a heavier relatively short period of time. As such, patients with
smoking history.20 Similarly, in the NELSON screening study, cancer have much more frequent contact with medical
participants in the control arm were younger, were better providers than the general population, and this provides an
educated, were former smokers, and expressed a greater ideal platform to address tobacco use at multiple visits and
willingness to quit smoking.21 Established evidence dem- support patients with cancer in an effective quit attempt.29-31
onstrates that smoking is associated with having a lower The concept of a teachable moment is ideal, in which
socioeconomic status, having lower education, being un- patients with newly diagnosed cancer are faced with a life-
married, and having poor health in part attributed to poorer changing situation and providers can take advantage of this
health-promoting activities.2,6 Participating in a LDCT pro- opportunity to assist patients in a beneficial change in health
gram could reflect increased interest in quitting or habits. Reviews above and the American Society for Clinical
following a healthier lifestyle. Data suggest that non- Oncology tobacco resources34 discuss these issues in greater
participants express feelings of protection based on personal detail.
or family health status, indicate that they are too old for Despite the clinical need and rationale to address tobacco
screening, and express sentiments of fatalism, all charac- use, recent surveys demonstrate that although approxi-
teristics unlikely to compel action.22 Consideration must be mately 90% of oncologists ask about tobacco use and ap-
given as to how to recruit hard-to-reach populations. proximately 80% advise patients to quit smoking, only 30%
When considering the effect of LDCT on smoking cessa- to 40% provide assistance with quitting.3,4 Given the evi-
tion, data suggest that participation in screening is associ- dence, one might question why many clinicians and on-
ated with increased motivation to quit smoking.23 Although cologists do not readily incorporate tobacco cessation into
NLST participants perceived a high risk of cancer as a result of effective clinical practice. However, it is important to note
smoking, smokers doubted the benefits of quitting and had that approximately 90% reported believing that smoking
low confidence in their ability to quit.24 A review of nine caused adverse outcomes for patients with cancer and that
cross-sectional, longitudinal and randomized controlled smoking cessation should be a standard part of cancer care,
studies examined the effect of CT screening for lung cancer which suggests that efforts to convert physicians knowledge
on smoking behaviors and found quit rates ranging from base to a better understanding of the adverse effects of
6.6% to 42%.25 Investigators for the Danish Lung Cancer smoking may not prove fruitful. A recent survey of LDCT
Screening Trial found no difference between study arms in screening programs shows very similar findings, with 99%
smoking rates at 5 years, although the number of ex-smokers reporting asking about tobacco use and 91% advising to quit,
increased significantly in both arms and a stated motivation but only 50% to 60% providing assistance with cessation and

226 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


ADDRESSING TOBACCO WITH CANCER SCREENING AND TREATMENT

38% discussing cessation medications.35 Analysis of predictive cessation support themselves or refer to existing com-
barriers to providing cessation support suggests that a lack of munity resources. Considering the location where ces-
time, lack of cessation resources, and lack of education on sation support is provided is important to consider.
cessation methods are dominant barriers to providing ces- Providing cessation support in a busy clinical setting
sation support.5 However, the concern that patients do not may be met with resistance, such as where patient wait
want help with tobacco cessation has been refuted in studies times for oncology care are often delayed because of
demonstrating that approximately 80% to 90% of patients with limited clinical space. This is particularly salient in clinics
cancer automatically referred to receive smoking cessation where multiple clinicians use a single clinic room.
support (an opt-out approach to tobacco cessation) are
receptive to intervention36,37 and patients who quit smoking How will tobacco be assessed? Reviews above discuss
methods to assess tobacco. However, it is important to
had significant improvements in survival.38 Collectively, these
consider how tobacco use assessments will be collected.
data support a substantial need to provide evidence-based
cessation support to participants in LDCT screening programs For instance, tobacco assessments collected by nurses
as well as to patients with cancer to manifest in the numerous or other staff can permit identification of tobacco use
health benefits associated with smoking cessation. However, that can be presented to a physician prior to entering a
despite the potential public health benefits for further re- patient room. It also permits consideration of scheduling a
duction in tobacco-related morbidity and mortality, it is un- patient for cessation support while a physician is
known whether screening and treatment centers will commit interviewing a patient for LDCT screening or cancer care.
the time and resources needed to ensure delivery of evidence- The use of electronic medical records or paper charts is
based tobacco cessation treatment.35 also important and clinic specific. Clinical infrastructure
and development of efficient assessment and treatment
PRACTICAL CONSIDERATIONS TO IMPLEMENT systems can be critical to gaining the support of clinical
CHANGE and administrative staff in a department. However, once a
Simply stated, asking patients about tobacco use and ad- patients tobacco use has been identified, all clinicians
vising them to quit is performed by most clinicians and is should advise patients to quit smoking and either provide
necessary, but patients have higher smoking cessation rates cessation assistance or refer/connect patients to dedi-
when providers help them quit smoking. Resources noted cated cessation support programs.
above expound on common evidence-based methods to
address tobacco use and assist patients in quitting
How will tobacco cessation support be delivered? The
basic elements of providing tobacco cessation support
smoking.2,14,29-34 Readers are encouraged to review these include assessing a patients willingness to quit and de-
resources as needed. However, implementation of veloping an individualized tobacco cessation approach
evidence-based care requires careful consideration of clin-
that incorporates counseling, pharmacotherapy, and
ical resources, environment, and expertise. Several practical
follow-up. Specialized training is available. However,
considerations are important to consider when developing
cessation support can be delivered by a broad variety of
approached to addressing tobacco use; unfortunately, the
evidence base is lacking as to the best approach. When providers, including physicians, physician assistants, nurse
addressing tobacco use with LDCT or cancer care, it is im- practitioners, nurses, pharmacists, psychologists, and
portant to consider the following: other providers who have received tobacco treatment
What is the clinical setting? Consider opportunities to
integrate tobacco treatment with medical management
education. Advice to quit smoking should be repeatedly
delivered by all clinical staff. Support in the United States
and engage patients in tobacco treatment efforts. Pa- and many other countries can also be delivered using
tients presenting for LDCT screening typically return quitlines (1-800-QUIT-NOW in the United States) and
annually for repeat assessments unless an abnormal community cessation programs. In the experience of the
finding requires further work-up. In this setting, LDCT authors, many institutions may already have programs or
participants may be reticent to return for in-person clinicians available that are often underutilized.
clinical support for tobacco cessation alone. In con-
trast, patients diagnosed with cancer often return fre-
How will a tobacco cessation program be maintained? In
the experience of the authors, successful implementa-
quently and multiple times for work-up, management, tion of a tobacco cessation effort by clinicians or ded-
and follow-up. Considering delivery of tobacco cessation icated cessation programs includes buy-in and
in coordination with repeated cancer care visits may be participation from patients, clinical staff, providers, and
helpful to both patients and providers. administration. Educating about the importance of to-
What resources are available to assess and address
tobacco use? One size does not fit all. Larger institutions
bacco across the clinic, cancer center, or institution can
increase awareness and help sustain a tobacco cessation
may have more resources to support development of initiative. Considering issues related to billing and re-
dedicated tobacco cessation programs, whereas small imbursement can facilitate implementation in many
clinics and individual providers may need to provide cases, but given the high cost associated with cancer

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 227


WARREN, OSTROFF, AND GOFFIN

care, the cost savings associated with cessation by tobacco use will continue to promote adverse health risks.
helping prevent costs associated with increased cancer This simple relationship is easy to apply to a chronic disease
treatment toxicity or progression to next-line cancer state such as cancer. Addressing tobacco by assessing tobacco
therapy are likely substantial. Thus, perhaps billing and use and providing evidence-based tobacco cessation support
reimbursement on the front end of clinical care should is extremely important across the continuum of cancer
not be the foremost concerns when implementing a screening, diagnosis, and treatment. Although many evidence-
cessation approach. As noted above, most clinicians based approaches exist, most providers do not assist patients
fully support addressing tobacco but lack the education, with quitting smoking. However, smoking cessation is the best
time, and resources to implement cessation support.5 method to reduce or eliminate the adverse health effects of
tobacco. Combining practical clinical considerations with
CONCLUSION evidence-based tobacco cessation methods should result in
Overall, the principles of tobacco use are relatively simple. substantially increased access to cessation support and ulti-
Tobacco causes adverse health effects and continued mately result in improved patient outcomes.

References
1. U.S. Department of Health and Human Services. Tobacco. www. 13. National Comprehensive Cancer Center. NCCN Clinical Practice
surgeongeneral.gov/priorities/tobacco/. Accessed February 1, 2016. Guidelines in Oncology (NCCN Guidelines): Lung Cancer Screening.
2. U.S. Department of Health and Human Services. The Health Consequences www.nccn.org/professionals/physician_gls/pdf/lung_screening.pdf.
of Smoking50 Years of Progress: A Report of the Surgeon General. Accessed February 1, 2016.
Atlanta, GA: U.S. Department of Health and Human Services, Centers for 14. National Comprehensive Cancer Center. NCCN Clinical Practice Guidelines
Disease Control and Prevention, National Center for Chronic Disease in Oncology (NCCN Guidelines): Smoking Cessation. www.nccn.org/
Prevention and Health Promotion, Office on Smoking and Health; 2014. professionals/physician_gls/pdf/smoking.pdf. Accessed February 1, 2016.
3. Warren GW, Marshall JR, Cummings KM, et al. Addressing tobacco use 15. Tanner NT, Kanodra NM, Gebregziabher M, et al. The association be-
in patients with cancer: a survey of American Society of Clinical On- tween smoking abstinence and mortality in the National Lung Screening
cology members. J Oncol Pract. 2013;9:258-262. Trial. Am J Respir Crit Care Med. Epub 2015 Oct 26.
4. Warren GW, Marshall JR, Cummings KM, et al; IASLC Tobacco Control 16. Black WC, Gareen IF, Soneji SS, et al. Cost-effectiveness of CT screening in
and Smoking Cessation Committee. Practice patterns and perceptions the National Lung Screening Trial. N Engl J Med. 2014;371:1793-1802.
of thoracic oncology providers on tobacco use and cessation in cancer 17. Cornuz J, Gilbert A, Pinget C, et al. Cost-effectiveness of pharmaco-
patients. J Thorac Oncol. 2013;8:543-548. therapies for nicotine dependence in primary care settings: a multi-
5. Warren GW, Dibaj S, Hutson A, et al. Identifying targeted strategies to national comparison. Tob Control. 2006;15:152-159.
improve smoking cessation support for cancer patients. J Thorac Oncol. 18. Goffin JR, Flanagan WM, Miller AB, et al. Cost-effectiveness of lung
2015;10:1532-1537. cancer screening in Canada. JAMA Oncol. 2015;1:807-813.
6. American Cancer Society. The Tobacco Atlas. www.cancer.org/aboutus/ 19. Sobus SL, Warren GW. The biologic effects of cigarette smoke on cancer
globalhealth/cancerandtobaccocontrolresources/the-tobacco-atlas. cells. Cancer. 2014;120:3617-3626.
Accessed February 1, 2016. 20. Tammemagi MC, Berg CD, Riley TL, et al. Impact of lung cancer screening
7. Warren GW, Cummings KM. Tobacco and lung cancer: risks, trends, and results on smoking cessation. J Natl Cancer Inst. 2014;106:dju084.
outcomes in patients with cancer. Am Soc Clin Oncol Educ Book. 2013; 21. Yousaf-Khan U, Horeweg N, van der Aalst C, et al. Baseline charac-
33:359-364. teristics and mortality outcomes of control group participants and
8. Thun MJ, Carter BD, Feskanich D, et al. 50-year trends in smoking- eligible non-responders in the NELSON lung cancer screening study.
related mortality in the United States. N Engl J Med. 2013;368:351-364. J Thorac Oncol. 2015;10:747-753.
9. Brandon TH, Goniewicz ML, Hanna NH, et al. Electronic nicotine delivery 22. Patel D, Akporobaro A, Chinyanganya N, et al; Lung-SEARCH In-
systems: a policy statement from the American Association for Cancer vestigators. Attitudes to participation in a lung cancer screening trial:
Research and the American Society of Clinical Oncology. J Clin Oncol. a qualitative study. Thorax. 2012;67:418-425.
2015;33:952-963. 23. Ostroff JS, Buckshee N, Mancuso CA, et al. Smoking cessation following CT
10. Goldstraw P, Crowley J, Chansky K, et al; International Association for the screening for early detection of lung cancer. Prev Med. 2001;33:613-621.
Study of Lung Cancer International Staging Committee; Participating In- 24. Park ER, Streck JM, Gareen IF, et al. A qualitative study of lung cancer
stitutions. The IASLC Lung Cancer Staging Project: proposals for the risk perceptions and smoking beliefs among national lung screening trial
revision of the TNM stage groupings in the forthcoming (seventh) participants. Nicotine Tob Res. 2014;16:166-173.
edition of the TNM Classification of malignant tumours. J Thorac 25. Poghosyan H, Kennedy Sheldon L, Cooley ME. The impact of computed
Oncol. 2007;2:706-714. tomography screening for lung cancer on smoking behaviors:
11. Humphrey LL, Teutsch S, Johnson M; U.S. Preventive Services Task a teachable moment? Cancer Nurs. 2012;35:466-475.
Force. Lung cancer screening with sputum cytologic examination, chest 26. Ashraf H, Saghir Z, Dirksen A, et al. Smoking habits in the randomised
radiography, and computed tomography: an update for the U.S. Pre- Danish Lung Cancer Screening Trial with low-dose CT: final results
ventive Services Task Force. Ann Intern Med. 2004;140:740-753. after a 5-year screening programme. Thorax. 2014;69:574-579.
12. Aberle DR, Adams AM, Berg CD, et al; National Lung Screening Trial 27. Anderson CM, Yip R, Henschke CI, et al. Smoking cessation and relapse
Research Team. Reduced lung-cancer mortality with low-dose com- during a lung cancer screening program. Cancer Epidemiol Biomarkers
puted tomographic screening. N Engl J Med. 2011;365:395-409. Prev. 2009;18:3476-3483.

228 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


ADDRESSING TOBACCO WITH CANCER SCREENING AND TREATMENT

28. Park ER, Gareen IF, Japuntich S, et al. Primary care provider-delivered smoking 34. American Society of Clinical Oncology. Tobacco cessation and control
cessation interventions and smoking cessation among participants in the resources. www.asco.org/practice-research/tobacco-cessation-and-
National Lung Screening Trial. JAMA Intern Med. 2015;175:1509-1516. control-resources. Accessed February 1, 2016.
29. Warren GW, Sobus S, Gritz ER. The biological and clinical effects of 35. Ostroff JS, Copeland A, Borderud SP, et al. Readiness of lung cancer
smoking by patients with cancer and strategies to implement evidence- screening sites to deliver smoking cessation treatment: current prac-
based tobacco cessation support. Lancet Oncol. 2014;15:e568-e580. tices, organizational priority, and perceived barriers. Nicotine Tob Res.
30. Gritz ER, Toll BA, Warren GW. Tobacco use in the oncology setting: advancing Epub 2015 Sep 7.
clinical practice and research. Cancer Epidemiol Biomarkers Prev. 2014;23:3-9. 36. Warren GW, Marshall JR, Cummings KM, et al. Automated tobacco
31. Warren GW, Ward KD. Integration of tobacco cessation services into assessment and cessation support for cancer patients. Cancer. 2014;
multidisciplinary lung cancer care: rationale, state of the art, and future 120:562-569.
directions. Transl Lung Cancer Res. 2015;4:339-352. 37. Tang MW, Oakley R, Dale C, et al. A surgeon led smoking cessation
32. Karam-Hage M, Cinciripini PM, Gritz ER. Tobacco use and cessation for cancer intervention in a head and neck cancer centre. BMC Health Serv Res.
survivors: an overview for clinicians. CA Cancer J Clin. 2014;64:272-290. 2014;14:636.
33. Fiore MC, Jaen CR, Baker TB, et al. Treating Tobacco Use and De- 38. Dobson Amato KA, Hyland A, Reed R, et al. Tobacco cessation may
pendence: 2008 Update. Clinical Practice Guideline. Rockville, MD: U.S. improve lung cancer patient survival. J Thorac Oncol. 2015;10:
Public Health Service; 2008. 1014-1019.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 229


LUNG CANCER

Lung Cancer, Small Cell Lung


Cancer: On the Move (Again?)

CHAIR
Walter J. Curran Jr., MD
Winship Cancer Institute
Atlanta, GA

SPEAKERS
Solange Peters, MD, PhD
Lausanne University
Lausanne, Switzerland

Maria Catherine Pietanza, MD


Memorial Sloan Kettering Cancer Center
New York, NY
NEW APPROACHES TO PATIENTS WITH SMALL CELL LUNG CANCER

Seeking New Approaches to Patients With Small Cell Lung


Cancer
Marie Catherine Pietanza, MD, Stefan Zimmerman, MD, Solange Peters, MD, PhD, and Walter J. Curran Jr., MD

OVERVIEW

The fundamental approach to the treatment of small cell lung cancer (SCLC) has not changed in the last several decades, with
most advances being restricted to improved radiation approaches. The standard first-line chemotherapy regimen in the
United States and Europe remains cisplatin or carboplatin plus etoposide in the treatment of limited stage (LS-SCLC) and
extensive stage (ES-SCLC) disease. Radiation therapy is administered to those patients with LS-SCLC, whose cancer is
confined to the chest in a single tolerable radiation field. This article will summarize a number of exciting observations
regarding the biology of SCLC and how a deeper understanding of newly integrated targets and target pathways may lead to
new and better therapeutic approaches in the near future.

T he fundamental approach to the treatment of SCLC has


not changed in the last several decades, with most ad-
vances being restricted to improved radiation approaches.
patients are defined as having sensitive or refractory disease.
Those patients who maintain a response to initial treatment
of 3 months or longer are defined as having sensitive disease;
The standard first-line chemotherapy regimen in the United additional chemotherapy yields response rates of 25% and
States and Europe remains cisplatin or carboplatin plus median survival from the time of relapse of approximately
etoposide in the treatment of LS-SCLC and ES-SCLC disease. 6 months. Patients with refractory disease are those who
Radiation therapy is administered to those patients with either have no response to initial therapy or progress within
LS-SCLC, whose cancer is confined to the chest in a single 3 months; these patients rarely benefit from additional
tolerable radiation field: data support initiation of radiation treatment, with response rates less than 10% and median
during cycle one or two and use of hyperfractionated radi- survival of 4 months.
ation therapy,1-8 though there is an ongoing large national Only one agent has been approved by the U.S. Food and
cooperative group study addressing the question of stan- Drug Administration for recurrent or progressive SCLC:
dard hyperfractionation versus a higher total dose radiation topotecan.13-15 Unfortunately, for patients with SCLC whose
using modern radiation techniques (NCT00632853). More disease has progressed after first- and second-line treat-
recently, thoracic radiation therapy is being extended to ments, there are no accepted regimens. Although inves-
patients with ES-SCLC after response to chemotherapy, as a tigators have studied a wide range of rationally based
phase III trial showed considerable reduction in intratho- therapies in the hopes of improving outcomes, results have
racic recurrence and in 2-year overall survival.9 Prophy- not been favorable (reviewed in Pietanza et al16).
lactic cranial irradiation is recommended to patients with However, more recent insights generated from genomic
LS-SCLC and ES-SCLC demonstrating a response to front- and proteomic studies, as well as pathway-specific investi-
line platinum-based therapy, which has been shown to gations, as indicated above, may lead to the development of
decrease the risk of intracranial recurrence and improve effective therapies, which are critically needed in SCLC.
overall survival.10,11
Although the response rates to first-line treatment are as
high as 60%80% in SCLC, cure occurs only in approximately POTENTIALLY TARGETABLE SCLC MOLECULAR
20% of patients, restricted to those with LS-SCLC.12 Essen- ALTERATIONS
tially all patients with ES-SCLC, and the majority of patients The MYC gene family members of transcription factors,
with LS-SCLC, suffer relapse within months of completing which are contributors to oncogenesis, have been described
initial therapy. Depending on the length of time a response in approximately 20% of SCLCs.17 As stated above, previous
is maintained, which is the strongest predictor of outcome, efforts to inhibit MYC activity proved to be difficult; however,

From the Merck Research Laboratories, Rahway, NJ; Department of Oncology, HFR Fribourg, Fribourg, Switzerland; Department of Oncology, Centre Hospitalier Universitaire Vaudois,
Lausanne, Switzerland; Winship Cancer Institute of Emory University, Atlanta, GA.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Walter J. Curran Jr., MD, Winship Cancer Institute of Emory University, 1365 Clifton Rd. NE, Room A1356, Atlanta, GA 30322; email: curran04@gmail.com.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e477


PIETANZA ET AL

further understanding of the biology of the disease has 42756493 (a pan-FGFR inhibitor; NCT01703481),30 BIBF1120
revealed specific mechanisms by which MYC can be po- (a multitargeted drug that inhibits FGFR, vascular endo-
tentially targeted. MYC is a transcriptional regulator of thelial growth factor receptor, and platelet-derived growth
aurora kinases A and B, which, in the absence of p53, factor receptor; NCT02152059); and lucitinib (a potent
provide a growth advantage,18-21 and as such, favorable inhibitor of FGFR1/2, vascular endothelial growth factor
antitumor activity may be demonstrated by inhibiting aurora receptor 13, and platelet-derived growth factor receptor
kinases.22 Multiple preclinical studies have suggested that a/b; NCT02109016).
SCLCs with MYC alterations are sensitive to aurora kinase
inhibitors.18,22,23 Alisertib, a selective aurora kinase A
inhibitor, has been evaluated in a phase II clinical trial of PROBING THE EPIGENOME
patients with recurrent or progressive SCLC and demon- Epigenetic processes, such as gene promoter methylation
strated a response rate of 21% among patients with both and histone acetylation, are known to be dysregulated in
sensitive and refractory disease.24 To augment these results, SCLC and lead to alterations in chromatin and other asso-
there is an ongoing clinical trial evaluating paclitaxel with or ciated factors that modify the ability of genes to be tran-
without alisertib for the second-line treatment of patients scribed.31 Acetyltransferases and histone deacetylases
with SCLC (NCT02038647), as aurora kinase A has a key role in regulate histone acetylation, which leads to increased ac-
mitotic spindle assembly. cessibility of promoter regions and increased transcription
Inhibiting key regulators of MYC transcription may be of genes.32,33 Vorinostat and belinostat, which are histone
another potential mechanism in SCLC. MYC transcription in deacetylase inhibitors, have synergistic activity when added
SCLC and other tumors is regulated by the bromodomain and to topotecan and cisplatin/etoposide, respectively.34,35
extraterminal (BET) family of bromodomain-containing Clinical trials investigating the combination of vorinostat
proteins, which bind acetylated lysines on histone tails (NCT00702962) and belinostat (NCT00926640) with plati-
and recruit key transcriptional proteins.25,26 Inhibiting BET num and etoposide in the first-line treatment of patients
has been shown to prevent interaction of BET proteins with with ES-SCLC are in progress.
acetylated histones and halt assembly of an active gene
transcriptional complex, leading to focal chromatin re-
modeling and decreased inhibition of c-Myc. GSK525762 is a DNA REPAIR
small-molecule inhibitor of BET being evaluated in a phase I SCLC has been characterized by aberrant expression of
clinical trial that includes patients with SCLC (NCT01587703). DNA repair proteins, including O6 alkyl-guanine, DNA
The genomic studies completed in SCLC thus far have alkyltransferase (MGMT), poly (ADP-ribose) polymerase-1
found amplification of fibroblast growth factor receptor (PARP-1), checkpoint kinase-1 (Chk-1), BRCA-1, and RAD51.36,37
(FGFR) 1 in approximately 6% of cases,27-29 and sensitivity to Therefore, multiple DNA repair pathways represent attractive
FGFR inhibitors has been described in some, but not all, SCLC targets in SCLC.
tumors.22 Although the magnitude to which this subset of Temozolomide is an oral alkylating agent with improved
SCLC is dependent on the FGFR pathway remains unknown, outcomes in the presence of MGMT promoter hyper-
there are clinical studies evaluating drugs targeting the methylation.38,39 In a phase II clinical trial of patients with
FGFR family members for patients with SCLC: including JNJ relapsed sensitive or refractory SCLC, temozolomide dem-
onstrated an overall response rate of 20%, and responses
were noted in patients in need of third-line treatment and in
those with brain metastases.39 These findings led to the
KEY POINTS addition of temozolomide to compendia of agents recom-
mended for use in the treatment of SCLC.40
SCLC is an aggressive malignancy, uniformly linked to PARP activity is essential for the repair of single-stranded
tobacco use, and has a unique biology. DNA breaks through the base excision repair pathway.41,42 In
The fundamental approach to the treatment of SCLC has preclinical SCLC models, PARP inhibitors have demonstrated
not changed in nearly 4 decades and incorporates use of activity whether evaluated alone or in combination with
a platinum doublet and, when possible, radiation DNA-damaging agents, including temozolomide.37,43-46
therapy. Talazoparib (BMN673), a PARP inhibitor that both mediates
As the biology of this disease is elucidated, multiple catalytic PARP1 inhibition and traps PARP1 and PARP2 en-
targets are being identified and new trials have been zymes at damaged DNA sites,47 has shown single-agent activity
developed.
in patients with sensitive relapsed SCLC, with a 10% overall
The MYC gene family of transcription factors may be
a targetable entity to test new therapies against, in
response rate and 25% clinical benefit response rate.48 The
particular, aurora kinase inhibitors. PARP inhibitors, talazoparib, veliparib, and olaparib, are being
Epigentic processes, such as gene promoter methylation evaluated in combination with etoposide/platinum or temo-
and histone acetylation, may be candidate targets to zolomide for patients with newly diagnosed or relapsed SCLC,
test in SCLC. respectively (NCT01286987, NCT01642251, NCT02289690,
NCT01638546, NCT02446704, and NCT02049593). Phase I

e478 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


NEW APPROACHES TO PATIENTS WITH SMALL CELL LUNG CANCER

data reveal that veliparib can safely be administered with response rate of 23% in those evaluable.62 Notably, among
etoposide and platinum in patients with untreated SCLC.49 the 27 patients with confirmed DLL3-high tumor expression,
Among the multiple functions of Chk-1 are activation of the response rates were 44% and 45% in the second- and
S and G2/M phase checkpoints, preventing entry into mi- third-line settings, respectively, which were durable.62
tosis, and coordination of homologous repair.50,51 Additional A phase II trial evaluating rovalpituzumab tesirine for third-
proteins necessary at the G2/M checkpoint include ataxia line treatment of patients with SCLC with DLL3-expressing
telangiectasia and Rad3-related (ATR) and Wee-1. Inhibition tumors (TRINITY; NCT02674568) has begun accrual and, if
of Chk-1, ATR, or Wee-1 leads to G2 checkpoint abrogation positive, will fulfill an unmet need, as there are no ap-
and, in p53-negative tumors, such as SCLC, sensitizes the proved agents in this setting for this malignancy.
cancer cells to DNA-damaging agents, leading to mitotic ca- A phase II study evaluating sacituzumab govitecan
tastrophe and cell death.50 The Chk-1 inhibitors LY2606368 (IMMU-132), an antibody-drug conjugate comprising SN-38,
and AZD7762 have demonstrated activity when administered the active metabolite of the topoisomerase I inhibitor,
alone or with olaparib and cisplatin in preclinical SCLC irinotecan, conjugated to an antiTrop-2 humanized anti-
models.50,52,53 Numerous inhibitors of Chk-1 (LY2606368 and body has been enrolling patients with previously treated
AZD7762), ATR (AZD6738, VX970, and Wee-1; AZD1775) are in SCLC and non-SCLC (NCT01631552). Interim results have
clinical development, and two phase I trials are actively shown a response rate of 30% in 20 evaluable patients with
recruiting patients with SCLC: topotecan with VX970 in SCLC who had received a median of 2.5 prior lines of therapy
ES-SCLC (NCT02487095) and AZD1775 with olaparib in re- and median progression-free survival of 2.4 months.63 As
fractory solid tumors (NCT02511795). Trop-2 is a human trophoblast cell-surface glycoprotein
overexpressed in multiple epithelial tumors, including SCLC,
this antibody drug conjugate is being studied in other ma-
THE NOTCH DEVELOPMENTAL PATHWAY lignancies as well.
The Notch pathway is essential in early lung development
and regulates stem cell self-renewal; thus, when abnormally
activated, it can cause neoplastic proliferation, representing
an early event in tumorigenesis.54-56 Recent comprehensive
IMMUNOTHERAPY
Modulating the immune response may represent another
genomic analyses have found that the NOTCH family genes
treatment modality for patients with SCLC, as supported by
are mutated in 25% of human SCLC. Thus, this pathway is a
clinical and preclinical data indicated above (and reviewed
potential target in SCLC. Importantly, however, Notch sig-
in Pietanza et al64): the disease is associated with immu-
naling can have oncogenic or tumor-suppressive effects
nogenic effects; PD-L1 expression on SCLC tumors corre-
depending on the cellular content and can influence multiple
lates with the presence of tumor-infiltrating lymphocytes;
other oncogenic pathways.57
and the malignancy is nearly entirely related to smoking
Overexpression of Notch2 and Notch3 receptors and
and characterized by an elevated mutation burden, which
target genes has been noted in SCLC. In preclinical SCLC
are known to be markers of response to immune check-
models, tarextumab (OMP-59R5), which is a fully human
point inhibitors.65-71
monoclonal antibody that selectively inhibits Notch2 and
Patients with untreated stage IIIB/IV NSCLC or ES-SCLC
Notch3 receptor function, has demonstrated delayed tu-
were administered paclitaxel and carboplatin alone or with
mor recurrence following the discontinuation of chemo-
the addition of ipilimumab, a humanized IgG1 monoclonal
therapy, as well as a reduction in cancer stem cell frequency
antibody against CTLA-4, given on two dosing schedules, in a
and tumorigenicity.58 A phase I study of tarextumab with
randomized, double-blind, three-arm phase II trial.72 Based
etoposide/platinum in patients with ES-SCLC has been
on the results from the patient with SCLC cohort that re-
completed,59 and a randomized phase II study is ongoing
ceived the phased dosing schedule of ipilimumab, admin-
(NCT01859741).
istered with cycle three of paclitaxel and carboplatin, who
appeared to have improved immune-related progression-
ANTIBODY-DRUG CONJUGATES free survival compared with those treated in the other two
Antibody-drug conjugates, composed of an antibody di- arms,73 a randomized, multicenter, double-blind phase III
rected to a well-characterized antigen on cancer cells, a trial comparing the efficacy of platinum/etoposide with
linker, and cytotoxic agent (payload), represent an ef- or without ipilimumab in patients with newly diagnosed
fective mechanism of targeted drug delivery, resulting in ES-SCLC, with OS as the primary endpoint, has completed
decreased toxicity and improved therapeutic index.60 accrual, and results are anticipated (NCT01450761).
Rovalpituzumab tesirine exploits the Notch pathway as the Nivolumab, with and without ipilimumab, has been
humanized monoclonal antibody portion targets the cell evaluated in heavily pretreated patients with relapsed
surface available Notch ligand delta-like protein 3 (DLL3), SCLC as part of a phase I trial evaluating the agents in
which is overexpressed in SCLC tumor-initiating cells but various tumor types (NCT01928394). Early results indi-
not in normal tissue.61 The phase I trial of rovalpituzumab cate response rates of 13% and 31%, when nivolumab
tesirine enrolled 73 recurrent patients with SCLC in need is administered alone or with ipilimumab, respectively,
of second- or third-line treatment and demonstrated a regardless of PD-L1 tumor expression, previous lines of

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e479


PIETANZA ET AL

therapy, and platinum sensitivity.74 Notably, the re- Finally, durvalumab (MEDI4736, the humanized IgG1k
sponses are durable, with 1-year overall survival of 27% monoclonal antibody directed against PD-L1) is being eval-
and 48% for patients treated with nivolumab and nivo- uated alone (NCT01693562) and with tremelimumab, a hu-
lumab with ipilumumab, respectively.74 These results manized IgG2 monoclonal CTLA-4 antibody (NCT02658214)
have led to the development of two phase III studies among patients with relapsed SCLC.
among patients with SCLC evaluating nivolumab, nivo- As seen, the immune system is a reasonable target in SCLC,
lumab with ipilimumab, or placebo (NCT02538666) in the with promising results from early-phase trials. Currently,
maintenance setting, in which there is no defined treat- multiple approaches are being developed to use immune
ment modality, and nivolumab compared with chemo- checkpoint inhibitors in an attempt to best exploit this
therapy for those in need of second-line treatment treatment modality in SCLC.
(NCT02481830).
As part of a phase IB multicohort study, pembrolizumab
has been evaluated among patients with relapsed SCLC with CONCLUSION
PD-L1positive tumors (NCT02054806). The response rate SCLC has a unique biology, driven by multiple aberrant
was 29%, with a median duration of response of 29 weeks.75 pathways and mutations, and distinct clinical features. The
There are several ongoing trials of pembrolizumab in patients mechanisms that lead to the shift from initial therapeutic
with SCLC, including a phase II study of the agent as main- sensitivity to ultimate therapeutic resistance are not well
tenance therapy after the completion of standard, first-line understood. Recent genomic, proteomic, and preclinical
chemotherapy in extensive-stage disease (NCT02359019), studies have identified novel therapeutic strategies cur-
a phase I trial evaluating pembrolizumab with concurrent rently being evaluated in clinical trials. Ongoing efforts to
chemoradiation therapy (NCT02402920), and others in- collect and analyze samples in the setting of these studies,
vestigating the drug alone (NCT02628067), with various with use of improved research tools, will allow us to continue
chemotherapy agents (NCT02551432, NCT02580994, and to develop rational clinical trials with the potential to ad-
NCT02331251), or with novel biologics (NCT02661100). vance the field.

References
1. Amini A, Byers LA, Welsh JW, et al. Progress in the management of 10. Auperin A, Arriagada R, Pignon JP, et al; Prophylactic Cranial Irradiation
limited-stage small cell lung cancer. Cancer. 2014;120:790-798. Overview Collaborative Group. Prophylactic cranial irradiation for pa-
2. Spiro SG, James LE, Rudd RM, et al; London Lung Cancer Group. Early tients with small-cell lung cancer in complete remission. N Engl J Med.
compared with late radiotherapy in combined modality treatment for 1999;341:476-484.
limited disease small-cell lung cancer: a London Lung Cancer Group 11. Slotman B, Faivre-Finn C, Kramer G, et al; EORTC Radiation Oncology
multicenter randomized clinical trial and meta-analysis. J Clin Oncol. Group and Lung Cancer Group. Prophylactic cranial irradiation in ex-
2006;24:3823-3830. tensive small-cell lung cancer. N Engl J Med. 2007;357:664-672.
3. Fried DB, Morris DE, Poole C, et al. Systematic review evaluating the 12. Simon GR, Turrisi A. American College of Chest P. Management of small
timing of thoracic radiation therapy in combined modality therapy for cell lung cancer: ACCP evidence-based clinical practice guidelines (2nd
limited-stage small-cell lung cancer. J Clin Oncol. 2004;22:4837-4845. edition). Chest. 2007;132:324S-339S (suppl 3).
4. De Ruysscher D, Pijls-Johannesma M, Bentzen SM, et al. Time between 13. von Pawel J, Schiller JH, Shepherd FA, et al. Topotecan versus cyclo-
the first day of chemotherapy and the last day of chest radiation is the phosphamide, doxorubicin, and vincristine for the treatment of recurrent
most important predictor of survival in limited-disease small-cell lung small-cell lung cancer. J Clin Oncol. 1999;17:658-667.
cancer. J Clin Oncol. 2006;24:1057-1063. 14. OBrien ME, Ciuleanu TE, Tsekov H, et al. Phase III trial comparing
5. De Ruysscher D, Pijls-Johannesma M, Vansteenkiste J, et al. Systematic supportive care alone with supportive care with oral topotecan in
review and meta-analysis of randomised, controlled trials of the timing patients with relapsed small-cell lung cancer. J Clin Oncol. 2006;24:
of chest radiotherapy in patients with limited-stage, small-cell lung 5441-5447.
cancer. Ann Oncol. 2006;17:543-552. 15. Eckardt JR, von Pawel J, Pujol JL, et al. Phase III study of oral compared
6. Pijls-Johannesma MC, De Ruysscher D, Lambin P, et al. Early versus late with intravenous topotecan as second-line therapy in small-cell lung
chest radiotherapy for limited stage small cell lung cancer. Cochrane cancer. J Clin Oncol. 2007;25:2086-2092.
Database Syst Rev. 2005;1:CD004700. 16. Pietanza MC, Krug LM, Wu A, et al. Small cell and neuroendocrine
7. Huncharek M, McGarry R. A meta-analysis of the timing of chest ir- tumors of the lung. In DeVita VT, Lawrence TS, Rosenberg SA (eds).
radiation in the combined modality treatment of limited-stage small cell DeVita, Hellman, and Rosenbergs Cancer: Principles and Practice of
lung cancer. Oncologist. 2004;9:665-672. Oncology. 10th ed. Philadelphia: Lippincott, Williams, and Wilkins;
8. Turrisi AT III, Kim K, Blum R, et al. Twice-daily compared with once- 2015;536-559.
daily thoracic radiotherapy in limited small-cell lung cancer treated 17. Hwang DH, Sun H, Rodig SJ, et al. Myc protein expression correlates with
concurrently with cisplatin and etoposide. N Engl J Med. 1999;340: MYC amplification in small-cell lung carcinoma. Histopathology. 2015;
265-271. 67:81-89.
9. Slotman BJ, van Tinteren H, Praag JO, et al. Use of thoracic radiotherapy 18. Brockmann M, Poon E, Berry T, et al. Small molecule inhibitors of
for extensive stage small-cell lung cancer: a phase 3 randomised aurora-a induce proteasomal degradation of N-myc in childhood
controlled trial. Lancet. 2015;385:36-42. neuroblastoma. Cancer Cell. 2013;24:75-89.

e480 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


NEW APPROACHES TO PATIENTS WITH SMALL CELL LUNG CANCER

19. den Hollander J, Rimpi S, Doherty JR, et al. Aurora kinases A and B are 41. Schreiber V, Ame JC, Dolle P, et al. Poly(ADP-ribose) polymerase-2
up-regulated by Myc and are essential for maintenance of the malig- (PARP-2) is required for efficient base excision DNA repair in association
nant state. Blood. 2010;116:1498-1505. with PARP-1 and XRCC1. J Biol Chem. 2002;277:23028-23036.
20. Lu LY, Wood JL, Ye L, et al. Aurora A is essential for early embry- 42. Memisoglu A, Samson L. Base excision repair in yeast and mammals.
onic development and tumor suppression. J Biol Chem. 2008;283: Mutat Res. 2000;451:39-51.
31785-31790. 43. Tamborini E, Bonadiman L, Negri T, et al. Detection of overexpressed
21. Vader G, Lens SM. The Aurora kinase family in cell division and cancer. and phosphorylated wild-type kit receptor in surgical specimens of
Biochim Biophys Acta. 2008;1786:60-72. small cell lung cancer. Clin Cancer Res. 2004;10:8214-8219.
22. Sos ML, Dietlein F, Peifer M, et al. A framework for identification of 44. Palma JP, Wang YC, Rodriguez LE, et al. ABT-888 confers broad in vivo
actionable cancer genome dependencies in small cell lung cancer. Proc activity in combination with temozolomide in diverse tumors. Clin
Natl Acad Sci USA. 2012;109:17034-17039. Cancer Res. 2009;15:7277-7290.
23. Hook KE, Garza SJ, Lira ME, et al. An integrated genomic approach to 45. Cardnell RJ, Feng Y, Diao L, et al. Proteomic markers of DNA repair and
identify predictive biomarkers of response to the aurora kinase inhibitor PI3K pathway activation predict response to the PARP inhibitor BMN
PF-03814735. Mol Cancer Ther. 2012;11:710-719. 673 in small cell lung cancer. Clin Cancer Res. 2013;19:6322-6328.
24. Melichar B, Adenis A, Lockhart AC, et al. Safety and activity of alisertib, 46. Feng Y, Post LE, Cardnell R, et al. BMN 673 as single agent and in
an investigational aurora kinase A inhibitor, in patients with breast combination with temozolomide or PI3K pathway inhibitors in small cell
cancer, small-cell lung cancer, non-small-cell lung cancer, head and neck lung cancer and gastric cancer models. Eur J Cancer. 2014;50:81.
squamous-cell carcinoma, and gastro-oesophageal adenocarcinoma: 47. Murai J, Huang SY, Das BB, et al. Trapping of PARP1 and PARP2 by
a five-arm phase 2 study. Lancet Oncol. 2015;16:395-405. Clinical PARP Inhibitors. Cancer Res. 2012;72:5588-5599.
25. Helin K, Dhanak D. Chromatin proteins and modifications as drug 48. Wainberg Z, Rafii S, Ramanathan R. Safety and antitumor activity of the
targets. Nature. 2013;502:480-488. PARP inhibitor BMN673 in a phase 1 trial recruiting metastatic small-cell
26. Delmore JE, Issa GC, Lemieux ME, et al. BET bromodomain inhibition as a lung cancer (SCLC) and germline BRCA-mutation carrier cancer patients.
therapeutic strategy to target c-Myc. Cell. 2011;146:904-917. J Clin Oncol. 2014;32:15s (suppl; abstr 7522).
27. Rudin CM, Durinck S, Stawiski EW, et al. Comprehensive genomic 49. Owonikoko TK, Dahlberg SE, Khan SA, et al. A phase 1 safety study of
analysis identifies SOX2 as a frequently amplified gene in small-cell lung veliparib combined with cisplatin and etoposide in extensive stage small
cancer. Nat Genet. 2012;44:1111-1116. cell lung cancer: A trial of the ECOG-ACRIN Cancer Research Group
28. Peifer M, Fernandez-Cuesta L, Sos ML, et al. Integrative genome an- (E2511). Lung Cancer. 2015;89:66-70.
alyses identify key somatic driver mutations of small-cell lung cancer. 50. Syljuasen RG, Hasvold G, Hauge S, et al. Targeting lung cancer through
Nat Genet. 2012;44:1104-1110. inhibition of checkpoint kinases. Front Genet. 2015;6:70.
29. George J, Lim JS, Jang SJ, et al. Comprehensive genomic profiles of small 51. Maugeri-Sacc`a M, Bartucci M, De Maria R. Checkpoint kinase 1 in-
cell lung cancer. Nature. 2015;524:47-53. hibitors for potentiating systemic anticancer therapy. Cancer Treat Rev.
30. Tabernero J, Bahleda R, Dienstmann R, et al. Phase I dose-escalation 2013;39:525-533.
study of JNJ-42756493, an oral pan-fibroblast growth factor receptor 52. Valliani AA, Sen T, Masrorpour F, et al. Check point kinase 1 targeting
inhibitor, in patients with advanced solid tumors. J Clin Oncol. 2015;33: as a novel therapeutic strategy in small cell lung cancer. Cancer Res.
3401-3408. 2015;75 (suppl 15; abstr 5316).
31. Jones PA, Baylin SB. The epigenomics of cancer. Cell. 2007;128:683-692. 53. Hsu W-H, Hsu S-T, Rao G, et al. Chk1 Inhibition Enhances Cisplatin
32. Gregory PD, Wagner K, Horz W. Histone acetylation and chromatin Cytotoxicity Regardless of p53 Status in Human Small Cell Lung Cancer
remodeling. Exp Cell Res. 2001;265:195-202. Cells. J Thorac Oncol. 2015;10:S399 (suppl 2).
33. Lane AA, Chabner BA. Histone deacetylase inhibitors in cancer therapy. 54. Peacock CD, Watkins DN. Cancer stem cells and the ontogeny of lung
J Clin Oncol. 2009;27:5459-5468. cancer. J Clin Oncol. 2008;26:2883-2889.
34. Bruzzese F, Rocco M, Castelli S, et al. Synergistic antitumor effect 55. Pardal R, Clarke MF, Morrison SJ. Applying the principles of stem-cell
between vorinostat and topotecan in small cell lung cancer cells is biology to cancer. Nat Rev Cancer. 2003;3:895-902.
mediated by generation of reactive oxygen species and DNA damage- 56. Rubin LL, de Sauvage FJ. Targeting the Hedgehog pathway in cancer. Nat
induced apoptosis. Mol Cancer Ther. 2009;8:3075-3087. Rev Drug Discov. 2006;5:1026-1033.
35. Luchenko VL, Salcido CD, Zhang Y, et al. Schedule-dependent synergy of 57. Takebe N, Nguyen D, Yang SX. Targeting notch signaling pathway in
histone deacetylase inhibitors with DNA damaging agents in small cell cancer: clinical development advances and challenges. Pharmacol Ther.
lung cancer. Cell Cycle. 2011;10:3119-3128. 2014;141:140-149.
36. Toyooka S, Toyooka KO, Maruyama R, et al. DNA methylation profiles of 58. Yen W-C, Fischer MM, Axelrod F, et al. Targeting Notch signaling with a
lung tumors. Mol Cancer Ther. 2001;1:61-67. Notch2/Notch3 antagonist (tarextumab) inhibits tumor growth and
37. Byers LA, Wang J, Nilsson MB, et al. Proteomic profiling identifies decreases tumor-initiating cell frequency. Clin Cancer Res. 2015;21:
dysregulated pathways in small cell lung cancer and novel therapeutic 2084-2095.
targets including PARP1. Cancer Discov. 2012;2:798-811. 59. Pietanza MC, Spira AI, Jotte RM, et al. Final results of phase Ib of
38. Hegi ME, Diserens AC, Gorlia T, et al. MGMT gene silencing and tarextumab (TRXT, OMP-59R5, anti-Notch2/3) in combination with
benefit from temozolomide in glioblastoma. N Engl J Med. 2005;352: etoposide and platinum in patients with untreated extensive-stage
997-1003. small-cell lung cancer. J Clin Oncol. 2015;33 (suppl; abstr 7508).
39. Pietanza MC, Kadota K, Huberman K, et al. Phase II trial of temozolomide 60. Zolot RS, Basu S, Million RP. Antibody-drug conjugates. Nat Rev Drug
in patients with relapsed sensitive or refractory small cell lung cancer, Discov. 2013;12:259-260.
with assessment of methylguanine-DNA methyltransferase as a po- 61. Saunders LR, Bankovich AJ, Anderson WC, et al. A DLL3-targeted
tential biomarker. Clin Cancer Res. 2012;18:1138-1145. antibody-drug conjugate eradicates high-grade pulmonary neuroen-
40. Kalemkerian GP, Loo BW, Akerley W, et al. NCCN Guidelines Version 1.2016 docrine tumor-initiating cells in vivo. Sci Transl Med. 2015;7:302ra136.
Small Cell Lung Cancer. http://www.nccn.org/professionals/physician_gls/ 62. Pietanza MC, Spigel D, Bauer TM, et al. Safety, activity, and response
recently_updated.asp. Accessed April 7, 2016. durability assessment of single agent rovalpituzumab tesirine, a

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e481


PIETANZA ET AL

delta-like protein 3-targeted antibody drug conjugate, in small cell lung 70. Herbst RS, Soria JC, Kowanetz M, et al. Predictive correlates of response
cancer. Eur J Cancer. 2015;51:S712 (suppl 3). to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature.
63. Camidge DR, Starodub AN, Ocean A, et al. Therapy of advanced met- 2014;515:563-567.
astatic lung cancers with an anti-Trop-2-SN-38 antibody-drug conju- 71. Rizvi NA, Hellmann MD, Snyder A, et al. Cancer immunology. Mutational
gate, IMMU-132: interim phase II clinical results. J Thorac Oncol. 2015; landscape determines sensitivity to PD-1 blockade in non-small cell lung
10:S173 (suppl 2). cancer. Science. 2015;348:124-128.
64. Pietanza MC, Byers LA, Minna JD, et al. Small cell lung cancer: will recent 72. Lynch TJ, Bondarenko I, Luft A, et al. Ipilimumab in combination with
progress lead to improved outcomes? Clin Cancer Res. 2015;21: paclitaxel and carboplatin as first-line treatment in stage IIIB/IV non-
2244-2255. small-cell lung cancer: results from a randomized, double-blind, mul-
65. Graus F, Dalmou J, Ren~e R, et al. Anti-Hu antibodies in patients with ticenter phase II study. J Clin Oncol. 2012;30:2046-2054.
small-cell lung cancer: association with complete response to therapy 73. Reck M, Bondarenko I, Luft A, et al. Ipilimumab in combination with
and improved survival. J Clin Oncol. 1997;15:2866-2872. paclitaxel and carboplatin as first-line therapy in extensive-disease-
66. Schultheis AM, Scheel AH, Ozretic L, et al. PD-L1 expression in small cell small-cell lung cancer: results from a randomized, double-blind, mul-
neuroendocrine carcinomas. Eur J Cancer. 2015;51:421-426. ticenter phase 2 trial. Ann Oncol. 2013;24:75-83.
67. Soria J-C, Marabelle A, Brahmer JR, et al. Immune checkpoint modulation 74. Calvo E, Lopez-Martin JA, Bendell J, et al. Nivolumab monotherapy or in
for non-small cell lung cancer. Clin Cancer Res. 2015;21:2256-2262. combination with ipilimumab for treatment of recurrent small cell lung
68. Hellmann MD, Creelan BC, Woo K, et al. Smoking history and response to cancer. Eur J Oncol. 2015;51:S633 (suppl 3).
nivolumab in patients with advanced NSCLCs. Ann Oncol. 2014;25:iv429. 75. Ott PA, Elez E, Hiret S, et al. Pembrolizumab for extensive stage disease
69. Garon EB, Rizvi NA, Hui R, et al; KEYNOTE-001 Investigators. SCLC: efficacy and relationship with PD-L1 expression abstr 3285.
Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Presented at: 16th World Conference on Lung Cancer; September 2015;
Med. 2015;372:2018-2028. Denver, CO.

e482 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


LUNG CANCER

Treatment of Lung Cancer in


Medically Compromised Patients

CHAIR
Jeffrey Crawford, MD
Duke University Medical Center
Durham, NC

SPEAKERS
Josephine Louella Feliciano, MD
University of Maryland Greenebaum Cancer Center
Baltimore, MD

Paul Wheatley-Price, MBChB, FRCP, MD


Ottawa Hospital Research Institute
Ottawa, ON, Canada
CRAWFORD, WHEATLEY-PRICE, AND FELICIANO

Treatment of Lung Cancer in Medically Compromised Patients


Jeffrey Crawford, MD, Paul Wheatley-Price, MBChB, FRCP, MD, and Josephine Louella Feliciano, MD

OVERVIEW

Outcomes for patients with lung cancer have been improved substantially through the integration of surgery, radiation, and
systemic therapy for patients with early-stage disease. Meanwhile, advances in our understanding of molecular mechanisms
have substantially advanced our treatment of patients with advanced lung cancer through the introduction of targeted
therapies, immune approaches, improvements in chemotherapy, and better supportive care. However, the majority of these
advances have occurred among patients with good functional status, normal organ function, and with the social and
economic support systems to be able to benefit most from these treatments. The aim of this article is to bring greater
attention to management of lung cancer in patients who are medically compromised, which remains a major barrier to care
delivery. Impaired performance status is associated with poor outcomes and correlates with the high prevalence of cachexia
among patients with advanced lung cancer. CT imaging is emerging as a research tool to quantify muscle loss in patients with
cancer, and new therapeutics are on the horizon that may provide important adjunctive therapy in the future. The benefits of
cancer therapy for patients with organ failure are poorly understood because of their exclusion from clinical trials. The
availability of targeted therapy and immunotherapy may provide alternatives that may be easier to deliver in this pop-
ulation, but clinical trials of these new agents in this population are vital. Patients with lower socioeconomic status are
disproportionately affected by lung cancer because of higher rates of tobacco addiction and the impact of socioeconomic
status on delay in diagnosis, treatment, and outcomes. For all patients who are medically compromised with lung cancer,
multidisciplinary approaches are particularly needed to evaluate these patients and to incorporate rapidly changing
therapeutics to improve outcomes.

S ince the pioneering work of Karnofsky, the relationship


between impaired functional status and poor outcome
in patients with lung cancer has been well established.1 For
develop in approximately two-thirds of patients during
treatment. It is also clear that muscle wasting, or sarcopenia,
is common in patients with lung cancer, regardless of body
patients with advanced cancer, a grading system including weight, and may exist even among patients with obesity.
both body mass index and percent weight loss is highly This population of patients with sarcopenic obesity is at
predictive of survival outcomes across multiple tumor types particular high risk of complications, including impaired
including lung cancer. 2 Weight loss per se is the result of functional status.4
the process of cancer cachexia, which has been defined To better understand the impact of muscle wasting on
as a multifactorial syndrome associated with ongoing loss patients with cancer techniques have been developed using
of skeletal muscle mass (with or without fat mass), which standardized CT, which allows quantitative assessment of
cannot be fully reversed through conventional nutritional skeletal muscle and other body tissues.5 By using this
support and leads to progressive functional impairment.3 technique for patients with advanced lung and gastroin-
This definition has been very useful as a framework for testinal cancers, virtually 100% of patients with a body mass
better understanding of cachexia, both at the molecular and index below 20 will have muscle wasting, 30%50% will have
clinical levels, with a focus on muscle and muscle wasting. muscle wasting with a normal body mass index, and as many
as 20% with obesity will have overt muscle wasting. Looking
CANCER CACHEXIA across all patients with sarcopenia, the incidence of dose-
Cancer-induced muscle wasting results in accelerated limiting chemotherapy toxicity is increased, and time to
muscle loss and a decline in physical function. Approximately progression and overall survival is reduced.
half of the patients with advanced lung cancer have severe For the patient with cancer the disease process leads to a
muscle wasting at diagnosis, and muscle wasting will decline in muscle mass and associated anorexia, fatigue, and

Department of Medicine and Solid Tumor Therapeutics Program, Duke Cancer Institute, Duke University Medical Center, Durham, NC; Division of Medical Oncology, University of Ottawa,
The Ottawa Hospital Cancer Centre, Ottawa, Ontario, Canada; University of Maryland Greenebaum Cancer Center, Baltimore, MD.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Jeffrey Crawford, MD, Duke Cancer Institute, DUMC 3476, Durham, NC 27710; email: crawf006@mc.duke.edu.

2016 by American Society of Clinical Oncology.

e484 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


CACHEXIA, FATIGUE, AND PATIENTS WITH LUNG CANCER

reduced quality of life, with subsequent weakness, declining functional improvement, including the duration of therapy,
muscle strength, and reduced mobility, disability, and im- the choice of the functional tests, and other factors. How-
pact on physical performance. At a molecular level, this loss ever, these agents and others in development represent
of muscle mass is associated with changes in protein stores, exciting possibilities for future therapeutics in the field of
including enzymes and other regulatory proteins, altered cancer cachexia.11
metabolism, such as insulin resistance, and impaired At present, most importantly for us as clinicians is rec-
immunity. ognition of the presence of cachexia in our patients. Weight
The molecular mechanisms behind the muscle loss that loss remains the primary clinical criteria. By definition, any
occurs in cancer cachexia and other forms of cachexia is an patient with greater than 5% of body weight loss has ca-
area of extreme active investigation. Other factors that chexia regardless of their overall body weight. Although this
may lead to direct muscle atrophy through cytokines, such only represents a portion of patients with muscle loss as
as tumor necrosis factoralpha and interleukin-6, as well defined by CT imaging, weight loss at least allows us to
as myostatin and activin, are also under active investigation. recognize this subset of patients who require additional
A number of agents that affect these cytokine-mediated supportive care. Although current pharmacologic interventions
pathways are in development.6 A second approach has have not been established to be effective, exercise12 and
been to directly increase muscle growth signals through nutritional support13 continue to be important for the
androgenic anabolic steroids or selective androgen recep- multimodality treatment of patients with lung cancer at
tor modulators (SARM), as well as ghrelin mimetics. A first- all stages. It is hoped that in the years ahead, CT imaging
in-class SARM, enobosarm, showed promising results on of muscle will move from an investigational tool to a
improvement in muscle and physical function among pa- clinical measurement to enable us to assess the impact of
tients with cancer cachexia.7 Subsequent phase III trials our interventions for our patients, which should be and
among patients with advanced lung cancer receiving che- include pharmacologic approaches, nutritional support, and
motherapy showed an increase in muscle mass in the exercise.
enobosarm treatment group compared with overall decline
in muscle mass in the placebo population, but effects on
physical function using stair-climb power were inconsistent TREATMENT OF PATIENTS WITH ORGAN
between the trials.8 Phase II trials of anamorelin, a ghrelin FAILURE
receptor agonist for patients with cancer cachexia, again For patients with lung cancer, choices of systemic therapy
showed improvement in skeletal muscle mass.9 Phase III are routinely informed by, and based on, protocols emerging
trials have also been performed with this agent for patients from clinical research. These guide the patient and clinician
with advanced lung cancer and cachexia and have shown about the gold standard options when making decisions
improvement in skeletal muscle mass, compared with pla- regarding optimal therapy. However, in reality, many pa-
cebo controls. In addition, this agent has positive effects on tients seen daily in the clinic may not be eligible for a clinical
improving appetite and overall weight gain. Improvement trial because of a variety of factors ranging from poor
in function as measured by hand-grip strength, however, functional status, comorbidities, other malignancies, non-
was not observed.10 Multiple factors may explain the lack measurable disease, or blood work falling outside of specific
of correlation between increase in muscle and lack of parameters, to name a few. It is reasonable, therefore, to
question the transferability of clinical trial results to a
general population, and it potentially means that the
standards of care options actually have a less solid evidence
KEY POINTS base for many patients. In a recent analysis of 528 patients
who were newly diagnosed with stage IV nonsmall cell lung
Patients with lung cancer who are compromised cancer (NSCLC) and seen in consultation by medical on-
physically, medically, or socioeconomically are often cologists, only 55% received systemic treatment.14 Further,
excluded from clinical trials and have poorer treatment when simple and limited generic clinical trial inclusion cri-
outcomes. teria were applied to these patients, only 27% would have
Cancer cachexia is clinically recognized by weight loss, been trial-eligible.15 In additional analysis of this dataset,
but the primary process is muscle loss, which occurs in the average survival of patients who received systemic
the majority of patients with advanced lung cancer, palliative chemotherapy was not significantly different
regardless of body mass index. among patients who were considered trial eligible or trial
Care for patients with cancer and organ failure requires
ineligible. An interpretation may be that clinical trial eligi-
close integration with disease subspecialists.
Socioeconomic disparities affect lung cancer risk,
bility is too strict and clinical judgment is more important.
diagnosis, therapy, and outcomes. Despite this, there is another small but important cohort of
New paradigms are needed that include patients with major medical comorbidities, or a formal di-
multidisciplinary approaches to improving outcomes of agnosis of organ failure, who appropriately are excluded
medically compromised patients. from clinical trials and for whom systemic therapy poses
specific and challenging problems. In a review of selected

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e485


CRAWFORD, WHEATLEY-PRICE, AND FELICIANO

TABLE 1. Selected Recent Practice-Changing Trials in Lung Cancer and Organ Function Exclusions
Trial Author Year Renal Failure Liver Failure Heart Failure
First-Line Chemotherapy Schiller et al16 2002 Excluded Excluded Unstated
17
Second-Line Chemotherapy Hanna et al 2004 Excluded Excluded Unstated
Chemotherapy + Bevacizumab Sandler et al18 2006 Excluded Excluded Excluded
Maintenance Ciuleanu et al19 2009 Excluded Excluded Excluded
20
EGFR + Gefitinib Mok et al 2009 Unstated Excluded Unstated
ALK + Crizotinib Shaw et al21 2013 Excluded Excluded Excluded
Nivolumab Brahmer et al22 2015 Excluded Excluded Unstated

recent practice-changing chemotherapy, targeted therapy, noncancer illnesses, Salpeter et al evaluated heart failure,
and immunotherapy trials, patients with renal impairment, dementia, geriatric failure-to-thrive syndrome, hepatic cir-
hepatic impairment, or cardiac impairment would have been rhosis, chronic obstructive pulmonary disease, and end-stage
excluded (Table 1).16-23 Therefore, how should clinicians renal disease. This list represented approximately 70% of the
and patients approach decisions about systemic therapy in noncancer diagnoses on admission to hospice.24 Clearly, not
the presence of organ failure, given the lack of available all patients with these conditions die within 6 months, but the
evidence? authors identified common and disease-specific prognostic
This article provides guidance on a reasonable and pragmatic indicators, including poor performance status (PS), malnu-
approach to making systemic therapy decisions for patients trition, comorbid illness, and organ dysfunction.
with lung cancer and organ failure. In recognizing and accepting In the cancer clinic, the clinician must understand the
that there is an absence of published data in the medical lit- natural course of the organ failure pathology. For patients
erature to provide clarity, these recommendations are the with liver, kidney, or heart failure who may be waiting for
considered views of the authors, but other reasonable and organ transplantation, the diagnosis of lung cancer un-
well-planned approaches may have equal validity. It is also fortunately makes them ineligible for the transplant pro-
important to recognize that there is a spectrum ranging from gram. Regarding prognosis of advanced organ failure, the
normal organ function, through stages of organ impairment United States Renal Data System Annual Data Report, de-
or dysfunction to organ failure. This article is restricted to scribes 3-year survival as 52% for patients receiving he-
discussing implications of systemic therapy for patients with modialysis for end-stage renal disease, and 61% for patients
lung cancer and established organ failure. receiving peritoneal dialysis. The risk of death is particularly
The first recommendation is that these issues should high in the first year of hemodialysis, with rates reported up
be discussed in a multidisciplinary format, and specific in- to 25%. The Canadian Organ Replacement Register Annual
teraction with specialists related to the particular organ Report describes a 5-year survival for patients on dialysis of
failure is advised (e.g., nephrology, hepatology, cardiology, approximately 43%. For patients with end-stage heart fail-
etc.). Further, additional consultation with a specialist on- ure, the 1-year survival is approximately 50%,25 which is not
cology pharmacist is advised, particularly if the decision is dramatically different from patients with stage IV NSCLC
made to proceed with therapy. Patients should be fully receiving first-line chemotherapy. The prognosis of patients
informed regarding relative benefits and harms from ther- with liver cirrhosis is variable, depending on severity, eti-
apy, the consequences of declining therapy, and that pro- ology, and the presence or absence of complications. The
ceeding with treatment will almost certainly not be based Model for End-Stage Liver Disease (MELD) score is used to
on level-one evidence. For patients with advanced disease, assess the severity of chronic liver disease26 as an alternative
consideration should be given to early palliative care spe- to the Child-Pugh scoring system. Salpeter et al24 reported
cialist input and advanced care planning. that patients with decompensated liver failure (the presence
Understanding the cause and prognosis of organ failure is of complications of cirrhosis) may have a median survival of
self-evidently important. Some patients are diagnosed less than 6 months if associated with high MELD scores.
with an acute organ failure directly related to the lung An understanding of competing morbidities therefore
cancer, and, if considered reversible, this should be ad- clearly plays an important role in understanding the role
dressed. For example in a patient presenting with acute systemic therapy plays in lung cancer. In assessing the need
renal failure caused by hydronephrosis from retroperitoneal for adjuvant chemotherapy for patients with early-stage
lymphadenopathy, by inserting a nephrostomy tube, the disease, it is highly likely that any benefit from chemother-
renal failure can be reversed and the barrier to systemic apy (approximately 5%) will be outweighed by the competing
therapy is removed. However, the remainder of this article risks of the comorbid condition for patients with organ failure.
will address patients with pre-existing organ failure, rather As an illustration, the adjuvant online program is used to
than organ failure secondary to the malignancy. In a recent model potential benefits from adjuvant chemotherapy in
review of clinical indicators of 6-month mortality in advanced resected NSCLC. Figure 1, adapted from the adjuvant online

e486 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


CACHEXIA, FATIGUE, AND PATIENTS WITH LUNG CANCER

FIGURE 1. The Benefit of Adjuvant Chemotherapy for hepatic impairment and for patients receiving dialysis.
a Patient With Stage IIA NonSmall Cell Lung Cancer, Tabular information is taken from product monographs and
With or Without Organ Failure selected references.28,29 Data on efficacy for these drugs in
these scenarios are largely limited to case reports.
In conclusion, patients with lung cancer and organ failure
represent a population excluded from clinical trials and
with a limited evidence base. The competing morbidity and
mortality mitigate against potential benefits from anti-
cancer systemic therapy. The newer generations of targeted
therapies and immunotherapies may be easier to deliver,
but limited data exist. Clinicians should discuss these cases
in a multidisciplinary environment, and early intervention
from palliative care specialists may be particularly appro-
priate. Finally, as a community, we should seek to perform
clinical trials in this population of patients with lung cancer.

TREATMENT OF LUNG CANCER IN MEDICALLY


COMPROMISED PATIENTS: COMPROMISED
SOCIAL AND ECONOMIC STATUS IN LUNG
website, demonstrated that the absolute benefit of chemo-
CANCERIS IT REAL AND DOES IT MATTER?
therapy (notwithstanding the challenges of actually admin-
Many factors can make a patient vulnerable to poor out-
istering the treatment) is only modestly reduced but is now
comes with lung cancer. Biologic and patient-related factors
seen in the context of lung cancer no longer being the primary
such as poor nutrition, functional status, or organ function
health threat in any event. In contrast to the curative situation
undoubtedly contribute to vulnerability to poor outcomes.
in which the prize of successful therapy is great, for patients
However, it is critical to recognize that poor socioeconomic
with metastatic disease the role of competing morbidities
status (SES) can render a patient equally vulnerable to poor
and organ failure is even more striking. When the absolute
outcomes from lung cancer. Socioeconomic status is often
benefits, measured in terms of prolonged overall survival
defined as ones class or standing measured by education,
from systemic therapy, may only be measured in a few
income, and occupation.30 Poor SES can affect the disease at
months in a healthy population, the benefits from palliative
various stages including development, diagnosis, treatment,
systemic therapy in patients with significant comorbidities or
and, ultimately, outcomes from lung cancer.7 There are
organ failure are likely to be further diminished. These issues
many confounders such as race, education level, and in-
must be openly disclosed and discussed with patients prior to
surance status when evaluating socioeconomic disparities,
any decision to attempt systemic therapy.
but these variables are intimately connected. Interventions
For many (or indeed the majority of) patients with advanced
that target these vulnerable populations may minimize the
lung cancer and coexisting organ failure, a palliative approach
socioeconomic disparities that exist in lung cancer.
may be the most appropriate. Based on the seminal publication
Socioeconomic disparities have also been shown to af-
by Temel et al, early palliative care intervention is associated
fect lung cancer risk, diagnosis, therapy, and outcomes in-
with improved quality of life and longer survival.27 Therefore
ternationally. It is often difficult to separate socioeconomic
a recommendation would be to initiate a palliative approach
disparities from other sources of disparities in lung cancer
to management of these patients, including discussions re-
such as race and insurance. Here, we will discuss the role of
garding advanced care planning and place of care. While
socioeconomic factors, with a focus on income, in the de-
a cliche, for many patients facing an incurable lung cancer,
velopment, treatment, and outcomes of lung cancer in the
and barriers to therapy because of organ failure, we maybe
United States.
should consider care as being more important than treatment.
Specifically, patients with rapid decline, hospitalization, frailty,
and poor functional status should have serious consideration to Socioeconomic Factors Contribute to Disparities
a purely palliative treatment plan. in Lung Cancer Risk
After assessing patients with lung cancer, in the multi- Tobacco is the leading risk factor for lung cancer develop-
disciplinary context and taking into account the issues dis- ment, and approximately 80%90% of lung cancers are
cussed, the decision may still be to proceed with therapy. related to tobacco.31 Earlier age of smoking initiation has
This should be on the understanding of the relative lack been associated with a higher prevalence of smoking
of data, and then a choice of regimen based on an un- through adulthood, lower cessation rates, and increased risk
derstanding of the drug metabolism, with appropriate dose of lung cancer. It is indisputable that these factors dispro-
adjustments after dialogue with an oncology pharmacist. portionately affect lower-income populations.32-35
Table 2 outlines common lung cancer drugs and their route Childhood poverty, for example, has been linked to as
of elimination and recommendations for their use in renal or much as a 33% higher likelihood of initiation and earlier

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e487


CRAWFORD, WHEATLEY-PRICE, AND FELICIANO

TABLE 2. Common Lung Cancer Drugs and Recommendations for Use in Renal or Hepatic Impairment and for
Patients Receiving Dialysis

Drug Elimination Liver Renal Dialysis


Cisplatin Renal Not applicable Decrease depending on CrCl 50% post-HD or non-HD days
Carboplatin Renal Not applicable Calvert formula Calverts formula give on non-HD day
Docetaxel Liver Adjust Not applicable Before or after HD not removed by HD
Pemetrexed Renal Caution in severe dysfunction Avoid if CrCl # 45 Avoid
Paclitaxel Liver Adjust Not applicable Not removed by HD
Gemcitabine Urine (inactive Consider adjustment if bilirubin Caution (D/C if HUS) Give 6-12 hours before HD
metabolites) . 27
Vinorelbine Liver Adjust depending on bilirubin Not applicable Limited data, consider 20 mg/m2 on
non-HD day
Etoposide Liver/Renal Adjust depending on bilirubin Adjust 50% before or after HD
Gefitinib Liver Caution Caution if CrCl , 20 No information
Erlotinib Liver Caution Not applicable No information
Afatinib Liver Caution Caution if CrCl , 30 No information
Crizotinib Liver Adjust Caution if CrCl , 30 No information
Ceritnib Liver Adjust Caution if CrCl , 30 No information
Bevacizumab Reticulo-endothelial Not involved Not involved No information
system
Nivolumab Biochemical degrada- No effect in mild impairment No effect if CrCl $ 15 mL/min No information
tion; no liver or kidney (not studied in severe) (not studied if less)
involvement
Abbreviations: CrCl, creatinine clearance; HD, hemodialysis; HUS, hemolytic uremic syndrome.

onset of smoking (odds ratio [OR] 1.33; 95% CI, 1.081.63).34 provided extra support to vulnerable populations have had
The prevalence of smoking is significantly higher in people alleviated, but not eliminated, smoking-related disparities in
who live below the poverty versus those who live above the low-income populations.44,45 It is crucial to address smoking
poverty level. At the census track level in the United States, uptake and prevalence in lower-income populations to fur-
smoking rates in populations with incomes below $20,000 ther reduce disparities in the development of lung cancer.
per year is estimated at 38.2% compared with 17.3%29.6%
in populations who have incomes above $20,000 per
year.36,37 In addition, it has been recognized that people in Socioeconomic Status Affects Stage at Diagnosis and
lower-income groups are less likely to use clinically proven Receipt of Stage-Appropriate Therapy for Lung Cancer
smoking cessation treatments and have less successful quit Lung cancer prognosis depends on numerous factors including
attempts.38,39 stage at diagnosis and receipt of stage-appropriate therapy.
Many factors contribute to this disproportionate tobacco Unfortunately, socioeconomic factors, including lower income,
epidemic. One factor is the tobacco industrys marketing are associated with diagnosis of lung cancer at later, less
campaigns. The tobacco industry has marketed smoking curable stages.46,47 Numerous studies have demonstrated
and tobacco products to target young, lower-income pop- that patients with lower income and Medicaid insurance or
ulations for decades, and these campaigns have been re- no insurance are more likely to have advanced disease.7,48
markably successful.40,41 Adolescents who are receptive to For example, in areas where there is less than 10% poverty,
tobacco advertising, such as those who can recognize to- 52%54.6% of patients have distant metastases compared with
bacco advertising messages or who identify a favorite areas with more than 20% poverty, where 56.8%59% of
tobacco advertisement, are 2.33.9 times as likely to begin patients have metastatic lung cancer.2 Similarly, in an analysis
smoking in adolescence compared with those who are not of the Florida Cancer Registry, 25% of individuals from areas
receptive to tobacco advertising.42 Other factors such as with less than 5% poverty have localized disease compared
placement of low-cost tobacco outlets in low-income areas with only 22% of those from areas with more than 15% poverty
and environmental or social stressors exacerbate this epi- (p , .001).49 Presentation at later stages may be related to
demic as well.43 numerous factors, such as a lower likelihood of being under the
Despite antitobacco campaigns such as clean environment care of a health provider who could potentially recognize the
bills and advertisements, the decline in smoking rates has signs of lung cancer or economic stressors that prevent in-
been larger among people above the poverty level. Pro- dividuals from seeking care.50
grams like those that have raised the price of cigarettes, One potential factor that may widen the disparity in stage at
reduced advertising to lower-income populations, or have diagnosis in lower-income populations may be with the use and

e488 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


CACHEXIA, FATIGUE, AND PATIENTS WITH LUNG CANCER

uptake of lung cancer screening. The National Lung Screening significantly associated with lower rates of hospice utilization
Trial (NLST) has demonstrated that lung cancer screening with in elderly patients with advanced lung cancer.60
low-dose CT scan can reduce lung cancer mortality by 20% and
that more lung cancer cases were diagnosed at earlier stages of
disease for patients who underwent a low-dose CT.51 However, We Can Learn From Other Cancers Where
this research population of over 53,000 individuals represented a Socioeconomic Disparities Have Been Reduced
population of largely white, highly-educated former smokers.51 As health care providers, we have an opportunity to learn
It is largely unknown how these results translate or can be from successful programs that have reduced cancer dis-
applied to lower-income populations who are more likely to be parities by targeting vulnerable populations. One highly
current smokers and less likely to be under the care of a health successful program implemented by the Delaware Cancer
care provider. Furthermore, it is largely unknown what health Consortium, for example, mandated a statewide program
care providers beliefs and attitudes are toward lung cancer to increase colorectal cancer (CRC) screening in high-risk
screening and how often screening guidelines are adhered to in populations (African Americans and under- or uninsured
the era after the National Lung Screening Trial results were patients) and demonstrated remarkable success.61 This
published in various practice settings.52,53 It is likely that as lung program used nurse navigators and care coordinators to
cancer screening becomes more refined and used, similar so- recruit vulnerable populations, funded screening for
cioeconomic disparities in screening will become apparent, and treatment for CRC, and accomplished its goal of reducing
efforts to empirically reduce this are important. CRC screening, treatment, and survival disparities. In 9 years,
Lower socioeconomic groups are more likely to experience the disparities in screening rates, stage at diagnosis, and
delays in therapy from the time of diagnosis and less likely to mortality were almost eliminated. Similarly, because the CRC
undergo stage-appropriate therapy for lung cancer.54,55 For program has been so successful, Delaware has also recently
example, in a large population-based Surveillance, Epidemi- launched a comprehensive program for lung cancer screening.62
ology, and End Results (SEER)-Medicare analysis, those with In this program, education about screening is targeted
incomes of less than $25,000 were more likely to have delays in toward health care providers and patients. Patients who
guideline-concordant lung cancer care (hazard ratio [HR] 0.89; are potentially eligible for screening are given access to
95% CI, 0.80.98; p # .05).54 patient navigators and care coordinators who assist them
As has been observed for other malignancies such as breast with receiving counseling and assistance with smoking
or colon cancer, in which disparities have become apparent, cessation. Eligible participants are also able to establish
lack of awareness, lack of access, or mistrust of the health care care with a primary care provider who will order screening if
system may contribute to socioeconomic and racial disparities appropriate and recommend follow-up or referrals to
in screening rates, stage at diagnosis, therapy, and survival. providers for further workup. Furthermore, participants
Provider factors, such as whether a health care provider has who are diagnosed with lung cancer will also be able to
knowledge or understanding about the diagnosis or man-
agement of lung cancer, can also affect this receipt of therapy. TABLE 3. Contributing Factors to Economic Disparities
For example, even after the NLST results were published, many and Possible Interventions
physicians are not aware of who would be eligible for lung
cancer screening, even at an NLST site.52 In addition, lower Contributing Factor Intervention
income has been associated with a lower likelihood of referral Age at Smoking Initiation Interventions to target peer
to an oncologist, as well as lower likelihood of receipt of groups 43,63,64
guideline-appropriate therapy for locally advanced and Smoking Prevalence Clean air laws62
advanced NSCLC.56 Furthermore, health system barriers Smoke-free multi-unit housing62
such as lack of access to specialists can also influence the Prohibit tobacco sales near schools62
diagnosis and receipt of therapy for lung cancer. Prohibit outdoor advertisements62
Prohibit distribution of free or low-
cost tobacco 43,62
Lower Income Associated With Poorer Outcomes Smoking Cessation Reduce low-cost tobacco sale
From Lung Cancer outlets43
Although there are many confounders when evaluating so- Financial incentives65
cioeconomic disparities in lung cancer outcomes, such as Increased access to smoking cessation
smoking status, race, or medical comorbidities, individuals support65,66
with lower incomes have inferior outcomes from lung cancer. Stage at Diagnosis and Receipt Education to providers61
In population-based studies, even when controlling for other of Therapy
prognostic factors, lower income (, $46,000 compared Awareness (Patient and Education on screening61
with . $46,000) was associated with inferior 30-day and Provider)
long-term survival after surgical resection.55,57 Low income Access to Care Payment for therapy61
has also been independently associated with inferior survival Fear of Diagnosis Patient and nurse navigators61,62
in advanced lung cancer.58,59 Furthermore, lower income was Establish care with a provider61,62

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e489


CRAWFORD, WHEATLEY-PRICE, AND FELICIANO

receive therapy for their diagnosis with minimal to no widen. It is crucial that as health care providers, we ac-
cost. knowledge that socioeconomic disparities contribute to infe-
Despite improvements in early detection of lung cancer rior outcomes for lung cancer and that we can learn from
and in therapeutics, the toll from lung cancer remains dismal the past and from other malignancies in which disparities
and disparities at all stages of lung cancer diagnosis and have been reduced to guide current and future identifi-
management persist and, in many situations, continue to cation and management of lung cancer (Table 3).

References
1. Karnofsky DA, Burchenal JH. The clinical evaluation of chemotherapeutic 16. Schiller JH, Harrington D, Belani CP, et al. Comparison of four che-
agents in cancer. In MacLeod CM (ed). Evaluation of Chemotherapeutic motherapy regimens for advanced non-small-cell lung cancer. N Engl J
Agents. New York: Columbia University Press, 1949;191-205. Med. 2002;346:92-98.
2. Martin L, Senesse P, Gioulbasanis I, et al. Diagnostic criteria for the 17. Hanna N, Shepherd FA, Fossella FV, et al. Randomized phase III trial of
classification of cancer-associated weight loss. J Clin Oncol. 2015;33: pemetrexed versus docetaxel in patients with non-small-cell lung cancer
90-99. previously treated with chemotherapy. J Clin Oncol. 2004;22:1589-1597.
3. Fearon K, Strasser F, Anker SD, et al. Definition and classification of 18. Sandler A, Gray R, Perry MC, et al. Paclitaxel-carboplatin alone or with
cancer cachexia: an international consensus. Lancet Oncol. 2011;12: bevacizumab for non-small-cell lung cancer. N Engl J Med. 2006;355:
489-495. 2542-2550.
4. Prado CM, Lieffers JR, McCargar LJ, et al. Prevalence and clinical im- 19. Ciuleanu T, Brodowicz T, Zielinski C, et al. Maintenance pemetrexed
plications of sarcopenic obesity in patients with solid tumours of the plus best supportive care versus placebo plus best supportive care for
respiratory and gastrointestinal tracts: a population-based study. non-small-cell lung cancer: a randomised, double-blind, phase 3 study.
Lancet Oncol. 2008;9:629-635. Lancet Oncol. 2009;374:1432-1440.
5. Prado CM, Antoun S, Sawyer MB, et al. Two faces of drug therapy in 20. Mok TS, Wu YL, Thongprasert S, et al. Gefitinib or carboplatin-paclitaxel
cancer: drug-related lean tissue loss and its adverse consequences to in pulmonary adenocarcinoma. N Engl J Med. 2009;361:947-957.
survival and toxicity. Curr Opin Clin Nutr Metab Care. 2011;14:250-254. 21. Shaw AT, Kim DW, Nakagawa K, et al. Crizotinib versus chemo-
6. Cohen S, Nathan JA, Goldberg AL. Muscle wasting in disease: molecular therapy in advanced ALK-positive lung cancer. N Engl J Med. 2013;
mechanisms and promising therapies. Nat Rev Drug Discov. 2015;14: 368:2385-2394.
58-74. 22. Brahmer J, Reckamp KL, Baas P, et al. Nivolumab versus docetaxel in
7. Dobs AS, Boccia RV, Croot CC, et al. Effects of enobosarm on muscle advanced squamous-cell non-small-cell lung cancer. N Engl J Med. 2015;
wasting and physical function in patients with cancer: a double-blind, 373:123-135.
randomised controlled phase 2 trial. Lancet Oncol. 2013;14:335-345. 23. Gettinger SN, Horn L, Gandhi L, et al. Overall survival and long-term
8. Crawford J, Dalton JT, Hancock ML, et al. LATE BREAKING ABSTRACT: safety of nivolumab (anti-programmed death 1 antibody, BMS-936558,
Results from two Phase 3 randomized trials of enobosarm, selective ONO-4538) in patients with previously treated advanced non-small-cell
androgen receptor modulator (SARM), for the prevention and treat- lung cancer. J Clin Oncol. 2015;33:2004-2012.
ment of muscle wasting in NSCLC. Presented at: European Cancer 24. Salpeter SR, Luo EJ, Malter DS, Stuart B. Systematic review of noncancer
Congress; 2013 Amersterdam; Abstract 21. presentations with a median survival of 6 months or less. Am J Med.
9. Garcia JM, Boccia RV, Graham CD, et al. Anamorelin for patients with 125:512 e1-6.
cancer cachexia: an integrated analysis of two phase 2, randomised, 25. Friedrich EB, Bohm M. Management of end stage heart failure. Heart.
placebo-controlled, double-blind trials. Lancet Oncol. 2015;16:108-116. 2007;93:626-631.
10. Temel JS, Currow DC, Fearon K, et al. Phase III trials of anamorelin in 26. Kamath PS, Kim WR; Advanced Liver Disease Study Group. The model
patients with advanced non-small cell lung cancer (NSCLC) and cachexia for end-stage liver disease (MELD). Hepatology. 2007;45:797-805.
(ROMANA 1 and 2). J Clin Oncol. 2015;33(suppl; abstr 9500). 27. Temel JS, Greer JA, Muzikansky A, et al. Early palliative care for patients with
11. Fearon K, Argiles JM, Baracos VE, et al. Request for regulatory guidance metastatic nonsmall-cell lung cancer. N Engl J Med. 2010;363:733-742.
for cancer cachexia intervention trials. J Cachexia Sarcopenia Muscle. 28. Janus N, Thariat J, Boulanger H, et al. Proposal for dosage adjustment
2015;6:272-274. and timing of chemotherapy in hemodialyzed patients. Ann Surg Oncol.
12. Alves CR, da Cunha TF, da Paix~ao NA, et al. Aerobic exercise training as 2010;21:1395-1403.
therapy for cardiac and cancer cachexia. Life Sci. 2015;125:9-14. 29. Brandes JC, Grossman SA, Ahmad H. Alteration of pemetrexed excretion
13. Bourdel-Marchasson I, Blanc-Bisson C, Doussau A, et al. Nutritional in the presence of acute renal failure and effusions: presentation of a
advice in older patients at risk of malnutrition during treatment for case and review of the literature. Cancer Invest. 2006;24:283-287.
chemotherapy: a two-year randomized controlled trial. PLoS One. 2014; 30. Baker EH. Socioeconomic status, definition. The Wiley Blackwell
9:e108687. Encyclopedia of Health, Illness, Behavior, and Society. Hoboken, NJ:
14. Brule S, Al-Baimani K, Jonker H, et al. Palliative chemotherapy (CT) for Wiley-Blackwell; 2014.
advanced non-small cell lung cancer (NSCLC): investigating disparities 31. U.S. Department of Health and Human Services. The Health Conse-
between patients who are treated versus those who are not. J Clin quences of Smoking: A Report of the Surgeon General. Atlanta, GA: U.S.
Oncol. 2015;33(suppl; abstr e17681). Department of Health and Human Services, Centers for Disease Control
15. Al-Baimani K, Jonker H, Zhang T, et al. Are clinical trial eligibility criteria and Prevention, National Center for Chronic Disease Prevention and
an accurate reflection of a real world population of advanced lung Health Promotion, Office on Smoking and Health; 2004.
cancer patients. Presented at: World Conference on Lung Cancer; 2015 32. Chen J, Millar WJ. Age of smoking initiation: Implications for quitting.
Denver, CO; Abstract 1398. Health Rep. 1998;9:39-46.

e490 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


CACHEXIA, FATIGUE, AND PATIENTS WITH LUNG CANCER

33. Wiencke JK, Thurston SW, Kelsey KT, et al. Early age at smoking initiation 51. Aberle DR, Adams AM, Berg CD, et al; National Lung Screening Trial
and tobacco carcinogen DNA damage in the lung. J Natl Cancer Inst. Research Team. Reduced lung-cancer mortality with low-dose com-
1999;91:614-619. puted tomographic screening. N Engl J Med. 2011;365:395-409.
34. Gilman SE, Abrams DB, Buka SL. Socioeconomic status over the life 52. Lewis JA, Petty WJ, Tooze JA, et al. Low-dose CT lung cancer
course and stages of cigarette use: initiation, regular use, and cessation. screening practices and attitudes among primary care providers at
J Epidemiol Community Health. 2003;57:802-808. an academic medical center. Cancer Epidemil Biomarkers Prev.
35. Hegmann KT, Fraser AM, Keaney RP, et al. The effect of age at smoking 2015;24:664-670.
initiation on lung cancer risk. Epidemiology. 1993;4:444-448. 53. Lam VK, Miller M, Dowling L, et al. Community low-dose CT lung cancer
36. King BA, Dube SR, Tynan MA. Current tobacco use among adults in the screening: a prospective cohort study. Lung. 2015;193:135-139.
United States: findings from the National Adult Tobacco Survey. Am J 54. Nadpara P, Madhavan SS, Tworek C. Guideline-concordant timely
Public Health. 2012;102:e93-e100. lung cancer care and prognosis among elderly patients in the United
37. Agaku IT, King BA, Dube SR; Centers for Disease Control and Prevention. States: a population-based study. Cancer Epidemiol. 2015;39:
Current cigarette smoking among adults - United States, 2005-2012. 1136-1144.
MMWR Morb Mortal Wkly Rep. 2014;63:29-34. 55. Melvan JN, Sancheti MS, Gillespie T, et al. Nonclinical factors associated
38. Burns ME, Fiore MC. Under-use of tobacco dependence treatment with 30-day mortality after lung cancer resection: an analysis of 215,000
among Wisconsins fee-for-service Medicaid recipients. WMJ. 2001; patients using the national cancer data base. J Am Coll Surg. 2015;221:
100:54-58. 550-563.
39. Fu SS, Kodl MM, Joseph AM, et al. Racial/Ethnic disparities in the use of 56. Goulart BH, Reyes CM, Fedorenko CR, et al. Referral and treatment
nicotine replacement therapy and quit ratios in lifetime smokers ages patterns among patients with stages III and IV nonsmall-cell lung
25 to 44 years. Cancer Epidemiol Biomarkers Prev. 2008;17:1640-1647. cancer. J Oncol Pract. 2013;9:42-50.
40. Hackbarth DP, Silvestri B, Cosper W. Tobacco and alcohol billboards in 57. Khullar OV, Gillespie T, Nickleach DC, et al. Socioeconomic risk factors
50 Chicago neighborhoods: market segmentation to sell dangerous for long-term mortality after pulmonary resection for lung cancer: an
products to the poor. J Public Health Policy. 1995;16:213-230. analysis of more than 90,000 patients from the national cancer data
41. Yerger VB, Przewoznik J, Malone RE. Racialized geography, corporate base. J Am Coll Surg. 2015;220:156-168.
activity, and health disparities: tobacco industry targeting of inner 58. Tannenbaum SL, Koru-Sengul T, Zhao W, et al. Survival disparities in
cities. J Health Care Poor Underserved. 2007;18:10-38. non-small cell lung cancer by race, ethnicity, and socioeconomic status.
42. Evans N, Farkas A, Gilpin E, et al. Influence of tobacco marketing and Cancer J. 2014;20:237-245.
exposure to smokers on adolescent susceptibility to smoking. J Natl 59. Erhunmwunsee L, Joshi MB, Conlon DH, et al. Neighborhood-level
Cancer Inst. 1995;87:1538-1545. socioeconomic determinants impact outcomes in nonsmall cell lung
43. Cantrell J, Anesetti-Rothermel A, Pearson JL, et al. The impact of the cancer patients in the Southeastern United States. Cancer. 2012;118:
tobacco retail outlet environment on adult cessation and differences by 5117-5123.
neighborhood poverty. Addiction. 2015;110:152-161. 60. Mack JW, Chen K, Boscoe FP, et al. Underuse of hospice care by
44. Vijayaraghavan M, Messer K, White MM, et al. The effectiveness of Medicaid-insured patients with stage IV lung cancer in New York and
cigarette price and smoke-free homes on low-income smokers in the California. J Clin Oncol. 2013;31:2569-2579.
United States. Am J Public Health. 2013;103:2276-2283. 61. Grubbs SS, Polite BN, Carney J Jr, et al. Eliminating racial disparities in
45. Haas JS, Linder JA, Park ER, et al. Proactive tobacco cessation outreach colorectal cancer in the real world: it took a village. J Clin Oncol. 2013;31:
to smokers of low socioeconomic status: a randomized clinical trial. 1928-1930.
JAMA Intern Med. 2015;175:218-226. 62. Healthy Delaware. https://www.healthydelaware.org/lung. Accessed
46. Halpern MT, Ward EM, Pavluck AL, et al. Association of insurance status January 13, 2016.
and ethnicity with cancer stage at diagnosis for 12 cancer sites: a ret- 63. Lipperman-Kreda S, Friend KB, Grube JW. Rating the effectiveness of
rospective analysis. Lancet Oncol. 2008;9:222-231. local tobacco policies for reducing youth smoking. J Prim Prev. 2014;35:
47. Schwartz KL, Crossley-May H, Vigneau FD, et al. Race, socioeconomic 85-91.
status and stage at diagnosis for five common malignancies. Cancer 64. Lee YO, Jordan JW, Djakaria M, et al. Using peer crowds to segment
Causes Control. 2003;14:761-766. Black youth for smoking intervention. Health Promot Pract. 2014;15:
48. Slatore CG, Au DH, Gould MK; American Thoracic Society Disparities in 530-537.
Healthcare Group. An official American Thoracic Society systematic 65. Parks MJ, Slater JS, Rothman AJ, et al. Interpersonal communication and
review: insurance status and disparities in lung cancer practices and smoking cessation in the context of an incentive-based program: survey
outcomes. Am J Respir Crit Care Med. 2010;182:1195-1205. evidence from a telehealth intervention in a low-income population.
49. Yang R, Cheung MC, Byrne MM, et al. Do racial or socioeconomic J Health Commun. 2016;21:125-133.
disparities exist in lung cancer treatment? Cancer. 2010;116:2437-2447. 66. Christiansen BA, Reeder KM, TerBeek EG, et al. Motivating low socio-
50. Pateman K, Ford P, Fitzgerald L, et al. Stuck in the catch 22: attitudes economic status smokers to accept evidence-based smoking cessation
towards smoking cessation among populations vulnerable to social treatment: a brief intervention for the community agency setting.
disadvantage. Addiction. Epub 2015 Nov 28. Nicotine Tob Res. 2015;17:1002-1011.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e491


MELANOMA/SKIN CANCERS

Clinical Conundrums in
Melanoma Therapy

CHAIR
Janice M. Mehnert, MD
Rutgers Cancer Institute of New Jersey
New Brunswick, NJ

SPEAKERS
Robyn Saw, MBBS, FRACS, MS
Melanoma Institute Australia and The University of Sydney
North Sydney, Australia

Sapna Pradyuman Patel, MD


The University of Texas MD Anderson Cancer Center
Houston, TX
BIOMARKERS FOR IMMUNOTHERAPY

Biomarkers for Immunotherapy: Current Developments and


Challenges
Kristen R. Spencer, DO, MPH, Jianfeng Wang, MD, Ann W. Silk, MD, Shridar Ganesan, MD, PhD,
Howard L. Kaufman, MD, and Janice M. Mehnert, MD

OVERVIEW

Immunotherapy has revolutionized cancer therapy and has been named the cancer advance of the year for 2016. Checkpoint
inhibitors have demonstrated unprecedented rates of durable responses in some of the most difficult-to-treat cancers;
however, many treated patients do not respond, and the potential for serious side effects exists. There is a growing need to
identify biomarkers that will improve the selection of patients who will best respond to therapy, further elucidate drug
mechanisms of action, and help tailor therapy regimens. Biomarkers are being explored at the soluble, cellular, and genomic
levels, and examples in immunotherapy include serum proteins, tumor-specific receptor expression patterns, factors in the
tumor microenvironment, circulating immune and tumor cells, and host genomic factors. The search for reliable biomarkers
is limited by our incomplete understanding of how immunotherapies modify the already complex immune response to
cancer, as well as the contribution of immuno-editing to a dynamic and inducible tumor microenvironment and immune
milieu. Furthermore, there has been little extension of any candidate assay into large, prospective studies, and the lack of
standardization in measurement and interpretation restricts their validity. Both tumor-infiltrating lymphocytes and PD-L1
expression within the tumor microenvironment have been recognized as having both prognostic and predictive value for
patients treated with immunotherapy. Alternately, a larger panel of gene signatures, chemokines, and other factors that
correlate with response has been proposed. In this article, we will explore the status of current biomarker candidates.

S ince ipilimumab entered the treatment landscape in


2011, immunotherapy has continued to revolutionize
cancer therapy. In fact, immunotherapy was recently named
and patient subsets who will respond, illuminate the
mechanism of action of novel immunotherapeutic ap-
proaches, and potentially inform which patients require
the American Society of Clinical Oncologys top cancer ad- single-agent versus various combination strategies, which
vance of the year for 2016.1 A number of U.S. Food and Drug are increasing in availability. This review focuses on the de-
Administration (FDA)approved agents are available for an velopment of biomarkers for immunotherapy at the soluble,
increasing number of difficult-to-treat cancers, such as cellular, and genomic levels. Examples of biomarkers in the
melanoma, renal cell carcinoma (RCC), and lung cancer, immunotherapy landscape include (1) soluble factors such
among others. In contrast with most chemotherapy and as serum proteins, (2) tumor-specific factors such as receptor
targeted therapies, immunotherapy offers the possibility expression patterns and components of the microenviron-
of durable response, sometimes even without continued ment, and (3) host genomic factors.6-8 Despite the interest in
treatment.2-4 However, objective responses among patients biomarker development for immunotherapy, validated bio-
treated with single-agent regimens are seen in less than one- markers have remained an elusive goal.
half of patients treated. Combination immune checkpoint
inhibitor therapy raises response rates but also increases
toxicity and cost.5 Thus, to optimize selection of appropriate WHY ARE BIOMARKERS CHALLENGING IN
patients for immunotherapy and avoid unnecessary toxi- IMMUNOTHERAPY?
city and health care costs, there is a clear need to identify Our incomplete understanding of the mechanisms of action
truly predictive, and not simply prognostic, biomarkers of of specific immunotherapies makes it difficult to identify a
response. surrogate marker that adequately captures the process
Understanding which factors predict clinical benefit with across different classes of drugs.9 Many published analyses
immunotherapy can improve the selection of tumor types of potential predictive biomarkers for immunotherapy are

From the Rutgers Cancer Institute of New Jersey, New Brunswick, NJ.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Janice M. Mehnert, MD, Rutgers Cancer Institute of New Jersey, 195 Little Albany St., New Brunswick, NJ 08901; email: mehnerja@cinj.rutgers.edu.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e493


SPENCER ET AL

retrospective, with limited extension into large prospective associated with more favorable outcomes, although this has
trials. In addition, there has been substantial variability in not been uniformly demonstrated. Other groups have used
standardization, measurement, and interpretation of early a larger panel of gene signatures, including T helper (Th)
biomarker assays.10 Furthermore, biomarker development 1 cytokines, chemokines, and other factors, that correlate with
in immunotherapy is challenged by the fact that immuno- therapeutic responses. These studies collectively suggest
therapy targets are often inducible and dynamic over time that there may be host, tumor, and immune factors that can
and location. This is a function of the complex tumor mi- be used for biomarker development. The importance of the
croenvironment and the contribution of immuno-editing tumor microenvironment has been appropriately stressed,
to the immune milieu. The tumor microenvironment in- but, practically, the ability to use serum or peripheral blood
volves complicated interactions between several types of biomarkers is of major clinical utility. We will explore the
infiltrating immune cells such as monocytes, neutrophils, status of current biomarker candidates from both com-
dendritic cells, T and B cells, eosinophils, basophils, mast partments (Table 1).
cells, and natural killer (NK) cells as well as the heteroge-
neous tumor cells themselves and their companion stromal
cells, including tumor-associated macrophages, fibroblasts,
PERIPHERAL BLOOD BIOMARKERS
Candidate biomarkers from serum, plasma, or peripheral
adipocytes, and endothelial and other cells.11 The local
blood must be accurately and reproducibly measurable,
environment is further complicated by microniches cre-
clinically feasible, cost-effective, and prospectively validated
ated by alterations in perfusion, oxygenation, electrolyte
in randomized clinical trials. Putative biomarkers in blood
levels, and the subsequent development of resistant tumor
might be soluble factors such as serum proteins, circulating
cells surviving in nutrient- and oxygen-deprived condi-
tumor DNA, or other cellular factors such as tumor cells,
tions.11,12 Thus, these microniches likely represent distinct
T-cell subsets, or other immune cell populations. The serum
microenvironments with different cell types and factors,
factors may be single or could include a panel of factors
all within one tumor deposit. Finally, incomplete immuno-
preferably measured by a single, validated assay. To date,
editing may result in selective pressure on tumor cells,
most published analyses of peripheral blood biomarkers in
resulting in resistant tumor cell clones and immune
immunotherapy have been retrospective and hypothesis
escape. 11
generating, although important information has been gained
Despite these challenges, clinical research of immuno-
that illuminates the mechanisms of clinical benefit with some
therapy over the last several years has confirmed the im-
approaches and has helped inform subsequent clinical trial
portance of tumor-infiltrating lymphocytes within the tumor
design.
microenvironment as having both prognostic and predictive
value for patients with cancer and for treatment with im-
munotherapy, respectively. There have also been several Serum Soluble Biomarkers
trials of T-cell checkpoint inhibitors in which PD-L1 ex- The potential role of soluble serum proteins was first sug-
pression in the tumor microenvironment has been gested by studies of high-dose interleukin-2 (HD IL-2) among
patients with advanced melanoma and RCC in the 1990s.
High pretreatment serum levels of IL-6 and C-reactive
protein (CRP) were identified as possible prognostic markers
KEY POINTS for treatment failure and shorter overall survival (OS) in
metastatic RCC after IL-2 therapy.13 Later, a French multi-
Checkpoint inhibitors have demonstrated
institutional study revealed that high pretreatment serum
unprecedented rates of durable responses in difficult-
to-treat tumors; however, many treated patients still do
values of CRP were independent predictors of resistance to
not respond. IL-2 therapy among patients with metastatic melanoma.14
Biomarkers remain a critical missing link in attempting More recently, in a retrospective analysis of patients with
to identify appropriate candidates for immunotherapy advanced melanoma, pretreatment serum VEGF and fi-
and tailoring immunotherapy treatment regimens. bronectin, as measured by a customized proteomics array,
Examples of biomarkers being explored in were found to inversely correlate with clinical response to
immunotherapy include serum proteins, tumor-specific IL-2 treatment.15 In this trial, high levels of these proteins
receptor expression patterns, factors in the tumor were associated with lack of clinical response and decreased
microenvironment, circulating immune and tumor cells, OS. A prospective study of patients with melanoma being
and host genomic factors. treated with IL-2 (Melanoma SELECT) has been completed
The identification of appropriate biomarkers has been
and will help to confirm the validity of these markers as
limited by little extension into large prospective trials
that can validate their use and variability in assay
predictive biomarkers.
development and interpretation. Elevated levels of VEGF and CRP have similarly been as-
Tumor-infiltrating lymphocytes, tumor PD-L1 sociated with clinical outcomes for patients treated with
expression, tumor mutational burden, and several other ipilimumab, an antibody that targets the T-cell checkpoint
factors are being considered as candidate biomarkers. CTLA-4 and was approved by the FDA in 2011 for the
treatment of patients with advanced melanoma. In an

e494 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


BIOMARKERS FOR IMMUNOTHERAPY

TABLE 1. Potential Predictive Biomarkers for Immunotherapy


Type Source Biomarker Clinical Significance
Soluble Serum IL-6 High levels are prognostic for HD IL-2 treatment failure and shorter OS
in metastatic RCC13
CRP High levels predict resistance to HD IL-214; decreasing levels during ipilimumab
therapy are associated with disease control and survival18
VEGF High levels are an independent predictor for lack of response to HD IL-215 and are
associated with decreased OS16
LDH Low pretreatment levels predict benefit from ipilimumab17; decreasing levels
during ipilimumab therapy are associated with disease control and survival18
sCD25 High levels predict resistance to ipilimumab19
NY-ESO-1 antibody Seropositivity has greater likelihood to respond to CTLA-4 blockade29,30
Cellular Peripheral blood Neutrophils/leukocytes High counts are prognostic for HD IL-2 treatment failure and shorter OS21
Lymphocytes Immediate lymphocytosis is associated with response to HD IL-2 therapy26
+
CD8 T cells Presence is associated with clinical benefit to CTLA-4 blockade30
ALCs Increasing counts during ipilimumab therapy are associated with an improved
OS18,23; however, this may occur in all patients regardless of benefit27
Eosinophils Increasing counts during ipilimumab therapy are associated with an improved OS23
+ +
CD4 ICOS T cells Increase in frequency after ipilimumab46
MDSCs Low frequency predicts benefit from ipilimumab therapy25
Tumor PD-L1 Refer to Table 2
TILs CD4+ICOShigh T cells Increased frequency correlates with clinical benefit in ipilimumab41,44-46
+
CD8 T cells PD-1/PD-L1 expression on these cells predicts response to PD-1 blockade47,55,57
Genomic Tumor Tumor mutation loads Predict clinical benefit to ipilimumab8,75 and PD-1 blockade7
MMR Predicts clinical benefit to PD-1 blockade76,77
Abbreviations: IL, interleukin; HD, high-dose; OS, overall survival; RCC, renal cell carcinoma; CRP, C-reactive protein; LDH, lactate dehydrogenase; NY-ESO-1, NY-esophageal
cancer 1; ALC, absolute lymphocyte count; ICOS, inducible T-cell costimulator; MDSC, myeloid-derived suppressor cell; TIL, of tumor-infiltrating lymphocyte; MMR, mismatch
repair.

analysis of sera collected from 176 patients with melanoma whether these biomarkers are predictive or merely prog-
treated with ipilimumab, pretreatment VEGF of 43 pg/mL nostic factors. The prospective SELECT trial may help clarify
or greater was associated with decreased OS.16 In addition, this important distinction.
elevated serum lactate dehydrogenase (LDH) levels have
demonstrated a negative predictive value, with limited long-
Peripheral Blood Cellular Biomarkers
term benefit from ipilimumab treatment found among
Similar to serum soluble factors, cells in the peripheral blood,
patients with baseline serum LDH greater than twice the
such as T cells, NK cells, dendritic cells, macrophages, and
upper limit of normal in a cohort of 166 patients from the
tumor cells, have been studied as both prognostic and
Netherlands.17 These findings were also reported in an in- predictive factors in multiple clinical trials. Elevated pe-
dependent cohort of 64 patients from the United Kingdom. ripheral blood neutrophil and monocyte counts were as-
Simeone et al18 also found that decreasing levels of serum sociated with poor survival among patients with metastatic
LDH and CRP between baseline and the end of ipilimumab melanoma20 and were an independent prognostic factor for
treatment at week 12 were significantly associated with short OS among patients with stage IV melanoma un-
both disease control and survival for 95 patients treated with dergoing IL-2based immunotherapy in the retrospective
ipilimumab (p , .0001). Although they are often commer- European Organisation for the Research and Treatment of
cially available, these laboratory assessments are not fac- Cancer 18951 study.21 Interestingly, neutrophil and mono-
tored into current clinical algorithms for making treatment cyte counts did not predict clinical benefit in a recent ret-
decisions. Another soluble factor that has been proposed is rospective trial of 36 patients treated with ipilimumab,
baseline serum concentrations of soluble CD25, the alpha although an early increase in eosinophil count was associ-
chain of the IL-2 receptor, which independently predicted ated with improved clinical benefit,22 a finding reported
OS in a large retrospective trial of 262 patients. Low levels previously23 that can perhaps be explained as a sign of a
of CD25 were associated with favorable outcomes and ele- developing inflammatory response. In this analysis, myeloid-
vated levels predicted resistance to ipilimumab therapy, a derived suppressor cells (MDSCs), a heterogeneic immu-
finding rooted in preclinical studies demonstrating that noregulatory population of immune cells, were also negatively
blockade of the IL-2 receptor subunits abrogated the anti- correlated with clinical benefit. This finding was consistent with
tumor efficacy of CLTA-4 blockade.19 It is not currently clear reports of poorer prognosis for patients with increased MDSCs

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e495


SPENCER ET AL

during the first 24 weeks of treatment and improved outcome large numbers of T cells at the tumor periphery, with in-
with low baseline MDSC counts.24,25 creased expression of T-cell activation markers, type 1 in-
Lymphocytes are perhaps the most frequently studied terferon signatures, and high levels of Th1 cytokines and
peripheral blood component as predictors of response to chemokines that can effectuate T-cell recruitment and ef-
immunotherapy. It is well established that initial lympho- fector functions. In contrast, the nonT cellinflamed tumor
penia and rebound lymphocytosis are hematologic side microenvironment has few, if any, effector T cells, although
effects of IL-2. A strong positive association between clinical these tumors may contain evidence of chronic inflammation
response and the degree of lymphocytosis immediately after with tumor-associated macrophages, MDSCs, Th2 cytokines,
therapy has been reported.26 In a study of ipilimumab and chemokines, resulting in an immunosuppressed mi-
treatment, decreasing regulatory CD4+FoxP3+ T cells and croenvironment that allows tumor progression. The precise
increasing absolute lymphocyte counts (ALCs) during underlying mechanisms that mediate the type of immune
treatment were significantly associated with both disease microenvironment in cancer are not completely understood.
control and survival.18 This same trend of increasing ALCs Nonetheless, there have been several tumor-derived soluble
correlating with improved clinical benefit has been reported and cell membrane factors that may be responsible for the
elsewhere,23 but a pooled analysis of data from several observed phenotypic differences. For example, some tumor
studies noted this increase among all patients treated re- cells in an inflamed environment will express high levels of
gardless of benefit.27 Martens et al28 recently reported a T-cell checkpoints, such as PD-L1, B7H4, Tim-3, Lag-3, and
composite analysis suggesting that a signature of low LDH, others that can disable tumor-infiltrating effector cells.
absolute monocyte counts, high relative lymphocyte counts, Furthermore, various metabolic changes within inflamed
eosinophil counts, and regulatory T cells are associated with tumors, such as beta-catenin expression and intracellular
favorable outcomes among 209 patients, suggesting that hypoxia and metabolic demand, or release of soluble
evaluating multiple dynamic cell populations may be nec- factors, such as indoleamine-2,3-dioxygenase, IL-10, VEGF,
essary to achieve predictive power. and transforming growth factor-beta may result in in-
Finally, the presence of induced autoimmunity has long hibition of effector T cells. Some tumors may also exhibit
been hypothesized to predict clinical benefit from immu- high levels of CD4+FoxP3+ regulatory T cells. These events
notherapy. Metastatic melanoma has been associated with have a cumulative effect of ultimately preventing effector
spontaneous antibody formation to a variety of common T-cell function despite their abundance (Fig. 1).31 The
tumor antigens, including differentiation antigens gp100, importance of T cells within the tumors has been under-
tyrosinase, and Melan-A/melanoma antigen recognized by scored by the prognostic influence of such cells in a variety of
T-cells 1 (MART-1), as well as cancer/testis antigens mela- solid tumors, including colorectal, hepatocellular, gallblad-
noma associated antigen 3 (MAGE-3) or NY-esophageal der, pancreatic, esophageal, ovarian, endometrial, cervical,
cancer 1 (NY-ESO-1). As many as 50% of patients with ad- bladder, nonsmall cell lung, prostate, head and neck, and
vanced melanoma with NY-ESO-1expressing tumors spon- breast cancer.33-36
taneously develop antibody.29 The patients with NY-ESO-1 Currently, there is some controversy about which immune
seropositivity at baseline showing responses or with sero- cells are important in terms of promoting antitumor im-
negativity showing a seroconversion had a greater likelihood munity versus tumor progression. The most important cells
of experiencing clinical benefit 24 weeks after treatment for promoting antitumor immunity are likely CD8+ cytotoxic
with ipilimumab (an antiCTLA-4 antibody) than their se- T cells, CD4+ Th1 cells, NK cells, and mature dendritic cells.
ronegative counterparts.30 The tumors with this lymphocyte-predominant infiltrate
pattern do seem to benefit more from tumor immunotherapy
than cancers without these tumor-infiltrating lymphocytes
TUMOR CELL PHENOTYPE AND TUMOR (Fig. 1).31 The elegant work of Galon et al34 suggests that when
MICROENVIRONMENT BIOMARKERS colorectal cancer lesions were analyzed for simply CD3+ T cells
Although blood is more easily accessible, there is clear
or CD8+ T cells, a significant improvement in overall survival
evidence that activity within the tumor microenvironment
was seen. In a different series, T cellinfiltrated melanomas,
provides important clues as to how an individual tumor and
especially those with high CD8+ T-cell content, were more
patient may respond to immunotherapy.
likely to be associated with high PD-L1 expression, improved
prognosis, and longer time to development of brain metas-
Tumor-Infiltrating Immune Cells as Biomarkers tases.37 The presence of intratumoral CD3+ T cells is also
Tumors with an active immune microenvironment, as ex- correlated with improved progression-free survival (PFS)
emplified by infiltration of activated, effector T cells, may be and OS among patients with advanced ovarian carcinoma.38
better primed to respond to immunotherapy. Indeed, an- High peritumoral densities of infiltration of lymphocytes
alyses of melanoma samples revealed that two distinct expressing the T-cell activation markers CD25 or OX40 were
phenotypic classes of tumor microenvironments exist: those associated with longer survival of patients with cutaneous
with a high prevalence of T cells (T-cell predominant or T-cell malignant melanoma.39 Concurrent high CD8+ and CD4+ T-cell
inflamed) and those without T cells (T-cell poor or nonT-cell infiltration in nonsmall cell lung carcinoma was found to be
inflamed; Fig. 1).31,32 The T cellinflamed tumors harbor an independent favorable prognostic factor.40

e496 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


BIOMARKERS FOR IMMUNOTHERAPY

FIGURE 1. Tumors Exhibiting a T CellInflamed or Non-T-Cell-Inflamed Phenotype

(Top) Some tumors exhibit a T cellinflamed phenotype. In these tumors, a large number of tumor-infiltrating lymphocytes (TILs) and chemokines that recruit T cells are found.
Negative immune regulators including Fox P3+ regulatory T cells, PD-L1, and indoleamine-2,3-dioxygenase (IDO) are present as well; a type 1 interferon signature may also be present.
(Bottom) In contrast, some tumors exhibit a non-T-cellinflamed phenotype with an inverse pattern in which TILs are absent but chronic inflammation, as evidenced by common
suppressive cytokines, tumor-associated macrophages, and myeloid-derived suppressor cells (MDSCs), exists.31,73

Additional studies of markers of immune activation, such prognosis or lack of response to immunotherapy. Th17 cells
as inducible T-cell costimulator (ICOS), a T cellspecific are also controversial with mixed correlations in clinical
molecular that belongs to the CD28/CTLA-4 family and is reports.
expressed only after T-cell activation, have been performed.
ICOS is thought to play an especially important role in T-cell
PD-1/PD-L1 Biomarkers in Development
survival, proliferation, and generation of memory T cells. In a Among the most important potential biomarkers of the last
neoadjuvant trial of 12 patients with bladder cancer treated few years are PD-1 and PD-L1 (Table 2). PD-1 is expressed in
with ipilimumab, investigators demonstrated an increase in the majority of tumor-infiltrating T cells, including antigen-
peripheral and intratumoral ICOShighCD4+ T cells, which may specific CD8+ T cells. PD-1 expression occurs after T-cell
have an association with good clinical responses to ipili- activation and has been used as a marker of T-cell ex-
mumab therapy.41 Similar findings have been reported in haustion. The PD-1 receptor binds to two ligands, which have
patients with prostate cancer, breast cancer, or mesothe- been designated PD-L1 and PD-L2. When PD-1 is engaged, it
lioma treated with ipilimumab or tremelimumab.42-45 A results in T-cell elimination and PD-1 is thus considered a
statistically significant increase in the frequency of ICOS+ T-cell checkpoint whose physiologic function is likely designed
CD4+ T cells measured by flow cytometry in the peripheral to eliminate activated T cells once they have completed their
blood from patients with melanoma was found after ipili- effector functions. When the interaction between PD-1 and
mumab treatment.46 These data suggest that the frequency PD-L1 is inhibited (for example, with antiPD-1 or antiPD-L1
of ICOS+ CD4+ T cells may be useful in monitoring the biologic antibodies), T cells remain activated, mediating antitumor
activity of antiCTLA-4 therapy. activity.47 Interestingly, PD-L1 expression has been detected
In contrast with the lymphocytes and activation markers in several different types of tumor cells, including mela-
mentioned thus far, the presence of CD4+FoxP3+ regulatory noma, nonsmall cell lung cancer, RCC, bladder cancer,
T cells, MDSCs, tumor-associated macrophages, and Th2- gastric cancer, ovarian cancer, B-cell lymphoma cells, Merkel
type cytokine profiles has been associated with poor cell carcinoma, and Hodgkin lymphoma (Table 2).

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e497


SPENCER ET AL

TABLE 2. Summary of PD-L1 Expression and Response to Therapy in Various Clinical Trials

First Author Tumor Type Therapy Cutoff (%) Biomarker Results


48
Topalian Advanced melanoma, NSCLC, Pembrolizumab 5 0 of 17 patients with PD-L1negative tumors had
CRPC, RCC, and CRC objective response
Borghaei51 Advanced nonsquamous NSCLC Nivolumab vs. 1, 5, and 10 Nivolumab had superior efficacy to docetaxel, greater
docetaxel with higher tumor membrane PD-L1 expression
Muro80 Gastric Pembrolizumab 1 Tumor PD-L1 expression was associated with ORR
Taube53 Melanoma, NSCLC, RCC, Nivolumab 5 Tumor cell PD-L1 expression correlated with objective
CRC, CRPC response
Disis68 Recurrent/refractory ovarian Avelumab 1 Trend toward better response rates in PD-L1positive
cancer tumors
Garon49 Advanced NSCLC Pembrolizumab 50 PD-L1 expression in at least 50% of tumor cells correlated
with improved efficacy
Powles69 Bladder Atezolizumab 1, 5, and 10 PD-L1positive tumors at . 5% had particularly high
(antiPD-L1) response rates
Weber66 Advanced melanoma progressed Nivolumab vs. 5 Higher response rates with nivolumab correlated with
on antiCTLA-4 therapy investigators positive tumor PD-L1 expression, but patients with
choice PD-L1negative tumors still had benefit
Weber65 Advanced melanoma progressed Nivolumab 1 and 5 PD-L1 positivity correlated significantly with better
on prior therapy/CTLA-4 therapy response but negativity did not rule out response
Kefford67 Melanoma Pembrolizumab 1 PD-L1 positivity associated with improved ORR and
PFS, but activity observed in patients with low PD-L1
expression
Robert70 Metastatic melanoma Nivolumab vs. 5 Nivolumab-treated patients had improved objective
dacarbazine response rate and overall survival, regardless of
PD-L1 status
Motzer71 Metastatic RCC Nivolumab 1 and 5 Response rates were higher with greater PD-L1
expression ($ 5%), but those with lower expression
(, 5%) also had meaningful responses
Brahmer50 Advanced progressed squamous Nivolumab vs. 1, 5, and 10 Expression of PD-L1 was neither prognostic nor
NSCLC docetaxel predictive of benefit
Herbst57 Advanced melanoma, NSCLC, Atezolizumab 5 Response correlated with PD-L1 expression by
RCC, and other tumor-infiltrating immune cells, but correlation
between response and PD-L1 expression by tumor cells
was not significant
Abbreviations: NSCLC, nonsmall cell lung cancer; CRPC, castration-resistant prostate cancer; RCC, renal cell carcinoma; CRC, colorectal cancer; ORR, overall response rate; PFS,
progression-free survival.

In early phase I clinical studies of PD-1 blockade, clinical There are multiple challenges with using PD-1 and PD-L1
responses were noted among patients with melanoma, RCC, expression as biomarkers. First, PD-L1 expression is heter-
and nonsmall cell lung cancer.48 In many studies, a cor- ogenous and dynamic within an individual. PD-L1 expression
relation between clinical response and the presence of can be induced by activated tumor-specific T cells, further
PD-L1 expression on tumor cells was reported, although demonstrating that PD-L1 expression is a dynamic, not a
this was not seen in every trial (Table 2). Based on this static, process.52,53 PD-L1 expression is often heterogeneous
unexpected finding, some clinical trials used baseline within a sample, thus using focal expression may reflect the
PD-L1 expression as a criterion for study participation. biology of the tumor or may simply be sampling error.53
A clinical trial of pembrolizumab, a monoclonal antibody Furthermore, PD-L1 expression is often discordant between
that targets PD-1, was conducted in previously treated the primary lesion and its metastases, and a positive marker
patients with PD-L1 expressing nonsmall cell lung cancer in one may not be reflective of the other.48,52,54 Additionally,
and demonstrated an objective response rate of 19%, multiple cell types within a tumor besides the tumor cells
leading to FDA approval of the agent but only for patients themselves may express PD-L1, such as lymphocytes and
whose tumors stain positive for PD-L1. 49 Other clinical macrophages, potentially altering readings,55 especially if
studies using nivolumab, a comparable antiPD-1 agent, these inflammatory cells represent the host immune re-
also showed significant clinical responses in patients sponse to tumor.56 A higher number of PD-1 and PD-L1
with nonsmall cell lung cancer resulting in FDA approval expressing tumor-infiltrating CD8+ T cells was associated
without requiring pretreatment PD-L1 expression test- with clinical responses among patients with metastatic
ing because this was not used as a study eligibility melanoma treated with pembrolizumab.55 In a study eval-
criterion. 50,51 uating MPDL3280A, an antiPD-L1 antibody, in multiple

e498 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


BIOMARKERS FOR IMMUNOTHERAPY

cancer types, responses were observed in patients with assay as a companion diagnostic for the indication of
tumor-infiltrating immune cells expressing high levels of identifying patients with nonsmall cell lung cancer for
PD-L1.57 These results suggest that PD-1/PD-L1expressing treatment with pembrolizumab.49,61,62
tumor-infiltrating immune cells may be predictive of tumors The antiPD-1 antibody nivolumab also has a companion
likely to be responsive to T-cell checkpoint blockade and may diagnostic PD-L1 assay. PD-L1 IHC 28-8 pharmDx uses the
serve as a predictive biomarker. In most of these studies, monoclonal antibody 28-8 to score tumor cell membranes in
however, there was a low rate of clinical response observed formalin-fixed, paraffin-embedded tissue. Although the
in patients with tumors having low levels of PD-1/PD-L1 original CheckMate 057 study of nivolumab versus docetaxel
expression. Thus, the sensitivity and specificity must be was conducted among an unselected population of patients
better defined to avoid overtreatment of patients positive with advanced nonsquamous nonsmall cell lung cancer, a
for PD-1/PD-L1 who will not respond and undertreatment of retrospective analysis of the data showed that patients
patients negative for PD-1/PD-L1 who might respond. whose tumors expressed PD-L1 at a low threshold value of
1% or greater had a more significant improvement over
docetaxel than PD-L1negative patients across all end-
SPECIFIC PD-1/PD-L1 BIOMARKER ASSAYS points. However, the retrospective PD-L1 testing was only
Although the immunohistochemistry (IHC) assay for PD-L1 is performed on 78% of the patient specimens, which may
affordable and relatively easy to perform on tissue, it is introduce selection bias.51,61 This may be reflected in the
limited by factors such as adequacy and abundance of the premarket approval from the FDA, which states that PD-L1
sample. The PD-L1 ligand has only two hydrophilic regions expression as detected by the 28-8 pharmDx assay in non-
where antibodies bind, and IHC antibodies seemingly bind squamous nonsmall cell lung cancer may be associated
at a different site than therapeutic antibodies, possibly with enhanced survival from nivolumab.61,63 This assay was
compromising the efficacy of the technique.52,58,59 Different approved by the FDA in late 2015 in association with the
IHC antibodies and staining techniques introduce further expanded approval of nivolumab in previously treated
variability into the test and may influence positive or neg- nonsmall cell lung cancer.
ative test rates.52 Without standardization across assays, it The POLAR study examined the efficacy of the PD-L1 in-
becomes difficult to compare clinical trial results, especially hibitor atezolizumab versus docetaxel among patients with
if the assays details are not easily available. Indeed, in a advanced nonsmall cell lung cancer of squamous and non-
recent study by McLaughlin et al60 testing two antiPD-L1 squamous histologies. This study used an SP142 antibody to
rabbit monoclonal antibodies, nearly 25% of lung cancer the membranes of tumor cells and tumor-infiltrating immune
samples that were positive for PD-L1 with one antibody tested cells. The data confirmed the efficacy of atezolizumab in-
negative with the second. Especially pertinent to recent in- dependently of PD-L1 expression, but the superiority of
vestigations, different assessment methods such as reading atezolizumab over docetaxel was more pronounced with
tumor cells or tumor-infiltrating lymphocytes or both, dif- higher expressions of PD-L1 as measured by the SP142 assay.61,64
ferent scoring methods such as percent staining positive or As noted elsewhere, clinically meaningful responses have
H score, and different cutoff values set for positive or negative been seen among patients whose tumors have been defined
results (1%, 5%, 10%, or even 50%) prevent standardization as negative for PD-L149,65-71 (Table 2). In addition, among
across tumor types and same therapies.52 patients receiving combination checkpoint inhibitor ther-
In October 2015, the FDA gave the first regulatory approval apy, responses among patients with PD-L1positive and
to an immunotherapy biomarker to Mercks PD-L1 com- PD-L1negative results were similar.72 Given that a negative
panion diagnostic assay. This assay, PD-L1 IHC 22C3 result may still have considerable variability and may
pharmDx, uses the monoclonal antibody clone 22C3 for exclude a patient from a therapy that could confer significant
membrane staining of tumor and/or infiltrating immune clinical benefit, perhaps the more appropriate immediate
cells in formalin-fixed, paraffin-embedded samples, with use of a PD-L1 biomarker is not to exclude patients with
a positivity cutoff of 1% or greater. The biomarker was negative results from treatment, but to better refine pre-
evaluated in the KEYNOTE-001 study of pembrolizumab in dictors of response by evaluating PD-L1 expression in
patients with advanced nonsmall cell lung cancer. To combination with other putative predictive markers to
clinically validate the assay, 495 patients were assigned better improve the sensitivity and specificity.73
either to a training group (one-third) or a validation group
(two-thirds). Data obtained from the training group were
used to establish the cutoff for PD-L1 positivity, which was TUMOR GENOMIC BIOMARKERS
ultimately defined as expression in 50% of tumor cells. The The ability of T-cell checkpoint inhibitors to induce an ef-
outcomes data from the validation group were analyzed fective immune response in some cancers raises the critical
using the 50% cutoff defined by the training group. Patients question of what antigen(s) are being targeted by the ac-
with a score of 50% or greater had a higher response rate, tivated T cells. It was originally hypothesized that the
longer PFS, and longer OS. Given the link between PD-L1 mechanism of antitumor activity might be through stoking
expression defined by the PD-L1 IHC 22C3 pharmDx assay pre-existing, ineffective tumor-reactive T cells. In several
and efficacy with pembrolizumab, the FDA approved the recent studies, however, it has become clear that clinical

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e499


SPENCER ET AL

responses to T-cell checkpoint inhibitors are correlated with with MMR-proficient colorectal cancer responded to ther-
overall mutation burden in the tumor cell.7,8,74,75 This apy, 40% of patients (four of 10) with MMR-deficient co-
finding suggested that the T-cell response may be targeted lorectal cancer and 71% of patients (five of seven) with
not to established antigens, but to so-called neoantigens, MMR-deficient noncolorectal cancer had immune-related
that evolve as the mutation rate increases in the tumor cell. objective responses.76,77 Further WES revealed much higher
Most of these mutations are likely passenger mutations somatic mutation loads with a mean of 1,782 mutations per
that may be influencing the immuno-editing process and tumor in MMR-deficient tumors, compared with a mean of
placing selective pressure on the immune system to pro- only 73 mutations per tumor in MMR-proficient tumors.
mote tumor elimination through new, previously unedited Mutations in other enzymes involved in DNA replication and
antigens. Although this is an intriguing hypothesis, further repair, such as the DNA polymerase epsilon gene (POLE) and
work is needed to firmly establish whether mutation burden DNA polymerase delta 1 (POLD1), have been implicated in
or neoantigen emergence can be predictive biomarkers for the generation of a high mutation burden and associated
tumor response to immunotherapy agents. with response to immunotherapy. The POLE mutation is
A recent study using whole-exome sequencing (WES) to associated with disruption of the exonuclease activity re-
analyze the potential effect of cancer genomes on the re- quired for proofreading function and results in a high mu-
sponse to ipilimumab among 64 patients with melanoma tational burden or ultramutator phenotype.78 Both POLE
confirmed that a high mutational load and the number of and POLD1 mutations have been noted among patients with
nonsynonymous mutations per exome were significantly nonsmall cell lung cancer who are responsive to pem-
correlated with improved OS.75 Using a bioinformatics brolizumab, and POLE and POLD1 are associated with high
pipeline, the authors identified 101 tetrapeptide (four amino mutational burdens.7 In addition, a case of an exceptional
acid) motifs within the nonamer (nine amino acid) peptides response to pembrolizumab for a patient with endometrial
sitting in the peptide-binding grooves formed by major cancer whose tumor had a somatic POLE mutation was
histocompatibility complex class I molecules. These tetra- recently reported.79 These data suggest that the presence of
peptides were shared exclusively among patients in the MMR deficiency or POLE mutation may identify a subset of
discovery cohort who had long-term clinical benefit. A re- seemingly diverse cancers that are especially vulnerable to
cent correction to the original report clarified that this immune checkpoint blockade, although this approach must
analysis did not have a true independent validation set be validated in further prospective studies.
because all samples were used in the process of identifying
the final set of neoantigens.74
Indeed, mutation burden may be as predictive as neo- FUTURE DIRECTIONS
antigen load in the landscape of response to immuno- The progress in tumor immunotherapy over the last 5 years
therapy. Rizvi et al7 observed that a high mutational burden has been truly remarkable, with several monotherapy and
may be predictive of response to immunotherapy. Using combination therapy regimens demonstrating impressive
WES of nonsmall cell lung cancers treated with pem- clinical benefit across numerous solid and hematologic
brolizumab, this study revealed higher nonsynomymous malignancies. The ability to have a predictive biomarker to
mutation burden in tumors, and clinical responses were better select appropriate patients for specific treatment
correlated with molecular signature characteristics of to- regimens and help define prognosis and other important
bacco carcinogen-related mutagenesis, higher neoantigen patient outcomes is a high priority in the field. Although
burden, and DNA repair pathway mutations. In one re- several soluble factors and cell-surface receptors in the
sponder, pembrolizumab enhanced neoantigen-specific peripheral blood and tumor microenvironments have been
CD8+ T-cell reactivity that was associated with tumor re- proposed, none have yet been fully validated in prospective,
gression. In a series of 110 patients with melanoma, overall randomized clinical trials. Putative biomarkers of interest
mutation load, neoantigen load, and expression of immune include the number and location of tumor-infiltrating im-
microenvironment cytolytic markers were associated with mune cells, PD-1 and PD-L1 expression, and host genomic
clinical benefit, but no recurrent neoantigen peptide se- factors, such as mutation burden and neoantigen emer-
quences predicted responder patient populations.8 There- gence. Biomarker development also depends on having stan-
fore, although it is intriguing, the neoantigen hypothesis dardized, reproducible, and feasible assays that can be rapidly
needs further validation in other cohorts. completed with clearly defined cutoff values and with high
Although heavy mutation burdens may be seen in lung and sensitivity and specificity. Although it may be premature to
melanoma tumors, which are often generated by exogenous recommend any of the current biomarkers for routine clinical
environmental insults such as tobacco and ultraviolet light, practice, there should be a high priority to include biomarkers
gene mutations that beget mutations, such as DNA mis- in clinical trial design for ongoing investigations of tumor im-
match repair (MMR) gene deficiency, can also result in a munotherapy regimens. Combination strategies and/or larger
heavy mutational burden and may be useful to predict re- panels may be of special interest for improving the predictive
sponse to immunotherapy. A phase II study evaluated the power of contemporary biomarkers. As the use of immu-
response to pembrolizumab therapy among patients with notherapy is further optimized and novel combination
MMR-deficient cancers. Although 0% of patients (zero of 18) approaches are developed, biomarker investigation will

e500 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


BIOMARKERS FOR IMMUNOTHERAPY

assume a critical role to inform the rational selection of ACKNOWLEDGMENT


patients for this treatment approach. K.R.S. and J.W. contributed equally to this work.

References
1. Dizon DS, Krilov L, Cohen E, et al. Clinical cancer advances 2016: annual 19. Hannani D, Vetizou M, Enot D, et al. Anticancer immunotherapy by
report on progress against cancer from the American Society of Clinical CTLA-4 blockade: obligatory contribution of IL-2 receptors and negative
Oncology. J Clin Oncol. 2016;34:987-1011. prognostic impact of soluble CD25. Cell Res. 2015;25:208-224.
2. Ansell SM, Lesokhin AM, Borrello I, et al. PD-1 blockade with nivolumab 20. Schmidt H, Bastholt L, Geertsen P, et al. Elevated neutrophil and
in relapsed or refractory Hodgkins lymphoma. N Engl J Med. 2015;372: monocyte counts in peripheral blood are associated with poor survival
311-319. in patients with metastatic melanoma: a prognostic model. Br J Cancer.
3. Gettinger SN, Horn L, Gandhi L, et al. Overall survival and long-term 2005;93:273-278.
safety of nivolumab (anti-programmed death 1 antibody, BMS-936558, 21. Schmidt H, Suciu S, Punt CJ, et al; American Joint Committee on Cancer
ONO-4538) in patients with previously treated advanced non-small-cell Stage IV Melanoma; EORTC 18951. Pretreatment levels of peripheral
lung cancer. J Clin Oncol. 2015;33:2004-2012. neutrophils and leukocytes as independent predictors of overall survival
4. Larkin J, Lao CD, Urba WJ, et al. Efficacy and safety of nivolumab in in patients with American Joint Committee on Cancer Stage IV Mela-
patients with BRAF V600 mutant and BRAF wild-type advanced mel- noma: results of the EORTC 18951 Biochemotherapy Trial. J Clin Oncol.
anoma: a pooled analysis of 4 clinical trials. JAMA Oncol. 2015;1: 2007;25:1562-1569.
433-440. 22. Gebhardt C, Sevko A, Jiang H, et al. Myeloid cells and related chronic
5. Postow MA, Chesney J, Pavlick AC, et al. Nivolumab and ipilimumab inflammatory factors as novel predictive markers in melanoma treat-
versus ipilimumab in untreated melanoma. N Engl J Med. 2015;372: ment with ipilimumab. Clin Cancer Res. 2015;21:5453-5459.
2006-2017. 23. Delyon J, Mateus C, Lefeuvre D, et al. Experience in daily practice with
6. Schumacher TN, Kesmir C, van Buuren MM. Biomarkers in cancer ipilimumab for the treatment of patients with metastatic melanoma: an
immunotherapy. Cancer Cell. 2015;27:12-14. early increase in lymphocyte and eosinophil counts is associated with
7. Rizvi NA, Hellmann MD, Snyder A, et al. Cancer immunology. Mutational improved survival. Ann Oncol. 2013;24:1697-1703.
landscape determines sensitivity to PD-1 blockade in non-small cell lung 24. Kitano S, Postow MA, Cortez C, et al. Myeloid-derived suppressor cell
cancer. Science. 2015;348:124-128. quantity prior to treatment with ipilimumab at 10mg/kg to predict for
8. Van Allen EM, Miao D, Schilling B, et al. Genomic correlates of response overall survival in patients with metastatic melanoma. J Clin Oncol.
to CTLA-4 blockade in metastatic melanoma. Science. 2015;350: 2012;30 (suppl; abstr 2518).
207-211. 25. Meyer C, Cagnon L, Costa-Nunes CM, et al. Frequencies of circulating
9. Strimbu K, Tavel JA. What are biomarkers? Curr Opin HIV AIDS. 2010;5: MDSC correlate with clinical outcome of melanoma patients treated
463-466. with ipilimumab. Cancer Immunol Immunother. 2014;63:247-257.
10. Fox BA, Schendel DJ, Butterfield LH, et al. Defining the critical hurdles in 26. Phan GQ, Attia P, Steinberg SM, et al. Factors associated with response
cancer immunotherapy. J Transl Med. 2011;9:214. to high-dose interleukin-2 in patients with metastatic melanoma. J Clin
11. Nelson D, Fisher S, Robinson B. The Trojan Horse approach to tumor Oncol. 2001;19:3477-3482.
immunotherapy: targeting the tumor microenvironment. J Immunol 27. Postow MA, Chasalow SD, Yuan J, et al. Pharmacodynamic effect of
Res. 2014;2014:789069. ipilimumab on absolute lymphocyte count (ALC) and association with
12. Das B, Tsuchida R, Malkin D, et al. Hypoxia enhances tumor stemness by overall survival in patients with advanced melanoma. J Clin Oncol. 2013;
increasing the invasive and tumorigenic side population fraction. Stem 31 (suppl; abstr 9052).
Cells. 2008;26:1818-1830. 28. Martens A, Wistuba-Hamprecht K, Geukes Foppen MH, et al. Baseline
13. Blay JY, Negrier S, Combaret V, et al. Serum level of interleukin 6 as a peripheral blood biomarkers associated with clinical outcome of ad-
prognosis factor in metastatic renal cell carcinoma. Cancer Res. 1992; vanced melanoma patients treated with ipilimumab. Clin Cancer Res.
52:3317-3322. Epub 2016 Jan 19.
14. Tartour E, Blay JY, Dorval T, et al. Predictors of clinical response to 29. Gnjatic S, Nishikawa H, Jungbluth AA, et al. NY-ESO-1: review of an
interleukin-2based immunotherapy in melanoma patients: a French immunogenic tumor antigen. Adv Cancer Res. 2006;95:1-30.
multiinstitutional study. J Clin Oncol. 1996;14:1697-1703. 30. Yuan J, Adamow M, Ginsberg BA, et al. Integrated NY-ESO-1 antibody
15. Sabatino M, Kim-Schulze S, Panelli MC, et al. Serum vascular endothelial and CD8+ T-cell responses correlate with clinical benefit in advanced
growth factor and fibronectin predict clinical response to high-dose melanoma patients treated with ipilimumab. Proc Natl Acad Sci USA.
interleukin-2 therapy. J Clin Oncol. 2009;27:2645-2652. 2011;108:16723-16728.
16. Yuan J, Zhou J, Dong Z, et al. Pretreatment serum VEGF is associated 31. Gajewski TF, Schreiber H, Fu YX. Innate and adaptive immune cells in the
with clinical response and overall survival in advanced melanoma tumor microenvironment. Nat Immunol. 2013;14:1014-1022.
patients treated with ipilimumab. Cancer Immunol Res. 2014;2: 32. Spranger S, Spaapen RM, Zha Y, et al. Up-regulation of PD-L1, IDO, and T
127-132. (regs) in the melanoma tumor microenvironment is driven by CD8(+)
17. Kelderman S, Heemskerk B, van Tinteren H, et al. Lactate de- T cells. Sci Transl Med. 2013;5:200ra116.
hydrogenase as a selection criterion for ipilimumab treatment in 33. Mlecnik B, Tosolini M, Kirilovsky A, et al. Histopathologic-based
metastatic melanoma. Cancer Immunol Immunother. 2014;63:449-458. prognostic factors of colorectal cancers are associated with the state
18. Simeone E, Gentilcore G, Giannarelli D, et al. Immunological and bi- of the local immune reaction. J Clin Oncol. 2011;29:610-618.
ological changes during ipilimumab treatment and their potential 34. Galon J, Costes A, Sanchez-Cabo F, et al. Type, density, and location of
correlation with clinical response and survival in patients with advanced immune cells within human colorectal tumors predict clinical outcome.
melanoma. Cancer Immunol Immunother. 2014;63:675-683. Science. 2006;313:1960-1964.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e501


SPENCER ET AL

35. Pag`es F, Berger A, Camus M, et al. Effector memory T cells, early for anti-PD-1/PD-L1 clinical trials. Pigment Cell Melanoma Res. 2015;28:
metastasis, and survival in colorectal cancer. N Engl J Med. 2005;353: 245-253.
2654-2666. 55. Tumeh PC, Harview CL, Yearley JH, et al. PD-1 blockade induces re-
36. Jochems C, Schlom J. Tumor-infiltrating immune cells and prognosis: the sponses by inhibiting adaptive immune resistance. Nature. 2014;515:
potential link between conventional cancer therapy and immunity. Exp 568-571.
Biol Med (Maywood). 2011;236:567-579. 56. Bhaijee F, Anders RA. PD-L1 expression as a predictive biomarker: is
37. Kluger HM, Zito CR, Barr ML, et al. Characterization of PD-L1 expression absence of proof the same as proof of absence? JAMA Oncol. 2016;2:
and associated T-cell infiltrates in metastatic melanoma samples from 54-55.
variable anatomic sites. Clin Cancer Res. 2015;21:3052-3060. 57. Herbst RS, Soria J-C, Kowanetz M, et al. Predictive correlates of response
38. Zhang L, Conejo-Garcia JR, Katsaros D, et al. Intratumoral T cells, re- to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature.
currence, and survival in epithelial ovarian cancer. N Engl J Med. 2003; 2014;515:563-567.
348:203-213. 58. Patel SP, Kurzrock R. PD-L1 expression as a predictive biomarker in
39. Ladanyi A, Somlai B, Gilde K, et al. T-cell activation marker expression on cancer immunotherapy. Mol Cancer Ther. 2015;14:847-856.
tumor-infiltrating lymphocytes as prognostic factor in cutaneous ma- 59. Sznol M, Chen L. Antagonist antibodies to PD-1 and B7-H1 (PD-L1) in the
lignant melanoma. Clin Cancer Res. 2004;10:521-530. treatment of advanced human cancer. Clin Cancer Res. 2013;19:
40. Hiraoka K, Miyamoto M, Cho Y, et al. Concurrent infiltration by CD8+ 1021-1034.
T cells and CD4+ T cells is a favourable prognostic factor in non-small-cell 60. McLaughlin J, Han G, Schalper KA, et al. Quantitative assessment of the
lung carcinoma. Br J Cancer. 2006;94:275-280. heterogeneity of PD-L1 expression in non-small-cell lung cancer. JAMA
41. Carthon BC, Wolchok JD, Yuan J, et al. Preoperative CTLA-4 blockade: Oncol. 2016;2:46-54.
tolerability and immune monitoring in the setting of a presurgical 61. Jrgensen JT. Companion diagnostic assays for PD-1/PD-L1 checkpoint
clinical trial. Clin Cancer Res. 2010;16:2861-2871. inhibitors in NSCLC. Expert Rev Mol Diagn. 2016;16:131-133.
42. Calabro` L, Maio M. Immune checkpoint blockade in malignant meso- 62. U.S. Food and Drug Administration. Premarket approval (PMA) for
thelioma. Semin Oncol. 2015;42:418-422. PD-L1 IHC 22C3 pharmDx. http://www.accessdata.fda.gov/scripts/
43. Vonderheide RH, LoRusso PM, Khalil M, et al. Tremelimumab in cdrh/cfdocs/cfpma/pma.cfm?id=P150013. Accessed October 2, 2015.
combination with exemestane in patients with advanced breast cancer 63. U.S. Food and Drug Administration. Premarket approval (PMA) for
and treatment-associated modulation of inducible costimulator ex- PD-L1 IHC nivolumab pharmDx. http://www.accessdata.fda.gov/scripts/
pression on patient T cells. Clin Cancer Res. 2010;16:3485-3494. cdrh/cfdocs/cfpma/pma.cfm?id=P150025. Accessed October 9, 2015.
44. Chen H, Liakou CI, Kamat A, et al. Anti-CTLA-4 therapy results in higher 64. Spira AI, Park K, Mazieres J, et al. Efficacy, safety and predictive bio-
CD4+ICOShi T cell frequency and IFN-g levels in both nonmalignant and marker results from a randomized phase II study comparing
malignant prostate tissues. Proc Natl Acad Sci USA. 2009;106: MPDL3280A vs docetaxel in 2L/3L NSCLC (POPLAR). J Clin Oncol. 2015;
2729-2734. 33 (suppl; abstr 8010).
45. Liakou CI, Kamat A, Tang DN, et al. CTLA-4 blockade increases 65. Weber JS, Kudchadkar RR, Yu B, et al. Safety, efficacy, and biomarkers of
IFNgamma-producing CD4+ICOShi cells to shift the ratio of effector to nivolumab with vaccine in ipilimumab-refractory or -naive melanoma.
regulatory T cells in cancer patients. Proc Natl Acad Sci USA. 2008;105: J Clin Oncol. 2013;31:4311-4318.
14987-14992. 66. Weber JS, DAngelo SP, Minor D, et al. Nivolumab versus chemotherapy
46. Ng Tang D, Shen Y, Sun J, et al. Increased frequency of ICOS+ CD4 T cells in patients with advanced melanoma who progressed after anti-CTLA-4
as a pharmacodynamic biomarker for anti-CTLA-4 therapy. Cancer treatment (CheckMate 037): a randomised, controlled, open-label,
Immunol Res. 2013;1:229-234. phase 3 trial. Lancet Oncol. 2015;16:375-384.
47. Ahmadzadeh M, Johnson LA, Heemskerk B, et al. Tumor antigen-specific 67. Kefford R, Ribas A, Hamid O, et al. Clinical efficacy and correlation with
CD8 T cells infiltrating the tumor express high levels of PD-1 and are tumor PD-L1 expression in patients (pts) with melanoma (MEL) treated
functionally impaired. Blood. 2009;114:1537-1544. with the anti-PD-1 monoclonal antibody MK-3475. J Clin Oncol. 2014;
48. Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune 32:5s (suppl; abstr 3005).
correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366: 68. Disis ML, Patel MR, Pant S, et al. Avelumab (MSB0010718C), an anti-PD-
2443-2454. L1 antibody, in patients with previously treated, recurrent or refractory
49. Garon EB, Rizvi NA, Hui R, et al; KEYNOTE-001 Investigators. Pem- ovarian cancer: a phase Ib, open-label expansion trial. J Clin Oncol. 2015;
brolizumab for the treatment of non-small-cell lung cancer. N Engl J 33 (suppl; abstr 5509).
Med. 2015;372:2018-2028. 69. Powles T, Eder JP, Fine GD, et al. MPDL3280A (anti-PD-L1) treatment
50. Brahmer J, Reckamp KL, Baas P, et al. Nivolumab versus docetaxel in leads to clinical activity in metastatic bladder cancer. Nature. 2014;515:
advanced squamous-cell nonsmall-cell lung cancer. N Engl J Med. 558-562.
2015;373:123-135. 70. Robert C, Long GV, Brady B, et al. Nivolumab in previously untreated
51. Borghaei H, Paz-Ares L, Horn L, et al. Nivolumab versus docetaxel in melanoma without BRAF mutation. N Engl J Med. 2015;372:320-330.
advanced nonsquamous nonsmall-cell lung cancer. N Engl J Med. 71. Motzer RJ, Rini BI, McDermott DF, et al. Nivolumab for metastatic renal
2015;373:1627-1639. cell carcinoma: results of a randomized phase II trial. J Clin Oncol. 2015;
52. Meng X, Huang Z, Teng F, et al. Predictive biomarkers in PD-1/PD-L1 33:1430-1437.
checkpoint blockade immunotherapy. Cancer Treat Rev. 2015;41: 72. Wolchok JD, Kluger H, Callahan MK, et al. Nivolumab plus ipilimumab in
868-876. advanced melanoma. N Engl J Med. 2013;369:122-133.
53. Taube JM, Anders RA, Young GD, et al. Colocalization of inflammatory 73. Mahoney KM, Atkins MB. Prognostic and predictive markers for the new
response with B7-h1 expression in human melanocytic lesions supports immunotherapies. Oncology (Williston Park). 2014;28:39-48.
an adaptive resistance mechanism of immune escape. Sci Transl Med. 74. Chan TA, Wolchok JD, Snyder A. Genetic basis for clinical response to
2012;4:127ra37. CTLA-4 blockade in melanoma. N Engl J Med. 2015;373:1984.
54. Madore J, Vilain RE, Menzies AM, et al. PD-L1 expression in melanoma 75. Snyder A, Makarov V, Merghoub T, et al. Genetic basis for clinical response
shows marked heterogeneity within and between patients: implications to CTLA-4 blockade in melanoma. N Engl J Med. 2014;371:2189-2199.

e502 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


BIOMARKERS FOR IMMUNOTHERAPY

76. Kelderman S, Schumacher TN, Kvistborg P. Mismatch repair-deficient 79. Mehnert JM, Panda A, Zhong H, et al. Exceptional response to PD-1
cancers are targets for anti-PD-1 therapy. Cancer Cell. 2015;28:11-13. antibody treatment in a POLE-mutant endometrial cancer (Abstract).
77. Le DT, Uram JN, Wang H, et al. PD-1 blockade in tumors with mismatch- Mol Cancer Ther. 2015;14:PR05.
repair deficiency. N Engl J Med. 2015;372:2509-2520. 80. Muro K, Bang YJ, Shankaran V, et al. Relationship between PD-L1 expression
78. Hussein YR, Weigelt B, Levine DA, et al. Clinicopathological analysis of and clinical outcomes in patients (Pts) with advanced gastric cancer treated
endometrial carcinomas harboring somatic POLE exonuclease domain with the anti-PD-1 monoclonal antibody pembrolizumab (Pembro;
mutations. Mod Pathol. 2015;28:505-514. MK-3475) in KEYNOTE-012. J Clin Oncol. 2015;33 (suppl 3; abstr 3).

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e503


MELANOMA/SKIN CANCERS

Curing High-Risk Melanoma: Are


We There Yet?

CHAIR
Sanjiv S. Agarwala, MD
St. Lukes Medical Center
Easton, PA

SPEAKERS
Alexander C.J. van Akkooi, MD, PhD
Netherlands Cancer Institute Antoni van Leeuwenhoek
Amsterdam, Netherlands

Michael B. Atkins, MD
Lombardi Comprehensive Cancer Center
Washington, DC

Paul Lorigan, MD
Institute of Cancer Sciences, University of Manchester
Manchester, United Kingdom
SURGICAL MANAGEMENT OF MELANOMA

Surgical Management and Adjuvant Therapy for High-Risk and


Metastatic Melanoma
Alexander C.J. van Akkooi, MD, PhD, Michael B. Atkins, MD, Sanjiv S. Agarwala, MD, and Paul Lorigan, MD

OVERVIEW

Wide local excision is considered routine therapy after initial diagnosis of primary melanoma to reduce local recurrences, but
it does not impact survival. Sentinel node staging is recommended for melanomas of intermediate thickness, but it has also
not demonstrated any indisputable therapeutic effect on survival. The prognostic value of sentinel node staging has been
long established and is therefore considered routine, especially in light of the eligibility criteria for adjuvant therapy (trials).
Whether completion lymph node dissection after a positive sentinel node biopsy improves survival is the question of current
trials. The MSLT-2 study is best powered to show a potential benefit, but it has not yet reported any data. Another study, the
German DECOG study, presented at the 2015 American Society of Clinical Oncology (ASCO) Annual Meeting did not show any
benefit but is criticized for the underpowered design and insufficient follow-up. There is no consensus on the use of adjuvant
interferon in melanoma. This topic has been the focus of many studies with different regimens (low-, intermediate-, or high-
dose and/or short- or long-term treatment). Adjuvant interferon has been shown to improve relapse-free survival but failed
to improve overall survival. More recently, adjuvant ipilimumab has also demonstrated an improved relapse-free survival.
Overall survival data have not yet been reported due to insufficient follow-up. Currently, studies are ongoing to analyze the
use of adjuvant antiPD-1 and molecular targeted therapies (vemurafenib, dabrafenib, and trametinib). In the absence of
unambiguously positive approved agents, clinical trial participation remains a priority. This could change in the near future.

S urvival rates differ according to the stage of melanoma,


and, even for the low stages of melanoma, the 10-year
survival rates differ largely, from 93% in stage IA to 39%
overall survival (OS). This was the topic of a few prospective
randomized controlled trials comparing 1-cm with 3-cm,
2-cm with 5-cm, and 2-cm with 4-cm resection margins.13-15
in stage IIC.1 Since the last update in the American Joint The World Health Organization trial by Cascinelli et al
Committee on Cancer (AJCC) staging system, a number of compared 1-cm margins with 3-cm margins for two Breslow
treatment breakthroughs have occurred in terms of im- thickness groups, less than 1-mm Breslow and 1.012.00-mm
provement in systemic therapy (e.g., immune checkpoint Breslow, and did not find any significant difference in local
inhibitors, BRAF/MEK inhibitors [BRAFi/MEKi]),2-12 which recurrence rate or in OS after a median follow-up of 12 years
may ultimately affect these survival rates. Nevertheless, among a total of 612 patients.13,14 The Swedish group
these survival rates illustrate the fact that already some compared 2-cm with 5-cm margins for 0.82.0 mm Breslow
patients with melanoma and only primary tumors without thickness tumors in 989 patients and found a nonsignificant
(nodal) metastases will be likely to progress and can be hazard ratio (HR) of 0.96 (95% CI, 0.751.24) for OS and 1.02
considered high risk. Moreover, it demonstrates the fact that (95% CI, 0.801.30) for recurrence-free survival after a me-
surgery can be curative for the majority of patients with dian follow-up of 11 years (range 717 years).15 Similar results
early-stage melanoma. are reported in the other studies by Khayat et al, Karakousis
et al, Thomas et al, and Gillgren et al.16-19
SURGICAL TREATMENT OF THE HIGH-RISK In contrast to these randomized controlled trials, Haydu
MELANOMA PATIENT: IS THERE A STANDARD? et al analyzed 2,131 patients with pT2 tumors from a single
Primary Tumor institution.20 They found that there was a significantly worse
After diagnosis, most physicians recommend performing disease-free survival for patients with a margin less than
a wide local excision with a certain margin. An important 8 mm on pathologic analysis (corresponding to , 1 cm
question is whether an increasing margin will improve clinically) compared with a 8- to 16-mm margin (corresponding

From the Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam, Netherlands; Georgetown Lombardi Comprehensive Cancer Center, Washington, DC; St. Lukes
University Hospital, Temple University, Allentown, PA; University of Manchester, The Christie NHS Foundation Trust, Manchester, United Kingdom.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Alexander C.J. van Akkooi, MD, PhD, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, Netherlands;
email: a.v.akkooi@nki.nl.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e505


VAN AKKOOI ET AL

to 12 cm clinically; p = .044), however, this did not translate disease-free survival and melanoma-specific survival for
into any OS difference.20 patients with nodal metastases from intermediate-thickness
Interestingly, an update of the prospective randomized melanomas.28 This is formally a completely correct con-
controlled trial by Thomas19 discussed at the 2015 ASCO clusion because there is a clear benefit of 12.3% (69.8%
Annual Meeting by Hayes et al21,22 did show, for the first [6 4.4%] for sentinel nodepositive versus 57.5% [6 5.4%]
time, a difference in melanoma-specific survival (HR 1.27; for observation nodepositive; HR 0.56; 95% CI, 0.370.84;
95% CI, 1.021.59; p = .036) in favor of the 3-cm margin p = .006) in melanoma-specific survival after 10 years.28
group compared with the 1-cm margin group. All patients in However, the study has been criticized for a number of
this U.K. study had melanomas with a Breslow thickness reasons. First, the benefit was only found in a post hoc
of greater than 2 mm, and 453 patients underwent a 1-cm subgroup analysis, but no difference in survival was seen
resection margin compared with 457 patients undergo- between the primary randomized study groups (wide local
ing a 3-cm resection margin. Although there were more excision plus observation versus wide local excision plus
melanoma-related deaths in the 1-cm resection margin sentinel node biopsy), with a 10-year melanoma-specific
group, the number of nonmelanoma-related deaths increased survival of 78.3% (6 2.0%) compared with 81.4% (6 1.5%),
in the 3-cm group. Therefore, the OS did not significantly differ respectively (p = .18).28,34 This is reflected in potential
between the two groups.22 At the same time, this study has overtreatment (minimal sentinel node tumor burden; prog-
also been criticized for the fact that patients were not staged nostic false positivity) and under-treatment (worse outcome
by sentinel node procedure and, therefore, an undetected of patients with false-negative disease compared with pa-
misbalance in this important staging factor might explain tients with sentinel nodepositive disease), which do not
the imbalance in the survival outcomes.23 Thus, the authors influence the subgroup analysis but do influence the pri-
practice-changing conclusion that a lesser margin has a det- mary randomized study groups and primary endpoint of the
rimental effect on a patients survival cannot be simply ac- study.32-34
cepted yet. Therefore, the true therapeutic benefit of a sentinel node
procedure remains a matter of debate. Nonetheless, today
most surgeons, dermatologists, and other melanoma spe-
Sentinel Node cialists worldwide consider sentinel node biopsy a standard-
Multiple institutional and multicenter studies have verified of-care procedure for optimal staging and, therefore, part of
the prognostic information collected from the sentinel node the workup of intermediate thickness and thick melanomas
status of patients, not only for intermediate thickness but ($ pT1b = $ 1.00-mm Breslow or in case of , 1.00-mm
also for thick melanoma.24-30 There is ongoing debate on the Breslow when the tumor is ulcerated or if there is one or
interpretation of the results with respect to the interim and more dermal mitosis present), who do not yet have clini-
final data of the Multicenter Selective Lymphadenectomy cally apparent detectable metastases. With the potential
Trial-1 (MSLT-1).28,31-34 The authors of the MSLT-1 study for more effective adjuvant therapy options as approved
concluded that: Biopsy-based staging of intermediate- therapies in the stage IV setting complete testing in the
thickness or thick primary melanomas provides important stage III population, this need for optimal staging will only
prognostic information and identifies patients with nodal increase.
metastases who may benefit from immediate complete
lymphadenectomy. Biopsy-based management prolongs
disease-free survival for all patients and prolongs distant Lymph Node Dissections
Lymph node dissections can be divided into three categories:
(1) elective (or prophylactic) lymph node dissection, (2) ther-
KEY POINTS apeutic lymph node dissection, and (3) completion lymph
node dissection.
Sentinel node staging provides optimal staging Elective lymph node dissection is currently an abandoned
information and is required for participation in adjuvant procedure. Four prospective randomized controlled trials
therapy trials. from the 1970s and 1990s have sought to find a survival
Surgery alone can be curative for primary melanomas, benefit for elective lymph node dissection but have
locoregional disease, and in selected stage IV cases. failed.35-38 The morbidity of elective lymph node dissection
Adjuvant interferon has not demonstrated unequivocal was high, and, in the case of trunk or head and neck mel-
results but is the only approved adjuvant therapy in anomas, it could be unclear which nodal basin should be
many countries.
prophylactically removed. Subgroup analyses from the
Adjuvant therapy with checkpoint inhibitors (antiCLTA-
4 and antiPD-1) and targeted therapies are ongoing,
Cascinelli et al and Balch et al studies did seem to indicate a
and the results are pending. potential benefit for patients with intermediate thickness
AntiCTLA-4 has demonstrated a progression-free melanoma, who did indeed have nodal involvement.37,38
survival benefit, but its effect on overall survival is not This was the starting point for the MSLT-1 study.
clear yet. Therapeutic lymph node dissection is performed for clini-
cally apparent detectable nodal metastases at presentation

e506 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


SURGICAL MANAGEMENT OF MELANOMA

or during follow-up. If staging imaging examinations (CT, radiotherapy, isolated limb perfusion/infusion, systemic
PET-CT, and/or MRI) do not indicate any distant metastases, therapy). However, regardless of this heterogeneity, most
surgeons worldwide will propose this to patients. Five-year surgeons and other melanoma-treating specialists will
survival rates differ depending on the type of dissection (neck, concur that, in the absence of distant metastases on dis-
axilla, or groin), the number of lymph nodes involved, and the semination examination (CT, PET-CT, or MRI), a locoregional
presence of extracapsular extension. The AJCC database re- treatment with curative intent is the preferred approach.
ports survival between 59% for stage IIIB and 40% for stage However, with the advent of more effective systemic
IIIC.1 Some institutional databases report even worse out- therapies, it may become reasonable to first treat such
come of 20%40%.39-45 patients with systemic treatment (particularly immuno-
One prospective randomized controlled trial, MSLT-2 therapy) and save surgical resection as a salvage procedure
(NCT00297895), is evaluating the therapeutic value of com- for residual disease.
pletion lymph node dissection after a positive sentinel node Surgical treatment of distant metastases is a rare phe-
biopsy, which compares with nodal observation by periodic nomenon because usually the amount of lesions is too
ultrasound.46,47 This study has completed accrual but needs many for complete resection of all with any potential clin-
to complete follow-up before initial outcomes can be re- ical benefit. However, there are a few scenarios where one
ported. Therefore, there is no consensus yet on this topic, might consider surgery for a patient with stage IV disease.
which is illustrated by several studies demonstrating a dis- Obvious reasons are in the case of symptoms such as bowel
tribution of around 50% for completion lymph node dissec- obstruction, bleeding, and pain for palliative reasons. But
tion compared with 50% for nodal observation in the United there is potential for cure in a highly selected patient cohort.
States/North America, Europe, and Australia.48,49 Typical selection criteria are one, two, or three or more
At the 2015 ASCO Annual Meeting, Leiter et al reported on lesions; M1a, M1b, or M1c disease; and the disease-free
the results of the prospective randomized controlled trial interval before the development of these distant metasta-
German DECOG group study, which also compared com- ses. Howard et al demonstrated in these cases a 4-year
pletion lymph node dissection with nodal observation. In a survival rate of 20.8% for patients undergoing surgery as part
total of 483 patients (242 of whom underwent completion of their treatment compared with 7.0% for patients who
lymph node dissection versus 241 of whom underwent nodal were not eligible for surgery and received systemic therapy
observation), with a median follow-up of 34 months, there only (prior to checkpoint inhibitors/BRAFi/MEKi).57 A se-
were no differences in recurrence-free survival, distant lected subgroup of patients who only received surgery could
metastasesfree survival, or melanoma-specific survival.50 achieve even a 4-year survival of 45.7%.57 Similar rates have
However, this trial was criticized for the lack of mature been shown by Sosman et al (31% at 4 years).58 In general,
follow-up and was likely to be underpowered compared with surgical resections with curative intent are reserved for this
the MSLT-2 trial, which accrued nearly 2,000 patients. The highly selected (biased) patient cohort.
very recent results of the Sunbelt Melanoma Trial confirm
the DECOG results, by showing again that there was no
Future Projections
difference in overall survival after CLND. However, this was
With the introduction of effective systemic therapy (e.g.,
not the primary focus of this study and potentially un-
immune checkpoint inhibitors, BRAFi/MEKi), the role of
derpowered to look at that.51 Finally, there is a question if all
surgery will be subject to change. Primarily, staging for
patients with tumor-bearing sentinel nodes might benefit
adjuvant therapy through standard of care sentinel-node
from completion lymph node dissection or if some patients
biopsy will no longer be a matter of debate. The question of
with minimal sentinel node tumor burden might not benefit
whether completion lymph node dissection is necessary
as their prognosis is comparable to patients with sentinel
after a positive sentinel node will be a matter of debate at
nodenegative disease.52-54 This topic is being addressed by
first (upcoming 1020 years) but is likely to evolvesimilarly
the EORTC 1208 Minitub (NCT01942603) prospective
to the situation in breast cancer, where it is reserved for
registry.
cases who did not respond to the systemic therapy or as a
salvage procedure after progression. At the same time,
surgery for patients with stage IV disease will also evolve.
In-Transit/Distant Metastases
Patients with solitary residual disease (near complete re-
The latest AJCC staging system considers in-transit meta-
sponse to systemic therapy) or with a mixed response
stases as stage IIIC, and they have a poor 5-year survival rate
(progression of solitary lesion, when all others show re-
of 46% or 69% (with and without regional lymph node
sponse) will be the target of surgical resection. Finally, locally
involvement, respectively).1 Read et al reported rates of
advanced (unresectable) disease will be the target of neo-
40% or 59% (with and without regional lymph node in-
adjuvant induction treatment to allow complete resection.
volvement, respectively),55 and Pawlik et al reported a rate
of 54%.56 None of these studies reported the type of in-
transit metastases (cutaneous or subcutaneous) or number Surgical Conclusions
of metastases (single or multiple), and they also did not Wide local excision is recommended to reduce local re-
report on the type of treatment (e.g., surgery, amputation, currence rates, but it does not influence survival. Worldwide,

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e507


VAN AKKOOI ET AL

there is no consensus on whether performing a sentinel node the time of its publication; however, it is at the mercy of
procedure and/or completion lymph node dissection has a rapidly advancing technology and the inconsistent appli-
proven therapeutic benefit. Sentinel node staging and sub- cation of this both across and within trials. Sentinel node
sequent completion lymph node dissection in the case of biopsy has been clearly established as a very strong prog-
positive sentinel node biopsy provides optimal staging in- nostic factor for primary melanoma.1,28 However, the
formation and is required to allow patients to be included in emergence of molecular-based diagnostic techniques means
adjuvant therapy trials. Locoregional treatment is considered that much a smaller tumor burden can be detected in the
with curative intent for in-transit metastases. Highly selected lymph node, and the clinical implications and optimum
patients with stage IV disease might benefit from surgical treatment of microscopic nodal disease has not yet been
resection. The role of surgery for melanoma will evolve be- established.40 At a more basic level, not all older trials man-
cause of the introduction of effective systemic therapy, with dated sentinel node biopsy, so the prognosis for patients with
an increasing need for optimal staging, increasing salvage stage II disease varies greatly, making this the most hetero-
surgery after near complete response or in case of mixed geneous stage group. The use of sentinel node biopsy and
response, and neoadjuvant approaches after induction treat- more sensitive imaging for detection of distant metastatic
ment of locally advanced disease. disease have resulted in stage migration with improvement in
the median survival for patients with stage II, III, and IV disease,
making it difficult to compare trials where different staging
ADJUVANT THERAPY FOR PATIENTS WITH techniques were performed.
RESECTED HIGH-RISK AND STAGE IV MELANOMA Tumor ulceration is an independent prognostic factor for
The past 5 years have seen dramatic improvements in the
survival across all stage groups. Although there is an ac-
outcomes for patients with metastatic melanoma, evi-
cepted definition of tumor ulceration, there remains the
denced by an improvement in the expected median survival
potential for differences in interpretation of the morpho-
of patients from 9 months to approximately 2530 months
logic features defining tumor ulceration. Newer clinical trials
and durable treatment-free responses from 10% to as high
require central review of pathology specimens to minimize
as 50% in this short period of time.59,60 Linked to this, a
uncertainty and standardize staging; this is particularly im-
number of new immunotherapy and targeted therapy agents
portant for ulceration and sentinel node biopsy involvement.
have been licensed both as single agents and in combinations.
However, older studies on which many management guide-
There is a high expectation that outcomes for patients will
lines are based did not require this.
continue to improve based on the evaluation of new agents
Identifying the appropriate clinical endpoint for an adju-
and the rapidly increasing understanding of the molecular
vant therapy study in melanoma has become increasingly
basis of resistance to both targeted therapy and immuno-
difficult. Until recently, there was no concern that a potential
therapy and how this can be circumvented.
benefit in an adjuvant therapy study could be confounded by
Advances in the field of adjuvant therapy of melanoma
subsequent therapy treatment in the advanced disease
have been significantly less dramatic than for metastatic
setting; this is clearly not the case at present. There remains
melanoma. Despite the approval of two new agents (ipili-
major discussion about the validity of recurrence-free sur-
mumab and pegylated interferon [PEG-IFN]), the past
vival as a clinically useful endpoint in its own right.61 It is clear
5 years have not seen any improvement in OS in any adjuvant
that patients do value time without disease, even if this is
therapy study. However, it is too early to draw any con-
at the expense of treatment-related toxicity.62 Increas-
clusions about the use of targeted therapy or checkpoint
ingly, there is a phenomenon of patients wishing to remain
inhibitors as adjuvant therapy, as the studies are either still
disease-free as long as possible in an attempt to bridge to
recruiting or have not yet been fully reported. Until then, IFN
emerging new and potentially more effective agents and/or
remains a treatment option in the United States and in many
clinical trials for the treatment of metastatic disease. Other
European countries, although there is no consensus on the
questions that must be considered include the potential
exact indication group, dose, and/or duration of treatment.
toxicity of adjuvant therapy, the cost implications of this, and
The U.S. Food and Drug Administration (FDA) approved
whether these treatments are more effective and/or cost-
ipilimumab in October 2015 based on the improvement in
effective in the adjuvant or metastatic disease settings.
relapse-free survival (RFS), but with absent OS data at this
Although some of these challenges are easily addressed,
moment, a definitive recommendation for its use cannot yet
others will require clinical trials that include quality of life
be made.
and cost-effectiveness endpoints.
We discuss the current role of adjuvant therapy in mel-
anoma and highlight potential developments in the next
5 years.
Adjuvant Interferon
The role of IFN as adjuvant treatment of melanoma remains
Challenges in Evaluating Adjuvant Trials unclear, leading to a lack of consensus across Europe and the
A number of challenges arise when examining individual United States. A number of randomized trials have in-
studies and comparing different adjuvant therapy trials. The vestigated the use of adjuvant IFN-a for the treatment
AJCC staging system reflects the understanding of staging at of patients with high-risk melanoma following surgical

e508 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


SURGICAL MANAGEMENT OF MELANOMA

resection. The results of these trials have, however, been high-dose IFN. The concept of the study, when designed in
inconsistent, with some suggesting a survival benefit for 1997, was to find out if patients with minimal disease
IFN-a and others showing no difference. High-dose IFN-a (sentinel node negative, but RT-PCR positive) would be the
has been approved since 1996 based on the results of the group with the most benefit. The trial did not demonstrate
ECOG 1684 trial of 287 patients with stage IIB/III melanoma, any OS benefit.51 The subgroup of patients with ulceration
which showed a benefit for high-dose IFN-a on both RFS and did show a benefit for disease-free survival but not for OS.
OS.63 Updated results with a median follow-up of 12.6 years The amount of ulcerated patients in this study was minimal,
showed that the RFS benefit was maintained (HR 0.72; which might be the explanation for the lack of OS benefit in
p = .02), but the HR for OS had decreased from 0.67 to 0.82 and this group.
was no longer significant (p = .18), though clearly the effect In Europe, low-dose IFN-a was approved based on a
of competing causes of death on survival may have con- French trial of 499 patients with stage II disease, which
tributed to this.64 The ECOG 1690 trial compared high- and showed an RFS benefit (p = .035) and a trend toward an
low-dose IFN-a with observation in 624 patients with stage improvement in OS (p = .059).67 More recently, in 2011, the
IIB/III disease and showed only a marginal benefit of RFS for FDA approved PEG-IFN for patients with stage III melanoma
both regimens and no OS benefit for either treatment.65 As based on the EORTC 18991 trial of 1,256 patients.68 The trial
this trial included patients with T4 primary tumors (stage II) first reported in 2007 with a median follow-up of 3.8 years.
and did not require regional nodal staging, many patients There was a significant improvement in RFS in favor of PEG-
relapsed in regional nodes. Crossover of patients in the IFN-a-2b (HR 0.82; p = .011), however, there was no sig-
observation arm to the then FDA-approved high-dose IFN nificant effect on either distant metastasis-free survival or
regimen after lymph node recurrence was cited as a possible OS. Subgroup analysis showed that the benefit was only seen
contributing cause for the lack of difference. Intergroup in patients with microscopic nodal disease, and there was
E1694 compared high-dose IFN-a-2b with the GM2-KLH also an improvement in distant metastasis-free survival.
vaccine in patients with resected stage IIB and III Updated results with a median follow-up of 7.6 years
melanomathe same population studied in ECOG 1684 and showed the effect on RFS had deteriorated from an HR of
1690.66 The trial was closed prematurely when interim 0.82 to 0.87.69 In addition, the greatest benefit was seen for
analysis disclosed a significantly worse RFS and OS for pa- the patients with microscopic nodal disease (N1) who had
tients treated with the GM2-KLH vaccine compared with the ulcerated primary tumors (RFS HR 0.72; OS HR 0.59), with no
high-dose IFN-a (HR 1.47 and 1.52, respectively). Although benefit seen in nonulcerated tumors.
there was some initial concern that patients who were Other studies have looked at dose and duration of
administered the GM2-KLH vaccine fared worse than they treatment, and many did not use a no-treatment control
would have on observation, a subsequent trial comparing arm. Shortening the regimen to a month of high-dose therapy
the vaccine with observation in patients with stage II mel- administered either once or multiple times has not been
anoma showed no significant difference in OS, perhaps successful.70-73 Five meta-analyses have been reported.74-78
mitigating this concern. The two meta-analyses by Mocellin included studies without
Very recently, the results of the Sunbelt Melanoma Trial an active treatment control arm.74,75 A recent meta-analysis
were published. This trial examined high-dose adjuvant IFN reported on 17 trials published between 1995 and 2011.76
after sentinel nodepositive disease in one node versus Adjuvant IFN was associated with a significantly improved
observation versus high-dose IFN in the case of more than disease-free survival (HR 0.83; 95% CI, 0.7887; p = .00001)
one positive lymph node. Also, patients with sentinel node and OS (HR 0.91; 95% CI, 0.8597; p = .003). This translated
immunohistochemistrynegative but reverse transcription- into an improvement in OS from 46.1% to 49.1% at 5 years and
polymerase chain reactionpositive disease were randomly from 37.1% to 39.9% at 10 years. There was no clear evidence
assigned into three groups: observation, complete lymph that a particular dose or duration of treatment or patient
node dissection, or complete lymph node dissection plus subgroup benefitted differently. However, ulceration status
affected IFN benefit on event-free survival and OS. For
ulcerated melanoma, IFN increased event-free survival from
TABLE 1. Outcome According to Dose of Interferon 26.9% to 32.9% at 5 years and from 20.4% to 27.3% at 10 years
(Table 1).
Overall Risk (95% CI)
The interaction between ulceration and response to IFN is
Dose EFS OS
not universally accepted. No interaction has been seen in the
High (1,196 patients) 0.83 (0.720.96) 0.93 (0.801.08) studies from ECOG (Kirkwood, personal communication) or
PEG-IFN (1,256 patients) 0.83 (0.761.00) 0.96 (0.821.11) from the Nordic group.79 This is being examined pro-
Intermediate (2,243 patients) 0.84 (0.740.95) 0.91 (0.791.04) spectively in an EORTC trial 18081 (NCT01502696).
Low (2,732 patients) 0.85 (0.770.94) 0.86 (0.770.96) To look further at whether progression-free survival
Very low (484 patients) 0.99 (0.801.23) 0.96 (0.761.21) predicts for OS with IFN treatment, Suciu and colleagues
Overall (95% CI) 0.86 (0.810.91) 0.90 (0.850.97) examined 12 studies involving 6,708 patients for which in-
Abbreviations: EFS, event-free survival; OS, overall survival; PEG-IFN, pegylated-
dividual patient data were available to produce a training
interferon. set. This was then validated on published data from 5,774

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e509


VAN AKKOOI ET AL

patients from a further 13 studies, including studies with an patients who were HLA-A2negative received only GM-CSF or
active treatment control arm. The results showed that both placebo. There was no benefit associated with the use of the
individual and trial level correlations were close, suggesting peptide vaccine.83 There was no significant difference in ei-
that for adjuvant IFN trials, there is a strong surrogacy of RFS ther OS (5-year survival rate 52% vs. 49%; HR 0.94; 95% CI,
for OS.80 0.771.15) or RFS (median RFS 11.4 vs. 8.8 months, and 5-year
RFS rate 31% vs. 27%; HR 0.88; 95% CI, 0.741.04; p = .131),
when GM-CSF was compared with placebo. Exploratory an-
Non-IFNBased Adjuvant Therapy Approaches alyses showed a trend toward improved OS in patients with
Other approaches to adjuvant treatment have been pursued resected visceral metastases who received GM-CSF, but this
over the past two decades with most showing mixed re- observation required independent validation especially given
sults. These have included biochemotherapy, granulocyte- that this was not a protocol stratification factor.83 Given these
macrophage colony-stimulating factor with or without mixed results, GM-CSF is generally not recommended for use
peptide vaccinations, and bevacizumab. More encouraging in the adjuvant setting.
data are emerging with the checkpoint inhibitor ipilimumab,
and there is great optimism surrounding the potential value Bevacizumab. Bevacizumab was reported to have some
of the PD-1 pathway inhibitors. These approaches are de- activity in patients with advanced melanoma, prompting its
scribed below. study in the adjuvant setting. In a phase III multicenter trial
conducted in the United Kingdom, 1,343 patients with
Biochemotherapy. Cisplatin and interleukin-2 (IL-2)based resected stage IIB, IIC, or III disease were randomly assigned
biochemotherapy produced encouraging results in a series to receive 1 year of bevacizumab treatment (7.5 mg/kg every
of phase II trials in patients with stage IV melanoma. These 3 weeks) or to observation.84 At a median follow-up of
results prompted the Southwest Oncology Group (SWOG) to 25 months, there was no significant difference in OS, which
perform a phase III cooperative group trial (S0008) in which was the primary endpoint of the trial (HR 0.97; 95% CI,
biochemotherapy was compared with high-dose IFN.81 The 0.781.22; p = .76).84 However, there was a significant in-
trial randomly assigned 432 patients with high-risk mela- crease in the disease-free interval (1-year and 2-year
noma to either three cycles of cisplatin, vinblastine, disease-free rates 77% vs. 70% and 59% vs. 57%, re-
dacarbazine, IL-2, and IFN-a given over a 9-week period or spectively; HR 0.83; 95% CI, 0.700.98), and an insignificant
to high-dose IFN-a for 1 year. At a median follow-up of trend toward improved distant metastasis-free survival (HR
7.2 years, the biochemotherapy regimen significantly pro- 0.88; 95% CI, 0.731.06; p = .18).84 Interpretation of the trial
longed RFS (median 4.0 vs. 1.9 years, and 5-year RFS rate 48% and the potential role of bevacizumab will require further
vs. 39%; HR 0.75; 95% CI, 0.580.97).81 However, there was no follow-up to assess the 5-year OS rate. Until then, the use of
significant difference in the OS (5-year rate 56% for both bevacizumab in the adjuvant setting for patients with ad-
treatment arms; HR 0.98; 95% CI, 0.741.31).81 The bio- vanced melanoma is not recommended.
chemotherapy regimen was substantially more toxic, with
grade 3 or 4 side effects (consisting primarily of hematologic Ipilimumab. Ipilimumab was approved for use in patients
and gastrointestinal toxicity) observed in 76% of participants. with stage IV disease in 2011 based on its being able to
Neurologic, psychiatric, and hepatic toxicities were the most prolong median OS.2 The approved regimen in the stage IV
frequent with high-dose IFN-a. Biochemotherapy toxicities setting (3 mg/kg every 3 weeks for four doses) was also
were limited to the 9-week treatment period, while IFN-a shown to produce a durable effect on OS extending out to
toxicities were distributed across the year of treatment. Even 10 years.85 The encouraging initial studies with ipilimumab
though the biochemotherapy regimen was the first to in the stage IV setting prompted its study in the adjuvant
produce a significant improvement in RFS compared with an setting. Two randomized phase III trials compared with
active control arm, the lack of OS benefit coupled with the placebo (EORTC 18071) or high-dose IFN (ECOG 1609) have
failure of this regimen to show an OS benefit relative to completed accrual. In the EORTC 18071 trial, patients with
chemotherapy alone in patients with stage IV disease has resected stage III melanoma were randomly assigned to
limited its acceptance in the adjuvant setting. receive ipilimumab 10 mg/kg or placebo every 3 weeks for
four doses and then every 3 months for up to 3 years.86
Granulocyte-macrophage colony-stimulating factor. Results showed improved median recurrence-free survival
Granulocyte-macrophage colony-stimulating factor (GM-CSF) of 26.1 months with ipilimumab compared with 17.1 months
has been reported to prolong survival in patients with with placebo (HR 0.75; p = .0013).86 In subgroup analyses,
resected stage IV disease in a single-arm trial,82 prompting its patients with microscopic lymph node disease or ulcerated
evaluation in a randomized controlled trial (E4697).83 In this primary lesions demonstrated the most benefit. Impor-
multicenter trial, 815 patients with high-risk stage III or tantly, there was a high rate of grade 3-5 immune-related
completely resected stage IV melanoma were randomly adverse events in patients receiving ipilimumab (43%
assigned to receive GM-CSF or placebo. Patients who were compared with 2% with placebo).86 These included five
HLA-A2positive were randomly assigned to peptide vacci- treatment-related deaths, despite treatment with immu-
nation or placebo, in combination with GM-CSF or placebo; nomodulatory therapy. Although these data are provocative

e510 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


SURGICAL MANAGEMENT OF MELANOMA

and have led to the recent FDA approval of ipilimumab for stage III and IV melanoma are currently underway. These
patients with resected stage III melanoma, it is as yet unclear include Checkmate-238 (NCT02388906) comparing ipilimu-
whether this reduction in recurrence rate with ipilimumab mab 10 mg/kg (according to the EORTC 18071 schedule) to
represents an improvement over adjuvant IFN therapy and nivolumab 3 mg/kg administered intravenously every
whether this benefit will translate into an improvement in 2 weeks; EORTC 1325 (KEYNOTE-054; NCT02362594) protocol
OS. Furthermore, the approved adjuvant dose of 10 mg/kg is comparing pembrolizumab (200-mg flat dose) with placebo;
over three times higher than the approved dose in meta- and the SWOG S1404 protocol comparing pembrolizumab
static melanoma, with the potential for higher toxicity. These (200-mg flat dose) with high-dose IFN (NCT02506153).
questions are being addressed by the U.S. Intergroup E1609 Checkmate-238 completed accrual in September 2015.
study, which randomly assigned patients with resected stage EORTC 1325 and SWOG S1404 are actively accruing patients.
III-IV melanoma to ipilimumab 10 mg/kg or 3 mg/kg or high-
dose IFN.87 The study completed accrual of more than 1,500 Molecularly targeted therapies. The selective inhibitors of
patients in the summer of 2014 and is pending analysis for the mutant BRAF kinase (vemurafenib and dabrafenib) have
coprimary endpoints of RFS and OS. Long-term survival data been shown to be superior to dacarbazine in patients with
from both of these adjuvant studies will ultimately be nec- stage IV BRAFV600-mutant melanoma, leading to their FDA
essary to determine the true effectiveness of adjuvant approval.3,8 More recently, the combination of dabrafenib
ipilimumab therapy relative to either high-dose IFN or and the MEK inhibitor trametinib have been proven to
observation. produce superior response rate, median progression-free
survival, and median OS compared with single-agent dab-
MAGE-A3 vaccine. The DERMA (ADjuvant ImmunothERapy rafenib or vemurafenib in this patient population.7,8 Based
with MAGE3 in MelanomA) was a double-blind, randomized, on these encouraging results, clinical trials have begun testing
placebo-controlled phase III study of recombinant MAGE-A3 these agents in patients with high-risk resected BRAF-mutant
with AS15 antigen-specific cancer immunotherapeutic (ASCI) melanoma. In the COMBI-AD trial, approximately 850 patients
in patients with MAGE-A3positive resected stage IIIB/C are to be randomly assigned to receive either dabrafenib
disease. MAGE-A3 is expressed on approximately 60% of with trametinib or placebo and treated for up to 1 year
melanoma specimens. The study was based on an EORTC (NCT01682083). This trial completed accrual in December
phase II study of patients with stage IV M1A disease that 2014 and is pending analysis. In the BRIM 8 study, up to
identified a superior survival benefit for patients receiving 775 patients with stage IIC to stage IIIC resected melanoma
the MAGE-A3 vaccine treated with the AS15 rather than will be randomly assigned to receive vemurafenib or placebo
the ASO2B adjuvant (HR 0.55; 95% CI, 0.281.06).88 for up to 1 year. The study is currently recruiting at 187 sites in
Furthermore, a genetic predictor identified a group of pa- 29 countries (NCT01667419).
tients receiving MAGE-A3 with AS15 ASCI with a better OS The results of these trials are eagerly awaited, especially
(HR 0.27; 95% CI, 0.080.89).88 The study screened 3,914 the assessment of whether these active agents can eliminate
patients and randomly assigned 1,344 patients 2:1 to 13 the last tumor cell in the low-volume adjuvant setting and
intramuscular injections of vaccine or placebo. The study thereby actually prevent, not just delay, disease recurrence.
failed to meet its coprimary endpoint of disease-free survival
in either the overall population of patients studied or in
those with the potential predictive gene signature.88 Adjuvant Therapy Conclusions
The role of adjuvant therapy in patients with resected high-
risk melanoma remains unsettled. Although several ap-
Ongoing Adjuvant Studies proaches have shown improvements in RFS, only the high-dose
PD-1 pathway blockers. In the meantime, the field has IFN regimen has shown an improvement in OS. The extent of
moved on to examine potentially more exciting checkpoint this survival benefit remains controversial. Although multiple
inhibitors: those blocking the PD-1 pathway. PD-1 inhibitors meta-analyses have suggested that the benefit of IFN on OS is
have proven to be less toxic and more active than ipili- small (under 10%), these usually (1) include studies using
mumab in patients with established, unresectable meta- different dose levels of IFN rather than simply the high-dose
static melanoma.9,89 Given their favorable therapeutic index regimen, (2) include the E1690 study data despite its survival
and ability to produce durable off-treatment benefit in endpoint being potentially confounded by crossover to then
patients with advanced melanoma, there is much interest in FDA-approved IFN use, and (3) exclude the overwhelmingly
developing this class of therapies as adjuvant treatment for positive E1694 trial because it involved a vaccine control arm.
patients with high-risk resected melanoma. Results from a The unresolved question is: do you base a decision on the
phase I trial of nivolumab plus a multipeptide vaccine in results of the individual trials or the meta-analyses? The choice
33 patients with resected stage IIIC or IV melanoma showed a for some is further complicated by the toxicity of the high-
relatively low relapse rate (30%) during a median follow-up dose IFN regimen, which is clearly more than desirable for the
period of 32.1 months from trial enrolment. Median RFS adjuvant setting.
was estimated to be 47.1 months.90 Phase III trials with The benefit of ipilimumab on RFS appears to be equivalent
nivolumab and pembrolizumab in patients with resected at least to that of high-dose IFN and was sufficiently robust as

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e511


VAN AKKOOI ET AL

to garner FDA approval. Given that the major effect of are highly likely to prolong RFS compared with placebo in the
ipilimumab in patients with stage IV disease is on OS, it is ongoing clinical trials. However, in contrast to immuno-
likely that the RFS benefit of ipilimumab will ultimately therapy, they typically do not produce durable off-treatment
translate into a survival benefit. Nonetheless, many will be responses in the metastatic setting and, therefore, may only
unwilling to prescribe ipilimumab given the high incidence of delay, rather than prevent, relapse in the adjuvant setting.
high-grade toxicity, including five deathsan outcome that The current trials will eventually answer these questions,
is difficult to swallow in a population in whom as much but, until then, the use of these agents should be reserved
as half might remain disease-free in the absence of any for patients with measurable disease where the effect of
treatment. tumor volume and its duration can be monitored. Although
Absent an unambiguously positive approved agent for questions remain about the ability of PD-1 blockers to ef-
adjuvant therapy in melanoma, clinical trial participation fectively activate T cells in the absence of measurable
remains the priority. The most promising approaches cur- metastatic disease, it is hoped that, as in the stage IV setting,
rently under study include BRAF inhibitors and PD-1 pathway these agents will show more improvement over existing
blockers. The BRAF inhibitors have powerful antitumor ef- treatments and finally yield universally accepted adjuvant
fects in patients with BRAF-mutant melanoma. Thus, they treatment approaches for patients with high-risk melanoma.

References
1. Balch CM, Gershenwald JE, Soong SJ, et al. Final version of 2009 AJCC versus 5-cm resection margins for patients with cutaneous melanoma
melanoma staging and classification. J Clin Oncol. 2009;27:6199-6206. with a tumor thickness of 0.8-2.0 mm. Cancer. 2000;89:1495-1501.
2. Hodi FS, ODay SJ, McDermott DF, et al. Improved survival with ipili- 16. Gillgren P, Drzewiecki KT, Niin M, et al. 2-cm versus 4-cm surgical
mumab in patients with metastatic melanoma. N Engl J Med. 2010;363: excision margins for primary cutaneous melanoma thicker than 2 mm:
711-723. a randomised, multicentre trial. Lancet. 2011;378:1635-1642.
3. Chapman PB, Hauschild A, Robert C, et al. Improved survival with 17. Karakousis CP, Balch CM, Urist MM, et al. Local recurrence in malignant
vemurafenib in melanoma with BRAF V600E mutation. N Engl J Med. melanoma: long-term results of the multiinstitutional randomized
2011;364:2507-2516. surgical trial. Ann Surg Oncol. 1996;3:446-452.
4. Flaherty KT, Infante JR, Daud A, et al. Combined BRAF and MEK in- 18. Khayat D, Rixe O, Martin G, et al. Surgical margins in cutaneous mel-
hibition in melanoma with BRAF V600 mutations. N Engl J Med. 2012; anoma (2 cm versus 5 cm for lesions measuring less than 2.1-mm thick).
367:1694-1703. Cancer. 2003;97:1941-1946.
5. Flaherty KT, Robert C, Hersey P, et al. Improved survival with MEK 19. Thomas JM, Newton-Bishop J, AHern R, et al. Excision margins in high-
inhibition in BRAF-mutated melanoma. N Engl J Med. 2012;367: risk malignant melanoma. N Engl J Med. 2004;350:757-766.
107-114. 20. Haydu LE, Stollman JT, Scolyer RA, et al. Minimum safe pathologic
6. Hauschild A, Grob JJ, Demidov LV, et al. Dabrafenib in BRAF-mutated excision margins for primary cutaneous melanomas (1-2 mm in
metastatic melanoma: a multicentre, open-label, phase 3 randomised thickness): analysis of 2131 patients treated at a single center. Ann Surg
controlled trial. Lancet. 2012;380:358-365. Oncol. Epub 2015 May 9.
7. Long GV, Stroyakovskiy D, Gogas H, et al. Combined BRAF and MEK 21. Hayes AJ, Maynard L, AHern R, et al. Long term follow up of survival in a
inhibition versus BRAF inhibition alone in melanoma. N Engl J Med. randomised trial of wide or narrow excision margins in high risk primary
2014;371:1877-1888. melanoma. J Clin Oncol. 2015;33 (suppl; abstr 9001).
8. Robert C, Karaszewska B, Schachter J, et al. Improved overall survival in 22. Hayes AJ, Maynard L, Coombes G, et al. Wide versus narrow excision
melanoma with combined dabrafenib and trametinib. N Engl J Med. margins for high-risk, primary cutaneous melanomas: long-term
2015;372:30-39. follow-up of survival in a randomised trial. Lancet Oncol. 2016;17:
9. Larkin J, Chiarion-Sileni V, Gonzalez R, et al. Combined nivolumab and 184-192.
ipilimumab or monotherapy in untreated melanoma. N Engl J Med. 23. Moncrieff M. Excision margins for melanomas: how wide is enough?
2015;373:23-34. Lancet Oncol. 2016;17:127-128.
10. Postow MA, Chesney J, Pavlick AC, et al. Nivolumab and ipilimumab 24. Cascinelli N, Bombardieri E, Bufalino R, et al. Sentinel and nonsentinel
versus ipilimumab in untreated melanoma. N Engl J Med. 2015;372: node status in stage IB and II melanoma patients: two-step prognostic
2006-2017. indicators of survival. J Clin Oncol. 2006;24:4464-4471.
11. Robert C, Long GV, Brady B, et al. Nivolumab in previously untreated 25. Doubrovsky A, De Wilt JH, Scolyer RA, et al. Sentinel node biopsy
melanoma without BRAF mutation. N Engl J Med. 2015;372:320-330. provides more accurate staging than elective lymph node dissection in
12. Wolchok JD, Kluger H, Callahan MK, et al. Nivolumab plus ipilimumab in patients with cutaneous melanoma. Ann Surg Oncol. 2004;11:
advanced melanoma. N Engl J Med. 2013;369:122-133. 829-836.
13. Bono A, Bartoli C, Clemente C, et al. Ambulatory narrow excision for thin 26. Estourgie SH, Nieweg OE, Valdes Olmos RA, et al. Review and evaluation
melanoma (, or = 2 mm): results of a prospective study. Eur J Cancer. of sentinel node procedures in 250 melanoma patients with a median
1997;33:1330-1332. follow-up of 6 years. Ann Surg Oncol. 2003;10:681-688.
14. Cascinelli N. Margin of resection in the management of primary mel- 27. Gershenwald JE, Thompson W, Mansfield PF, et al. Multi-institutional
anoma. Semin Surg Oncol. 1998;14:272-275. melanoma lymphatic mapping experience: the prognostic value of
15. Cohn-Cedermark G, Rutqvist LE, Andersson R, et al. Long term results sentinel lymph node status in 612 stage I or II melanoma patients. J Clin
of a randomized study by the Swedish Melanoma Study Group on 2-cm Oncol. 1999;17:976-983.

e512 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


SURGICAL MANAGEMENT OF MELANOMA

28. Morton DL, Thompson JF, Cochran AJ, et al. Final trial report of sentinel- from the multicenter selective lymphadenectomy trial-1 results in
node biopsy versus nodal observation in melanoma. N Engl J Med. 2014; melanoma? Melanoma Res. 2013;23:181-184.
370:599-609. 48. Bilimoria KY, Balch CM, Bentrem DJ, et al. Complete lymph node dis-
29. Roulin D, Matter M, Bady P, et al. Prognostic value of sentinel node section for sentinel node-positive melanoma: assessment of practice
biopsy in 327 prospective melanoma patients from a single institution. patterns in the United States. Ann Surg Oncol. 2008;15:1566-1576.
Eur J Surg Oncol. 2008;34:673-679. 49. Pasquali S, Spillane AJ, de Wilt JH, et al. Surgeons opinions on lym-
30. van der Ploeg AP, van Akkooi AC, Schmitz PI, et al. EORTC Melanoma phadenectomy in melanoma patients with positive sentinel nodes:
Group sentinel node protocol identifies high rate of sub- a worldwide web-based survey. Ann Surg Oncol. 2012;19:4322-4329.
micrometastases according to Rotterdam Criteria. Eur J Cancer. 2010; 50. Leiter U, Stadler R, Mauch C, et al. Survival of SLNB-positive melanoma
46:2414-2421. patients with and without complete lymph node dissection: a multi-
31. Morton DL, Thompson JF, Cochran AJ, et al. Sentinel-node biopsy or center, randomized DECOG trial. J Clin Oncol. 2015;33 (suppl; abstr
nodal observation in melanoma. N Engl J Med. 2006;355:1307-1317. LBA9002).
32. Thomas JM. Prognostic false-positivity of the sentinel node in mela- 51. McMasters KM, Egger ME, Edwards MJ, et al. Fianl results of the Sunbelt
noma. Nat Clin Pract Oncol. 2008;5:18-23. Melanoma Trial: a multi-institutional prospective randomized phase III
33. Torjesen I. Sentinel node biopsy for melanoma: unnecessary treat- study evaluating the role of adjuvant high-dose interferon alfa-2b and
ment? BMJ. 2013;346:e8645. completion lymph node dissection for patients staged by sentinel lymph
34. van Akkooi AC. Sentinel node followed by completion lymph node node biopsy. J Clin Oncol. Epub 2016 Feb 8.
dissection versus nodal observation: staging or therapeutic? Contro- 52. van Akkooi AC, de Wilt JH, Verhoef C, et al. Clinical relevance of
versy continues despite final results of MSLT-1. Melanoma Res. 2014; melanoma micrometastases (,0.1 mm) in sentinel nodes: are these
24:291-294. nodes to be considered negative? Ann Oncol. 2006;17:1578-1585.
35. Veronesi U, Adamus J, Bandiera DC, et al. Inefficacy of immediate node 53. van der Ploeg AP, van Akkooi AC, Haydu LE, et al. The prognostic
dissection in stage 1 melanoma of the limbs. N Engl J Med. 1977;297: significance of sentinel node tumour burden in melanoma patients: an
627-630. international, multicenter study of 1539 sentinel node-positive mela-
36. Sim FH, Taylor WF, Ivins JC, et al. A prospective randomized study of the noma patients. Eur J Cancer. 2014;50:111-120.
efficacy of routine elective lymphadenectomy in management of 54. van der Ploeg AP, van Akkooi AC, Rutkowski P, et al. Prognosis in pa-
malignant melanoma. Preliminary results. Cancer. 1978;41:948-956. tients with sentinel node-positive melanoma is accurately defined by
37. Cascinelli N, Morabito A, Santinami M, et al. Immediate or delayed the combined Rotterdam tumor load and Dewar topography criteria.
dissection of regional nodes in patients with melanoma of the trunk: J Clin Oncol. 2011;29:2206-2214.
a randomised trial. WHO Melanoma Programme. Lancet. 1998;351: 55. Read RL, Haydu L, Saw RP, et al. In-transit melanoma metastases: in-
793-796. cidence, prognosis, and the role of lymphadenectomy. Ann Surg Oncol.
38. Balch CM, Soong S, Ross MI, et al. Long-term results of a multi- 2015;22:475-481.
institutional randomized trial comparing prognostic factors and sur- 56. Pawlik TM, Ross MI, Johnson MM, et al. Predictors and natural history of
gical results for intermediate thickness melanomas (1.0 to 4.0 mm). in-transit melanoma after sentinel lymphadenectomy. Ann Surg Oncol.
Intergroup Melanoma Surgical Trial. Ann Surg Oncol. 2000;7:87-97. 2005;12:587-596.
39. Badgwell B, Xing Y, Gershenwald JE, et al. Pelvic lymph node dissection is 57. Howard JH, Thompson JF, Mozzillo N, et al. Metastasectomy for distant
beneficial in subsets of patients with node-positive melanoma. Ann metastatic melanoma: analysis of data from the first Multicenter Se-
Surg Oncol. 2007;14:2867-2875. lective Lymphadenectomy Trial (MSLT-I). Ann Surg Oncol. 2012;19:
40. Balch CM, Gershenwald JE, Soong SJ, et al. Multivariate analysis of 2547-2555.
prognostic factors among 2,313 patients with stage III melanoma: 58. Sosman JA, Moon J, Tuthill RJ, et al. A phase 2 trial of complete resection
comparison of nodal micrometastases versus macrometastases. J Clin for stage IV melanoma: results of Southwest Oncology Group Clinical
Oncol. 2010;28:2452-2459. Trial S9430. Cancer. 2011;117:4740-06.
41. Jonk A, Kroon BB, Rumke P, et al. Results of radical dissection of the 59. Daud A, Ribas A, Robert C, et al. Long-term efficacy of pembrolizumab
groin in patients with stage II melanoma and histologically proved (pembro; MK-3475) in a pooled analysis of 655 patients (pts) with
metastases of the iliac or obturator lymph nodes, or both. Surg Gynecol advanced melanoma (MEL) enrolled in KEYNOTE-001. J Clin Oncol. 2015;
Obstet. 1988;167:28-32. 33 (suppl; abstr 9005).
42. Karakousis CP, Driscoll DL. Positive deep nodes in the groin and survival 60. Grob JJ, Amonkar MM, Karaszewska B, et al. Comparison of dabrafenib
in malignant melanoma. Am J Surg. 1996;171:421-422. and trametinib combination therapy with vemurafenib monotherapy
43. Karakousis CP, Driscoll DL, Rose B, et al. Groin dissection in malignant on health-related quality of life in patients with unresectable or
melanoma. Ann Surg Oncol. 1994;1:271-277. metastatic cutaneous BRAF Val600-mutation-positive melanoma
44. Spillane AJ, Pasquali S, Haydu LE, et al. Patterns of recurrence and (COMBI-v): results of a phase 3, open-label, randomised trial. Lancet
survival after lymphadenectomy in melanoma patients: clarifying the Oncol. 2015;16:1389-1398.
effects of timing of surgery and lymph node tumor burden. Ann Surg 61. Flaherty KT, Hennig M, Lee SJ, et al. Surrogate endpoints for overall
Oncol. 2014;21:292-299. survival in metastatic melanoma: a meta-analysis of randomised
45. van der Ploeg AP, van Akkooi AC, Schmitz PI, et al. Therapeutic surgical controlled trials. Lancet Oncol. 2014;15:297-304.
management of palpable melanoma groin metastases: superficial or 62. Kilbridge KL, Cole BF, Kirkwood JM, et al. Quality-of-life-adjusted sur-
combined superficial and deep groin lymph node dissection. Ann Surg vival analysis of high-dose adjuvant interferon alpha-2b for high-risk
Oncol. 2011;18:3300-3308. melanoma patients using intergroup clinical trial data. J Clin Oncol.
46. Morton DL. Overview and update of the phase III Multicenter Selective 2002;20:1311-1318.
Lymphadenectomy Trials (MSLT-I and MSLT-II) in melanoma. Clin Exp 63. Kirkwood JM, Strawderman MH, Ernstoff MS, et al. Interferon alfa-2b
Metastasis. 2012;29:699-706. adjuvant therapy of high-risk resected cutaneous melanoma: the
47. van Akkooi AC. Comments and controversies: sentinel node and lym- Eastern Cooperative Oncology Group Trial EST 1684. J Clin Oncol. 1996;
phadenectomy in melanoma: staging or therapeutical?: what to expect 14:7-17.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e513


VAN AKKOOI ET AL

64. Kirkwood JM, Manola J, Ibrahim J, et al. A pooled analysis of eastern 79. Hansson J, Aamdal S, Bastholt L, et al. Two different durations of ad-
cooperative oncology group and intergroup trials of adjuvant high-dose juvant therapy with intermediate-dose interferon alfa-2b in patients
interferon for melanoma. Clin Cancer Res. 2004;10:1670-1677. with high-risk melanoma (Nordic IFN trial): a randomised phase 3 trial.
65. Kirkwood JM, Ibrahim JG, Sondak VK, et al. High- and low-dose in- Lancet Oncol. 2011;12:144-152.
terferon alfa-2b in high-risk melanoma: first analysis of intergroup trial 80. Suciu S, Eggermont A, Lorigan P, et al. Relapse-free survival (RFS) as a
E1690/S9111/C9190. J Clin Oncol. 2000;18:2444-2458. surrogate endpoint for overall survival in adjuvant Interferon trials in
66. Kirkwood JM, Ibrahim JG, Sosman JA, et al. High-dose interferon alfa-2b patients with resectable cutaneous melanoma: an individual patient
significantly prolongs relapse-free and overall survival compared with data meta-analysis. In: ESMO 2014 Congress. Madrid, Spain; 2014.
the GM2-KLH/QS-21 vaccine in patients with resected stage IIB-III Abstract 1089PD.
melanoma: results of intergroup trial E1694/S9512/C509801. J Clin 81. Flaherty LE, Othus M, Atkins MB, et al. Southwest Oncology Group
Oncol. 2001;19:2370-2380. S0008: a phase III trial of high-dose interferon Alfa-2b versus cisplatin,
67. Grob JJ, Dreno B, de la Salmoni`ere P, et al. Randomised trial of in- vinblastine, and dacarbazine, plus interleukin-2 and interferon in pa-
terferon alpha-2a as adjuvant therapy in resected primary melanoma tients with high-risk melanomaan intergroup study of cancer and
thicker than 1.5 mm without clinically detectable node metastases. leukemia Group B, Childrens Oncology Group, Eastern Cooperative
Lancet. 1998;351:1905-1910. Oncology Group, and Southwest Oncology Group. J Clin Oncol. 2014;32:
68. Eggermont AM, Suciu S, Santinami M, et al. Adjuvant therapy with 3771-3778.
pegylated interferon alfa-2b versus observation alone in resected stage 82. Spitler LE, Weber RW, Allen RE, et al. Recombinant human granulocyte-
III melanoma: final results of EORTC 18991, a randomised phase III trial. macrophage colony-stimulating factor (GM-CSF, sargramostim) ad-
Lancet. 2008;372:117-126. ministered for 3 years as adjuvant therapy of stages II(T4), III, and IV
69. Eggermont AM, Suciu S, Testori A, et al. Long-term results of the melanoma. J Immunother. 2009;32:632-637.
randomized phase III trial EORTC 18991 of adjuvant therapy with 83. Lawson DH, Lee S, Zhao F, et al. Randomized, placebo-controlled, phase
pegylated interferon alfa-2b versus observation in resected stage III III trial of yeast-derived granulocyte-macrophage colony-stimulating
melanoma. J Clin Oncol. 2012;30:3810-3818. factor (GM-CSF) versus peptide vaccination versus GM-CSF plus peptide
70. Agarwala SS, Lee SJ, Flaherty K, et al. Randomized phase III trial of high- vaccination versus placebo in patients with no evidence of disease after
dose interferon alfa-2b (HDI) for 4 weeks induction only in patients with complete surgical resection of locally advanced and/or stage IV mel-
intermediate- and high-risk melanoma (Intergroup trial E 1697). J Clin anoma: a trial of the Eastern Cooperative Oncology Group-American
Oncol. 2011;29 (suppl; abstr 8505). College of Radiology Imaging Network Cancer Research Group (E4697).
71. Payne MJ, Argyropoulou K, Lorigan P, et al. Phase II pilot study of in- J Clin Oncol. 2015;33:4066-4076.
travenous high-dose interferon with or without maintenance treatment 84. Corrie PG, Marshall A, Dunn JA, et al. Adjuvant bevacizumab in patients
in melanoma at high risk of recurrence. J Clin Oncol. 2014;32:185-190. with melanoma at high risk of recurrence (AVAST-M): preplanned in-
72. Pectasides D, Dafni U, Bafaloukos D, et al. Randomized phase III study of terim results from a multicentre, open-label, randomised controlled
1 month versus 1 year of adjuvant high-dose interferon alfa-2b in phase 3 study. Lancet Oncol. 2014;15:620-630.
patients with resected high-risk melanoma. J Clin Oncol. 2009;27: 85. Schadendorf D, Hodi FS, Robert C, et al. Pooled analysis of long-term
939-944. survival data from phase II and phase III trials of ipilimumab in unre-
73. Hauschild A, Weichenthal M, Rass K, et al. Efficacy of low-dose in- sectable or metastatic melanoma. J Clin Oncol. 2015;33:1889-1894.
terferon alpha2a 18 versus 60 months of treatment in patients with 86. Eggermont AM, Chiarion-Sileni V, Grob JJ, et al. Adjuvant ipilimumab
primary melanoma of .= 1.5 mm tumor thickness: results of a ran- versus placebo after complete resection of high-risk stage III melanoma
domized phase III DeCOG trial. J Clin Oncol. 2010;28:841-846. (EORTC 18071): a randomised, double-blind, phase 3 trial. Lancet Oncol.
74. Mocellin S, Lens MB, Pasquali S, et al. Interferon alpha for the adjuvant 2015;16:522-530.
treatment of cutaneous melanoma. Cochrane Database Syst Rev. 2013; 87. Tarhini AA. Melanoma and Skin Cancer: Adjuvant Therapy for High-Risk
6:CD008955. Melanoma. Physicians Education Resource, LLC. http://www.gotoper.
75. Mocellin S, Pasquali S, Rossi CR, et al. Interferon alpha adjuvant therapy com/publications/ajho/2014/2014nov/adjuvant-therapy-for-high-risk-
in patients with high-risk melanoma: a systematic review and meta- melanoma. Accessed March 16, 2016.
analysis. J Natl Cancer Inst. 2010;102:493-501. 88. Dreno B, Thompson JA, Smithers BM. DERMA, a double-blind, placebo
76. Suciu S, Ives N, Eggermont A, et al. Predictive importance of ulceration controlled Phase III study to assess the efficacy of MAGE-A3 cancer
on the efficacy of adjuvant interferon-a (IFN): An individual patient data immunotherapeutic adjuvant therapy in patients with resected MAGE-
(IPD) meta-analysis of 15 randomized trials in more than 7,500 mel- A3 positive Stage III melanoma. Proc SMR. 2015.
anoma patients (pts). J Clin Oncol. 2014;32:5s (suppl; abstr 9067). 89. Robert C, Schachter J, Long GV, et al; KEYNOTE-006 investigators.
77. Wheatley K, Ives N, Eggermont A, et al. Interferon-alpha as adjuvant Pembrolizumab versus ipilimumab in advanced melanoma. N Engl J
therapy for melanoma: an individual patient data meta-analysis of Med. 2015;372:2521-2532.
randomised trials. J Clin Oncol. 2007;25 (suppl; abstr 8526). 90. Gibney GT, Kudchadkar RR, DeConti RC, et al. Safety, correlative
78. Wheatley K, Ives N, Hancock B, et al. Does adjuvant interferon-alpha for markers, and clinical results of adjuvant nivolumab in combination with
high-risk melanoma provide a worthwhile benefit? A meta-analysis of vaccine in resected high-risk metastatic melanoma. Clin Cancer Res.
the randomised trials. Cancer Treat Rev. 2003;29:241-252. 2015;21:712-720.

e514 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


PATIENT AND SURVIVOR CARE

Cancer Survivorship: Is Every


Patients Journey the Same?

CHAIR
Michael T. Halpern, MD, PhD
University of Arizona College of Public Health
Tucson, AZ

SPEAKERS
Mary S. McCabe, RN, MA
Memorial Sloan Kettering Cancer Center
New York, NY

Mary Ann Burg, PhD, MSW, LCSW


University of Florida
Gainesville, FL
THE CANCER SURVIVORSHIP JOURNEY

The Cancer Survivorship Journey: Models of Care, Disparities,


Barriers, and Future Directions
Michael T. Halpern, MD, PhD, Mary S. McCabe, RN, MA, and Mary Ann Burg, PhD, MSW, LCSW

OVERVIEW

Although the number of long-term cancer survivors has increased substantially over past years, the journey of survivorship
does not always include high-quality, patient-centered care. A variety of survivorship care models have evolved based on
who provides this care, the survivor population, the site of care, and/or the capacity for delivering specific services. Other
areas of survivorship care being explored include how long follow-up care is needed, application of a risk-based approach to
survivorship care, and the role of the survivor in his or her own recovery. However, there is little evidence indicating whether
any models improve clinical or patient-reported outcomes. A newer focus in survivorship care has included assessment of
potential disparities; the sociodemographic characteristics of population subgroups associated with barriers to receiving
high-quality cancer treatment may also affect the survivorship period. Developing policies and programs to address dis-
parities in survivorship care is not simple, and examining how financial hardship affects cancer outcomes, reducing economic
barriers to care, and increasing incorporation of patient-centered strategies may be important components. Here too, there
is little evidence regarding the best strategies to address these disparities. Barriers to providing high-quality, patient-
centered survivorship care include lack of evidence, lack of a trained survivorship workforce, lack of reimbursement
structures/insurance coverage, and lack of a health care system that reduces fragmented care. Future research needs to
focus on developing a survivorship care evidence base, exploring strategies to facilitate provision of survivorship care, and
disseminating best survivorship care practices to diverse and international audiences.

A s of January 2014, there were approximately 14.5 million


cancer survivors in the United States; this number is
expected to grow to 19 million by 2024.1 Substantial in-
to identify challenges or barriers to providing high-quality
survivorship care, and to suggest potential directions for
future research to enhance the survivorship experience.
creases in the number of cancer survivors have also been
reported in other countries2-4; for example Maddams et al
projected that the number of cancer survivors in the United DEFINITION OF SURVIVORSHIP
Kingdom will increase by approximately one million per In exploring causes of variation in the survivorship journey, a
decade.5 The rapidly increasing numbers of cancer survivors key issue is how the term cancer survivor is defined. As
is the intended consequence of improvements in early with other aspects of survivorship and survivorship care,
cancer diagnosis and cancer treatment. As these two aspects there is not consensus on this definition. Some organiza-
of cancer care will likely continue to improve, the number of tions, such as the National Coalition for Cancer Survivorship
cancer survivors will correspondingly continue to increase. and National Cancer Institutes Office of Cancer Survivor-
The title of the corresponding American Society of Clinical ship, define survivors as individuals with cancer from the
Oncology (ASCO) 2016 Annual Meeting Education Session, time of diagnosis and for the balance of life, and include in
Cancer Survivorship: Is Every Patients Journey the Same? their definition family, friends, and caregivers of individuals
is largely rhetorical. Clearly, the journey for every cancer with cancer. In contrast, others define survivors as in-
survivor is not the same. However, this raises the question: dividuals who are diagnosed with cancer and have com-
why doesnt every cancer survivor receive high-quality and pleted potentially curative treatment, until either death or
comprehensive care tailored to his or her needs, situation, recurrence. The Institute of Medicines (IOMs) 2005 report
and risk factors? The goal of this article is to explore some of From Cancer Care to Cancer Survivor: Lost in Transition
the factors that lead to differences in the survivorship journey, acknowledges the definition of survivors used by the National

From the Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ; Memorial Sloan Kettering Cancer Center, New York, NY; University of Central Florida School
of Social Work, Orlando, FL.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Michael T. Halpern, MD, PhD, University of Arizona, Mel and Enid Zuckerman College of Public Health, 1295 North Martin Ave., Tucson, AZ 85724; email:
mthalpern@email.arizona.edu.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 231


HALPERN, MCCABE, AND BURG

Cancer Institutes Office of Cancer Survivorship but then follow-up care for the period following treatment.9,10 As one
focuses on the period following first diagnosis and treatment might expect, a variety of models have evolved based on the
and prior to the development of a recurrence of the initial provider, survivor population, site of care, and capacity for the
cancer or death.6 This report highlights the potential delivery of specific services. In addition, the type of survi-
distinction between a survivor and the period of care re- vorship care model is influenced by the design of the health
ferred to as survivorship. Jefford et al reports that the care delivery system(s) and payment mechanisms for health
definition of survivorship used in the United Kingdom is the care. Despite these differences, the major goals of care for
period after completion of initial treatment, regardless of cancer survivors transcend national health care systems and
whether the person is free from cancer at that time.7 focus on improved survival, long-term health, and the highest
More broadly, there is disagreement regarding whether quality of life possible, including both physical and psychosocial
the term cancer survivor should be used at all. As discussed well-being.11 However, many recommendations for survivor-
by Bell and Ristovski-Slijepcevic, some disagree with the ship care are largely based on expert consensus. At present,
term survivor being used for individuals who have expe- there is only limited evidence regarding services that improve
rienced cancer.8 This term is not used in other medical outcomes for cancer survivors, and there is essentially no in-
conditions; people who have had heart attacks are not called formation regarding the costs of providing these services
heart attack survivors.8 As an alternative, the U.K. National relative to their benefits.
Health Services has used the term living beyond cancer in In the United States and elsewhere, the IOM report, From
some of their materials. Cancer Patient to Cancer Survivor: Lost in Transition, has
served as the foundational document for establishing the
essential elements of care to be included in any survivorship
MODELS OF SURVIVORSHIP CARE care model: surveillance for recurrence, screening for sec-
As survivorship takes its place as a formal part of the cancer ond cancers, assessment and intervention of long-term/late
care continuum, there are increasing international efforts di- effects, counseling for healthy living, and communication/
rected toward the development and, most recently, the coordination with primary care.6 However, since this doc-
evaluation of models of care delivery. This activity has resulted ument was published in 2006, a number of countries, such as
in a growing body of literature describing and comparing the United Kingdom and Canada, have developed a national
various configurations of health care services and models of strategy for cancer survivorship. In the United Kingdom, the
National Cancer Survivorship Initiative was launched in 2008
as a partnership between the Department of Health, which
KEY POINTS has responsibility for the National Health Service, and
Macmillan Cancer Support, a large British charity.12 To date,
There are increasing international efforts directed the National Cancer Survivorship Initiative has promoted
toward models of care delivery for survivorship. Models novel, proactive approaches to identifying groups at in-
may be based on the care provider, survivor population, creased risk and customizing interventions for them.12 In
care site, service delivery capacity, follow-up care Canada, although the provision of health care is a provin-
duration, and the survivors role in recovery. cial responsibility, the Canadian Partnership Against Cancer
There are four categories of care: nurse-led survivorship implemented a national strategy in 2007.13 Similar to the
care models, risk-based care plans, rehabilitation- United Kingdom, Canadian Partnership Against Cancer has
focused care models, and self-management techniques focused on the concept of risk stratification to inform care
for post-treatment recovery.
pathways. Other nations, such as Australia, have developed
The burden of cancer is disproportionate across
subpopulations, with gender, race, ethnicity, age,
regional plans with the state government organizing the
income, health insurance, place of residence, and access initiative. This allows for pilot projects that are specific to the
to health care contributing to cancer disparities. These patient population of the region.7 In both examples of na-
factors may lead to disparities in survivorship care and tional and regional planning, there has been a population
influence survivorship experiences. focus on survivorship care with an expectation and often a
Developing policies and programs to address disparities requirement for the implementation of pilot models that will
in survivorship care may include examining how be assessed and revised as necessary.
financial hardship affects cancer outcomes, reducing In addition to the growing international acknowledgment
economic barriers to care, and increasing incorporation that individuals treated for cancer continue to have health
of patient-centered, integrated strategies into oncology care needs, there has been an evolution in thinking about
care.
who provides this care, how long follow-up is needed, and
There are four main barriers to the provision of
survivorship care: lack of evidence, lack of a trained
the role of the survivor in his or her own recovery. Initially,
survivorship care workforce, lack of reimbursement the focus of post-treatment services was generic, with a
structures/insurance coverage for survivorship care spotlight on surveillance for recurrence and the oncology
services, and lack of a cohesive health care system that specialist providing the follow up. But more recently, there
would reduce fragmented care. have been proposals and studies conducted across the globe
where models apply a risk-based approach to care so that

232 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


THE CANCER SURVIVORSHIP JOURNEY

the unique needs of survivors are addressed while effi- included face-to-face supportive care visits with the nurse
ciently applying health care resources. Although there are an and a 6-week education program.17
expanding number of survivorship models to consider, some Another important approach to survivorship care is one
important types of models are worth highlighting because that focuses not on the oncology providers but rather on the
they are applicable across countries and have the potential PCP. Using a risk-based approach to care where survivors are
to transform the delivery of survivorship care. This article stratified as being at low, medium, or high risk for recurrence
will briefly highlight four important categories of care: (1) and late effects, one can integrate the PCP in a shared-care
nurse-led survivorship care models as an important way to model or transition all the post-treatment care to the PCP at
redesign the survivorship work force; (2) risk-based care varying points based on the potential for cancer-related
plans where survivorship services are stratified based on risks. Having the PCP as a focus of the post-treatment care
need and the primary care provider (PCP) is a partner in care; has been integral to survivorship planning countries
(3) care models focused on a rehabilitation; and (4) efforts to throughout Europe and in Canada where the pan-Canadian
introduce self-management techniques for post-treatment Guideline for Survivorship services recommends that the
recovery. PCP be integrated early to provide survivorship services.18
First, there has been an increasing interest internationally Increasingly, this model is gaining traction in the United
in the development of nurse-led survivorship clinics where States for reasons of manpower concerns, a better un-
nurses are responsible for a variety of services. Although the derstanding of the shared care model successfully used by
nurse is the health care provider common to this model, the other specialties, and the growing number of older cancer
services range from active responsibility for the medical survivors for whom the PCP is essential.
follow up to providing counseling and education about late As with other survivorship models, the availability and
effects. In the United States, these clinics have been focused uptake of a rehabilitation care model will depend on the
primarily on the nurse practitioner (NP) as either an in- health care system of the country and how well established
dependent provider of the post-treatment care or as an this service is as usual care. Because rehabilitation services
individual who provides a onetime consultation visit. In the after illness are well developed throughout much of Europe,
independent model, the survivor transitions from the on- survivorship services can and are applied within this con-
cologist to the NP who is then responsible for all the ele- struct and include psychosocial care, exercise, and physical
ments included in the IOM report and provides the patient therapy. For example, in Germany, rehabilitation has long
and PCP with the treatment summary and survivorship care been established as standard care to promote recovery after
plan. In contrast, the consultation visit allows the oncologist acute illness, but it has focused on returning to work and is
to continue providing follow-up care, but the NP provides a done as an inpatient service. However, studies have shown
one-time visit focused on the psychosocial issues, makes that patients prefer outpatient services, which may make
appropriate referrals, and provides a survivorship care plan them more accessible in the future.19 In Belgium, a study was
to the survivor and PCP. conducted to offer adult cancer survivors access to a re-
Watts and colleagues have published an important de- habilitation program that included physical training and
scription of the NP role in implementing Wagners chronic psychosocial support during a 12-week program. Repeated
care model that includes shared visits and group visits, measures showed improvements in physical ability and
both of which are relevant to oncology follow-up care.14 In quality of life and a decrease in fatigue.20 In contrast, in the
Australia, research is ongoing to evaluate nurse-led survi- United States, rehabilitation services are separate and re-
vorship care for specific patient populations. For example, ferrals are made primarily to meet physical and vocational
researchers at the Peter MacCallum Cancer Centre conducted a needs. The current state of rehabilitation across the globe
randomized study comparing the effect of their SurvivorCare requires greater evaluation where these services are well
(SC) intervention with usual care for colorectal cancer survi- developed but not yet routinely offered to individuals after
vors. This new intervention includes the provision of educa- cancer treatment.
tional materials, a survivorship care plan, and a tailored, nurse- Finally, self-management is increasingly discussed in re-
led post-treatment consultation followed by three telephone lationship to cancer survivorship care. It is a component of
calls. The addition of SurvivorCare to usual care did not have a chronic care management that offers methods for patient
beneficial effect on distress, supportive care needs, or quality empowerment and prepares individuals by providing them
of life, but patients who received SurvivorCare were more with the skills and knowledge to play a role in optimizing
satisfied.15 Researchers at Peter MacCallum Cancer Centre also their health. In both the United Kingdom and Canada, na-
implemented and evaluated a nurse-led consultation that tional guidance has promoted such an approach to care
includes a tailored education package, a psychosocial assess- using a variety of interventions such as counseling, psycho-
ment, and the provision of a tailored survivorship care plan for education, and other forms of professional ad peer sup-
long-term survivors of Hodgkin lymphoma.16 In the United port.18,21 Many of the self-management programs in the
Kingdom, a nurse-led service model has been successfully United States build on the successful chronic disease self-
implemented for men treated for prostate cancer. During the management model developed by Lorig and Barlow, with a
evaluation period, the researchers found that 90% of the in- focus on promoting lifestyle change and psychosocial
vited men participated in the survivorship services, which health.22 Although it is a concept well suited to survivorship

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 233


HALPERN, MCCABE, AND BURG

care, self-management, to date, has not become a well- type of treatment received. Black men are known to have
evaluated component of survivorship care in the United lower odds of having radical prostatectomy than white
States. men, and they also report lower quality of life related to
physical (urinary, bowel, and sexual dysfunction) and psy-
chological functioning post-treatment compared with white
CONTRIBUTING FACTORS TO DISPARITIES IN men.26,29 The type of treatment a patient with cancer re-
CANCER SURVIVORSHIP ceives is an important factor in what survivors experience
We know that the burden of cancer is disproportionate in
after treatment, and the type of treatment received is a
the population. Evidence for population health disparities
function of clinical considerations and current treatment
in cancer incidence, morbidity, and mortality are indisput-
options. However, treatment choice also varies by more
able, and some of these disparities are projected to increase
modifiable factors, including what the provider commu-
in the future.23-26 Gender, race, ethnicity, age, income, health
nicates to the patient about treatment options, patient
insurance, place of residence, and access to health care are all
preferences, patient access to treatment centers, and
factors that contribute to cancer disparities. Being black in
patient ability to cover the costs of treatment. In the case of
America is particularly associated with higher risk for poor
race disparities in prostate cancer post-treatment, symp-
outcomes in cancer. For example, it is well documented that
toms apparently cannot be explained by clinical factors as
black women present with later-stage breast cancers and
much as lower socioeconomic status, being uninsured, and
more aggressive tumors, which contributes to higher mor-
having treatment in a county hospital compared with a
tality and morbidity compared with white women, and black
private hospital.29
men have higher prostate cancer incidence, more aggressive
Race and ethnic differences in the survivorship experience
disease, and the highest mortality of any population group
may be more common than we currently know, and much
from prostate cancer.24 Overall, compared with the white
more research is needed to fully understand variability in
populations, the black population is less likely to survive most
cancer outcomes. For example, there is now evidence that
forms of cancer regardless of stage of diagnosis, tumor
black pediatric cancer survivors experience increased rates
characteristics, and comorbidity.24
of anthracycline-related cardiotoxicities compared with
Ethnic identity is associated with disparities in cancer
white pediatric cancer survivors30; vitamin D deficiency rates
morbidity and mortality, but the trends are not straight-
in black and Hispanic breast cancer survivors are more
forward. Cervical cancer is an example of the complex
prevalent than among white breast cancer survivors; and
contribution of ethnic status to cancer rates and outcomes.
minority women breast cancer survivors suffer from dis-
Cervical cancer incidence is higher in Hispanic women64%
turbed sleep at higher rates than white breast cancer
higher than for non-Hispanic white women. Cervical cancer
survivors.31
incidence is also 30% higher in Native Hawaiian/Pacific Is-
Obesity may be an important crosscutting factor in dif-
lander women compared with non-Hispanic white women.
ferential cancer outcomes and side effects. Overall, cancer
Although American Indian/Alaskan Native women have
survival rates are lowest among the black population and
lower incidence of cervical cancer than white women, they
Native American/Pacific Islanderstwo population subgroups
experience increased mortality from cervical cancer.27
with a high prevalence of obesity.28,32 Obesity may also
Cancer survival for almost all cancers is less for Alaska Na-
contribute to racial and ethnic differences in certain persistent
tives than for the white population for almost all cancers.28
adverse effects of cancer treatment, including lymphedema,
The reasons for race and ethnic disparities in cancer rates
cancer-related fatigue, and chemotherapy-induced peripheral
and outcomes are complex, with the main causes including
neuropathy.32
both biologic and social factors. For example, the dramatic
problem of cancer disparities in the Alaska Native population
is believed to be associated with this ethnic groups unique
exposure to multiple infectious and carcinogenic agents,
REDUCING DISPARITIES OVER THE
their unique geographic problems with access to care, and
SURVIVORSHIP CONTINUUM
Although it is clear that there are subgroups of patients with
their not so unique problems with obesity, smoking, physical
cancer who are experiencing poorer outcomes than others,
inactivity, and alcohol consumption.28
solutions to closing this gap are not simple. One important
avenue of research is examining how financial hardship
DISPARITIES IN THE SURVIVORSHIP EXPERIENCE affects risk for poor cancer outcomes. Minority patients are
Are there reasons to believe that there is diversity in how more likely to experience greater financial hardship in the
cancer is experienced across population subgroups, similar wake of cancer than other groups.33 Cancer treatment is
to the diversity in cancer incidence and outcomes? We still expensive, and out-of-pocket costs are particularly prob-
know very little about how racial and ethnic minorities lematic for those with public health insurance or no health
experience life after cancer compared with the white ma- insurance, those with incomes under 100% of the poverty
jority, but there is evidence that side effects from cancer level, and those age 50 to 64.34 Financial circumstances may
treatment are different to some degree. For example, side constitute a barrier to good follow-up care among survivors.
effect profiles for men with prostate cancer may differ by For example, decreased and discontinued hormonal therapy

234 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


THE CANCER SURVIVORSHIP JOURNEY

adherence in women with breast cancer, delayed medical new models of care. We need models of care that support
care and prescription renewals, and higher levels of unmet patient self-management within continued, patient-centered
needs in cancer survivors are associated with low income follow-up care services. To begin to reverse disparities in
status.35-37 cancer outcomes, we must provide cancer survivors with
National oncology organizations wide endorsement of rational and practical supportspecifically the services of
cancer treatment summaries and cancer survivor care plans integrated social work professionalsto help them secure
was based on the hope that these tools would improve the social services and basic need referrals and to provide
post-treatment quality of care and increase patient self-care psychosocial support to increase their opportunity to have
following cancer treatment.6 The study conducted by Kent similar experiences in cancer as the most privileged patients
et al in 2013 asked survivors if they had received a treatment with cancer.
summary; only 17.6% had received one whereas 62.6%
wanted but did not receive one. Those who did not receive a
treatment summary had a higher number of health in-
BARRIERS TO PROVIDING HIGH-QUALITY
formation needs.37 Would the receipt of cancer care sum-
SURVIVORSHIP CARE
There are multiple reasons that every survivors journey
maries or cancer treatment plans help reduce race and
is not the same. Earle and Ganz discuss an illuminating
ethnic disparities in cancer outcomes through improved
framework for understanding the barriers to providing high-
follow-up care? In qualitative studies, minority cancer sur-
quality survivorship care.48 In this framework, physicians
vivors believed that survivorship care plans would be useful
incorporate something new into their practice only if at least
in navigating post-treatment health care.38,39 However, to
one of three conditions is met: (1) it has clearly been shown
date, research has not conclusively demonstrated effect of
to be better for their patients; (2) they are specifically re-
survivorship care plans on survivors outcomes or on dis-
munerated for it; or (3) it is more efficient for them in their
parities in survivorship care. One systematic review of
practice. Earle and Ganz state that at the time of their
outcomes of survivorship care plan found they had no effect
manuscript, survivorship care as envisioned by the IOM
on survivor distress, satisfaction with care, cancer care co-
report does not readily meet any of these criteria.
ordination, or oncologic outcomes.40 And ultimately, there is
In the context of this framework, we discuss below four
limited use of cancer care plans in oncology settings for a
barriers to the provision of survivorship care: lack of evi-
variety of practical reasons.41
dence, lack of a trained survivorship care workforce, lack of
ASCOs Recommendations for Elimination of Cancer Care
reimbursement structures/insurance coverage for survi-
Disparities in the United States called for reducing economic
vorship care services, and lack of a cohesive health care
barriers to care along with increasing research on disparities,
system that would reduce fragmented care.
increasing minority enrollment into clinical trials, and in-
creasing diversity of the oncology workforce.42 How have we
done so far? The expansion of insured minority Americans Lack of Evidence for Survivorship Care
under the Affordable Health Care for America Act has not The basis for many of the barriers to providing high-quality
solved the problem of out-of-pocket and rising costs of cancer care to all survivors is the lack of evidence regarding best
care as a barrier to receiving high-quality survivorship care. practices for survivorship care. As discussed by McCabe et al,
Women and minorities remain severely under-represented in much of the literature on survivorship care is based on
cancer clinical trials.43 There have been gains in the numbers analyses of existing datasets or surveys.49 Few prospective
of minority medical school graduates, but there is still a severe comparative studies have examined the benefits (in health
under-representation of minority physicians in oncology: in or economic terms) of alternative programs or models of
2013, only 2.3% of oncologists were black.44 care for cancer survivors. Furthermore, even the available
The 2008 IOM report, Cancer Care for the Whole Patient: databases are limited. In describing programs for childhood
Meeting Psychosocial Health Needs, promoted a standard cancer survivors, many of which have been in existence
of care that incorporates patient-centered and integrated substantially longer than have programs for adult cancer
strategies into routine oncology care, including identifying survivors, Eshelman-Kent et al reported that only 41% of
psychosocial health needs, connecting patients and families participating Childrens Oncology Group centers indicated
to needed services, supporting patients in managing illness, having a database that tracked survivors health outcomes.50
and coordinating the psychosocial and biomedical health The evidence base demonstrated that improved outcomes
care services.45 New transdisciplinary research on cancer associated with survivorship care are extremely limited.
disparities supports the notion that environmental factors More importantly, there is almost no information on
are important in cancer outcomes, specifically the role of whether one type of survivorship care model results in
accumulated social stress (such as chronic exposure to better outcomes compared with another type of model, or
poverty and violence) in the alteration of an individuals even what a survivorship care model is.51 When assess-
neuroendocrine stress response and how it influences tumor ment of survivorship programs have been performed, these
biology and cancer progression.46,47 We cannot reverse our assessments have generally focused on patient-reported
patients life experiences, but we can incorporate knowledge outcomes such as quality of life. Although these are
of the role of the social environment and social stress in clearly important, there is a critical lack of information on

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 235


HALPERN, MCCABE, AND BURG

long-term outcomes, including survival and costs, associated A potential strategy to address oncology workforce short-
with survivorship care.51 In addition, although efforts to ages related to survivorship care is to enhance training in
incorporate survivorship care plans into survivorship care this area. Rowland et al highlighted the need to train the
are becoming more widespread, there is little evidence that next generation of health care providers about survivorship
these plans have substantial or sustained benefits among issues and to attract clinicians and researchers to this area.57
survivors.40 McCabe et al also discussed expanding and coordinat-
ing educational opportunities for medical professionals
as a strategy toward improving overall survivorship care.49
Lack of a Trained Survivorship Care Workforce Essential topics specified for survivorship education curricula
Reports have discussed current and projected future short- included survivors quality of life, health promotion, palliative
ages among the oncology workforce.52 This shortage is made care, and prevention, diagnosis, and treatment of recurrences,
more acute by the increase over time in resource utilization secondary cancers, and late-occurring complications.
intensity for cancer care. As discussed by Bunnell and Shul-
man, among individuals diagnosed with cancer, there has
been an increasing number of physician visits and infusion Lack of Reimbursement for Survivorship Care Services
visits per patient.53 Related to the lack of reimbursement Lack of reimbursement for survivorship care is largely a
for survivorship services (discussed later), this workforce problem in the United States, which lacks a national health
shortage highlights the opportunity costs associated with care system. However, many of the issues related to this
survivorship care; that is, oncologists have insufficient time barrier apply globally to physicians, when there is a need to
available to engage in all patient activities and may be able prioritize the cancer care services provided. Survivorship
to earn greater reimbursement by providing cancer treat- care can be time consuming. Eshelman-Kent et al reported
ment rather than survivorship care. Furthermore, developing, that an initial visit with survivors of childhood cancer re-
implementing, and operating survivorship programs is time quired 122 minutes, whereas subsequent annual visits were
consuming. Among institutions providing care for survivors 91 minutes.50 In a survey of Australian oncologists and
of childhood cancer, the most commonly reported barrier nurses who provided breast cancer survivorship care, one-
was lack of dedicated time for developing late effects care third of these specialists reported spending more than 25%
programs.50 of their clinical time providing follow-up care.58
Other models of survivorship care, including shared care Most U.S. insurers do not provide additional reimbursement
with primary care physicians and involvement of advanced for survivorship care, which may include psychosocial support
practice providers (physician assistants and NPs), have been and care planning services. As presented in the 2009 IOM
discussed as strategies to improve survivorship care in the report Ensuring Quality Cancer Care Through the Oncology
context of oncologist shortages. However, shortages among Workforce: Sustaining Research and Care in the 21st Century:
primary care physicians and increased numbers of patients Reimbursement for survivorship care does not cover the
with health insurance (following implementation of the costs of providing appropriate monitoring and support for
Affordable Care Act) who need primary care services may the physical, social, and emotional effects in the short and
limit the ability to implement shared care survivorship long term course for the disease and treatment of cancer. In
models.53 There are also substantial differences between order to offer these services, institutions that do provide this
PCPs and oncologists regarding the perceived role of PCPS in care absorb the cost or must seek funding from other
survivorship care. In a U.S. physician survey, most PCPs sources. Many community-level institutions and smaller-
scale providers simply cannot afford to do so.59, p. 32
supported a shared care model, whereas most oncologists
favored an oncologist-based model of survivorship care.54 A McCabe et al also discuss these barriers, commenting
majority of oncologists (87%) did not feel that PCPs should on the limited reimbursement for components of survi-
take on the primary role of cancer follow up. Similarly, vorship care.49 These authors highlight that this may result
Potosky et al reported that oncologists were less likely than in a misalignment of resource utilization, with revenue-
PCPs to believe that PCPs could conduct appropriate testing generating services (such as surveillance testing) being
for breast cancer recurrence or could provide care for late overused and non-revenue-generating survivorship care
effects of breast cancer.55 These findings suggest that de- services (such as care coordination) being underused. These
spite workforce shortages, U.S. oncologists may not be nonreimbursed services may be time intensive. However,
accepting of opportunities to share care or transition care for although care coordination between oncologists and PCPs
survivors to PCPs. However, involvement of other health is almost always highlighted as an important component of
care professionals in survivorship care may be more com- survivorship care, definitions for care coordination vary
mon in other countries. For example, in a survey of Italian greatly, and little evidence is available showing the health
oncologists, Numico et al reported that only 55% and 30% of or economic benefits of care coordination for cancer
oncologists in Italy indefinitely follow patients with breast survivors.
cancer and patients with colorectal cancer, respectively, Furthermore, it may be more efficient (and equally effec-
while other oncologists discharge survivors to PCPs usually tive) to provide certain survivorship care services by email or
after 5 years.56 telephone rather than in person, but many insurers reimburse

236 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


THE CANCER SURVIVORSHIP JOURNEY

only for face-to-face delivery of care, resulting in a financial programs among an oncology workforce that is already
disincentive for provision of remote services. experiencing substantial shortages
Effectiveness of care coordination (and development
of a common specification or specifications of care
Lack of a Cohesive Health Care System That Would coordination) for survivors
Reduce Fragmented Care
Approximately 75% of survivors experience long-term or late Costs and cost-effectiveness for survivorship care
Key structures and settings, processes and services, and
effects of cancer and cancer treatment.4 These long-term
outcomes for survivorship programs, including quality of
and late effects can involve all body systems and can affect
care metrics that can potentially be associated with
all aspects of health. As such, we need a diverse range of
reimbursement.
health care providers to support cancer survivors. As dis-
Other areas of focus for survivorship care research include
cussed by Landier, the complexity of survivorship care cor-
responds to the diversity of patient characteristics, diagnoses, the following:
treatments, and potential sequelae: Development of strategies to improve provision of
patient-centered survivorship care to individuals from
Consequences of cancer treatment span the medical, psy- underserved populations and address survivorship care
chosocial, socioeconomic and spiritual domains, and survi-
disparities
vors and their healthcare providers often lack awareness of
treatment-related risks and the potential interactions of
these risks with common comorbidities, such as obesity,
Empowerment of cancer survivors to be active partic-
ipants in their care
diabetes, cardiovascular disease, anxiety, and depression. 60 Approaches to integrate survivorship care in physician
practices, health care institutions, health care systems,
This diversity in symptoms experienced and health care
and broader communities, and to include diverse groups
services needed among cancer survivors almost guarantees
of health care providers
that their health care will be fragmented. Even dedicated
and well-staffed cancer survivorship programs and health
care systems may not be able to provide care for all the
Improvement of tools and support to facilitate efficient
provisioning of survivorship care by oncologists, PCPs,
possible needs of survivors in a timely manner. and other health care professionals
Consideration of how evidence-based interventions can
be disseminated to and implemented in other settings,
FUTURE AREAS OF FOCUS FOR SURVIVORSHIP in the United States and internationally
CARE RESEARCH
Based on the information summarized above, we recom-
Dissemination of information to government and pri-
vate sector health care decision makers regarding the
mend consideration of several areas for future research on need for high-quality survivorship care and for devel-
survivorship care. We must focus on the continued devel- opment of policies that facilitate implementation and
opment of an evidence base for survivorship care. This evaluation of evidence-based programs
should include rigorous assessments of the following: As cancer care continues to improve and the survivor
Effect of different types of models of care, including
personnel providing survivorship care, use of health
population continues to grow, conducting research to ad-
dress knowledge gaps and barriers in survivorship care will
information technologies and care not administered become even more urgent. This will be critical to enhance
face to face, and comorbidities and risk adjustment health outcomes and quality of life among survivors and to
Needed resources (including funding, time, and person-
nel) to implement different types of survivorship care
help ensure that every survivor experiences an optimal and
patient-centered journey.

References
1. DeSantis CE, Lin CC, Mariotto AB, et al. Cancer treatment and survi- 6. Hewitt M, Greenfield S, Stovall E (eds). From Cancer Patient to Cancer
vorship statistics, 2014. CA Cancer J Clin. 2014;64:252-271. Survivor: Lost in Transition. Washington, DC: National Academies Press; 2005.
2. De Angelis R, Grande E, Inghelmann R, et al. Cancer prevalence esti- 7. Jefford M, Rowland J, Grunfeld E, et al. Implementing improved post-
mates in Italy from 1970 to 2010. Tumori. 2007;93:392-397. treatment care for cancer survivors in England, with reflections from
3. Herrmann C, Cerny T, Savidan A, et al. Cancer survivors in Switzerland: Australia, Canada and the USA. Br J Cancer. 2013;108:14-20.
a rapidly growing population to care for. BMC Cancer. 2013;13: 8. Bell K, Ristovski-Slijepcevic S. Cancer survivorship: why labels matter.
287. J Clin Oncol. 2013;31:409-411.
4. Mattioli V, Montanaro R, Romito F. The Italian response to cancer 9. McCabe MS, Jacobs LA. Clinical update: survivorship caremodels and
survivorship research and practice: developing an evidence base for programs. Semin Oncol Nurs. 2012;28:e1-e8.
reform. J Cancer Surviv. 2010;4:284-289. 10. McCabe MS, Partridge AH, Grunfeld E, et al. Risk-based health care, the
5. Maddams J, Utley M, Mller H. Projections of cancer prevalence in the cancer survivor, the oncologist, and the primary care physician. Semin
United Kingdom, 2010-2040. Br J Cancer. 2012;107:1195-1202. Oncol. 2013;40:804-812.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 237


HALPERN, MCCABE, AND BURG

11. McCabe MS, Faithfull S, Makin W, et al. Survivorship programs and care and Cancer 9. Switzerland: Springer International Publishing, 2014;
planning. Cancer. 2013;119:2179-2186. 63-90.
12. Richards M, Corner J, Maher J. The National Cancer Survivorship Ini- 33. Yabroff KR, Dowling EC, Guy GP Jr, et al. Financial hardship associated
tiative: new and emerging evidence on the ongoing needs of cancer with cancer in the United States: findings from a population-based
survivors. Br J Cancer. 2011;105:S1-S4. sample of adult cancer survivors. J Clin Oncol. 2016;34:259-267.
13. Canadian Partnership Again Cancer. www.partnershipagainstcancer.ca. 34. Guy GP Jr, Yabroff KR, Ekwueme DU, et al. Healthcare expenditure
Accessed February 7, 2016. burden among non-elderly cancer survivors, 2008-2012. Am J Prev Med.
14. Watts SA, Gee J, ODay ME, et al. Nurse practitioner-led multidisci- 2015;49:S489-S497.
plinary teams to improve chronic illness care: the unique strengths of 35. Burg MA, Adorno G, Lopez ED, et al. Current unmet needs of cancer
nurse practitioners applied to shared medical appointments/group survivors: analysis of open-ended responses to the American Cancer
visits. J Am Acad Nurse Pract. 2009;21:167-172. Society Study of Cancer Survivors II. Cancer. 2015;121:623-630.
15. Jefford M, Gough K, Drosdowsky A, et al. A randomized controlled trial 36. Hershman DL, Tsui J, Wright JD, et al. Household net worth, racial
of a supportive care package (SurvivorCare) for survivors of colorectal disparities, and hormonal therapy adherence among women with early-
cancer. J Clin Oncol. 2016;34:3s (suppl; abstr 79). stage breast cancer. J Clin Oncol. 2015;33:1053-1059.
16. Gates P, Seymour JF, Krishnasamy M. Insights into the development of a 37. Kent EE, Forsythe LP, Yabroff KR, et al. Are survivors who report cancer-
nurse-led survivorship care intervention for long-term survivors of related financial problems more likely to forgo or delay medical care?
Hodgkins lymphoma. Aust J Cancer Nurs. 2012;13:4-10. Cancer. 2013;119:3710-3717.
17. Ferguson J, Aning J. Prostate cancer survivorship: a nurse-led service 38. Burg MA, Lopez ED, Dailey A, et al. The potential of survivorship care
model. Br J Nurs. 2015;24:S14-S21. plans in primary care follow-up of minority breast cancer patients. J Gen
18. Howell D, Hack TF, Oliver TK, et al. Survivorship services for adult cancer Intern Med. 2009;24:S467-S471.
populations: a pan-Canadian guideline. Curr Oncol. 2011;18:e265-e281. 39. Keesing S, McNamara B, Rosenwax L. Cancer survivors experiences of
19. Bergelt C, Lehmann C, Beierlein V, et al. Patients assessment of inpatient using survivorship care plans: a systematic review of qualitative studies.
and outpatient rehabilitation. Psychooncology. 2009;18:S48. J Cancer Surviv. 2015;9:260-268.
20. Dergent T, Troch E, Vandebroek A, et al. Finding a new balance: a cancer 40. Brennan ME, Gormally JF, Butow P, et al. Survivorship care plans in
rehabilitation program. Psychooncology. 2009;18:S120. cancer: a systematic review of care plan outcomes. Br J Cancer. 2014;
21. Department of Health. The Expert Patient: A New Approach to Chronic 111:1899-1908.
Disease Management for the 21st Century. London: Her Majestys 41. Salz T, McCabe MS, Onstad EE, et al. Survivorship care plans: is there
Stationery Office; 2001. buy-in from community oncology providers? Cancer. 2014;120:
22. Barlow J, Wright C, Sheasby J, et al. Self-management approaches for 722-730.
people with chronic conditions: a review. Patient Educ Couns. 2002;48: 42. Moy B, Polite BN, Halpern MT, et al. American Society of Clinical
177-187. Oncology policy statement: opportunities in the patient protection and
23. Aizer AA, Wilhite TJ, Chen MH, et al. Lack of reduction in racial dis- affordable care act to reduce cancer care disparities. J Clin Oncol. 2011;
parities in cancer-specific mortality over a 20-year period. Cancer. 2014; 29:3816-3824.
120:1532-1539. 43. Kwiatkowski K, Coe K, Bailar JC, et al. Inclusion of minorities and women
24. American Cancer Society. Cancer Facts and Figures for African in cancer clinical trials, a decade later: have we improved? Cancer. 2013;
Americans 2013-2014. http://www.cancer.org/acs/groups/content/ 119:2956-2963.
@epidemiologysurveilance/documents/document/acspc-036921. 44. Hamel LM, Chapman R, Malloy M, et al. Critical shortage of African
pdf. Accessed February 7, 2016. American medical oncologists in the United States. J Clin Oncol. 2015;
25. Blinder VS, Griggs JJ. Health disparities and the cancer survivor. Semin 33:3697-3700.
Oncol. 2013;40:796-803. 45. Institute of Medicine. Adler NE, Page AEK (eds). Cancer Care for the
26. Borysova ME, Sultan DH, Chornokur G, et al. Prostate cancer disparities Whole Patient: Meeting Psychosocial Health Needs. Washington, DC:
through the cancer control continuum. Soc Sci. 2013;2:247-269. The National Academies Press, 2008.
27. National Cancer Institute, Division of Cancer Control and Population 46. Gehlert S, Sohmer D, Sacks T, et al. Targeting health disparities: a model
Sciences, Surveillance Research Program, Surveillance Systems Branch. linking upstream determinants to downstream interventions. Health
Surveillance, Epidemiology, and End Results (SEER) Program. http:// Aff (Millwood). 2008;27:339-349.
seer.cancer.gov/statfacts/html/cervix.html. Accessed March 13, 2016. 47. Volden PA, Conzen SD. The influence of glucocorticoid signaling on
28. White MC, Espey DK, Swan J, et al. Disparities in cancer mortality and tumor progression. Brain Behav Immun. 2013;30:S26-S31.
incidence among American Indians and Alaska Natives in the United 48. Earle CC, Ganz PA. Cancer survivorship care: dont let the perfect be the
States. Am J Public Health. 2014;104:S377-S387. enemy of the good. J Clin Oncol. 2012;30:3764-3768.
29. Jenkins R, Schover LR, Fouladi RT, et al. Sexuality and health-related 49. McCabe MS, Bhatia S, Oeffinger KC, et al. American Society of Clinical
quality of life after prostate cancer in African-American and white men Oncology statement: achieving high-quality cancer survivorship care.
treated for localized disease. J Sex Marital Ther. 2004;30:79-93. J Clin Oncol. 2013;31:631-640.
30. Aminkeng F, Ross CJ, Rassekh SR, et al. Higher frequency of genetic 50. Eshelman-Kent D, Kinahan KE, Hobbie W, et al. Cancer survivorship
variants conferring increased risk for ADRs for commonly used drugs practices, services, and delivery: a report from the Childrens Oncology
treating cancer, AIDS and tuberculosis in persons of African descent. Group (COG) nursing discipline, adolescent/young adult, and late ef-
Pharmacogenomics J. 2014;14:160-170. fects committees. J Cancer Surviv. 2011;5:345-357.
31. Yao S, Ambrosone CB. Associations between vitamin D deficiency and 51. Halpern MT, Viswanathan M, Evans TS, et al. Models of cancer survi-
risk of aggressive breast cancer in African-American women. J Steroid vorship care: overview and summary of current evidence. J Oncol Pract.
Biochem Mol Biol. 2013;136:337-341. Epub 2014 Sep 9.
32. Schmitz KH, Agurs-Collins T, Neuhouse ML, et al. Impact of obesity, race 52. Erikson C, Salsberg E, Forte G, et al. Future supply and demand for
and ethnicity on cancer survivorship. In Bowen DJ, Denis GV, Berger NA oncologists : challenges to assuring access to oncology services. J Oncol
(eds). Impact of Energy Balance on Cancer Disparities, Energy Balance Pract. 2007;3:79-86.

238 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


THE CANCER SURVIVORSHIP JOURNEY

53. Bunnell CA, Shulman LN. Will we be able to care for cancer patients in 57. Rowland JH, Hewitt M, Ganz PA. Cancer survivorship: a new challenge in
the future? Oncology (Williston Park). 2010;24:1343-1348. delivering quality cancer care. J Clin Oncol. 2006;24:5101-5104.
54. Cheung WY, Aziz N, Noone AM, et al. Physician preferences and atti- 58. Brennan ME, Butow P, Spillane AJ, et al. Survivorship care after breast
tudes regarding different models of cancer survivorship care: a com- cancer: follow-up practices of Australian health professionals and atti-
parison of primary care providers and oncologists. J Cancer Surviv. 2013; tudes to a survivorship care plan. Asia Pac J Clin Oncol. 2010;6:116-125.
7:343-354. 59. Institute of Medicine. Ensuring Quality Cancer Care Through the On-
55. Potosky AL, Han PK, Rowland J, et al. Differences between primary care cology Workforce: Sustaining Research and Care in the 21st Century:
physicians and oncologists knowledge, attitudes and practices re- Workshop Summary. Washington, DC; The National Academies Press,
garding the care of cancer survivors. J Gen Intern Med. 2011;26: 2009.
1403-1410. 60. Landier W. Survivorship care: essential components and models of delivery.
56. Numico G, Pinto C, Gori S, et al. Clinical and organizational issues in the Oncology (Williston Park). 2009;23:46-53. http://www.cancernetwork.
management of surviving breast and colorectal cancer patients: atti- com/oncology-nursing/survivorship-care-essential-components-and-models-
tudes and feelings of medical oncologists. PLoS One. 2014;9:e101170. delivery. Accessed March 14, 2016.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 239


PATIENT AND SURVIVOR CARE

Cancer Treatment as an
Accelerated Aging Process

CHAIR
Arti Hurria, MD
City of Hope
Duarte, CA

SPEAKERS
Hyman B. Muss, MD
University of North Carolina at Chapel Hill School of Medicine
Chapel Hill, NC

Lee Jones, PhD


Memorial Sloan Kettering Cancer Center
New York, NY
HURRIA, JONES, AND MUSS

Cancer Treatment as an Accelerated Aging Process:


Assessment, Biomarkers, and Interventions
Arti Hurria, MD, Lee Jones, PhD, and Hyman B. Muss, MD

OVERVIEW

An accumulating body of evidence supports the hypothesis that cancer and/or cancer treatment is associated with
accelerated aging. The majority of these data come from the pediatric literature; however, a smaller yet growing body of
literature points toward similar findings in the geriatric population. This is a key survivorship issue the growing number of
older adults with cancer face, along with the short- and long-term impact of cancer therapy on the aging process. This article
will review clinical and biologic markers of aging in older adults with cancer, use cardiovascular disease as a model of
accelerated aging, and discuss potential interventions to decrease the risk.

T he U.S. population is aging with the number of in-


dividuals age 65 or older anticipated to double between
2010 and 2030.1 This growing population is at risk for cancer
two individuals who are chronologically age 75 can have very
different functional ages. At the extremes, one individual
could be wheelchair-bound in a nursing home, and another
because the majority of cancer incidence and mortality occurs may be a marathon runner; distinguishing the difference in
in individuals age 65 and older. Together, the aging of the U.S. functional age between these two individuals can be per-
population and the association of cancer and aging is cul- formed with the eyeball test from the door of the ex-
minating in a 67% increase in cancer incidence in individuals amination room. However, for most clinical situations, an
age 65 or older in the United States from 2010 to 2030.2 individuals outward appearance can be deceiving, and an
However, many of these individuals will survive cancer, and individuals functional age can be quite difficult to determine
the majority of cancer survivors are older adults. Presently, without a more detailed evaluation. There are two main
there are 8 million cancer survivors age 65 or older in the ways in which an oncologist can get a better sense of the
United States, and this number is anticipated to continue to functional age of an older adult. The first is through performing
grow to 11 million by 2020.3 a geriatric assessment, and the second is by assessing frailty.
A key survivorship issue facing these older adults is the short- Both of these methods are discussed below.
and long-term impact of cancer therapy on the aging process. A geriatric assessment identifies factors other than
It has been suggested that cancer and/or its treatment may chronological age that can predict the risk of morbidity and
contribute to an accelerated aging phenotype.4 The majority of mortality in older adults. These include functional status,
these data come from the pediatric literature, in which can- cognition, comorbidity, psychological state, social support,
cer survivors are more predisposed to the development of and nutritional status. There is a growing body of literature
frailty as well as chronic conditions such as myocardial in- regarding the benefits of performing a geriatric assessment
farction, congestive heart failure, and second cancers.5-7 There
in older adults with cancer.8 In patients with cancer, this
is a smaller yet growing body of literature pointing toward
assessment can identify areas of vulnerability not otherwise
similar findings in the geriatric population. This article will
detected in a routine history and physical examination,
review clinical and biologic markers of aging in older adults with
predict cancer treatment toxicity and survival, and serve as a
cancer, use cardiovascular disease as a model of accelerated
platform for interventions to decrease toxicity risk.8 Short
aging, and discuss potential interventions to decrease the risk.
geriatric assessment tools for oncologists have been de-
veloped that are feasible to implement in both daily clinical
THE CLINICAL ASSESSMENT OF AGING practice and among patients enrolled in clinical trials. Ge-
The aging process is unique to the individual, and chrono- riatric assessment tools that can be mailed to the patient and
logical age is a poor descriptor of an older adult. For example, completed prior to the office visit, as well as computerized

From the City of Hope Comprehensive Cancer Center and Beckman Research Institute, Duarte, CA; Memorial Sloan Kettering Cancer Center, New York, NY; The University of North
Carolina at Chapel Hill, Chapel Hill, NC.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Arti Hurria, MD, City of Hope, 1500 East Duarte Rd., Duarte, CA 91010; email: Ahurria@coh.org.

2016 by American Society of Clinical Oncology.

e516 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


CANCER TREATMENT AND ACCELERATED AGING

geriatric assessment tools, have also been evaluated.9-11 TABLE 1. The Frailty Phenotype21
Abbreviated geriatric assessment screening tools are avail-
able; however, a consensus has not been reached regarding Categorizations Criteria
which tool could identify those patients who would benefit Frailty Phenotype: Presents Unintentional weight loss
from a more detailed geriatric assessment.12 With 3 Criteria ($ 10 pounds in past year)
There are compelling geriatric assessment findings in Self-reported exhaustion
cancer survivors that support the hypothesis that cancer and Intermediate or Prefrail Weakness (lowest 20th percentile in
its treatment may impact the aging process. Cancer survivors Phenotype: Presents grip strength)
With 1 or 2 of the Criteria
are more likely to report poorer physical and mental health- Slow walking speed (lowest
20th percentile on a timed walk of
related quality of life compared with adults with cancer.13,14 15 feet)
Older cancer survivors are more likely to have limitations in
Low physical activity (lowest quintile of
performing activities of daily living as well as mobility lim- kilocalories per week)
itations compared with older adults without a history of
cancer.15 There is an increase in the number of comorbidities
in individuals who are cancer survivors compared with et al21 utilizing data from the Cardiovascular Health Study.
those without a history of cancer.16 Furthermore, specific They identified a phenotype for frailty among over 5,000
comorbid conditions may be linked to the treatment the community-dwelling men and women age 65 or older
patient has receivedfor example, congestive heart failure (Table 1), which consists of weight loss, exhaustion, slow
(among patients receiving anthracycline-based therapies),17 walking speed, weakness, and low physical activity. Patients
peripheral neuropathy (among patients receiving taxanes),18 who were defined as frail or prefrail, compared with nonfrail,
and declines in bone health (among patients receiving aro- were at increased risk for hospitalization, falls, decreased
matase inhibitors).19,20 A study of the neuropsychological mobility, decline in the ability to complete activities of daily
function of older patients (age 6070) with breast cancer living, and mortality over the subsequent 3 and 7 years.
demonstrated that those who were exposed to chemo- Rockwood and Mitnitski22 defined another method of
therapy were at higher risk for posttreatment cognitive de- assessing frailty that is based on the accumulation of deficits.
cline and factors associated with cognitive agingsuch as Deficits captured in a geriatric assessment are tallied and
lower cognitive capacity (low cognitive reserve) and apoli- provide a composite index of the degree of frailty. These
poprotein (ApoE4+) statusinteract with chemotherapy methods of describing frailty have mainly been used as
treatment to increase the risk of cognitive decline.8 research tools rather than tools used in daily clinical practice.
Another means of assessing the aging process, which
comes primarily from the geriatric literature, is measuring BIOMARKERS OF AGING AND CANCER THERAPY
frailty. Frailty can be defined as a decrease in physiologic Geriatric assessment is the cornerstone for assessing
reserve that places an individual at increased risk for adverse function in patients with cancer prior to treatment. It can be
events such as hospitalization, falls, and poorer overall helpful in predicting survival, treatment-related toxicity, and
survival. There are two main methods of assessing frailty in other outcomes. However, geriatric assessment can be time
the geriatric population. The first was proposed by Fried consuming, and many clinicians do not have the resources to
perform a geriatric assessment in daily practice. Biomarkers
of aging may help fill this gap.23 The term biomarker has
many definitions; the World Health Organization has
KEY POINTS defined a biomarker as almost any measurement reflecting
Cancer and/or its treatment may contribute to an
an interaction between a biological system and a potential
accelerated aging phenotype. hazard, which may be chemical, physical, or biological. The
A key survivorship issue facing older adults is the short- measured response may be functional and physiological,
and long-term impact of cancer therapy on the aging biochemical at the cellular level, or a molecular interaction.
process. A list of potential biomarkers is provided in Table 2. For this
Clinical (geriatric assessment) and biologic markers of review, we will focus on several categories of potential
aging hold great promise as independent predictors of biomarkers, including chronic inflammatory markers,
patient outcomes including toxicity, functional reserve, markers of cellular senescence, and sarcopenia to explore
and survival. how they might be further evaluated with the goal of de-
Standard cancer treatment causes significant and fining markers that can independently predict outcomes in
marked impairments in global cardiovascular function,
older patients with cancer. Several excellent reviews of this
which may persist years after the cessation of primary
therapy.
topic have been recently published.23,24
Structured exercise training may be an effective strategy Inflammatory markers have been extensively studied, and
to mitigate acute and long-term impairments in increased levels have been shown to correlate with frailty,
cardiovascular function in patients both during and functional decline, and survival.24 These markers now are
following primary adjuvant therapy. receiving wide attention, as there is good evidence that
chronically elevated levels may accelerate or exacerbate the

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e517


HURRIA, JONES, AND MUSS

TABLE 2. Potential Biomarkers of Treatment-Related Toxicity and Aging


Association With Association With
Marker Source Test Frailty/Function Mortality
Chronic Inflammatory Serum or plasma ELISA Yes (CRP, IL-6, TNF-a, Yes (CRP, IL-6,
Markers D-dimer, IL1RA) D-dimer, s-VCAM)
Telomere Length Leukocyte DNA q-PCR or southern blot Yes Yes
FISH
STELA
p16INK4a T lymphocyte RNA RT-qPCR Maybe Unknown
Sarcopenia DEXA scan Commercially available software Yes Yes
for body composition analysis
CT scan
Bioelectrical
Impedance
Maximal Oxygen O2 and CO2 of inhaled and exhaled air Incremental exercise testing Yes Yes
Consumption
(VO2max)
Abbreviations: ELISA, enzyme-linked immunosorbent assay; CRP, C-reactive protein; TNF-a, tumor necrosis factor a; IL1RA, IL-1 receptor agonist; s-VCAM, soluble vascular cell
adhesion molecule; q-PCR, quantitative polymerase chain reaction; FISH, fluorescent in situ hybridization; STELA, single telomere length analysis; RT-qPCR, quantitative reverse
transcription polymerase chain reaction; DEXA, dual-energy x-ray absorptiometry.
Modified from Hubbard et al.23

aging process. These markers, which include interleukins, Other markers of potential interest include the mea-
tumor necrosis factors, and others, have been studied surement of sarcopenia; recent data show the utility of
extensively in frail patients in whom they independently using CT scans of the abdomen as a valid measure of
correlate with other measures of physical function. muscle mass and a tool to predict cancer clinical out-
Interleukin-6 (IL-6) has probably been the most extensively comes.34 This is of great interest, as CT scans are widely
studied cytokine and has been shown to predict functional used in management of patients with cancer. The role of
decline, including a diminution in the ability to perform maximum oxygen consumption (VO2max) as a biomarker of
activities of daily living, poor ambulation, and decreased aging and treatment effect is discussed elsewhere in this
mobility.25 There also appears to be a major relationship review.
between inflammatory markers and cell senescence. Se-
nescent cells are viable and capable of secreting proin-
flammatory markers that have led to the definition of a THE EFFECT OF CANCER TREATMENT ON
senescence-associated secretory phenotype.26 To date, BIOMARKERS OF AGING
however, none of these markers has assumed a major role in There is little doubt that the treatment of cancer, especially
clinical care or further studies designed to see if any single radiation therapy and chemotherapy, greatly accelerates
marker or combination might have an independent role in aging.35,36 A recent overview of survivors of childhood
the management of the older patient with cancer. These studies cancer showed that these individuals were at greatly in-
would test whether such markers could be independent pre- creased risk for substantial comorbidity and premature
dictors of treatment tolerance, including acute and chronic death.35 Data from one of the large cohorts described in this
toxicities, functional loss, and cognitive decline. review demonstrated the cumulative prevalence for a se-
Telomere length as well as the proteins that play a role in rious or life-threatening chronic condition of 81% by age 45;
telomere length are also of great interest as markers that in addition, there was an extremely high incidence of second
may predict survival, functional status, and toxicity,27,28 and neoplasms that was directly related to the radiation dose. In
studies are underway to further define the potential role of another study of survivors of childhood cancer, the preva-
telomere length as a predictor of cancer-related outcomes. lence of prefrailty and frailty were 31.5 and 13.1% among
Another emerging marker of great potential benefit as a women and 12.9 and 2.7% among men, respectively. This
predictor of toxicity and outcomes is p16ink4a. This gene, in prevalence of frailty among young adult survivors of cancer
which expression increases 10-fold with aging, codes for a with a mean age of 34 years was similar to that of adults age
protein that blocks cyclin-dependent kinase, which leads to 65 or older.37
cell senescence.29 In animal models, there is a clear direct Operative procedures result in a cascade of cytokine and
relationship between p16ink4a expression and organ age.30 acute-phase responses including IL-6, white cell count, and
In addition, human studies have shown a strong association C-reactive protein. In a systematic review that included 164
of p16ink4a expression and T-cell aging in patients infected studies involving 14,362 patients, IL-6 and C-reactive protein
with HIV,31 senescence of human mesenchymal stem cells,32 responses were clearly associated with the magnitude and
and hospital stay after coronary artery bypass.33 invasiveness of the operative procedure. Colorectal cancer

e518 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


CANCER TREATMENT AND ACCELERATED AGING

surgery resection was associated with the highest acute ACCELERATED AGING IN PATIENTS WITH
increase in cytokines.38 Although of concern in the short run, CANCER: CARDIOVASCULAR AGING AS A MODEL
it is not likely that surgical procedures directly accelerate As described in the preceding sections, older patients with
aging. Radiation therapy is clearly related to the formation cancer are subject to the deleterious effects associated with
of proinflammatory cytokines and may result in progres- normal aging but experience the confounding effects of
sive and long-term tissue damage.39 As shown in survivors cancer treatment that can lead to an accelerated aging
of childhood cancer, radiation is associated with the ac- phenotype, characterized by a notable impairment in
celerated development of second malignancies as well as physical functioning and other parameters. In current
other comorbidities. Studies testing how biomarkers might practice, the extent of functional decline or physical func-
be used to predict radiation toxicity in individual patients tioning is typically assessed using performance status
and possible interventions to ameliorate radiation effects scoring systems, evaluated either by the Karnofsky Perfor-
are needed. As the childhood cancer survivorship studies mance Scale (KPS) or the Eastern Cooperative Oncology
show, chemotherapy also is a major cause of accelerated Group (ECOG) scale. However, performance status scoring
aging. This has been well demonstrated in vitro40 and in systems are subjective and lack sensitivity to discriminate
animal models.30 Chemotherapy has a major effect on between individuals, particularly those defined as having a
telomere length41 and has been associated with telomere good (i.e., KPS . 70; ECOG 0 to 1) performance status.46,47
shortening in hematopoietic stem cells42 as well as in pe- As a result, performance status measurements are often
ripheral blood mononuclear cells in patients given repetitive supplemented with the use of other more objective mea-
standard-dose chemotherapy for solid tumors.43 Further- sures of overall physical functioning such as the compre-
more, telomere shortening was shown to be greater in older hensive geriatric assessment or measurement of cardiac
patients given combined chemotherapy and radiation for and lung function via resting assessment of left ventricular
head and neck cancer when compared with younger ejection fraction and pulmonary function testing, re-
patients. 44 spectively. Although these tools provide valuable information
p16ink4a has major promise as a biomarker of chemo- regarding physical functioning, treatment eligibility, and risk
therapy toxicity. p16ink4a expression increases approxi- stratification, they do not provide evaluation of global car-
mately 10-fold between ages 20 and 80, and this dynamic diovascular function and reserve. Indeed, cardiac function is
range provides for a more robust marker as a predictor of only one organ component that contributes to the integrative
molecular aging. In one study of women receiving adjuvant capacity of the cardiovascular and musculoskeletal system to
chemotherapy for early-stage breast cancer, p16ink4a transport and use oxygen (O2) for adenosine triphosphate
expression measured in peripheral blood T cells increased resynthesis.48 The efficiency of O2 transport and utilization
by almost one log2 order of magnitude immediately after determines an individuals cardiovascular performance (or
treatment and remained elevated 12 months after treat- exercise capacity). An incremental cardiopulmonary exercise
ment. 45 This change corresponds to almost a 15-year test with gas-exchange measurement, to assess peak oxygen
increase in chronologic age. In this study, the cytokines consumption (VO2peak), provides the gold-standard assess-
VEGFA and monocyte chemotactic protein-1 also signifi- ment of exercise capacity. VO2peak is inversely correlated with
cantly increased and remained elevated at 12 months, cardiovascular and all-cause mortality in a broad range of
but telomere length was not affected. In a cross-sectional adult populations.49-53 On this basis, several groups have
cohort of patients in the same study, prior chemother- started to examine whether the cardiopulmonary exercise
apy exposure was independently associated with in- test provides a marker of physiologic aging in the oncology
creased p16ink4a expression comparable to 10 years of setting both during and following primary adjuvant therapy.
chronologic aging. Current studies are underway explor-
ing the potential role of p16ink4a as a predictor of toxic-
ity and subsequent comorbidity in patients receiving Cancer Therapy-Induced Changes in Exercise Capacity
chemotherapy. No prospective, observational studies have examined lon-
There is no doubt that chemotherapy and radiation gitudinal changes in exercise capacity during and following
therapy accelerate aging. This is an especially concerning adjuvant therapy in patients with solid tumors; nevertheless,
observation in younger patients, for whom the dramatically it is possible to glean initial data in the context of randomized
improved survival rates for many childhood cancers are controlled trials of exercise training among patients assigned
now associated with the early development of comor- to the usual care condition. For example, Courneya et al54
bidities and an increased risk of second cancers, and in performed an exercise randomized controlled trial among
older patients, in whom such changes may result in the 242 operable patients with breast cancer receiving standard
development of new comorbidities or accelerate previous adjuvant chemotherapy. Among the 82 women assigned to
noncancer-related illness. Several biomarkers of aging are usual care (i.e., chemotherapy only), exercise capacity as
now available that might prove to identify those patients measured by VO2peak declined approximately 5% from
most vulnerable to cancer treatment and allow for the earlier baseline (following the first cycle of chemotherapy) to fol-
testing of interventions that might minimize treatment related lowing the completion of chemotherapy (median 17 weeks).
toxicity. Similarly, van Waart et al55 reported that exercise capacity,

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e519


HURRIA, JONES, AND MUSS

as measured by a maximal cycle ergometer test, decreased difference = +2.90 mL $ kg21min21; 95% CI, 1.164.64),
approximately 18% from prechemotherapy to immediately compared with nonexercising sedentary control partici-
postchemotherapy. Finally, Hornsby et al56 reported that pants.62 However, the data from individual studies are more
12 weeks of standard neoadjuvant chemotherapy was as- heterogeneous. For instance, Courneya et al54 reported that
sociated with a 9.5% decline in VO2peak from pre- supervised aerobic training was superior to usual care
chemotherapy to postchemotherapy. The impairment in (chemotherapy only) for improving VO2peak in 242 patients
exercise capacity also appears to extend to other tumor sites with operable breast cancer receiving standard adjuvant
receiving other anticancer regimens. For example, Segal chemotherapy. Interestingly, aerobic training was associ-
et al57 reported that 6 months of androgen deprivation ated with a nonsignificant improvement in VO2peak (+0.5 mL
therapy with or without radiation therapy was associated $ kg21min21) but completely abrogated the VO2peak decline
with an approximately 10% decline in VO2peak among men observed in the usual-care group. Further work from this
with advanced prostate cancer. Similarly, West et al58 found group found that supervised exercise training following
that standard neoadjuvant chemoradiation in patients with standard (i.e., three times per week, at 25 to 30 minutes/
rectal cancer caused a 16% decline in VO2peak. The long-term session), higher volume (i.e., three times per week, at 50 to
clinical importance of this decline is not known; however, 60 minutes/session), or combined (i.e., three times per week
VO2peak typically declines 10% every decade in healthy of combined aerobic and resistance training) prescriptions
women, indicating that short-term chemotherapy may did not mitigate the considerable declines in VO2peak among
cause the equivalent of a decade of physiological aging.59 301 patients with breast cancer receiving conventional
Of importance, the decline in exercise capacity may not adjuvant therapy. In contrast, Jones et al63 tested the effi-
recover, even years following the cessation of primary cacy of supervised aerobic training consisting of three
therapy. For example, Jones et al59 found that despite sessions per week at 55%100% of VO2peak for 2060 min per
normal resting cardiac function (i.e., left ventricular ejection session following a nonlinear prescription in patients with
fraction $ 50%), VO2peak was, on average, 22% below that of breast cancer receiving neoadjuvant chemotherapy; spe-
age-matched sedentary women in 140 patients with early- cifically, in nonlinear prescriptions, aerobic training sessions
stage breast cancer a mean of 27 months following the are sequenced in such a fashion that training-induced
completion of primary adjuvant therapy. In corroboration, physiologic stress is continually altered in terms of in-
Khouri et al60 found that VO2peak was, on average, 20% below tensity and duration in conjunction with appropriate rest
that of age-matched sedentary women in 57 patients with and recovery sessions to optimize cardiovascular adapta-
early-stage breast cancer a mean of 26 months following the tion. Attendance and adherence rates to aerobic training
completion of primary therapy. The persistent impairment were 82 and 66%, respectively. Intention-to-treat analysis
in exercise capacity also appears to extend beyond operable indicated that VO2peak increased by 2.6 6 3.5 ml/kg/min
breast cancer to other cancer sites. For example, Adams (+13.3%) in the chemotherapy plus aerobic training group,
et al61 performed a study of survivors with Hodgkin disease whereas it decreased by 1.5 6 2.2 ml/kg/min (28.6%) in the
(48 patients, mean of 14 years after diagnosis) and found chemotherapy only group (between-group difference, p =
that VO2peak was significantly reduced in 30% of survivors. .001). In the oncology setting, approximately five additional
Again, the clinical and prognostic importance of these dec- studies, both during and following adjuvant therapy, have
rements is currently not known, but because exercise capacity examined the safety, tolerability, and preliminary efficacy of
is a strong independent predictor of both cardiovascular as nonlinear aerobic training, compared with a usual care (no
well as all-cause mortality in noncancer populations, the exercise training) control group. Overall, exercise pre-
observed impairments are alarming and create a strong ra- scriptions adhering to a nonlinear approach appear to be
tionale for the development and testing of interventions to safe (low adverse event rate), tolerable (mean adherence $
prevent and/or treat the observed impairments. 75% of prescribed sessions both during and after primary
adjuvant therapy), and efficacious, conferring favorable
improvements in VO2peak, quality of life, and other physi-
Efficacy of Exercise Training Countermeasures ologic outcomes.64
Aerobic (exercise) training is the most effective therapy to In summary, the extant evidence indicates that patients
improve VO2peak in healthy individuals given that it improves with cancer experience considerable and marked impair-
the reserve capacity of all O2 transport organs, which to- ments in exercise capacity during cancer therapy that appear
gether lead to favorable improvements in exercise capac- to persist even years following the completion of primary
ity,48 although fewer trials have examined the efficacy of treatmentsuch decrements are consistent with an
exercise on exercise capacity, as measured by VO2peak, in accelerated cardiovascular aging phenotype and may, in
patients with cancer, with the vast majority of work to date part, contribute to the increased risk of cardiovascular
in women with early-stage breast cancer. In a recent meta- disease, frailty, and functional dependence in certain cancer
analysis of six exercise training randomized controlled trials populations. Based on current data, supervised aerobic
(involving 571 patients) that assessed the effects of exercise exercise training appears to be a safe, tolerable, and effi-
training in adults with cancer, exercise training led to a cacious intervention strategy to potentially offset as well as
significant improvement in VO2peak (mean weighted recover impaired cardiopulmonary function in a broad range

e520 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


CANCER TREATMENT AND ACCELERATED AGING

of patients with cancer. The mechanisms, optimal timing, Improve or Maintain Quality of Survivorship in Older and/or
type, and schedule of exercise training, as well as the long- Frail Adults with Cancer, in which gaps in knowledge and
term clinical implications of declines and/or improvements research priorities to fill these gaps were recommended.
in exercise capacity, are a high research priority in geriatric Among the key recommendations was the need to expand
oncology. studies focusing on the survivorship issues facing older
adults with cancer, the impact of cancer on the aging pro-
CONCLUSION cess, as well as interventions to decrease the risk. Inclusion
An accumulating body of evidence is supporting the hy- of a geriatric assessment and biomarkers of aging in research
pothesis that cancer and/or cancer treatment is associated studies will be needed to accomplish these goals. In-
with accelerated aging; however, several gaps in knowledge terventions are needed to halt or modify the accelerated
remain, and future research is needed to understand the aging phenotype seen in survivors of cancer. The compelling
implications of these findings, as well as ways to decrease data with regard to exercise can serve as a model for future
the risk. This unmet need formed the basis for a research studies in the years to come.
conference of the Cancer and Aging Research Group, Na-
tional Institute on Aging, and National Cancer Institute, ti- ACKNOWLEDGMENT
tled Design and Implementation of Intervention Studies to All authors contributed equally.

References
1. Howlader N, Noone AM, Krapcho M, et al. SEER Cancer Statistics Re- 13. Reeve BB, Potosky AL, Smith AW, et al. Impact of cancer on health-
view, 1975-2012, National Cancer Institute. Bethesda, MD, http://seer. related quality of life of older Americans. J Natl Cancer Inst. 2009;101:
cancer.gov/csr/1975_2012/, based on November 2014 SEER data 860-868.
submission, posted to the SEER web site, April 2015. 14. Weaver KE, Forsythe LP, Reeve BB, et al. Mental and physical health-
2. Smith BD, Smith GL, Hurria A, et al. Future of cancer incidence in the related quality of life among U.S. cancer survivors: population estimates
United States: burdens upon an aging, changing nation. J Clin Oncol. from the 2010 National Health Interview Survey. Cancer Epidemiol
2009;27:2758-2765. Biomarkers Prev. 2012;21:2108-2117.
3. Parry C, Kent EE, Mariotto AB, et al. Cancer survivors: a booming 15. Keating NL, Nrredam M, Landrum MB, et al. Physical and mental health
population. Cancer Epidemiol Biomarkers Prev. 2011;20:1996-2005. status of older long-term cancer survivors. J Am Geriatr Soc. 2005;53:
4. Henderson TO, Ness KK, Cohen HJ. Accelerated aging among cancer 2145-2152.
survivors: from pediatrics to geriatrics. Am Soc Clin Oncol Educ Book. 16. Baker F, Haffer SC, Denniston M. Health-related quality of life of cancer
2014;34:e423-e430. and noncancer patients in Medicare managed care. Cancer. 2003;97:
5. Mulrooney DA, Yeazel MW, Kawashima T, et al. Cardiac outcomes in a 674-681.
cohort of adult survivors of childhood and adolescent cancer: retro- 17. Pinder MC, Duan Z, Goodwin JS, et al. Congestive heart failure in older
spective analysis of the Childhood Cancer Survivor Study cohort. BMJ. women treated with adjuvant anthracycline chemotherapy for breast
2009;339:b4606. cancer. J Clin Oncol. 2007;25:3808-3815.
6. Oeffinger KC, Mertens AC, Sklar CA, et al; Childhood Cancer Survivor 18. Lichtman SM, Hurria A, Cirrincione CT, et al; Cancer and Leukemia Group
Study. Chronic health conditions in adult survivors of childhood cancer. B. Paclitaxel efficacy and toxicity in older women with metastatic breast
N Engl J Med. 2006;355:1572-1582. cancer: combined analysis of CALGB 9342 and 9840. Ann Oncol. 2012;
7. Mohile SG, Xian Y, Dale W, et al. Association of a cancer diagnosis with 23:632-638.
vulnerability and frailty in older Medicare beneficiaries. J Natl Cancer 19. Eastell R, Adams JE, Coleman RE, et al. Effect of anastrozole on bone
Inst. 2009;101:1206-1215. mineral density: 5-year results from the anastrozole, tamoxifen, alone
8. Wildiers H, Heeren P, Puts M, et al. International Society of Geriatric or in combination trial 18233230. J Clin Oncol. 2008;26:1051-1057.
Oncology consensus on geriatric assessment in older patients with 20. Choksi P, Williams M, Clark PM, et al. Skeletal manifestations of
cancer. J Clin Oncol. 2014;32:2595-2603. treatment of breast cancer. Curr Osteoporos Rep. 2013;11:319-328.
9. Ingram SS, Seo PH, Martell RE, et al. Comprehensive assessment of the 21. Fried LP, Tangen CM, Walston J, et al; Cardiovascular Health Study
elderly cancer patient: the feasibility of self-report methodology. J Clin Collaborative Research Group. Frailty in older adults: evidence for a
Oncol. 2002;20:770-775. phenotype. J Gerontol A Biol Sci Med Sci. 2001;56:M146-M156.
10. Hurria A, Lichtman SM, Gardes J, et al. Identifying vulnerable older 22. Rockwood K, Mitnitski A. Frailty in relation to the accumulation of
adults with cancer: integrating geriatric assessment into oncology deficits. J Gerontol A Biol Sci Med Sci. 2007;62:722-727.
practice. J Am Geriatr Soc. 2007;55:1604-1608. 23. Hubbard JM, Cohen HJ, Muss HB. Incorporating biomarkers into cancer
11. McCleary NJ, Wigler D, Berry D, et al. Feasibility of computer-based self- and aging research. J Clin Oncol. 2014;32:2611-2616.
administered cancer-specific geriatric assessment in older patients with 24. Franceschi C, Campisi J. Chronic inflammation (inflammaging) and its
gastrointestinal malignancy. Oncologist. 2013;18:64-72. potential contribution to age-associated diseases. J Gerontol A Biol Sci
12. Decoster L, Van Puyvelde K, Mohile S, et al. Screening tools for mul- Med Sci. 2014;69(Suppl 1):S4-S9.
tidimensional health problems warranting a geriatric assessment in 25. Ferrucci L, Penninx BW, Volpato S, et al. Change in muscle strength
older cancer patients: an update on SIOG recommendations. Ann Oncol. explains accelerated decline of physical function in older women with
2015;26:288-300. high interleukin-6 serum levels. J Am Geriatr Soc. 2002;50:1947-1954.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e521


HURRIA, JONES, AND MUSS

26. Campisi J, Robert L. Cell senescence: role in aging and age-related contemporary palliative care clinical practice [ISRCTN81117481]. BMC
diseases. Interdiscip Top Gerontol. 2014;39:45-61. Palliat Care. 2005;4:7.
27. Chiodi I, Mondello C. Telomere and telomerase stability in human 48. Jones LW, Eves ND, Haykowsky M, et al. Exercise intolerance in cancer
diseases and cancer. Front Biosci (Landmark Ed). 2016;21:203-224. and the role of exercise therapy to reverse dysfunction. Lancet Oncol.
28. Jia H, Wang Z. Telomere Length as a Prognostic Factor for Overall Survival 2009;10:598-605.
in Colorectal Cancer Patients. Cell Physiol Biochem. 2016;38:122-128. 49. Kavanagh T, Mertens DJ, Hamm LF, et al. Prediction of long-term
29. Liu Y, Sanoff HK, Cho H, et al. Expression of p16(INK4a) in peripheral prognosis in 12 169 men referred for cardiac rehabilitation. Circula-
blood T-cells is a biomarker of human aging. Aging Cell. 2009;8:439-448. tion. 2002;106:666-671.
30. Krishnamurthy J, Torrice C, Ramsey MR, et al. Ink4a/Arf expression is a 50. Kavanagh T, Mertens DJ, Hamm LF, et al. Peak oxygen intake and cardiac
biomarker of aging. J Clin Invest. 2004;114:1299-1307. mortality in women referred for cardiac rehabilitation. J Am Coll Cardiol.
31. Nelson JA, Krishnamurthy J, Menezes P, et al. Expression of p16(INK4a) 2003;42:2139-2143.
as a biomarker of T-cell aging in HIV-infected patients prior to and 51. Kubozono T, Itoh H, Oikawa K, et al. Peak VO(2) is more potent than B-
during antiretroviral therapy. Aging Cell. 2012;11:916-918. type natriuretic peptide as a prognostic parameter in cardiac patients.
32. Shang J, Yao Y, Fan X, et al. miR-29c-3p promotes senescence of human Circ J. 2008;72:575-581.
mesenchymal stem cells by targeting CNOT6 through p53-p21 and p16- 52. Aaronson KD, Mancini DM. Is percentage of predicted maximal exercise
pRB pathways. Biochim Biophys Acta. 2016;1863:520-532. oxygen consumption a better predictor of survival than peak exercise
33. Pustavoitau A, Barodka V, Sharpless NE, et al. Role of senescence marker oxygen consumption for patients with severe heart failure? J Heart Lung
p16(INK4a) measured in peripheral blood T-lymphocytes in predicting Transplant. 1995;14:981-989.
length of hospital stay after coronary artery bypass surgery in older 53. Jones LW, Watson D, Herndon JE II, et al. Peak oxygen consumption and
adults. Exp Gerontol. 2016;74:29-36. long-term all-cause mortality in nonsmall cell lung cancer. Cancer. 2010;
34. Kazemi-Bajestani SM, Mazurak VC, Baracos V. Computed tomography- 116:4825-4832.
defined muscle and fat wasting are associated with cancer clinical 54. Courneya KS, Segal RJ, Mackey JR, et al. Effects of aerobic and resistance
outcomes. Semin Cell Dev Biol. Epub 2015 Sep 3. exercise in breast cancer patients receiving adjuvant chemotherapy:
35. Bhatia S, Armenian SH, Armstrong GT, et al. Collaborative Research in a multicenter randomized controlled trial. J Clin Oncol. 2007;25:
Childhood Cancer Survivorship: The Current Landscape. J Clin Oncol. 4396-4404.
2015;33:3055-3064. 55. van Waart H, Stuiver MM, van Harten WH, et al. Effect of low-intensity
36. Armstrong GT, Kawashima T, Leisenring W, et al. Aging and risk of physical activity and moderate- to high-intensity physical exercise
severe, disabling, life-threatening, and fatal events in the childhood during adjuvant chemotherapy on physical fitness, fatigue, and che-
cancer survivor study. J Clin Oncol. 2014;32:1218-1227. motherapy completion rates: results of the PACES randomized clinical
37. Ness KK, Krull KR, Jones KE, et al. Physiologic frailty as a sign of trial. J Clin Oncol. 2015;33:1918-1927.
accelerated aging among adult survivors of childhood cancer: a report 56. Hornsby WE, Douglas PS, West MJ, et al. Safety and efficacy of aerobic
from the St Jude Lifetime cohort study. J Clin Oncol. 2013;31:4496-4503. training in operable breast cancer patients receiving neoadjuvant
38. Watt DG, Horgan PG, McMillan DC. Routine clinical markers of the chemotherapy: a phase II randomized trial. Acta Oncol. 2014;53:65-74.
magnitude of the systemic inflammatory response after elective op- 57. Segal RJ, Reid RD, Courneya KS, et al. Randomized controlled trial of
eration: a systematic review. Surgery. 2015;157:362-380. resistance or aerobic exercise in men receiving radiation therapy for
39. Kim JH, Jenrow KA, Brown SL. Mechanisms of radiation-induced normal prostate cancer. J Clin Oncol. 2009;27:344-351.
tissue toxicity and implications for future clinical trials. Radiat Oncol J. 58. West MA, Loughney L, Lythgoe D, et al. Effect of prehabilitation on
2014;32:103-115. objectively measured physical fitness after neoadjuvant treatment in
40. Buttiglieri S, Ruella M, Risso A, et al. The aging effect of chemotherapy preoperative rectal cancer patients: a blinded interventional pilot study.
on cultured human mesenchymal stem cells. Exp Hematol. 2011;39: Br J Anaesth. 2015;114:244-251.
1171-1181. 59. Jones LW, Courneya KS, Mackey JR, et al. Cardiopulmonary function and
41. Beeharry N, Broccoli D. Telomere dynamics in response to chemo- age-related decline across the breast cancer survivorship continuum.
therapy. Curr Mol Med. 2005;5:187-196. J Clin Oncol. 2012;30:2530-2537.
42. Diker-Cohen T, Uziel O, Szyper-Kravitz M, et al. The effect of chemo- 60. Khouri MG, Hornsby WE, Risum N, et al. Utility of 3-dimensional
therapy on telomere dynamics: clinical results and possible mecha- echocardiography, global longitudinal strain, and exercise stress
nisms. Leuk Lymphoma. 2013;54:2023-2029. echocardiography to detect cardiac dysfunction in breast cancer pa-
43. Yoon SY, Sung HJ, Park KH, et al. Telomere length shortening of pe- tients treated with doxorubicin-containing adjuvant therapy. Breast
ripheral blood mononuclear cells in solid-cancer patients undergoing Cancer Res Treat. 2014;143:531-539.
standard-dose chemotherapy might be correlated with good treatment 61. Adams MJ, Lipsitz SR, Colan SD, et al. Cardiovascular status in long-term
response and neutropenia severity. Acta Haematol. 2007;118:30-37. survivors of Hodgkins disease treated with chest radiotherapy. J Clin
44. Unryn BM, Hao D, Gluck S, et al. Acceleration of telomere loss by Oncol. 2004;22:3139-3148.
chemotherapy is greater in older patients with locally advanced head 62. Jones LW, Liang Y, Pituskin EN, et al. Effect of exercise training on peak
and neck cancer. Clin Cancer Res. 2006;12:6345-6350. oxygen consumption in patients with cancer: a meta-analysis. Oncol-
45. Sanoff HK, Deal AM, Krishnamurthy J, et al. Effect of cytotoxic che- ogist. 2011;16:112-120.
motherapy on markers of molecular age in patients with breast cancer. 63. Jones LW, Fels DR, West M, et al. Modulation of circulating angiogenic
J Natl Cancer Inst. 2014;106:dju057. factors and tumor biology by aerobic training in breast cancer patients
46. Jones LW, Cohen RR, Mabe SK, et al. Assessment of physical functioning receiving neoadjuvant chemotherapy. Cancer Prev Res (Phila). 2013;6:
in recurrent glioma: preliminary comparison of performance status to 925-937.
functional capacity testing. J Neurooncol. 2009;94:79-85. 64. Sasso JP, Eves ND, Christensen JF, et al. A framework for prescription in
47. Abernethy AP, Shelby-James T, Fazekas BS, et al. The Australia-modified exercise-oncology research. J Cachexia Sarcopenia Muscle. 2015;6:
Karnofsky Performance Status (AKPS) scale: a revised scale for 115-124.

e522 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


PATIENT AND SURVIVOR CARE

New and Improved? Use of White


Blood Cell Growth Factors in
Oncology Practice

CHAIR
Thomas J. Smith, MD, FACP, FASCO, FAAHPM
Johns Hopkins University School of Medicine
Baltimore, MD

SPEAKERS
Gary H. Lyman, MD, MPH, FASCO, FACP, FRCP
Fred Hutchinson Cancer Research Center
Seattle, WA

James O. Armitage, MD
University of Nebraska Medical Center
Omaha, NE
SMITH AND HILLNER

Real-World Conundrums and Biases in the Use of White Cell


Growth Factors
Thomas J. Smith, MD, FACP, FASCO, FAAHPM, and Bruce E. Hillner, MD, FASCO

OVERVIEW

We present the 2015 American Society of Clinical Oncology (ASCO) white cell growth factors, or colony-stimulating factor (CSF),
guidelines, updated from 2006. One new indication has been addeddose-intense chemotherapy for bladder cancerto
accompany the existing use for dose-dense breast cancer chemotherapy. Colony-stimulating factors remain appropriate for
any regimen where the risk of febrile neutropenia is about 20% per cycle and dose reduction is not appropriate. Based on new
evidence from multiple trials, CSF use is no longer indicated in treatment of lymphoma unless there are special risk factors. The
United States accounts for 78% of the sales of CSF. The panel approved the use of all biosimilars, but the cost savings will
be small as the price is about 80% of the branded CSFs. More biosimilars at lower cost are awaited. Methods to reduce use
without harm to patients, by requiring justification according to accepted guidelines, are ongoing.

n 2015, the ASCO guideline on the use of CSFs was updated.1


I The panel reviewed 66 publications that met eligibility criteria,
including 41 randomized trials.
regular M-VAC) was found with the use of neoadjuvant
M-VAC with CSF support.3,4
There are now two clinical settings in which dose-dense
Since the preceding update in 2006, what has changed? Also, chemotherapy is indicated instead of usual-dose chemo-
what is likely to change in the near future? The short answer is therapy, breast cancer and uroepithelial cancer. As noted in
that, unless you treat bladder cancer or lymphoma, not much the guideline, dose-dense regimens that require a CSF
has changed, and there are no major practice-changing im- should only be used within an appropriately designed clinical
plications for most oncologists. The Sidebar summarizes the trial or if supported by convincing efficacy data.1
guideline report. The subsequent commentary is our own For those oncologists who use high-dose chemotherapy
perspective and is not necessarily that of ASCO or the panel. and stem-cell transplantation, CSFs may be used alone, after
chemotherapy, or with plerixafor to help mobilize stem cells.
This has been standard practice for many years.5,6
NEW REASONS AND SITUATIONS WHEN
COLONY-STIMULATING FACTORS ARE
INDICATED NEW REASONS AND SITUATIONS WHEN
There is now one additional regimen to add to the short list COLONY-STIMULATING FACTORS ARE NO
of indicated combinations for dose-dense chemotherapy, LONGER INDICATED
high-dose methotrexate/vinblastine/doxorubicin/cisplatin The major change in the 2015 guideline was a recommen-
(M-VAC) chemotherapy in urogenital cancers. In a phase dation to not routinely use CSFs in the treatment of diffuse
III clinical trial with 7.3 years of follow-up time, the median B-cell non-Hodgkin lymphoma in nonelderly patients (youn-
and 5-year overall survivals were 15.1 months and 21.8% in ger than age 65 years with no major comorbidities) on the
the high-dose M-VAC arm, compared with 14.9 months and basis of the lack of compelling benefit for 14-day CHOP
13.5% in the M-VAC arm (hazard ratio for mortality, 0.76).2 (cyclophosphamide, doxorubicin, vincristine, and prednisolone)
As the authors noted, the toxicity was not worse, fewer versus 21-day CHOP in two large definitive trials that had
people died of their cancers, and high-dose M-VAC median follow-up times of 5 and 4 years.7,8 The same
produces a borderline statistically significant relative re- conclusion held for a trial in older patients, in which there
duction in the risk of progression and death compared to was no benefit of 14-day versus 21-day R-CHOP (CHOP +
M-VAC.2 A similar result of equal efficacy and response rituximab).9 This guidance is in line with expert opinion ex-
rates with less toxicity (major toxicity rate, 32% vs. 55% for pressed elsewhere and with worldwide common practice.10

From the Harry J. Duffey Family Palliative Care Program of the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD; Hillner Consulting, LLC, Richmond, VA;
Massey Cancer Center, Virginia Commonwealth University, Richmond, VA.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Thomas J. Smith, MD, Sidney Kimmel Comprehensive Cancer Center, 600 North Wolfe St., Blalock 369, Baltimore, MD 21287; email: tsmit136@jhmi.edu.

2016 by American Society of Clinical Oncology.

e524 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


ISSUES ON THE USE OF WHITE CELL GROWTH FACTORS

FIGURE 1. Amgen Sales of Colony-Stimulating Factors Compared With World Population

NEW PRODUCTS THAT MAY REDUCE THE AMOUNT 78% of the worlds CSF sales. How much of the current U.S.
SOCIETY PAYS FOR COLONY-STIMULATING use is related to income generation from CSF sales compared
FACTORS with patient demand or physician training may shift and is
Filgrastim and peg-filgrastim are expensive. Despite in- worthy of study as large institutional providers and practices
troductions to the market more than 15 years ago, their begin switching to accountable care models with global
prices are actually increasing. As reported in the Amgen payments.
annual report, the 2014 Neulasta sales increased 7 percent What is the potential impact on the U.S. oncology mar-
year-over-year for the fourth quarter and 5 percent for the ketplace if we accept (as the ASCO panel and the U.S. Food
full year driven mainly by price.11 In 2014, the U.S. sales for and Drug Administration [FDA] have) that biosimilars are
Amgen were $4.488 billion from peg-filgrastim and fil- indeed biosimilar? As the guideline stated, pegfilgrastim,
grastim; in contrast, the sales to the rest of the world were filgrastim, tbo-filgrastim, and filgrastim-sndz (and other
just $1.267 billion (Fig. 1).11 biosimilars, as they become available) can be used for the
These are just revenues to Amgen and do not represent prevention of treatment-related febrile neutropenia. The
what patients and payers actually paid. The United States choice of agent depends on convenience, cost, and the
represents 4.5% of the world population12 and uses 20% of clinical situation. Tbo-filgrastim has captured approxi-
the worlds supply of energy13; the United States purchases mately 20% of the worldwide market for filgrastim.14 The
manufacturer, Teva, has stated that tbo-filgrastim now
claims 38% of the market share for in-hospital use in the
United States.15 Balugrastim (manufactured by Teva), a
recombinant protein that combines albumin and human
KEY POINTS granulocyte CSF (G-CSF), was compared with pegfilgrastim
CSFs are indicated to give dose-intense chemotherapy
in a trial with 256 patients who had breast cancer, and
for adjuvant breast cancer and neoadjuvant and balugrastim had noninferior results, maintained a 1-day
metastatic uroepithelial cancers using intensified duration of severe neutropenia, and had a slightly shorter
M-VAC. recovery of absolute neutrophil counts in cycle 1.16 How-
CSFs are indicated when the risk of febrile neutropenia is ever, Teva withdrew its U.S. application for FDA approval for
about 20% and dose reduction is not an appropriate balugrastim. Teva has released another long-acting bio-
strategy; most regimens have a risk of febrile similar CSF in Europe, lipegfilgrastim, that will be in direct
neutropenia that is less than 20%. competition with pegfilgrastim. Preliminary results showed
CSFs are not indicated in the routine treatment of equal efficacy.17,18 We could not find any estimates of the
lymphoma with regimens such as R-CHOP. cost impact.
The United States uses nearly 80% of the worlds supply
The economic impact of the biosimilars for this indication
of CSFs, far more than any nation, and the price
continues to rise, therefore attempts to reduce use will
has been modest; tbo-filgrastim was priced at approximately
increase. 80% of the price of its direct competitor.19 More direct ways
The use of biosimilars for CSFs was strongly endorsed by to reduce overall CSF use (specifically, peer-to-peer review
the committee and the U.S. Food and Drug anytime a CSF is ordered) are also successful,20 but most
Administration. U.S.insurers and Medicare have not taken that approach
to date. ASCO, in its initial wave of Choosing Wisely topic

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e525


SMITH AND HILLNER

SIDEBAR. 2015 Recommendations for the Use of White Blood Cell Growth Factors: ASCO Clinical Practice
Guideline Update*
Primary prophylaxis with a CSF starting with the first cycle and continuing through subsequent cycles of chemotherapy is rec-
ommended in patients who have an approximately 20% or higher risk for febrile neutropenia based on patient-, disease- and
treatment-related factors. Primary CSF prophylaxis should also be given in patients receiving dose-dense chemotherapy when
considered appropriate. Consideration should be given to alternative, equally effective and safe chemotherapy regimens not re-
quiring CSF support when available. (Type: evidence-based, benefits outweigh harms. Evidence quality: high. Strength of recom-
mendation: strong.)
Secondary prophylaxis with CSFs is recommended for patients who experienced a neutropenic complication from a prior cycle of
chemotherapy (for which primary prophylaxis was not received), in which a reduced dose or treatment delay may compromise
disease-free or overall survival or treatment outcome. In many clinical situations, dose reduction or delay may be a reasonable
alternative. (Type: evidence-based, benefits outweigh harms. Evidence quality: high. Strength of recommendation: strong.)
CSFs should not be routinely used for patients with neutropenia who are afebrile. (Type: evidence-based, benefits outweigh harms.
evidence quality: high. Strength of recommendation: strong.)
CSFs should not be routinely used as adjunctive treatment with antibiotic therapy for patients with fever and neutropenia. However,
CSFs should be considered in patients with fever and neutropenia who are at high-risk for infection-associated complications, or who
have prognostic factors that are predictive of poor clinical outcomes. (Type: evidence-based, benefits outweigh harms. Evidence
quality: high. Strength of recommendation: strong.)
Dose-dense regimens with CSF support should only be used if supported by convincing efficacy data or within an appropriately
designed clinical trial. Efficacy data support the use of dose-dense chemotherapy in the adjuvant treatment of high-risk breast cancer,
and the use of high-dose intensity methotrexate, vinblastine, doxorubicin, and cisplatin in urothelial cancer. There are limited and
conflicting data on the value of dose-dense regimens with CSF support in non-Hodgkin lymphoma, and it cannot routinely be
recommended at this time. (Type: evidence-based, benefits outweigh harms. Evidence quality: high for breast cancer and lymphoma,
intermediate for urothelial cancer. Strength of recommendation: strong for breast cancer and lymphoma, moderate for urothelial
cancer.)
CSFs may be used alone, after chemotherapy, or in combination with plerixafor to mobilize peripheral-blood progenitor cells.
Choice of mobilization strategy depends in part on type of cancer and type of transplantation. (Type: evidence-based, benefits
outweigh harms. Evidence quality: strong. Strength of recommendation: high.)
CSFs should be administered after autologous stem-cell transplantation to reduce the duration of severe neutropenia. (Type:
evidence-based, benefits outweigh harms. Evidence quality: high; Strength of recommendation: strong.)
CSFs may be administered after allogeneic stem-cell transplantation to reduce the duration of severe neutropenia. (Type: evidence-
based. Evidence quality: low. Strength of recommendation: weak).
Prophylactic CSF for patients with diffuse aggressive lymphoma age 65 years and older treated with curative chemotherapy (cy-
clophosphamide, doxorubicin, vincristine, prednisone and rituximab,) should be considered, particularly in the presence of
comorbidities. (Type: evidence-based, benefits outweigh harms. Evidence quality: intermediate. Strength of recommendation:
moderate.)
The use of CSFs in pediatric patients will almost always be guided by clinical protocols. As in adults, the use of CSFs is reasonable for the
primary prophylaxis of pediatric patients with a high likelihood of febrile neutropenia. Similarly, the use of CSFs for secondary
prophylaxis or for therapy should be limited to high-risk patients. (Type: evidence-based, benefits outweigh harms. Evidence quality:
high. Strength of recommendation: strong.)
For pediatric indications in which dose-intense chemotherapy is known to have a survival benefit, such as Ewing sarcoma, CSFs should
be used to enable the administration of these regimens. (Type: evidence-based, benefits outweigh harms. Evidence quality: high.
Strength of recommendation: strong.)
CSFs should not be used in pediatric patients with nonrelapsed acute lymphoblastic leukemia or nonrelapsed acute myeloid leukemia
who do not have an infection. (Type: informal consensus. Evidence quality: intermediate. Strength of recommendation: moderate.)
Pegfilgrastim, filgrastim, tbo-filgrastim, and filgrastim-sndz (and other biosimilars, as they become available) can be used for the
prevention of treatment-related febrile neutropenia. The choice of agent depends on convenience, cost, and the clinical situation.
There have been no further data comparing granulocyte-CSF (G-CSF) and granulocyte macrophageCSF since the 2006 update;
therefore, there is no change in the recommendation regarding their therapeutic equivalency.(Type: evidence-based, benefits
outweigh harms. Evidence quality: high. Strength of recommendation: strong.)
Current recommendations for the management of patients exposed to lethal doses of total-body radiotherapy, but not doses high
enough to lead to certain death due to injury to other organs, includes the prompt administration of CSF or pegylated G-CSF. (Type:
formal consensus (by others), benefits outweigh harms. Evidence quality: intermediate. Strength of recommendation: moderate.)

*Reproduced from the ASCO guidelines; bold and italics emphasis added by the authors.

e526 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


ISSUES ON THE USE OF WHITE CELL GROWTH FACTORS

recommendations, has promoted a reduction in use of in growth factor overuseuse in patients at low risk of
CSFs in support of noncurative-intent chemotherapy.21 febrile neutropenia.22 Less is known about the more
Overall, there has been no concerted effort by ASCO to consequential problem of CSF underusethat is, not
reduce use among its members; we could envision a using them in patients at high risk of febrile neutropenia.
concerted social marketing effort that involves patients, At a time when all other drug costs are rising exponen-
providers, and payers, much like survivorship initiatives. tially, it may be tempting to leave U.S. consumption of
A recent review noted that physician training, experi- CSFs as is, but there are multiple opportunities to improve
ence, and compensation all appeared to play large roles value without inducing harm.

References

1. Smith TJ, Bohlke K, Lyman GH, et al. Recommendations for the use of 11. Amgen. Amgens 2014 Revenues Increased 7 Percent To $20.1 Billion
WBC growth factors: American Society of Clinical Oncology Clinical And Adjusted Earnings Per Share (EPS) Increased 14 Percent To
Practice Guideline Update. J Clin Oncol. 2015;33:3199-3212. $8.70. http://investors.amgen.com/phoenix.zhtml?c=61656&p=irol-
2. Sternberg CN, de Mulder P, Schornagel JH, et al; EORTC Genito-Urinary newsArticle&ID=2010978. Accessed February 8, 2016.
Cancer Group. Seven year update of an EORTC phase III trial of high- 12. Worldometers. Current World Population. http://www.worldometers.
dose intensity M-VAC chemotherapy and G-CSF versus classic M-VAC in info/world-population/#top20. Accessed February 11, 2016, and March
advanced urothelial tract tumours. Eur J Cancer. 2006;42:50-54. 15, 2016.
3. van de Putte EE, Mertens LS, Meijer RP, et al. Neoadjuvant induction 13. World Population Balance. Population Energy and Consumption. www.
dose-dense MVAC for muscle invasive bladder cancer: efficacy and worldpopulationbalance.org/population_energy. Accessed February 11,
safety compared with classic MVAC and gemcitabine/cisplatin. World J 2016, and March 15, 2016.
Urol. 2016;34:157-162. 14. Renwick MJ, Smolina K, Gladstone EJ, et al. Postmarket policy con-
4. Choueiri TK, Jacobus S, Bellmunt J, et al. Neoadjuvant dose-dense siderations for biosimilar oncology drugs. Lancet Oncol. 2016;17:
methotrexate, vinblastine, doxorubicin, and cisplatin with pegfil- e31-e38.
grastim support in muscle-invasive urothelial cancer: pathologic, ra- 15. Trusted to take a bite out of G-CSF acquisition costs. U.S. Pharmacist.
diologic, and biomarker correlates. J Clin Oncol. 2014;32:1889-1894. January 2016. p HS 7. http://bt.e-ditionsbyfry.com/publication/?i=287196.
5. DiPersio JF, Micallef IN, Stiff PJ, et al; 3101 Investigators. Phase III Accessed February 11, 2016, and March 15, 2016.
prospective randomized double-blind placebo-controlled trial of pler- 16. Gladkov O, Moiseyenko V, Bondarenko IN, et al. A Phase III study of
ixafor plus granulocyte colony-stimulating factor compared with pla- balugrastim versus pegfilgrastim in breast cancer patients receiving
cebo plus granulocyte colony-stimulating factor for autologous stem- chemotherapy with doxorubicin and docetaxel. Oncologist. 2016;21:
cell mobilization and transplantation for patients with non-Hodgkins 7-15.
lymphoma. J Clin Oncol. 2009;27:4767-4773. 17. Ratti M, Tomasello G. Lipegfilgrastim for the prophylaxis and treat-
6. DiPersio JF, Stadtmauer EA, Nademanee A, et al; 3102 Investigators. ment of chemotherapy-induced neutropenia. Expert Rev Clin Pharmacol.
Plerixafor and G-CSF versus placebo and G-CSF to mobilize hemato- 2015;8:15-24.
poietic stem cells for autologous stem cell transplantation in patients 18. Bondarenko I, Gladkov OA, Elsaesser R, et al. Efficacy and safety of
with multiple myeloma. Blood. 2009;113:5720-5726. lipegfilgrastim versus pegfilgrastim: a randomized, multicenter, active-
7. Watanabe T, Tobinai K, Shibata T, et al. Phase II/III study of R-CHOP-21 control phase 3 trial in patients with breast cancer receiving doxorubicin/
versus R-CHOP-14 for untreated indolent B-cell non-Hodgkins lym- docetaxel chemotherapy. BMC Cancer. 2013;13:386.
phoma: JCOG 0203 trial. J Clin Oncol. 2011;29:3990-3998. 19. Royzman I. United States: the Value of Being Highly Similar: First U.S. Bio-
8. Cunningham D, Hawkes EA, Jack A, et al. Rituximab plus cyclophos- similar. http://www.mondaq.com/unitedstates/x/391742/food+drugs+law/
phamide, doxorubicin, vincristine, and prednisolone in patients with The+Value+of+Being+Highly+Similar+First+US+Biosimilar. Accessed February
newly diagnosed diffuse large B-cell non-Hodgkin lymphoma: a phase 3 12, 2016, and March 15, 2016.
comparison of dose intensification with 14-day versus 21-day cycles. 20. Fishman ML, Kumar A, Davis S, et al. Guideline-based peer-to-peer
Lancet. 2013;381:1817-1826. consultation optimizes pegfilgrastim use with no adverse clinical con-
9. Delarue R, Tilly H, Mounier N, et al. Dose-dense rituximab-CHOP sequences. J Oncol Pract. 2012;8:e14s-e17s (suppl).
compared with standard rituximab-CHOP in elderly patients with dif- 21. Schnipper LE, Smith TJ, Raghavan D, et al. American Society of Clinical
fuse large B-cell lymphoma (the LNH03-6B study): a randomised phase 3 Oncology identifies five key opportunities to improve care and reduce
trial. Lancet Oncol. 2013;14:525-533. costs: the top five list for oncology. J Clin Oncol. 2012;30:1715-1724.
10. Bennett CL, Djulbegovic B, Norris LB, et al. Colony-stimulating factors for 22. Barnes G, Pathak A, Schwartzberg L. G-CSF utilization rate and pre-
febrile neutropenia during cancer therapy. N Engl J Med. 2013;368: scribing patterns in United States: associations between physician and
1131-1139. patient factors and GCSF use. Cancer Med. 2014;3:1477-1484.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e527


GARY H. LYMAN

Issues on the Use of White Blood Cell Growth Factors in


Oncology Practice
Gary H. Lyman, MD, MPH

OVERVIEW

Appropriate use of myeloid growth factors may reduce the risk of neutropenic complications including febrile neutropenia (FN)
in patients receiving cancer chemotherapy. The recently updated American Society of Clinical Oncology (ASCO) Guidelines on
the Use of the White Blood Cell Growth Factors recommends routine prophylaxis with these agents starting in the first cycle
when the risk of FN is 20% or greater. However, the risks for neutropenic complications and the risk of serious adverse
consequences from FN vary considerably with different chemotherapy regimens as well as other disease-, treatment-, and
patient-specific risk factors. Considerably more information is now available on the major risk factors for FN. Multivariable risk
models combining factors look promising but require further validation. Most clinical studies of myeloid growth factor
prophylaxis assessed relative risk (RR) of FN but were not powered to evaluate the effect of prophylaxis on disease-free or
overall survival. Accumulating evidence suggests, however, that the appropriate use of these agents in selected patients may
improve both short-term and long-term survival by reducing the immediate risk of mortality accompanying patients with high-
risk disease developing FN as well as improving disease-free and overall survival by enabling the delivery of full dose intensity
chemotherapy and reducing the risk of disease recurrence in patients treated with curative intent. Further studies to evaluate
risk factors and models for FN are needed to guide clinical and shared decision making for the optimal personalized use of these
agents and offer patients at increased risk the best chance of long-term disease control.

rates in randomized controlled clinical trials (RCTs).11 A


M yelosuppression including neutropenia represents a
major toxicity associated with systemic cancer che-
motherapy and is associated with considerable morbidity,
threshold risk of FN of 20% for routine use of prophylactic
CSFs has been recommended by guidelines from ASCO,
mortality, and costs.1 Such complications may result in dose the National Comprehensive Cancer Network (NCCN), and
reductions or treatment delays, potentially compromising the European Organisation for the Research and Treat-
disease control and survival outcomes.2-6 Prophylactic my- ment of Cancer (EORTC) based on results from RCTs of
eloid growth factors may be used to reduce the risk of prophylactic use.7,12-14 However, the risk of chemotherapy-
neutropenic complications and enable safe delivery of induced neutropenia and its complications are likely under-
planned chemotherapy dose intensity on schedule when reported in RCTs, with reported rates varying considerably
indicated. Guidelines on the use of myeloid growth factors for common chemotherapy regimens.15,16 Rates of FN ob-
were recently updated by ASCO.7 The first question posed to served in observational or real-world patient populations
the panel related to what factors clinicians should consider appear to be consistently greater than those reported
when selecting patients for primary prophylaxis of FN with a among selected patients accrued to RCTs.17 Additional risk
colony-stimulating factor (CSF). Several studies demon- factors for chemotherapy-induced neutropenia and its
strated that the risk of an initial episode of FN is greatest complications have been identified based on patient char-
during the first cycle of treatment when patients are gen-
acteristics, cancer type, and variation in chemotherapy
erally receiving full dose intensity.8-10
treatment intensity and intent (cure vs. palliation).18-21 The
updated ASCO CSF guidelines distinguish between risk
RISK FACTORS AND MODELS FOR NEUTROPENIA factors other than the chemotherapy regimen that increase
COMPLICATONS the likelihood of FN (Sidebar 1) and those that also increase
The risk of FN for patients with cancer who are receiving the risk of serious medical consequences or death for pa-
systemic chemotherapy is generally based on reported tients who develop FN (Sidebar 2). Although there is some

From the Hutchinson Institute for Cancer Outcomes Research, Fred Hutchinson Cancer Research Center, and the University of Washington, Seattle, WA.

Disclosures of potential conflicts of interest provided by the author are available with the online article at asco.org/edbook.

Corresponding author: Gary H. Lyman, MD, MPH, Hutchinson Institute for Cancer Outcomes Research, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. North, M3-B232,
P.O. Box 19024, Seattle, WA 98109-1024; email: glyman@fhcrc.org.

2016 by American Society of Clinical Oncology.

e528 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


RISK FACTORS AND RISK MODELS FOR FEBRILE NEUTROPENIA

SIDEBAR 1. Patient Risk Factors for Febrile SIDEBAR 2. Patient Risk Factors for Poor Clinical
Neutropenia7 Outcomes From Febrile Neutropenia or
In addition to chemotherapy regimen and type of malig- Infection7
nancy, consider these factors when estimating a patients Sepsis syndrome
overall risk of febrile neutropenia19,26,34: Age 65 or older
Age 65 or older Severe neutropenia
Advanced disease Neutropenia expected to be longer than 10 days in
Previous chemotherapy or radiation therapy duration
Pre-existing neutropenia or bone marrow involvement Pneumonia
with tumor Invasive fungal infection
Infection Other clinically documented infections
Open wounds or recent surgery Hospitalization at the time of fever
Poor performance status or poor nutritional status Prior episode of febrile neutropenia
Poor renal function

Liver dysfunction, most notably elevated bilirubin

Cardiovascular disease

HIV infection for intercorrelation between risk factors, there has been
Multiple comorbid conditions an increasing interest in the development and validation
of multivariable risk models for chemotherapy-associated
neutropenia and its complications. There have been a
number of modeling efforts to predict the risk of serious
overlap between the individual factors, the latter are im- complications including death for patients with established
portant to note even among patients who are considered to FN.23-25 Although model performance has generally been
be at low or intermediate risk for developing FN. The NCCN limited and has challenged broad utilization of these models,
and EORTC guidelines also identify risk factors for developing some institutions and clinicians have used these to identify
FN for clinicians to consider in addition to the chemotherapy patients at low risk for FN for possible outpatient man-
regimen when estimating a patients individual risk of FN, agement. Attention has recently shifted to creating risk
supporting the concept of personalized supportive care in models for the development of FN in ambulatory patients
oncology.12,14,22 receiving cancer chemotherapy.26,27 Clinical practice guide-
Because of the number of identified risk factors, un- lines discuss the importance of patient-specific risk factors
certainty about their relative importance, and the potential and the need for prospective data on the risk of FN in the
unselected general cancer population.7,12
Lyman et al26 reported on a prospective cohort study of
patients with cancer who were treated at oncology practices
KEY POINTS throughout the United States. The study was undertaken to
explicitly evaluate the incidence of and risk factors for
Routine prophylaxis with the white blood cell growth neutropenic events among patients with cancer who were
factors is recommended starting in the first cycle of receiving systemic chemotherapy. Consecutive eligible pa-
cancer chemotherapy when the risk of febrile tients with solid tumors or malignant lymphoma initiating a
neutropenia (FN) is 20% or greater.
new chemotherapy regimen were enrolled. Patient de-
The risk of neutropenic complications and associated
infectious complications vary with different
mographics and clinical variables included age, sex, eth-
chemotherapy regimens and other disease-, treatment-, nicity, employment and educational status, performance
and patient-specific risk factors. status, body surface area, cancer type, disease stage, history
Risk models combining multiple individual risk factors of prior cancer and treatment, concomitant medications,
look promising but require further validation and baseline hematology and chemistry results, and planned
improvement. chemotherapy treatment including drugs, dose, and sched-
Appropriate use of the white blood cell growth factors ule. Neutropenic events reported included neutropenia
may improve survival by both reducing the risk of death (absolute neutrophil count [ANC] nadir , 1.0 3 103/mm3),
associated with FN and improving overall survival by severe neutropenia (ANC nadir , 0.5 3 103/mm3), and FN
enhancing delivery of full-dose chemotherapy on (fever/infection and ANC nadir , 1.0 3 103/mm3). The
schedule reducing the risk of disease recurrence and
primary outcome of the study was severe or FN in cycle 1
disease-specific mortality.
Further investigation is needed on risk factors and
owing to the dominant risk of events in the first cycle
models for FN to guide clinicians and patients on the and their major effect on subsequent risk and treatment
appropriate use of these agents and to provide optimal decisions.2,3,10 In the multivariable model based on nearly
treatment to affect long-term disease control and 2,500 patients, important prognostic factors for neutropenic
survival. complications included a history of previous chemotherapy
as well as baseline leukopenia, hepatic or renal dysfunction,

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e529


GARY H. LYMAN

FIGURE 1. Febrile Neutropenia in Patients With High- potentially life-threatening complications of cancer treat-
Risk and Low-Risk Disease Receiving Chemotherapy ment while allowing for the safe and adequate delivery of
effective chemotherapy dose intensity. Alternatively, the
identification of patients at low risk for early neutropenic
complications may provide reassurance and cost savings in
settings in which more aggressive supportive care is not
warranted. Importantly, among patients receiving chemo-
therapy who were considered to have an intermediate risk
for FN based on clinical guidelines, approximately one-
half were classified as high risk and one-half were classified
as low risk based on the risk model incorporating other
patient-, disease-, and treatment-related factors. In a sub-
sequent prospective cohort study of nearly 1,000 patients
being treated by 124 community oncologists, a priori
physician-assessed risk of FN as well as the decision to use
prophylactic CSF correlated poorly with the FN risk esti-
mated by the risk model.28
Personalization of supportive care strategies such as the
appropriate and targeted use of myeloid growth factors in a
fashion similar to the personalization of targeted cancer
therapies offers considerable potential for more effective,
safe, and cost-effective cancer care along with improved
survival and quality of life for patients with cancer.22

MYELOID GROWTH FACTORS AND SURVIVAL


Myeloid growth factors have been developed and approved
for reducing the risk of neutropenic complications including
severe neutropenia and FN. Nevertheless, FN can be a life-
threatening complication of cancer chemotherapy by di-
rectly leading to infectious complications and death or
indirectly by prompting a reduction in chemotherapy in-
tensity or duration, resulting in greater risk of disease re-
currence and cancer-related mortality (Fig. 2). In a recent
(A) KaplanMeier plot displaying the cumulative proportion of patients experiencing
one or more episodes of febrile neutropenia over time in days after chemotherapy
analysis of patients with cancer receiving systemic chemo-
initiation for patients at high risk and patients at low risk based on the risk model. (B) therapy who were included in a large U.S. health claims
Hazard function plot displaying the distribution of hazard rates for febrile neutropenia
over time in days after chemotherapy initiation for patients at high risk and patients at
database, Lyman et al29 estimated a 35% increase in early
low risk based on the risk model.
Abbreviation: HR, hazard ratio.
Reprinted from Lyman et al.26
FIGURE 2. Dual Impact of Neutropenia on Risk of
Infection and Chemotherapy Dose Intensity
delivered chemotherapy relative dose intensity, and the use
of a prophylactic myeloid growth factor. Based on the
median predicted risk of neutropenic events, 34% of patients
classified as high risk experienced events in cycle 1 compared
with 4% among patients at low risk. The cumulative risk of FN
with repeated cycles of chemotherapy for patients at high
and low risk is shown in Fig. 1. KaplanMeier estimates of the
cumulative risk of FN over the first three to four cycles of
chemotherapy were approximately 20% for patients at high
risk compared with 5% for patients at low risk. Additional
external validation of the risk model is currently being
pursued in different settings and patient populations. It is
anticipated that targeted application of a prophylactic Schematic diagram of the dual effects of neutropenic complications including
myeloid growth factor starting in cycle 1 among patients febrile neutropenia on both early and overall survival based on reductions in the
risk of febrile neutropenia and early infectious complications while enabling the
identified as being high risk for early events based on this delivery of full-dose chemotherapy on schedule and potentially reducing the risk
model will likely reduce the risk of these serious and of disease recurrence and cancer-associated mortality.

e530 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


RISK FACTORS AND RISK MODELS FOR FEBRILE NEUTROPENIA

mortality and a 15% increased risk of all-cause mortality the same time, the RR for all-cause mortality with and without
overall among patients experiencing FN. Although a favorable G-CSF support was 0.897 (95% CI, 0.8570.938; p , .001) with
effect of myeloid growth factors on survival of patients with an ARD of 3.40% (95% CI, 2.00%4.80%; p , .001),
cancer is therefore plausible, data directly addressing this representing a nearly 10-fold greater reduction in mortality
issue are limited. Most pivotal clinical trials of myeloid growth than the estimated increase in AML/MDS. Finally, among the
factors have been designed to evaluate their effect on severe 16 RCTs reporting to have delivered chemotherapy intensity
neutropenia or FN during the period of chemotherapy de- by treatment arm, significant associations were observed
livery, with limited follow-up beyond the treatment period. between reduced mortality and increases in both relative
Therefore, none of these trials were individually powered to (p = .0148) and absolute (p = .0266) delivered dose intensity
address the issue of disease-free or overall survival, and only in patients treated with G-CSF compared with controls. These
early mortality was reported. In a systematic review and results were subsequently extended in a systematic review of
meta-analysis of the 17 RCTs of primary prophylaxis with RCTs of patients receiving cancer chemotherapy and ran-
granulocyte-CSF (G-CSF) including about 3,500 patients with domly assigned to G-CSF or control and reporting all-cause
solid tumors or lymphoma, a significant reduction in the risk of mortality with a minimum follow-up of 24 months but re-
FN as seen in each individual trial was confirmed (RR, 0.54; moving the requirement for reporting of AML or MDS.33
95% CI, 0.43-0.67; p , .001).30 Importantly, in this pooled Among about 61 eligible RCTs involving nearly 10,000 pa-
analysis, reductions in the risk of infection-related mortality tients, the RR for all-cause mortality observed was 0.93 (95%
(RR 0.55; 95% CI, 0.330.90; p = .018) as well as early all-cause CI, 0.900.96; p , .001). Greater reductions in mortality were
mortality (RR, 0.60; 95% CI, 0.430.83; p = .002) were ob- observed among trials with longer follow-up, when treatment
served. Although a fourfold increase in reported bone pain was clearly for curative intent, and when survival was the
was noted, average delivered chemotherapy relative dose primary study outcome. In subgroup analyses by study design,
intensity was significantly greater for patients who were greater reductions in all-cause mortality were observed when
randomly assigned to receive prophylactic G-CSF. G-CSF support enabled chemotherapy treatment with a dose-
Concern has been raised about the potential for an in- dense schedule, greater treatment intensity, or the addition
creased risk for developing acute myeloid leukemia (AML) or of one or more additional myelosuppressive agents than
myelodysplastic syndrome (MDS) among patients treated when both study arms were intended to receive the same
with myeloid growth factors. In an analysis of data from the chemotherapy drugs, dose, and schedule.
Surveillance, Epidemiology, and End ResultsMedicare linked Although the potential effect of myeloid growth factors on
files, Hershman et al31 observed a doubling of the risk of overall survival has not been completely evaluated, the
subsequent AML or MDS, from 0.7% to 1.8% with CSF support available evidence suggests no deleterious effects of CSF
among older women diagnosed with early-stage breast support of cancer chemotherapy and raises the potential
cancer from 1991 to 1999 who received myeloid growth for a favorable effect on both early- and overall all-cause
factors with their adjuvant chemotherapy. A more recent mortality, most notably for patients treated with curative
systematic review of RCTs among patients receiving systemic intent and when CSF support enables treatment enhance-
chemotherapy and randomly assigned to receive G-CSF ment for patients with responsive and potentially curable
support or not and reporting on the occurrence of AML/ malignancies. Although an increased risk of AML or MDS
MDS or all second malignancies and a minimum follow-up of remains, available studies cannot clearly differentiate be-
24 months or more identified 25 eligible trials involving more tween the direct effects of the myeloid growth factor and
than 12,000 patients.32 With an average follow-up of the enabling effects of delivering more intensive treatment
60 months in these RCTs, AML, or MDS was observed in 0.36% with known leukemogenic chemotherapeutic agents. In
of controls and 0.79% of patients treated with G-CSF, with a total, the apparent reduction in all-cause mortality appears
RR of 1.92 (95% CI, 1.193.07; p = .007) and an absolute risk to overshadow the potential small increased risk associated
difference (ARD) of 0.41% (95% CI, 0.10%0.72%; p = .009). At with AML or MDS in this setting.

References
1. Kuderer NM, Dale DC, CrawfordJ, et al. Mortality, morbidity, and cost associated 5. Shayne M, Culakova E, Wolff D, et al. Dose intensity and hematologic
with febrile neutropenia in adult cancer patients. Cancer. 2006;106:2258-2266. toxicity in older breast cancer patients receiving systemic chemo-
2. Lyman GH, Dale DC, Crawford J. Incidence and predictors of low dose- therapy. Cancer. 2009;115:5319-5328.
intensity in adjuvant breast cancer chemotherapy: a nationwide study 6. Shayne M, Culakova E, Poniewierski MS, et al. Dose intensity and
of community practices. J Clin Oncol. 2003;21:4524-4531. hematologic toxicity in older cancer patients receiving systemic che-
3. Lyman GH, Dale DC, Friedberg J, et al. Incidence and predictors of low motherapy. Cancer. 2007;110:1611-1620.
chemotherapy dose-intensity in aggressive non-Hodgkins lymphoma: 7. Smith TJ, Bohlke K, Lyman GH, et al; American Society of Clinical On-
a nationwide study. J Clin Oncol. 2004;22:4302-4311. cology. Recommendations for the use of WBC growth factors: American
4. Lyman GH. Impact of chemotherapy dose intensity on cancer patient Society of Clinical Oncology clinical practice guideline update. J Clin
outcomes. J Natl Compr Canc Netw. 2009;7:99-108. Oncol. 2015;33:3199-3212.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e531


GARY H. LYMAN

8. Lyman GH, Morrison VA, Dale DC, et al; OPPS Working Group; ANC 22. Kuderer NM, Lyman GH. Personalized medicine and cancer supportive
Study Group. Risk of febrile neutropenia among patients with care: appropriate use of colony-stimulating factor support of chemo-
intermediate-grade non-Hodgkins lymphoma receiving CHOP che- therapy. J Natl Cancer Inst. 2011;103:910-913.
motherapy. Leuk Lymphoma. 2003;44:2069-2076. 23. Klastersky J, Paesmans M. The Multinational Association for Supportive
9. Lyman GH, Delgado DJ. Risk and timing of hospitalization for febrile Care in Cancer (MASCC) risk index score: 10 years of use for identifying
neutropenia in patients receiving CHOP, CHOP-R, or CNOP chemo- low-risk febrile neutropenic cancer patients. Support Care Cancer. 2013;
therapy for intermediate-grade non-Hodgkin lymphoma. Cancer. 2003; 21:1487-1495.
98:2402-2409. 24. Talcott JA, Siegel RD, Finberg R, et al. Risk assessment in cancer patients
10. Crawford J, Dale DC, Kuderer NM, et al. Risk and timing of neutropenic with fever and neutropenia: a prospective, two-center validation of a
events in adult cancer patients receiving chemotherapy: the results of a prediction rule. J Clin Oncol. 1992;10:316-322.
prospective nationwide study of oncology practice. J Natl Compr Canc 25. Carmona-Bayonas A, Jimenez-Fonseca P, Virizuela Echaburu J, et al.
Netw. 2008;6:109-118. Prediction of serious complications in patients with seemingly stable
11. Lyman GH. Risk assessment in oncology clinical practice. From risk factors febrile neutropenia: validation of the Clinical Index of Stable Febrile
to risk models. Oncology (Williston Park). 2003; 17(Suppl 11):8-13. Neutropenia in a prospective cohort of patients from the FINITE study.
12. National Comprehensive Cancer Network (ed). NCCN Clinical Practice J Clin Oncol. 2015;33:465-471.
Guidelines in Oncology: Myeloid Growth Factors. Fort Washington, PA: 26. Lyman GH, Kuderer NM, Crawford J, et al. Predicting individual risk of
National Comprehensive Cancer Network Inc; 2015. neutropenic complications in patients receiving cancer chemotherapy.
13. Schnipper LE, Smith TJ, Raghavan D, et al. American Society of Clinical Cancer. 2011;117:1917-1927.
Oncology identifies five key opportunities to improve care and reduce 27. Jenkins P, Scaife J, Freeman S. Validation of a predictive model that
costs: the top five list for oncology. J Clin Oncol. 2012;30:1715-1724. identifies patients at high risk of developing febrile neutropaenia fol-
14. Aapro MS, Bohlius J, Cameron DA, et al; European Organisation for lowing chemotherapy for breast cancer. Ann Oncol. 2012;23:
Research and Treatment of Cancer. 2010 update of EORTC guidelines 1766-1771.
for the use of granulocyte-colony stimulating factor to reduce the 28. Lyman GH, Dale DC, Legg JC, et al. Assessing patients risk of febrile
incidence of chemotherapy-induced febrile neutropenia in adult pa- neutropenia: is there a correlation between physician-assessed risk and
tients with lymphoproliferative disorders and solid tumours. Eur J model-predicted risk? Cancer Med. 2015;4:1153-1160.
Cancer. 2011;47:8-32. 29. Lyman GH, Michels SL, Reynolds MW, et al. Risk of mortality in patients
15. Dale DC, McCarter GC, Crawford J, et al. Myelotoxicity and dose in- with cancer who experience febrile neutropenia. Cancer. 2010;116:
tensity of chemotherapy: reporting practices from randomized clinical 5555-5563.
trials. J Natl Compr Canc Netw. 2003;1:440-454. 30. Kuderer NM, Dale DC, Crawford J, et al. Impact of primary prophylaxis
16. Kuderer NM, Wolff AC. Enhancing therapeutic decision making when with granulocyte colony-stimulating factor on febrile neutropenia and
options abound: toxicities matter. J Clin Oncol. 2014;32:1990-1993. mortality in adult cancer patients receiving chemotherapy: a systematic
17. Truong J, Lee EK, Trudeau ME, et al. Interpreting febrile neutropenia review. J Clin Oncol. 2007;25:3158-3167.
rates from randomized, controlled trials for consideration of primary 31. Hershman D, Neugut AI, Jacobson JS, et al. Acute myeloid leukemia or
prophylaxis in the real world: a systematic review and meta-analysis. myelodysplastic syndrome following use of granulocyte colony-
Ann Oncol. Epub 2015 Dec 27. stimulating factors during breast cancer adjuvant chemotherapy.
18. Laskey RA, Poniewierski MS, Lopez MA, et al. Predictors of severe and J Natl Cancer Inst. 2007;99:196-205.
febrile neutropenia during primary chemotherapy for ovarian cancer. 32. Lyman GH, Dale DC, Wolff DA, et al. Acute myeloid leukemia or
Gynecol Oncol. 2012;125:625-630. myelodysplastic syndrome in randomized controlled clinical trials of
19. Lyman GH, Abella E, Pettengell R. Risk factors for febrile neutropenia cancer chemotherapy with granulocyte colony-stimulating factor:
among patients with cancer receiving chemotherapy: a systematic a systematic review. J Clin Oncol. 2010;28:2914-2924.
review. Crit Rev Oncol Hematol. 2014;90:190-199. 33. Lyman GH, Dale DC, Culakova E, et al. The impact of the granulocyte
20. Weycker D, Li X, Barron R, et al. Importance of risk factors for febrile colony-stimulating factor on chemotherapy dose intensity and cancer
neutropenia among patients receiving chemotherapy regimens not survival: a systematic review and meta-analysis of randomized con-
classified as high-risk in guidelines for myeloid growth factor use. J Natl trolled trials. Ann Oncol. 2013;24:2475-2484.
Compr Canc Netw. 2015;13:979-986. 34. Hosmer W, Malin J, Wong M. Development and validation of a pre-
21. Weycker D, Li X, Edelsberg J, et al. Risk of febrile neutropenia in patients diction model for the risk of developing febrile neutropenia in the first
receiving emerging chemotherapy regimens. Support Care Cancer. cycle of chemotherapy among elderly patients with breast, lung, co-
2014;22:3275-3285. lorectal, and prostate cancer. Support Care Cancer. 2011;19:333-341.

e532 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


PATIENT AND SURVIVOR CARE

Optimizing Palliative and


End-of-Life Care: Evidence-Based
Practice Improvement

CHAIR
Thomas W. LeBlanc, MD, MA
Duke University Medical Center
Durham, NC

SPEAKERS
Jennifer S. Temel, MD
Massachusetts General Hospital Cancer Center
Boston, MA

Timothy D. Gilligan, MD, MSc


Cleveland Clinic
Cleveland, OH
NICKOLICH ET AL

Discussing the Evidence for Upstream Palliative Care in


Improving Outcomes in Advanced Cancer
Myles S. Nickolich, MD, Areej El-Jawahri, MD, Jennifer S. Temel, MD, and Thomas W. LeBlanc, MD, MA

OVERVIEW

Palliative care has received increasing attention at the American Society of Clinical Oncology (ASCO) Annual Meeting since
the publication of its provisional clinical opinion on the topic in 2012. Despite frequent discussion, palliative care remains a
source of some controversy and confusion in clinical practice, especially concerning who should provide it, what it en-
compasses, and when and how it can help patients and their families. In this article, we provide a formal definition of
palliative care and review the state of the science of palliative care in oncology. Several randomized controlled trials now
show that palliative care improves important outcomes for patients with cancer. Related outcome improvements include a
reduction in symptoms, improved quality of life, better prognostic understanding, less depressed mood, less aggressive end-
of-life care, reduced resource utilization, and even prolonged survival. As such, ASCO recommends early integration of
palliative care into comprehensive cancer care for all patients with advanced disease and/or significant symptom burden.
Our aim is that this summary will facilitate greater understanding about palliative care and encourage further integration of
palliative care services into cancer care. More research is needed to illuminate the mechanisms of action of palliative care
and to improve the specificity of palliative care applications to unique scenarios and populations in oncology.

P alliative care has received increasing attention at the


ASCO Annual Meeting since the publication of its
Provisional Clinical Opinion: The Integration of Palliative
Palliative care is specialized medical care for people with
serious illness. It focuses on providing patients with relief
from the symptoms, pain, and stress of a serious illness
Care Into Standard Oncology Care in 2012.1 In that paper, whatever the diagnosis. The goal is to improve quality of life
for both the patient and the family. It is appropriate at any
ASCO recommended early concurrent palliative care as part
age and at any stage of a serious illness. It can be provided at
of standard cancer care for all patients with metastatic the same time as disease treatment to help people live as
disease and/or significant symptom burden. This recom- well as possible while facing illness.2
mendation was based on a growing amount of evidence
about the many benefits of adding specialist palliative care Although other organizations, such as the National Com-
to standard oncology care, including data from several prehensive Cancer Network and the World Health Organi-
large randomized controlled trials. zation, have also posed definitions of palliative care, we prefer
Despite frequent discussion at ASCO Annual Meetings the clarity and inclusivity of the CAPC definition. Palliative care
since then, palliative care remains a source of some con- seeks to improve quality of life by providing additional sup-
troversy and confusion in clinical practice, especially con- port to the patient and caregiver(s), to assist with symptom
cerning who should provide it, what it encompasses, and burden, psychosocial needs, spiritual well being, communi-
when and how it can help patients and their families. cation, and understanding of prognosis, treatment decision
making, transitions of care, existential issues, and end-of-life
scenarios. Although palliative care is appropriate for many
WHAT IS PALLIATIVE CARE? patients with serious illness, this article focuses on patients
The term palliative care is sometimes misunderstood as with cancer.
being synonymous with end-of-life care. To address this There are different levels of palliative care, each of which is
misperception, the Center to Advance Palliative Care (CAPC) provided by different members of the care team. Oncolo-
conducted a public opinion polling study in 2011, to de- gists often provide basic palliative care, including pain and
velop a new definition of palliative care. According to this other symptom management. This type of palliative care is
definition: referred to as primary palliative care, generalist palliative

From Duke University Hospital, Durham, NC; Massachusetts General Hospital, Boston, MA; Duke Cancer Institute, Durham, NC.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Thomas W. LeBlanc, MD, MA, Duke University Medical Center, P.O. Box 2715, Durham, NC 27710; email: thomas.leblanc@duke.edu.

2016 by American Society of Clinical Oncology.

e534 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


WHATS THE EVIDENCE BASE FOR PALLIATIVE CARE IN ONCOLOGY?

care,3 or sometimes just supportive care.4 Although it is entirely with those randomly assigned to receive standard care
appropriate that the oncology care team provides primary (21.34 vs. +3.23; p = .05).6
palliative care, this is not always sufficient to meet the needs of
patients and families. In cases of complex and/or unmet needs,
Quality of Life
patients may benefit from a higher level of palliative care, often
Early palliative care also improves quality of life for patients
called specialty palliative care or secondary palliative care. This
with advanced cancer. In the ENABLE II study by Bakitas
type of palliative care is provided by specialists with formal
et al,7 patients with advanced gastrointestinal, breast, lung,
training in palliative care and related specialties. Specialty
or genitourinary cancer were randomly assigned to receive a
palliative care is akin to the expert care a cardiologist provides
nurse-led palliative care intervention or standard care within
to a patient with severe heart failure, even though the pa-
8 to 12 weeks of diagnosis. Patients who were randomly
tients primary care physician may also be monitoring the
assigned to receive early palliative care had better quality of
cardiac status of the patient.
life per the Functional Assessment of Cancer Therapy
This distinction is critical because some oncologists mis-
Palliative Care scale (mean difference of 4.6; p = .02).7
understand palliative care to suggest that they should pass off
Similarly, in a large randomized controlled trial by Temel
their primary palliative care responsibilities to someone else.
et al,8 patients with advanced nonsmall cell lung cancer
Rather, when we talk about specialty palliative care, we are
(NSCLC) who were randomly assigned to receive early
referring to care that is provided in addition to the primary
outpatient palliative care within 8 weeks of diagnosis had a
palliative care already being provided by the oncologist. This
significant improvement in quality of life, per the trial
type of care is additive, not duplicative, and should not be
outcome index subscale of the Functional Assessment of
misconstrued as any sort of transferal of responsibility. In-
Cancer TherapyLung scale at 12 weeks, compared with
deed, data from randomized controlled trials suggest that that
those assigned to usual care (59.0 vs. 53.0; p = .009).8 This
the focus of palliative care specialists is quite different from
finding was also confirmed in the trial by Zimmermann
that of oncologists.5 Together, oncologists and palliative care
et al6 where patients assigned to receive the palliative care
specialists working collaboratively can provide more com-
intervention per the Quality of Life at the End of Life scale
prehensive care for patients with cancer and their families.
experienced improvements seen at 3 months, compared
with those receiving standard care (2.33 vs. 0.06; p = .05).6
WHICH OUTCOMES IMPROVE WITH UPSTREAM Early palliative care also improves quality of life for pa-
PALLIATIVE CARE? tients in the emergency department setting.9 In a study by
Symptom Management Grudzen et al,9 patients with advanced cancer who pre-
In a large cluster-randomized controlled trial by Zimmermann sented to the emergency department at a quaternary care
et al6 with patients with advanced solid tumors (lung, gas- center were randomly assigned to receive a palliative care
trointestinal, genitourinary, breast, and gynecologic), patients consult from the inpatient team and subsequent follow-
who were randomly selected to receive early palliative care up visits in the outpatient palliative care clinic compared
had a significant reduction in symptom intensity at 4 months with usual care. Patients who were randomly assigned
by the Edmonton Symptom Assessment Scale, compared to palliative care experienced greater improvements in
quality of life scores by the Functional Assessment of
Cancer TherapyGeneral (FACT-G) scale at 12 weeks
(5.91 vs. 1.08; p = .03).9
KEY POINTS
Palliative care improves important outcomes for
Prognostic Understanding
patients with cancer, as shown in at least five Unfortunately, many patients with advanced, incurable
randomized controlled trials to date. cancer misunderstand their prognoses and the goals of their
ASCO recommends early integration of palliative care cancer treatments. In a study by Weeks et al,10 patients with
into comprehensive cancer care for all patients with stage IV lung or colorectal cancer were asked to identify the
advanced disease and/or significant symptom burden. goal of their prescribed chemotherapies. Sixty-nine percent
Outcome improvements seen with early palliative care of patients with lung cancer and 81% of those with colorectal
include a reduction in symptoms, improved quality of cancer mistakenly thought that their palliative chemotherapy
life, better prognostic understanding, less depressed regimens were prescribed with intent to cure.10 Interestingly,
mood, improved end-of-life outcomes, reduced early palliative care mitigates this problem. In the randomized
resource utilization, and even prolonged survival.
controlled trial by Temel et al,11 patients with advanced
More research is needed to illuminate the mechanisms
of action of palliative care and to improve the specificity
NSCLC who were randomly assigned to receive early palliative
of palliative care applications to unique scenarios and care were more likely to retain or develop an accurate un-
populations, such as patients with hematologic derstanding of their prognoses compared with patients who
malignancies, caregivers, and across various tumor received standard care (82.5% vs. 59.6%; p = .02).11 Of note,
types and treatments. patients in the early palliative care arm who reported an
accurate understanding of prognosis were also less likely to

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e535


NICKOLICH ET AL

receive chemotherapy near the end of life (9.4% vs. 50%; before death, no hospice care, or admission to hospice 3 or
p = .02), an indicator of higher-quality end-of-life care fewer days before death. In addition, more patients in the
among patients with advanced cancer. palliative care arm had documented resuscitation prefer-
ences in the outpatient setting (53% vs. 28%; p = .05).8
Caregiver Outcomes The use of intensive care unit (ICU) services for patients
In addition to direct patient benefits of early palliative care, with advanced cancer is another useful marker of aggressive
caregivers also appear to benefit from the early introduction end-of-life care, and palliative care appears to improve this
of palliative care. In the ENABLE III study by Bakitas et al,7 the outcome as well. For example, in a retrospective study in the
caregivers of patients assigned to early palliative care received Veterans Affairs system, patients who were observed by an
an additional caregiver-focused intervention. These care- inpatient palliative care team had a 42% lower ICU admission
givers reported better depression scores at 3 months, per rate than control patients.14 However, in the study by
the Center for Epidemiologic StudyDepression scale (mean Grudzen et al,9 emergency departmentinitiated pallia-
difference, 23.4; p = .02). Furthermore, caregivers whose tive care consultation (with outpatient follow-up there-
loved ones died during the study also had reduced stress after) did not yield any reduction in ICU admissions.9
burden, as measured on the Montgomery-Borgatta Caregiver In the ENABLE II study, there were no differences in
Burden stress subscale.12 The inclusion of a caregiver- number of days hospitalized, days in the ICU, or frequency of
specific intervention in this study is unique compared with emergency department visits.7 Similarly, ENABLE III found no
other randomized studies of early palliative care done to differences in relative rates of days hospitalized, days ad-
date, and results suggest that it is an important area for mitted to an ICU, emergency department visits, chemo-
future research and intervention development. therapy within the last 14 days of life, or death at home.15
However, both groups in this study received palliative care,
so it may be that late palliative care improves these out-
Mood comes similarly to earlier palliative care. Because there was
Mood disorders are major contributors to morbidity in not a control arm of patients who received no palliative care
patients with advanced cancer. Palliative care clinicians are in this study, a direct measure of the impact of palliative care
more likely than oncologists to focus on coping skills and on this outcome is not possible.
psychosocial concerns, which suggests that a role for early
palliative care is to improve mood among patients with
advanced cancer.5 Indeed, decreases in median survival Survival
have been observed in patients with advanced NSCLC and Beyond improvements in symptom burden and quality of
comorbid mood disturbance, with median survival times of life, early palliative care is also associated with improved
only 5.4 months for those with major depression compared survival. In the randomized controlled trial by Temel et al,8
with 10 months for those without (p = .001).13 In the ran- patients with metastatic NSCLC who received early palliative
domized controlled trial by Temel et al,8 patients with ad- care lived nearly 3 months longer than patients who received
vanced NSCLC who received early palliative care had lower standard care (median, 11.6 vs. 8.9 months; p = .02). This
rates of depression than patients who received usual care, difference was despite less aggressive end-of-life care.8
as measured by both the Hospital Anxiety and Depression Similarly, in the ENABLE III study, patients who received early
Scale (16% vs. 38%; p = .01) and the Patient Health palliative care had significantly longer 1-year survival rates
Questionnaire9 (4% vs. 17%; p = .04). Interestingly, 42.9% than those who received delayed palliative care; the dif-
of patients who received early palliative care also showed an ference was 15% at 1 year (63% vs. 48%; p = .038).15
improved depression response compared with 0% of pa-
tients in the usual-care control arm at 12 weeks. Of note, Resource Utilization
rates of new antidepressant prescriptions and mental health Palliative care also appears to improve resource utilization
visits did not differ significantly between the two groups, among patients with advanced cancer. However, these data
which suggests that early specialist palliative care indeed come from studies of hospitalized patients who received
affects mood.8 Similarly, in the ENABLE II study by Bakitas inpatient palliative care consultations. For example, in
et al,7 patients who received early palliative care had retrospective a study by May et al,16 inpatient palliative care
less depressed mood by Center for Epidemiologic Study consultation was associated with significant cost savings
Depression scale (mean difference, 21.8; p = .02).7 among patients with advanced cancer (p # .01), particularly
for those with more comorbidities. Overall, palliative care
End-of-Life Outcomes consultation yielded a 32% reduction in hospital costs when
In the randomized controlled trial by Temel et al,8 patients initiated within 2 days of admission for patients with cancer
who received early palliative care also received less aggres- and multiple comorbidities.16 A prospective cohort study, also
sive end-of-life care (palliative care vs. control, 33% vs. 54%; by May et al,17 demonstrated that cost savings may vary on
p = .05). Here, aggressive end-of-life care was a composite the basis of the timing of consultation. In this study, earlier
outcome defined by the presence of at least one of the palliative care consultation for hospitalized patients with
following criteria: receipt of chemotherapy within 14 days advanced cancer resulted in more cost reduction than did later

e536 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


WHATS THE EVIDENCE BASE FOR PALLIATIVE CARE IN ONCOLOGY?

consultation, with mean reductions in costs of 24% (day 2) and The hematologic malignancies population is a particularly
14% (day 6). Inpatient palliative care consultation within at least unique area for additional exploration. A growing amount of
6 days of admission had an estimated mean treatment effect literature demonstrates sizeable gaps in end-of-life quality
of 2$1,312 (p = .04) compared with usual care, and, within measures and unmet palliative care needs among patients
2 days of admission, palliative care consultation had a more with hematologic malignancies. For example, patients with
pronounced cost reduction (2$2,280; p = .002).17 Within the hematologic malignancies are more likely than patients with
Veterans Affairs setting, inpatient palliative care consultation solid tumors to be admitted to hospice services within the last
was associated with similar cost reductions in retrospective 3 days of life.20 Similarly, patients with hematologic malig-
analyses. Unadjusted total direct per day costs for patients nancies are more likely than patients with solid tumors to
who received a palliative care consultation were $891 receive inpatient or nursing facilitybased hospice care at the
versus $1,287.60 for usual care (p , .0001).14 end of life.21 Unique challenges exist to providing palliative
care to patients with hematologic malignancies because of
several complex factors, including differences in prognoses,
HOW DOES PALLIATIVE CARE IMPROVE THESE disease trajectories, treatment types and intensities, and
OUTCOMES? treating clinicians.22 Additional study is needed in the area of
There has been much discussion about the mechanism of
hematologic malignancies and bone marrow transplantation,
action of early palliative care in patients with advanced
and novel interventions to address the unique needs and
cancer. Part of the difficulty in understanding the mecha-
issues of these populations also are needed.
nism stems from the fact that the interventions studied thus
Although more research is needed to better understand
far have varied significantly. Some, such as ENABLE III, were
how palliative care improves outcomes and how to integrate
largely telephone based and provided by nurses,15 whereas
these services in different cancer populations, there is
others were based in the outpatient clinic and provided by
simultaneously a workforce crunch in the field. The palliative
specialist palliative care clinicians.6,8 Additional challenges
care workforce is unfortunately not large enough to meet
stem from the heterogeneity of populations included in
the needs of all patients with cancer. To date, there are
studies to date. Although palliative care mechanisms of
greater than 7,000 board-certified palliative care specialists
action are ultimately unknown, clues from published studies
in the United States, and there are greater than 100 fel-
point toward key ingredients in the interventions. For
lowship training programs. However, projections suggest
example, a qualitative analysis of documentation from one
that this structure is inadequate to meet the needs of the
early palliative care study demonstrated that palliative care
U.S. population.23,24 Furthermore, additional education is
clinicians spend much of their time on three primary areas:
needed in the provision of primary palliative care by on-
(1) managing symptoms, (2) facilitating coping, and (3)
cologists and others.25-27 Simply put, specialist palliative
serving as a bridge between the patient and oncologist.18
care is a scarce resource. As we develop a better un-
These elements map relatively nicely to the associated
derstanding of how palliative care improves outcomes, we
outcome improvements in symptoms and quality of life,
can better apply this scarce resource judiciously.
mood, and prognostic understanding that are seen with
Caregivers represent another key area of unmet need. To
early palliative care, and these areas are different from the
date, there is little caregiver-focused work in the palliative care
usual focus of oncologists in the clinic. However, more re-
literature, yet there are many reasons to think that caregivers
search is needed to better understand the most important
have unique needs that require novel solutions. In other
components of early palliative care interventions, which will
words, it is likely that specific interventions are needed to
thereby allow for more streamlined implementation and
support these important and often overlooked members of a
dissemination across cancer types and practices.
patients support system. The ENABLE III study is a wonderful
example of the promise for caregiver-specific interventions,12
FUTURE DIRECTIONS however, much more research is needed in this area.28
Although specialist palliative care is clearly helpful for pa- We must also develop innovative delivery models and
tients with advanced solid tumors, it is likely that there are reimbursement structures to facilitate early palliative care
differences in needs across tumor types and stages. Fur- for patients with cancer. Constraints within current models
thermore, differences in patient characteristics may herald for providing palliative care through hospice services often
different palliative care needs. For example, recent evidence creates an either/or mentality, wherein patients are re-
suggests that early palliative care in advanced lung cancer quired to forego cancer therapies to receive these services.
may be more effective among younger than older patients.19 This is particularly problematic in hematologic malignancies,
Differential efficacy may be true across other patient types for which beneficial palliative treatments, such as blood
and diseases as well. However, although it seems obvious product transfusions and antimicrobials, are often not
that patients with heart failure likely have different needs feasible within the hospice care setting.29-31 However, amid
than patients with advanced lung cancer, it is less clear whether the recent growth of targeted biologic therapies for advanced
such differences exist among patients with gastrointestinal solid tumors, this problem is increasing among patients with
cancers versus lung cancers, for example. More research is solid tumors as well. For example, we know that EGFR in-
needed in this area. hibitors may palliate bothersome symptoms and improve

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK e537


NICKOLICH ET AL

quality of life for patients with advanced lung cancer, yet CONCLUSION
these are not generally reimbursed under hospice care Palliative care is specialized care for people with serious
models. As data increasingly demonstrate the beneficial illnesses. When added to standard cancer care, specialty
impact of early concurrent palliative care, we must develop palliative care improves many outcomes for patients and
better models to facilitate its delivery as part of standard their caregivers, including symptom burden, quality of life,
cancer care, to supersede the barriers inherent in current mood, prognostic understanding, end-of-life outcomes,
reimbursement constraints. New, developing models of so- resource utilization, and even survival. However, more re-
called open access care promise to do just this and are a search is needed to illuminate mechanisms of action and to
beacon of hope that allow patients to receive both cancer improve the specificity of palliative care applications to
treatment and hospice care. unique scenarios and populations.

References

1. Smith TJ, Temin S, Alesi ER, et al. American Society of Clinical Oncology 16. May P, Garrido MM, Cassel JB, et al. Palliative care teams cost-saving
provisional clinical opinion: the integration of palliative care into effect is larger for cancer patients with higher numbers of comorbid-
standard oncology care. J Clin Oncol. 2012;30:880-887. ities. Health Aff (Millwood). 2016;35:44-53.
2. Center to Advance Palliative Care. 2011 Public Opinion Research on 17. May P, Garrido MM, Cassel JB, et al. Prospective cohort study of hospital
Palliative Care. https://www.capc.org/media/filer_public/18/ab/ palliative care teams for inpatients with advanced cancer: earlier
18ab708c-f835-4380-921d-fbf729702e36/2011-public-opinion- consultation is associated with larger cost-saving effect. J Clin Oncol.
research-on-palliative-care.pdf. Accessed March 20, 2016. 2015;33:2745-2752.
3. Quill TE, Abernethy AP. Generalist plus specialist palliative care: 18. Back AL, Park ER, Greer JA, et al. Clinician roles in early integrated
creating a more sustainable model. N Engl J Med. 2013;368:1173-1175. palliative care for patients with advanced cancer: a qualitative study.
4. Hui D, Finlay E, Buss MK, et al. Palliative oncologists: specialists in the J Palliat Med. 2014;17:1244-1248.
science and art of patient care. J Clin Oncol. 2015;33:2314-2317. 19. Nipp RD, Greer JA, El-Jawahri A, et al. Age and gender moderate the
5. Yoong J, Park ER, Greer JA, et al. Early palliative care in advanced lung impact of early palliative care in metastatic nonsmall-cell lung cancer.
cancer: a qualitative study. JAMA Intern Med. 2013;173:283-290. Oncologist. 2016;21:119-126.
6. Zimmermann C, Swami N, Krzyzanowska M, et al. Early palliative care 20. OConnor NR, Hu R, Harris PS, et al. Hospice admissions for cancer in the
for patients with advanced cancer: a cluster-randomised controlled final days of life: independent predictors and implications for quality
trial. Lancet. 2014;383:1721-1730. measures. J Clin Oncol. 2014;32:3184-3189.
7. Bakitas M, Lyons KD, Hegel MT, et al. Effects of a palliative care in- 21. LeBlanc TW, Abernethy AP, Casarett DJ. What is different about patients
tervention on clinical outcomes in patients with advanced cancer: the with hematologic malignancies? A retrospective cohort study of cancer
Project ENABLE II randomized controlled trial. JAMA. 2009;302: patients referred to a hospice research network. J Pain Symptom
741-749. Manage. 2015;49:505-512.
8. Temel JS, Greer JA, Muzikansky A, et al. Early palliative care for patients 22. LeBlanc TW, El-Jawahri A. When and why should patients with he-
with metastatic non-small-cell lung cancer. N Engl J Med. 2010;363: matologic malignancies see a palliative care specialist? Hematology (Am
733-742. Soc Hematol Educ Program). 2015;2015:471-478.
9. Grudzen CR, Richardson LD, Johnson PN, et al. Emergency 23. Lupu D, Friedman L, Alderman J, et al; American Academy of Hospice
departmentinitiated palliative care in advanced cancer: a randomized and Palliative Medicine Workforce Task Force. Estimate of current
clinical trial. JAMA Oncol. Epub 2016 Jan 14. hospice and palliative medicine physician workforce shortage. J Pain
10. Weeks JC, Catalano PJ, Cronin A, et al. Patients expectations about Symptom Manage. 2010;40:899-911.
effects of chemotherapy for advanced cancer. N Engl J Med. 2012;367: 24. Kamal AH, Bull J, Wolf S, et al. Characterizing the hospice and palliative
1616-1625. care workforce in the U.S.: clinician demographics and professional
11. Temel JS, Greer JA, Admane S, et al. Longitudinal perceptions of responsibilities. J Pain Symptom Manage. 2016;51:597-603.
prognosis and goals of therapy in patients with metastatic non-small- 25. Ramchandran K, Tribett E, Dietrich B, et al. Integrating palliative care
cell lung cancer: results of a randomized study of early palliative care. into oncology: a way forward. Cancer Contr. 2015;22:386-395.
J Clin Oncol. 2011;29:2319-2326. 26. Buss MK, Lessen DS, Sullivan AM, et al. A study of oncology fellows
12. Dionne-Odom JN, Azuero A, Lyons KD, et al. Benefits of early versus training in end-of-life care. J Support Oncol. 2007;5:237-242.
delayed palliative care to informal family caregivers of patients with 27. Buss MK, Lessen DS, Sullivan AM, et al. Hematology/oncology fellows
advanced cancer: outcomes from the ENABLE III randomized controlled training in palliative care: results of a national survey. Cancer. 2011;117:
trial. J Clin Oncol. 2015;33:1446-1452. 4304-4311.
13. Pirl WF, Greer JA, Traeger L, et al. Depression and survival in metastatic 28. Kamal AH, Dionne-Odom JN. A blue ocean strategy for palliative care:
non-small-cell lung cancer: effects of early palliative care. J Clin Oncol. focus on family caregivers. J Pain Symptom Manage. 2016;51:e1-e3.
2012;30:1310-1315. 29. Odejide OO. A policy prescription for hospice care. JAMA. 2016;315:
14. Penrod JD, Deb P, Luhrs C, et al. Cost and utilization outcomes of 257-258.
patients receiving hospital-based palliative care consultation. J Palliat 30. LeBlanc TW. Addressing end-of-life quality gaps in hematologic cancers:
Med. 2006;9:855-860. the importance of early concurrent palliative care. JAMA Intern Med.
15. Bakitas MA, Tosteson TD, Li Z, et al. Early versus delayed initiation of 2016;176:265-266.
concurrent palliative oncology care: patient outcomes in the ENABLE III 31. LeBlanc TW. Palliative care and hematologic malignancies: old dog, new
randomized controlled trial. J Clin Oncol. 2015;33:1438-1445. tricks? J Oncol Pract. 2014;10:e404-e407.

e538 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


PATIENT AND SURVIVOR CARE

Rehabilitation of Patients and


Survivors: Seizing the Opportunity

CHAIR
Catherine M. Alfano, PhD
American Cancer Society
Washington, DC

SPEAKERS
Andrea L. Cheville, MD, MSCE
Mayo Clinic
Rochester, MN

Karen Mustian, PhD, MPH


University of Rochester Medical Center
Rochester, NY
DEVELOPING HIGH-QUALITY CANCER REHABILITATION PROGRAMS

Developing High-Quality Cancer Rehabilitation Programs: A


Timely Need
Catherine M. Alfano, PhD, Andrea L. Cheville, MD, MSCE, and Karen Mustian, PhD, MPH

OVERVIEW

The number of survivors of cancer in the United States, already 14.5 million, is growing with improved cancer treatment and
aging of the population. Two-thirds of cancer survivors will be older than age 65 and are likely to enter cancer treatment
already deconditioned and with multiple comorbidities. Survivors of cancer face numerous adverse consequences of cancer
treatment that add to or exacerbate the effects of existing comorbidities and increase risk of functional decline. Many of
these problems are amenable to rehabilitation interventions, but referral to cancer rehabilitation professionals is not a
standard part of care. We present an expanded prospective model of surveillance, cancer rehabilitation assessment, and
referral efforts using a multidisciplinary team approach. In this model, cancer rehabilitation begins at the time of cancer
diagnosis and continues through and beyond cancer treatment. Physical impairments and psychosocial symptoms are
assessed and treated, and lifestyle and exercise interventions are provided to optimize functioning, health, and quality of
life. We present a stepped-care framework to guide decisions on when, how, and where to refer survivors to cancer
rehabilitation specialists depending on safety requirements and needs. This model has the potential to result in early
identification of symptoms and impairments, appropriate referral and timely treatment, and, in turn, will better address and
minimize both acute and long-term cancer morbidity.

survival15 and increased risk for exacerbation of comorbid


A n estimated 14.5 million survivors of cancer currently
live in the United States.1 This population will increase
dramatically in the coming decade because of increased
conditions or further declines in physical functioning post-
treatment.11,16,17
uptake of cancer screening, improved methods of early The United States is currently struggling to identify a
detection, better multimodal cancer treatments, and the coordinated medical and public health approach to meet
aging of the population.2,3 National reports have docu- the needs of our growing population of cancer survivors.
mented that survivors of cancer have complex needs.4-8 Predicted shortages in oncologists,18 primary care practi-
These include management of chronic and late effects of tioners,19 and other medical health professionals un-
cancer and comorbid conditions; surveillance and treatment derscore the need for effective and efficient care for cancer
of recurrence and second cancers; help with psychological, survivors. We have argued that a solution to this health care
social, economic, and family concerns; support to improve crisis lies in defining a new model of comprehensive cancer
lifestyle behaviors; and interventions to increase adherence rehabilitation, involving a multidisciplinary team of pro-
to long-term treatment and follow-up care guidelines. Given viders that aims to optimize the patients physical, psy-
that most cancers are diagnosed in older adults, an esti- chological, vocational, and social functioning given the limits
mated two-thirds of all survivors of cancer will be age 65 or imposed by the chronic or late-effects of cancer treatment
older by 2020.2 This growing number of older survivors of and other comorbid conditions.20
cancer presents a unique challenge to the health care system
because they are more likely to have multiple chronic dis-
eases and experience poorer physical functioning than THE CASE FOR COMPREHENSIVE CANCER
younger survivors of cancer.9,10 Cancer and chronic illness REHABILITATION
also may have interactive adverse effects on health and The Institute of Medicine has defined the four pillars of
psychosocial outcomes among older adult survivors of survivorship care: (1) surveillance for recurrence, second
cancer. Chronic conditions may limit the intensity and du- cancers, and late effects; (2) intervention for treatment
ration of cancer treatment,11-14 contributing to poorer consequences; (3) prevention of recurrence, new cancers,

From American Cancer Society, Washington, DC; Department of Physical Medicine and Rehabilitation, Program to Enhance Care Experiences Through Research, Center for the Science of
Health Care Delivery, Mayo Clinic, Rochester, MN; University of Rochester Medical Center, Wilmot Cancer Institute, Rochester, NY.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Catherine M. Alfano, PhD, American Cancer Society, 555 11th St. NW, Suite 300, Washington, DC 20004; email: catherine.alfano@cancer.org.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 241


ALFANO, CHEVILLE, AND MUSTIAN

late effects, and comorbidities where possible; and (4) co- potential to decrease the risk of further late effects (e.g.,
ordination of care between primary care, oncology, and cardiac, pulmonary, endocrine, or bone complications of
other specialists.4 Comprehensive cancer rehabilitation cancer treatment)21 and may even reduce the risk of
delivered using a multidisciplinary team is an essential second malignancies.23-29 Finally, comprehensive cancer
component of survivorship care that addresses these four rehabilitation focuses jointly on optimizing functional
pillars. First, it treats the acute and chronic effects of cancer status and quality of life, preserving the ability to remain in
and prevents or mitigates the effects of late-occurring life roles (including the workforce),30 and maximizing
problems. Survivors of cancer can face numerous adverse health and longevity.
consequences of cancer treatment, many of which are
amenable to rehabilitation interventions. These include
fatigue, cognitive dysfunction, pain syndromes, peripheral A PROSPECTIVE MODEL OF SURVEILLANCE FOR
neuropathy, sexual dysfunction, balance and gait problems, COMPREHENSIVE CANCER REHABILITATION
upper- or lower-quadrant mobility issues, lymphedema, A prospective model of surveillance has been developed by a
bladder and bowel problems, stoma care, problems with team of stakeholders as a best practice model for cancer
swallowing or dysphagia, communication difficulty, and rehabilitation.31 In this model, rehabilitative efforts begin at
psychosocial problems such as depression, anxiety, or fear of the time of cancer diagnosis and continue through treat-
recurrence, among others.21,22 In a comprehensive reha- ment and after treatment ends, using a multidisciplinary
bilitation model, the multidisciplinary team evaluates the team approach. The model specifies that a multidimen-
sum total of problems that a survivor faces and coordinates sional, comprehensive assessment is conducted at the
treatment rather than other fragmented models of care that preoperative evaluation to establish baseline functioning,
treat each symptom or impairment separately. Second, the identify patients with pre-existing conditions that may place
comprehensive cancer rehabilitation team can address pre- them at higher risk for the development of treatment
existing or treatment-related comorbid conditions. Bone toxicities and impairments during/after treatment, and refer
loss, diabetes, cardiovascular disease, congestive heart failure, patients with current problems for interventions to improve
adverse body composition, and renal disease are common in their symptoms and function. Ideally, this is a clinical
cancer survivors.4 These can be managed through rehabili- evaluation that includes a history focusing on symptoms and
tation interventions that include medication, counseling, current exercise and a physical examination that objectively
behavior change and promotion of healthy diets, physical assesses the patients range of motion, strength, sensation,
activity, and weight control.21 Third, these health promotion reflexes, and gait accompanied by performance testing (e.g.,
interventions, along with self-management skills provided in The Timed Up and Go, Repeated Sit to Stand tests, and Short
the context of comprehensive cancer rehabilitation, have the Physical Performance Battery). This may be obtained or
supplemented with patient-reported outcome (PRO)based
measurement of survivors symptoms and functioning, al-
though limited information is available on the sensitivity and
KEY POINTS specificity of PROs in identifying cancer survivors in need of
cancer rehabilitation services.32,33 After the preoperative
Survivors of cancer can face numerous adverse assessment, the prospective surveillance model specifies
consequences of cancer treatment, many of which are ongoing surveillance efforts with repeated assessments
amenable to rehabilitation interventions. postoperatively, throughout cancer therapies, and after
Using an expanded prospective model of surveillance for
treatment ends. This allows for monitoring of the devel-
comprehensive cancer rehabilitation, assessment and
referral efforts begin at the time of cancer diagnosis,
opment of treatment toxicities or impairments and fa-
continue through and beyond treatment, and use a cilitation of appropriate and timely referrals to cancer
multidisciplinary team approach. rehabilitation providers. The reassessments also provide
The model facilitates the identification and treatment of opportunities for assessment and referral for exercise,
physical impairments and psychosocial problems and nutrition, and weight management interventions. The
other problematic symptoms, and provides exercise, ongoing surveillance also allows the treatment team to
nutrition, and weight management interventions to ensure the resolution of problems over the long term.
optimize functioning, health, and quality of life. Though not explicitly stated by the original stakeholder
Survivors of cancer can be treated in one of four levels of group, a prospective surveillance model for comprehensive
stepped care or inpatient rehabilitation depending on cancer rehabilitation must incorporate assessment and re-
safety requirements and needs.
ferral for mental health problems and palliative care needs in
This expanded prospective model of surveillance for
cancer rehabilitation has the potential to result in early
addition to impairment-driven rehabilitation and health
identification of symptoms and impairments and behavior interventions to meet survivors ongoing needs.
appropriate referral and timely treatment and, in turn, Although this model has been outlined by a multidisciplinary
will better address and minimize both acute and long- group of stakeholder experts, it has not been extensively
term cancer morbidity. tested for effects on clinical outcome or health care costs.34
The model assumes that prospective surveillance will result

242 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


DEVELOPING HIGH-QUALITY CANCER REHABILITATION PROGRAMS

in early identification of symptoms and impairments, ap- educational print materials or videos. More formal coun-
propriate referral and timely treatment, and, in turn, will seling by an exercise or rehabilitation professional, enroll-
better address and minimize both acute and long-term ment in a conditioning program with clinical oversight, and
cancer morbidity. The model has the potential to achieve more intensive behavioral counseling represent the next
the triple aim set forth by the Institute for Healthcare Im- two levels of increasingly time- and resource-intensive re-
provement35 of facilitating better care for individuals, better habilitation. Efficacious models of cardiac and pulmonary
health for populations, and lower per capita costs. However, rehabilitation offer a potential framework for the latter, with
future health care delivery research must test these as- pilot studies of their utility for survivors of cancer showing
sumptions. Although early research suggests substantial promise.40,41 As yet, center-based conditioning programs for
cost savings of implementing this model early compared cancer survivors have not been developed in the United
with treating advanced-stage problems, at least in the case States, but successes in Canada and Europe attest to their
of breast cancerrelated lymphedema,36 future research value.42,43
must include assessment of cost outcomes as previously Several options are available for practitioners and in-
described.34 stitutions interested in making general conditioning guid-
ance and training available to survivors of cancer. Physical
therapists without specialized cancer rehabilitation training
LEVELS OF CANCER REHABILITATION STEPPED routinely deliver deconditioning-directed care, and they are
CARE well equipped to individualize programs and transition
The expanded prospective model of surveillance presents a
cancer survivors to home-based maintenance programs.
framework for evaluating the needs of people with cancer
However, in the absence of functional impairments or
and triaging them into appropriate pathways of care based
steady, ongoing improvement, payer coverage for physical
on their need for cancer rehabilitation. But what type of care
therapy may be limited. For survivors of cancer without
they should receive and who should deliver the care are
concerning impairments or comorbidities that would require
issues that continue to challenge the rehabilitation com-
physical therapy or medical oversight, the LIVESTRONG at the
munity. Below we present a stepped-care framework to
YMCA 12-week programs offer services and support de-
guide decisions on when, how, and where to refer survivors
livered by athletic trainers and exercise professionals. These
to cancer rehabilitation specialists depending on safety
services extend well beyond conditioning, although the re-
requirements and needs. Note that all levels may involve
covery of strength and aerobic fitness are principle program
physical, occupational, or speech therapy; psychosocial and
goals. There are currently approximately 460 LIVESTRONG
cognitive interventions; exercise prescription; and lifestyle
programs in the continental United States. Referral to
recommendations as dictated by the needs of the individual.
medically oriented gyms offers another option. Medically
The disciplines and services that provide these different
oriented gyms are facilities typically staffed by physical
interventions, the environment in which they are provided,
therapists and/or athletic trainers with expertise in chronic
and the degree of integration of the team will differ
illness and the care of frail patients, but they may lack
depending on numerous factors. These include the strengths
cancer specialization. Medically oriented gyms require out-
and expertise of individual providers and existing services,
of-pocket payments. However, some payers will cover the
geographic proximity and communication among services,
initial exercise prescription, professional oversight and
and facility and system resources.
monitoring, and other services provided in these facilities.
These programs can be supplemented with counseling to
Level I: General Conditioning Activities, Unspecialized support behavioral changes as needed.
In the absence of any cancer-specific morbidity, virtually all
survivors of cancer emerge from their cancer treatment with
poorer aerobic fitness and muscle quality than when they Level II: General Conditioning Activities, Specialized
began.37 For this reason aloneapart from secondary Higher or more specialized levels of cancer rehabilitation
cancer prevention38,39education and/or prescriptive along the stepped-care continuum require care from clini-
counseling on progressive aerobic conditioning, resistance cians with some degree of specialization in cancer re-
training, and the benefits of exercise in general should be habilitation. The most prevalent need at this level is for
integral to any cancer survivorship program. This should be general conditioning among survivors of cancer who lack
supplemented by psychosocial counseling to enhance par- specific cancer- or cancer treatmentrelated impairments
ticipation in activities and address participation barriers but have sufficient vulnerability to warrant specialist over-
where needed. This represents the most basic level of sight. Survivors of cancer frequently have or are at risk for
stepped cancer rehabilitation. In the future, efforts to create conditions potentially exacerbated by exercise. Theoretically,
more precision medicine approaches to exercise prescrip- these individuals should have a rehabilitation team with at
tion will help guide the type and dose of exercise chosen. For least one cancer rehabilitation specialist, although empirical
now, for survivors of cancer who have exercised in the validation of this need is lacking. Psychosocial counseling
past and lack concerning impairments or comorbidities, to address barriers to participation or self-management
counseling can be quite rudimentary, taking the form of strategies may be needed. Research has established the

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 243


ALFANO, CHEVILLE, AND MUSTIAN

beneficial and safe nature of resistive training when con- such as allogeneic bone marrow transplant recipients who
ducted under the close supervision of exercise professionals commonly have CIPN, steroid myopathy, joint contractures,
trained in lymphedema and other cancer treatmentrelated and generalized deconditioning.49 The need for a cancer
risks.44,45 In some clinical settings, there may not be therapists rehabilitation physician inevitably will vary across settings
with specific cancer rehabilitation training. In these cases, a because many quaternary cancer centers have seasoned
rehabilitation savvy oncologist or cancer rehabilitation phy- teams with therapists accustomed to managing challenging
sician may provide generally trained therapists with a detailed impairments. Level IV care is likely to involve coordinating
physical or occupational therapy prescription with clear care among more members of the multidisciplinary team as
precautions. They also may serve as a resource for these physical, occupational, speech and language, and psycho-
noncancer trained treating professionals. social therapies all may be needed. As in level III care, level IV
care focuses on treating impairments, ongoing symptom
management, and increasing activity levels to enable the
Level III: Impairment-Directed Care, Uncomplicated
survivor of cancer to safely transition to participate in
This level of cancer rehabilitation need is broad because it
lifestyle behaviors without oversight if possible.
encompasses focal cancer- and cancer treatmentrelated
impairments that limit function but are uncomplicated by
Inpatient Cancer Rehabilitation. Inpatient cancer re-
symptoms or other systemic concerns. These impairments
habilitation delivered by a multidisciplinary team of pro-
generally can be managed by physical and occupational
viders through consultation and liaison and postacute care
therapists with a moderate degree of cancer specialization.
services clearly is a vital component of comprehensive
Care focuses on treating the impairment(s) to optimize
cancer care. In fact, the overwhelming majority of physical
function and also on increasing activity levels with an end
impairments, at least among patients with stage IV disease,
goal of enabling the cancer survivors to safely participate in
are detected when patients are hospitalized.50 Goals of care
lifestyle behaviors without oversight if possible. This level of
are similar to higher outpatient levels: treating impairments,
care also is more likely to require psychosocial counseling to
psychosocial and other problematic symptoms, optimizing
help survivors cope with problematic symptoms, body image
functioning, and supporting exercise and dietary changes to
concerns, or other mental health issues and support be-
the extent possible. All hospitals and inpatient cancer
havior change efforts. The impairments that fall into this
centers offer physical and occupational therapy services.
category are as varied as they are numerous. They include,
Most have dietitians and some have rehabilitation-oriented
but are not limited to, cranial nerve 11 palsy, radiation-
mental health specialists, but the availability of a re-
induced fibrosis, axillary web syndrome (variant Mondor
habilitation physician specialized in cancer is less consistent.
disease), contractures from sclerodermiform graft versus
Even when lacking cancer rehabilitation expertise, the in-
host disease, persistent chemotherapy-induced peripheral
volvement of a rehabilitation physician in the management
neuropathy (CIPN), surgical donor site morbidity (e.g., post
of hospitalized patients offers salient advantages to a cancer
transverse rectus abdominis [TRAM] flap reconstruction),
rehabilitation program, providing particularly effective and
and secondary myofascial dysfunction. A rapidly growing
expeditious advocacy for appropriate postacute care ser-
body of literature describes the diagnosis and evidence-
vices.51 Such optimized discharge planning with the in-
based treatment of many of these impairments.46-48 Efforts
volvement of rehabilitation specialists has been shown to
are underway to make treatment guidelines widely available
reduce 30-day readmissions and costs per episode of care,
to practicing therapists. Fortunately, specialty conferences
issues that are becoming increasingly important with the
and other educational opportunities are a rapidly increasing
rapid adoption of bundled payment.
means by which therapists can refine their expertise and
benefit from collective experience. Ideally, a cancer reha-
bilitation physician specialist is available to consult on
complex or nonresponding cases, although in less complex
IMPLEMENTATION OF QUALITY CANCER
cases, a physician need not evaluate these impairments and
REHABILITATION CARE
The drive to prevent and reverse disablement has spurred
thereby potentially delay their timely treatment.
care providers to develop cancer rehabilitation programs
across diverse delivery settings. This drive has been further
Level IV: Impairment-Directed Care, Complicated intensified by the American College of Surgeons Commission
The most resource-intensive form of outpatient cancer re- on Cancer standard that accredited institutions provide
habilitation along the stepped-care continuum is specialist cancer rehabilitation services.52 Despite increased aware-
physiciandirected evaluation and treatment. This should be ness, patient access to appropriate cancer rehabilitation
reserved for patients with problematic symptoms (e.g., in- services remains hampered by an unfortunate legacy of the
tensely painful axillary cording or severe or refractory im- decades-long marginalization of cancer rehabilitationthe
pairments) because such care may prove to be time-consuming fragmentation of rehabilitation disciplines and the persis-
and costly for cancer survivors with depleted energy and tence of a severely limited workforce that has proven slow to
resources. At times, the sheer number of co-occurring im- expand. Because of low referral rates from oncology clinical
pairments may necessitate physician involvement in cases practices, rehabilitation medicine has not been incentivized

244 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


DEVELOPING HIGH-QUALITY CANCER REHABILITATION PROGRAMS

to develop cancer rehabilitation positions or training pro- body of literature demonstrates that maintaining moderate
grams. The limited availability of cancer rehabilitation to high levels of physical activity through formal exercise
programs, even in large, high-volume cancer centers,20 and interventions is an effective method for treating both the
the varied services provided by the few established pro- physical and psychological declines experienced by older
grams has afforded oncology clinicians minimal opportunity patients with cancer as well as younger cancer survivors.55-77
to learn how to effectively capitalize on the expertise of their Research shows that adults with cancer decrease their
rehabilitation medicine colleagues to improve patient out- physical activity levels after diagnosis and during treatment
comes. This self-perpetuating cycle has been hard to break. and often do not return to their pretreatment activity levels
Even today, oncology trainees seldom learn how and when without formal exercise interventions.78-80 This tendency
to refer their patients for rehabilitation services. toward declining physical activity is reinforced as pa-
Several approaches have been established and/or are tients often are advised to limit activities when they are
being actively tested to overcome workforce limitations. The older, fatigued, or experiencing other toxicities and side
first, structured rehabilitation provider trainings and certi- effects, especially cardiac, orthopedic, or functional
fications, have gained broad traction. CARF International comorbidities.9,75,77,81-83
introduced accreditation standards for cancer rehabilitation Exercise improves a wide array of physical and psycho-
specialty programs in 2014 that can be applied to hospitals, logical symptoms, including muscle atrophy and weakness,
health care systems, outpatient clinics, and community- fatigue, obesity, immune function, insomnia, anxiety, cognitive
based programs. The Survivorship Training and Rehab decline, and impaired quality of life, among others.55-74,84-88
(STAR) program, a commercial entity, offers web-based Epidemiologic data also suggest that increased physical activity
trainings to physical and occupational therapists linked to through regular exercise reduces the risk of cancer recurrence
examinations that, when passed, lead to institutional STAR and cancer mortality.39,89-90 The ACSM published public health
certification. The impact of CARF standards or STAR certi- recommendations for exercise among survivors of cancer.91
fication on care quality and effectiveness have not been Geriatric survivors of cancer should start low and progress to
characterized. The American College of Sports Medicine 150 minutes of moderate-intensity or 75 minutes of vigorous-
(ACSM) also has developed educational guidelines and a intensity aerobic exercise per week; 20 to 30 minutes of
certifying examination for cancer exercise trainers who work strength training across all the major muscle groups two to
with survivors. The certification provides a professional three times per week; and regular stretching daily each
competency benchmark for oncology professionals who are week.91-94 Despite these published public health guide-
looking for exercise-referral options in their local commu- lines, it is estimated up to 70% of survivors do not meet
nities.53 Again, the care implications of practitioner certifi- these ACSM public health recommendations.95 This lack of
cation are not known. However, the fact that a practitioner regular exercise is especially problematic for older survi-
or institution has gone to the trouble of seeking, identifying, vors of cancer when combined with the considerable
and achieving CARF, STAR, or ACSM certification attests to physical and psychological comorbidities, symptoms, and
their interest and potential skill in cancer rehabilitation. side effects they experience.
Another approach to workforce expansion is the enhance- Despite the numerous benefits of exercise as part of
ment of noncancer physical or occupational therapy services cancer rehabilitation, the majority of survivors of cancer
available in most U.S. communities through the direction report that they do not discuss exercise with their treating
and support of physician and therapist cancer rehabilitation oncologist or primary care physician during their cancer
specialists at tertiary cancer centers. Data collection for the treatment and recovery.96-98 However, research shows that
federally funded Collaborative Care to Preserve Perfor- cancer survivors of all ages want their oncologists to initiate
mance in Cancer (COPE) trial finished in 2015.54 COPE is discussions about exercise and make appropriate referrals.98
rigorously assessing whether a telemedicine approach that Survivors prefer to receive information on exercise early in
leverages limited cancer rehabilitation clinical expertise to the treatment trajectory and throughout the post-treatment
deliver evidence-based treatment by local generalist phys- period.99 Given the benefits of exercise during and after
ical therapists is a useful near-term and cost-sensitive means cancer treatments, routine discussions about exercise be-
of addressing current workforce limitations. tween oncologists and their older patients who are survivors
of cancer, along with appropriate referrals to a qualified
exercise physiologist or cancer rehabilitation therapist as
EXERCISE AS AN ESSENTIAL COMPONENT OF needed, could significantly improve prognosis, recovery, and
CANCER REHABILITATION, ESPECIALLY FOR multiple domains of quality of life for older individuals.
OLDER ADULTS
The team that created the prospective model of surveillance
for cancer rehabilitation described exercise as an essential Managing Risk and Contraindications for Exercise
component of cancer rehabilitation.31 Therefore, exercise is As part of the prospective surveillance model for cancer
integrated into each of the stepped-care levels described rehabilitation, it is essential to identify and address potential
above. This importance of exercise is especially true for contraindications (e.g., orthopedic, cardiopulmonary, on-
deconditioned and older adult survivors of cancer. A growing cologic) that might affect exercise safety and tolerance.53

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 245


ALFANO, CHEVILLE, AND MUSTIAN

Contraindications do not mean that a geriatric survivor of older patients with cancer and survivors of cancer.23,72,82,101
cancer cannot exercise at all. As described above in the levels Accumulating 30 minutes of daily activity through short bouts
of stepped care, contraindications simply require specific of aerobic exercise (3 to 10 minutes each), with rest in between,
modifications to the exercise regimen or care environment also can reduce symptoms in these patients.91 A moderate to
(e.g., facility-based vs. home) so that the individual can vigorously intense anaerobic resistance exercise prescriptions
exercise safely and still achieve the desired physical and schedule of three times per week and progressively increasing
mental health benefits.53 Although active exercisers or those up to as few as two sets and no more than four sets of 8 to 15
at low to moderate risk based on the ACSM guidelines are repetitions are effective at reducing symptoms.102-106 Com-
not required to obtain medical clearance to continue or bination exercise prescriptions that include both aerobic and
begin a low to moderate exercise program, most geriatric anaerobic exercise are safe and very effective for geriatric
survivors of cancer will need and benefit from further survivors of cancer. Low to moderately intense mindfulness-
medical assessment because their exercise risk will be based exercise prescriptions, including yoga and tai chi chuan,
moderate to high.53 These geriatric survivors of cancer will scheduled one to three times a week for 6090 minutes, are
benefit from additional medical assessment by qualified also highly effective in reducing symptom burden among older
medical professionals including oncologists, surgeons, adults.64,69,71,74,107,108 Additionally, those with advanced
cardiologists, orthopedists, and neurologists, among disease can safely perform, tolerate, and benefit from low-
others, because these consultations will provide important intensity exercise.109-114
information required by exercise physiologists to appro-
priately modify an exercise prescription and account for CONCLUSION
cancer-specific cardiovascular, pulmonary, metabolic, or- We have described an expanded prospective model of
thopedic, neurologic, or other comorbidities.53 These surveillance for cancer rehabilitation in which assessment
additional medical evaluations should be conducted and referral efforts begin at the time of cancer diagnosis,
before initiation of any baseline exercise testing that continue through and beyond treatment, and use a multi-
precedes developing an exercise prescription and sub- disciplinary team approach. This model focuses on iden-
sequent exercise participation.53 tification and treatment of physical impairments and
psychosocial problems and the provision of lifestyle and
Exercise Prescription for Older Adult Survivors of exercise interventions. The model proposes that patients
Cancer can be triaged into four levels of stepped care in addition to
Testing, prescription, and monitoring of exercise should be inpatient rehabilitation, depending on the degree of
done by qualified exercise professionals, especially for ge- comorbidities and impairments noted, safety concerns,
riatric survivors of cancer at moderate risk beginning or and other needs. This model has the potential to result in
continuing vigorous exercise and those at high risk beginning earlier identification of symptoms, appropriate referral,
or continuing any level of exercise.53 Exercise prescriptions timely symptom management, and ultimately lower
for geriatric survivors of cancer should be individualized and morbidity and mortality. However, several key research
tailored based on the older individuals health status, disease questions remain. Assessment methods must be developed
trajectory, previous and/or current treatment, current fitness to create an appropriate comprehensive measure of
level, past and present exercise participation, and individual symptoms, functioning, impairments, and needs. Modeling
preferences to be safe and effective.91,92 Moderately intense efforts must focus on identifying the algorithm to triage
aerobic exercise prescriptions (55%75% of heart rate maxi- survivors of cancer into the levels of care. Once these are in
mum100) scheduled over as little as 10 minutes and no more place, future research efforts must also test the impact of
than 90 minutes a day 3 to 7 days per week are effective at this model on patient outcomes, care coordination, and
reducing symptom burden and improving quality of life among health care costs.

References
1. American Cancer Society. Cancer Treatment and Survivorship Facts & 5. Hewitt M, Weiner SL, Simone JV. Childhood Cancer Survivorship:
Figures 2014-2015. Atlanta: American Cancer Society; 2014. Improving Care and Quality of Life. Washington, DC: National
2. Parry C, Kent EE, Mariotto AB, et al. Cancer survivors: a boom- Academies Press; 2003.
ing population. Cancer Epidemiol Biomarkers Prev. 2011;20: 6. Adler NE, Page A, National Institute of Medicine. (U. S.) Committee
1996-2005. on Psychosocial Services to Cancer Patients/Families in a Com-
3. de Moor JS, Mariotto AB, Parry C, et al. Cancer survivors in the United munity Setting. Cancer Care for the Whole Patient: Meeting
States: prevalence across the survivorship trajectory and implications Psychosocial Health Needs. Washington, DC: National Academies
for care. Cancer Epidemiol Biomarkers Prev. 2013;22:561-570. Press; 2008.
4. HewittM, GreenfieldS, Stovall E (eds). From Cancer Patient to Cancer 7. Centers for Disease Control and Prevention, Lance Armstrong
Survivor: Lost in Transition. Washington, DC: National Academies Foundation, U.S. Department of Health and Human Services. A
Press; 2006. National Action Plan for Cancer Survivorship: Advancing Public Health

246 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


DEVELOPING HIGH-QUALITY CANCER REHABILITATION PROGRAMS

Strategies. http://www.cdc.gov/cancer/survivorship/pdf/plan.pdf. 30. de Boer AG, Taskila T, Tamminga SJ, et al. Interventions to enhance
Accessed March 8, 2016. return-to-work for cancer patients. Cochrane Database Syst Rev.
8. Presidents Cancer Panel. Living Beyond Cancer: Finding a New 2011;2:CD007569.
Balance. Bethesda, MD: National Cancer Institute: 2004. 31. Stout NL, Binkley JM, Schmitz KH, et al. A prospective surveillance
9. Bellizzi KM, Mustian KM, Palesh OG, et al. Cancer survivorship and model for rehabilitation for women with breast cancer. Cancer. 2012;
aging: moving the science forward. Cancer. 2008;113(Suppl 12): 118(Suppl 8):2191-2200.
3530-3539. 32. Cheville AL, Yost KJ, Larson DR, et al. Performance of an item response
10. Avis NE, Deimling GT. Cancer survivorship and aging. Cancer. 2008;113 theory-based computer adaptive test in identifying functional decline.
(Suppl 12):3519-3529. Arch Phys Med Rehabil. 2012;93:1153-1160.
11. Yancik R, Wesley MN, Ries LA, et al. Effect of age and comorbidity in 33. Hahn EA, Devellis RF, Bode RK, et al; PROMIS Cooperative Group.
postmenopausal breast cancer patients aged 55 years and older. Measuring social health in the patient-reported outcomes mea-
JAMA. 2001;285:885-892. surement information system (PROMIS): item bank development and
12. Extermann M. Interaction between comorbidity and cancer. Cancer testing. Qual Life Res. 2010;19:1035-1044.
Contr. 2007;14:13-22. 34. Cheville AL, Nyman JA, Pruthi S, et al. Cost considerations regarding
13. Greenfield S, Blanco DM, Elashoff RM, et al. Patterns of care related to the prospective surveillance model for breast cancer survivors.
age of breast cancer patients. JAMA. 1987;257:2766-2770. Cancer. 2012;118(Suppl 8):2325-2330.
14. Ballard-Barbash R, Potosky AL, Harlan LC, et al. Factors associated with 35. Institute for Healthcare Improvement. IHI Triple Aim Initiative. http://
surgical and radiation therapy for early stage breast cancer in older www.ihi.org/engage/initiatives/tripleaim. Accessed February 12, 2016
women. J Natl Cancer Inst. 1996;88:716-726. 36. Stout NL, Pfalzer LA, Springer B, et al. Breast cancer-related
15. Piccirillo JF, Tierney RM, Costas I, et al. Prognostic importance of lymphedema: comparing direct costs of a prospective surveillance
comorbidity in a hospital-based cancer registry. JAMA. 2004;291: model and a traditional model of care. Phys Ther. 2012;92:152-163.
2441-2447. 37. Gilliam LA, St Clair DK. Chemotherapy-induced weakness and fatigue
16. Given CW, Given B, Azzouz F, et al. Comparison of changes in physical in skeletal muscle: the role of oxidative stress. Antioxid Redox Signal.
functioning of elderly patients with new diagnoses of cancer. Med 2011;15:2543-2563.
Care. 2000;38:482-493. 38. Lahart IM, Metsios GS, Nevill AM, et al. Physical activity, risk of death
17. Hewitt M, Rowland JH, Yancik R. Cancer survivors in the United States: and recurrence in breast cancer survivors: a systematic review and
age, health, and disability. J Gerontol A Biol Sci Med Sci. 2003;58: meta-analysis of epidemiological studies. Acta Oncol. 2015;54:635-654.
M82-M91. 39. Ballard-Barbash R, Friedenreich CM, Courneya KS, et al. Physical
18. Erikson C, Salsberg E, Forte G, et al. Future supply and demand for activity, biomarkers, and disease outcomes in cancer survivors:
oncologists: challenges to assuring access to oncology services. J Oncol a systematic review. J Natl Cancer Inst. 2012;104:815-840.
Pract. 2007;3:79-86. 40. Munro J, Adams R, Campbell A, et al. CRIBthe use of cardiac re-
19. Salsberg E, Grover A. Physician workforce shortages: implications and habilitation services to aid the recovery of patients with bowel cancer:
issues for academic health centers and policymakers. Acad Med. 2006; a pilot randomised controlled trial (RCT) with embedded feasibility
81:782-787. study. BMJ Open. 2014;4:e004684.
20. Alfano CM, Ganz PA, Rowland JH, et al. Cancer survivorship and cancer 41. Hubbard G, Adams R, Campbell A, et al. Is referral of postsurgical
rehabilitation: revitalizing the link. J Clin Oncol. 2012;30:904-906. colorectal cancer survivors to cardiac rehabilitation feasible and ac-
21. Stubblefield MD, ODell, MW. Cancer Rehabilitation Principles and ceptable? A pragmatic pilot randomised controlled trial with em-
Practice. New York: Demos Medical Publishing; 2009. bedded qualitative study. BMJ Open. 2016;6:e009284.
22. Silver JK, Baima J, Mayer RS. Impairment-driven cancer rehabilitation: 42. Chasen MR, Bhargava R. A rehabilitation program for patients with
an essential component of quality care and survivorship. CA Cancer J gastroesophageal cancera pilot study. Support Care Cancer. 2010;18
Clin. 2013;63:295-317. (Suppl 2):S35-S40.
23. Holmes MD, Chen WY, Feskanich D, et al. Physical activity and survival 43. Kjaer TK, Johansen C, Ibfelt E, et al. Impact of symptom burden on
after breast cancer diagnosis. JAMA. 2005;293:2479-2486. health related quality of life of cancer survivors in a Danish cancer
24. Pierce JP, Stefanick ML, Flatt SW, et al. Greater survival after breast rehabilitation program: a longitudinal study. Acta Oncol. 2011;50:
cancer in physically active women with high vegetable-fruit intake 223-232.
regardless of obesity. J Clin Oncol. 2007;25:2345-2351. 44. Schmitz KH, Ahmed RL, Troxel A, et al. Weight lifting in women with
25. Irwin ML, Smith AW, McTiernan A, et al. Influence of pre- and breast-cancer-related lymphedema. N Engl J Med. 2009;361:664-673.
postdiagnosis physical activity on mortality in breast cancer survivors: 45. Schmitz KH, Ahmed RL, Troxel AB, et al. Weight lifting for women at
the health, eating, activity, and lifestyle study. J Clin Oncol. 2008;26: risk for breast cancer-related lymphedema: a randomized trial. JAMA.
3958-3964. 2010;304:2699-2705.
26. Giovannucci E, Liu Y, Platz EA, et al. Risk factors for prostate cancer 46. McNeely ML, Parliament MB, Seikaly H, et al. Effect of exercise on
incidence and progression in the health professionals follow-up study. upper extremity pain and dysfunction in head and neck cancer sur-
Int J Cancer. 2007;121:1571-1578. vivors: a randomized controlled trial. Cancer. 2008;113:214-222.
27. Meyerhardt JA, Giovannucci EL, Holmes MD, et al. Physical activity and 47. Cho Y, Do J, Jung S, et al. Effects of a physical therapy program
survival after colorectal cancer diagnosis. J Clin Oncol. 2006;24: combined with manual lymphatic drainage on shoulder function,
3527-3534. quality of life, lymphedema incidence, and pain in breast cancer
28. Meyerhardt JA, Heseltine D, Niedzwiecki D, et al. Impact of physical patients with axillary web syndrome following axillary dissection.
activity on cancer recurrence and survival in patients with stage III colon Support Care Cancer. Epub 2015 Nov 5.
cancer: findings from CALGB 89803. J Clin Oncol. 2006;24:3535-3541. 48. De Groef A, Van Kampen M, Dieltjens E, et al. Effectiveness of
29. Irwin ML, McTiernan A, Manson JE, et al. Physical activity and survival postoperative physical therapy for upper-limb impairments after
in postmenopausal women with breast cancer: results from the breast cancer treatment: a systematic review. Arch Phys Med Rehabil.
Womens Health Initiative. Cancer Prev Res (Phila). 2011;4:522-529. 2015;96:1140-1153.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 247


ALFANO, CHEVILLE, AND MUSTIAN

49. Smith SR, Haig AJ, Couriel DR. Musculoskeletal, neurologic, and 67. Janelsins MC, Davis PG, Wideman L, et al. Effects of tai chi chuan on
cardiopulmonary aspects of physical rehabilitation in patients with insulin and cytokine levels in a randomized controlled pilot study on
chronic graft-versus-host disease. Biol Blood Marrow Transplant. breast cancer survivors. Clin Breast Cancer. 2011;11:161-170.
2015;21:799-808. 68. Peppone LJ, Janelsins MC, Kamen C, et al. The effect of YOCAS yoga
50. Cheville AL, Shen T, Chang M, et al. Appropriateness of the treatment for musculoskeletal symptoms among breast cancer survivors on
of fatigued patients with stage IV cancer. Support Care Cancer. 2013; hormonal therapy. Breast Cancer Res Treat. 2015;150:597-604.
21:229-233. 69. Peppone LJ, Mustian KM, Janelsins MC, et al. Effects of a structured
51. Yadav R. Rehabilitation of surgical cancer patients at University weight-bearing exercise program on bone metabolism among breast
of Texas M. D. Anderson Cancer Center. J Surg Oncol. 2007;95: cancer survivors: a feasibility trial. Clin Breast Cancer. 2010;10:
361-369. 224-229.
52. American College of Surgeons. The Commission on Cancers (CoC) 70. Kamen C, Heckler C, Janelsins MC, et al. A dyadic exercise intervention
Cancer Program Standards: Ensuring Patient-Centered Care. Chicago: to reduce psychological distress among lesbian, gay, and heterosexual
American College of Surgeons; 2016. cancer survivors. LGBT Health. 2016;3:57-64.
53. American College of Sports Medicine. ACSMs Guidelines for Exercise 71. Mustian KM, Katula JA, Gill DL, et al. Tai chi chuan, health-related
Testing and Prescription, 8th Ed. Philadelphia: Lippincott Williams & quality of life and self-esteem: a randomized trial with breast cancer
Wilkins; 2016. survivors. Support Care Cancer. 2004;12:871-876.
54. Cheville A. Collaborative Targeted Case Management in Improving 72. Mustian KM, Peppone L, Darling TV, et al. A 4-week home-based
Functional Status in Patients with Stage III-IV Cancer. https:// aerobic and resistance exercise program during radiation therapy:
clinicaltrials.gov/ct2/show/NCT01721343?term=cheville&rank=3. a pilot randomized clinical trial. J Support Oncol. 2009;7:158-167.
Accessed February 12, 2016 73. Mustian KM, Sprod LK, Janelsins M, et al. Multicenter, randomized
55. Demark-Wahnefried W, Clipp EC, Morey MC, et al. Lifestyle in- controlled trial of yoga for sleep quality among cancer survivors. J Clin
tervention development study to improve physical function in older Oncol. 2013;31:3233-3241.
adults with cancer: outcomes from Project LEAD. J Clin Oncol. 2006;24: 74. Mustian KM, Katula JA, Zhao H. A pilot study to assess the influence of
3465-3473. tai chi chuan on functional capacity among breast cancer survivors.
56. Morey MC, Snyder DC, Sloane R, et al. Effects of home-based diet and J Support Oncol. 2006;4:139-145.
exercise on functional outcomes among older, overweight long-term 75. Mustian KM, Sprod LK, Palesh OG, et al. Exercise for the management
cancer survivors: RENEW: a randomized controlled trial. JAMA. 2009; of side effects and quality of life among cancer survivors. Curr Sports
301:1883-1891. Med Rep. 2009;8:325-330.
57. Campo RA, Agarwal N, LaStayo PC, et al. Levels of fatigue and distress 76. Spence RR, Heesch KC, Brown WJ. Exercise and cancer rehabilitation:
in senior prostate cancer survivors enrolled in a 12-week randomized a systematic review. Cancer Treat Rev. 2010;36:185-194.
controlled trial of Qigong. J Cancer Surviv. 2014;8:60-69. 77. Mustian KM, Sprod LK, Janelsins M, et al. Exercise recommendations
58. Winters-Stone KM, Leo MC, Schwartz A. Exercise effects on hip bone for cancer-related fatigue, cognitive impairment, sleep problems,
mineral density in older, post-menopausal breast cancer survivors are depression, pain, anxiety, and physical dysfunction: a review. Oncol
age dependent. Arch Osteoporos. 2012;7:301-306. Hematol Rev. 2012;8:81-88.
59. Cormie P, Galv~ao DA, Spry N, et al. Can supervised exercise prevent 78. Irwin ML, Crumley D, McTiernan A, et al. Physical activity levels before
treatment toxicity in patients with prostate cancer initiating and after a diagnosis of breast carcinoma: the Health, Eating, Activity,
androgen-deprivation therapy: a randomised controlled trial. BJU Int. and Lifestyle (HEAL) study. Cancer. 2003;97:1746-1757.
2015;115:256-266. 79. Courneya KS, Friedenreich CM. Relationship between exercise pattern
60. Winters-Stone KM, Lyons KS, Dobek J, et al. Benefits of partnered across the cancer experience and current quality of life in colorectal
strength training for prostate cancer survivors and spouses: results cancer survivors. J Altern Complement Med. 1997;3:215-226.
from a randomized controlled trial of the Exercising Together project. 80. Mustian KM, Griggs JJ, Morrow GR, et al. Exercise and side effects
J Cancer Surviv. Epub 2015 Dec 29. among 749 patients during and after treatment for cancer: a Uni-
61. Winters-Stone KM, Dieckmann N, Maddalozzo GF, et al. Re- versity of Rochester Cancer Center Community Clinical Oncology
sistance exercise reduces body fat and insulin during androgen- Program Study. Support Care Cancer. 2006;14:732-741.
deprivation therapy for prostate cancer. Oncol Nurs Forum. 2015;42: 81. Mustian KM, Peppone LJ, Palesh OG, et al. Exercise and cancer-related
348-356. fatigue. US Oncol. 2009;5:20-23.
62. Sprod LK, Fernandez ID, Janelsins MC, et al. Effects of yoga on cancer- 82. Mustian KM, Morrow GR, Carroll JK, et al. Integrative non-
related fatigue and global side-effect burden in older cancer survivors. pharmacologic behavioral interventions for the management of
J Geriatr Oncol. 2015;6:8-14. cancer-related fatigue. Oncologist. 2007;12(Suppl 1):52-67.
63. Sprod LK, Mohile SG, Demark-Wahnefried W, et al. Exercise and 83. Bellizzi KM, Mustian KM, Bowen DJ, et al. Aging in the context of
cancer treatment symptoms in 408 newly diagnosed older cancer cancer prevention and control: perspectives from behavioral medi-
patients. J Geriatr Oncol. 2012;3:90-97. cine. Cancer. 2008;113(Suppl 12)3479-3483.
64. Sprod LK, Janelsins MC, Palesh OG, et al. Health-related quality of life 84. Brown JC, Huedo-Medina TB, Pescatello LS, et al. Efficacy of exercise
and biomarkers in breast cancer survivors participating in tai chi interventions in modulating cancer-related fatigue among adult
chuan. J Cancer Surviv. 2012;6:146-154. cancer survivors: a meta-analysis. Cancer Epidemiol Biomarkers Prev.
65. Sprod LK, Palesh OG, Janelsins MC, et al. Exercise, sleep quality, and 2011;20:123-133.
mediators of sleep in breast and prostate cancer patients receiving 85. Irwin ML, Alvarez-Reeves M, Cadmus L, et al. Exercise improves body
radiation therapy. Community Oncol. 2010;7:463-471. fat, lean mass, and bone mass in breast cancer survivors. Obesity
66. Janelsins MC, Peppone LJ, Heckler CE, et al. YOCAS Yoga reduces (Silver Spring). 2009;17:1534-1541.
self-reported memory difficulty in cancer survivors in a nationwide 86. Gleeson M, Bishop NC, Stensel DJ, et al. The anti-inflammatory effects
randomized clinical trial: investigating relationships between memory of exercise: mechanisms and implications for the prevention and
and sleep. Integr Cancer Ther. Epub 2015 Nov 29. treatment of disease. Nat Rev Immunol. 2011;11:607-615.

248 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


DEVELOPING HIGH-QUALITY CANCER REHABILITATION PROGRAMS

87. Tang M-F, Liou T-H, Lin C-C. Improving sleep quality for cancer pa- chemotherapy: a multicenter randomized controlled trial. J Clin
tients: benefits of a home-based exercise intervention. Support Care Oncol. 2007;25:4396-4404.
Cancer. 2010;18:1329-1339. 102. Ahmed RL, Thomas W, Yee D, et al. Randomized controlled trial of
88. Salmon P. Effects of physical exercise on anxiety, depression, and weight training and lymphedema in breast cancer survivors. J Clin
sensitivity to stress: a unifying theory. Clin Psychol Rev. 2001;21:33-61. Oncol. 2006;24:2765-2772.
89. Betof AS, Dewhirst MW, Jones LW. Effects and potential mechanisms 103. Courneya KS, Segal RJ, Gelmon K, et al. Six-month follow-up of patient-
of exercise training on cancer progression: a translational perspective. rated outcomes in a randomized controlled trial of exercise training
Brain Behav Immun. 2013;30(Suppl):S75-S87. during breast cancer chemotherapy. Cancer Epidemiol Biomarkers
90. Bittoni MA, Harris RE, Buckworth J, et al. Abstract 5043: Physical Prev. 2007;16:2572-2578.
activity and the risk of lung cancer death: results from the Third 104. Schmitz KH, Ahmed RL, Hannan PJ, et al. Safety and efficacy of weight
National Health and Nutrition Examination Survey. Cancer Res. 2014; training in recent breast cancer survivors to alter body composition,
74:5043. insulin, and insulin-like growth factor axis proteins. Cancer Epidemiol
91. Schmitz KH, Courneya KS, Matthews C, et al; American College of Biomarkers Prev. 2005;14:1672-1680.
Sports Medicine. American College of Sports Medicine roundtable on 105. Segal RJ, Reid RD, Courneya KS, et al. Resistance exercise in men
exercise guidelines for cancer survivors. Med Sci Sports Exerc. 2010; receiving androgen deprivation therapy for prostate cancer. J Clin
42:1409-1426. Oncol. 2003;21:1653-1659.
92. U.S. Department of Health and Human Services. Physical activity 106. Winters-Stone KM, Dobek J, Nail L, et al. Strength training stops bone
guidelines Advisory Committee Report, 2008. Part A: executive loss and builds muscle in postmenopausal breast cancer survivors:
summary. Nutr Rev. 2009;67:114-120. a randomized, controlled trial. Breast Cancer Res Treat. 2011;127:
93. Haskell WL, Lee IM, Pate RR, et al. Physical activity and public health: 447-456.
updated recommendation for adults from the American College of 107. Mustian KM, Janelsins M, Sprod L, et al. YOCAS Yoga significantly
Sports Medicine and the American Heart Association. Med Sci Sports improves circadian rhythm, anxiety, mood and sleep: a randomized,
Exerc. 2007;39:1423-1434. controlled clinical trial among 410 cancer survivors. Support Care
94. Haskell WL, Lee IM, Pate RR, et al; American College of Sports Cancer. 2011;19(suppl 2):S317-S318.
Medicine; American Heart Association. Physical activity and public 108. Reid-Arndt SA, Matsuda S, Cox CR. Tai chi effects on neuro-
health: updated recommendation for adults from the American psychological, emotional, and physical functioning following cancer
College of Sports Medicine and the American Heart Association. treatment: a pilot study. Complement Ther Clin Pract. 2012;18:26-30.
Circulation. 2007;116:1081-1093. 109. Crevenna R, Schmidinger M, Keilani M, et al. Aerobic exercise for a
95. Irwin ML. Physical activity interventions for cancer survivors. Br J patient suffering from metastatic bone disease. Support Care Cancer.
Sports Med. 2009;43:32-38. 2003;11:120-122.
96. Jones LW, Courneya KS. Exercise discussions during cancer treatment 110. Headley JA, Ownby KK, John LD. The effect of seated exercise on
consultations. Cancer Pract. 2002;10:66-74. fatigue and quality of life in women with advanced breast cancer.
97. Mustian KM, Griggs JJ, Morrow GR, et al. Exercise and side effects Oncol Nurs Forum. 2004;31:977-983.
among 749 patients during and after treatment for cancer: a Uni- 111. Porock D, Kristjanson LJ, Tinnelly K, et al. An exercise intervention for
versity of Rochester cancer center community clinical oncology advanced cancer patients experiencing fatigue: a pilot study. J Palliat
program study. Support Care Cancer. 2006;14:732-741. Care. 2000;16:30-36.
98. Yates JS, Mustian KM, Morrow GR, et al. Prevalence of complementary 112. Oldervoll LM, Loge JH, Paltiel H, et al. The effect of a physical exercise
and alternative medicine use in cancer patients during treatment. program in palliative care: a phase II study. J Pain Symptom Manage.
Support Care Cancer. 2005;13:806-811. 2006;31:421-430.
99. Sprod LK, Peppone LJ, Palesh OG, et al. Effect of timing of information 113. Adamsen L, Midtgaard J, Rorth M, et al. Feasibility, physical capacity,
on exercise behavior during cancer treatment: a URCC CCOP protocol. and health benefits of a multidimensional exercise program for cancer
J Clin Oncol. 28:15s (suppl; abstr 9138). patients undergoing chemotherapy. Support Care Cancer. 2003;11:
100. Borg GA. Psychophysical bases of perceived exertion. Med Sci Sports 707-716.
Exerc. 1982;14:377-381. 114. Oldervoll LM, Loge JH, Lydersen S, et al. Physical exercise for cancer
101. Courneya KS, Segal RJ, Mackey JR, et al. Effects of aerobic and patients with advanced disease: a randomized controlled trial.
resistance exercise in breast cancer patients receiving adjuvant Oncologist. 2011;16:1649-1657.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 249


PEDIATRIC ONCOLOGY

Controversies in Germline
Genetic Testing and Disclosure
in Pediatric Oncology

CHAIR
Kim E. Nichols, MD, PhD
Childrens Hospital of Philadelphia
Philadelphia, PA

SPEAKERS
Angela R. Bradbury, MD
Perelman School of Medicine at the University of Pennsylvania
Philadelphia, PA

Lainie Friedman Ross, MD, PhD


The University of Chicago Medicine
Chicago, IL
PEDIATRIC GENETIC TESTING FOR CANCER

The Advantages and Challenges of Testing Children for


Heritable Predisposition to Cancer
Chimene Kesserwan, MD, Lainie Friedman Ross, MD, PhD, Angela R. Bradbury, MD, and Kim E. Nichols, MD

OVERVIEW

The increased application of germline genetic testing is expanding our understanding of the risk factors associated with
childhood cancer development, and, in some cases, such testing is also informing clinical management. Nonetheless, the
incorporation of genetic testing into the pediatric oncology setting is complex and associated with many ethical and practical
challenges. The decision as to whether to pursue clinical genetic testing for hereditary cancer predisposition for children should
always be guided by the best interest of the child. Despite this fundamental ethical principle, patients, parents, and health care
providers may differ in their opinions. Clinical genetic testing to detect the presence of predisposition syndromes associated
with childhood-onset cancers, particularly those for which surveillance and preventive measures have proven to enhance
outcome, is currently well accepted. On the other hand, clinical genetic testing of children for syndromes associated with adult-
onset cancers has raised many concerns about the potential for psychological harm and disrespect of patient autonomy. As
a consequence, such testing is not encouraged. The challenges surrounding germline genetic testing are further complicated
when testing is done in the research setting and/or when it involves whole-exome or whole-genome sequencing approaches,
which can uncover genetic variants that may or may not be associated with the disease under study. Accordingly, there is great
debate around these processes and the most appropriate approaches regarding the return of test results. Future research is
needed to enhance knowledge about how best to incorporate genomic information into clinical practice.

T he increased incorporation of genetic and genomic


testing in the pediatric oncology setting, including the
use of larger and more comprehensive multigene panels and
medically best for the child, the parents calculation considers
what is best for the child by incorporating and balancing many
types of needs and interests. That is, parents consider not only
whole-exome sequencing, has greatly facilitated the iden- the childs medical needs but also the childs social, cultural,
tification of children with underlying predisposition syn- religious, and economic needs and interests. Ethical contro-
dromes. In this article, we discuss the advantages and versies related to clinical genetic testing of children include
complexities related to germline genetic testing, with an the following: (1) whether to focus exclusively on the childs
emphasis on the evaluation of children for heritable cancer self-regarding interests (e.g., concerns about the childs own
risk. We provide an ethical framework with which to ap- medical well-being) or to include other-regarding interests
proach clinical genetic testing in children and then discuss (e.g., concerns about the needs and interests of family
the advantages and challenges related to testing children in members who may be affected by the information learned
the clinical and research settings. from the childs genetic test results); (2) how to balance the
childs present-day needs and interests with his or her future
needs and interests; and (3) whether to include the childs
ETHICAL FRAMEWORK FOR APPROACHING present and future autonomy interests (as well as present
CLINICAL CANCER GENETIC TESTING IN and future privacy rights).8-10
PEDIATRICS In the context of clinical genetic testing of children, the
Professional guidelines regarding the clinical genetic testing childs best interest will focus on the diagnosis of conditions
of children all agree that the guiding principle should be this that require immediate attention. Traditionally, a physician
standard: the best interest of the child.1-8 What this principle orders one or more genetic tests targeted to diagnose a
means and how it should be applied in different clinical childs symptoms or phenotype. The increasing use of ge-
settings are less clear. Although physicians may know what is nomic sequencing has increased the number of successful

From the Department of Oncology, St. Jude Childrens Research Hospital, Memphis, TN; Departments of Pediatrics, Medicine, and Surgery, MacLean Center for Clinical Medical Ethics,
The University of Chicago, Chicago, IL; Department of Medicine, Department of Medical Ethics and Health Policy, Perelman School of Medicine of the University of Pennsylvania,
Philadelphia, PA.

Disclosures of potential conflicts of interest provided by the authors are available with the online article at asco.org/edbook.

Corresponding author: Kim E. Nichols, MD, 262 Danny Thomas Place, Memphis, TN 38105; email: kim.nichols@stjude.org.

2016 by American Society of Clinical Oncology.

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 251


KESSERWAN ET AL

diagnoses but has also resulted in discoveries of variants of 50% risk of inheriting a disease-causing mutation in RB1 or in one
unknown significance and secondary and incidental find- of the genes associated with HPPS. Clinically, one needs to know
ings. We provide an ethical framework for clinical genetic whether a child carries an RB1 mutation in infancy because the
and genomic testing in minors, examine scenarios that involve cancer usually presents before age 5. In contrast, tumors as-
predictive genetic testing in minors for cancer-predisposing sociated with HPPS do not generally appear before adolescence,
conditions by using single-gene testing, and then consider so genetic testing is recommended at approximately age 10 or 5
scenarios in which genomic sequencing, rather than targeted to 10 years before the earliest HPPS-associated cancer identified
testing, is used. in the family.
Consider, then, the case of a child whose father developed a
paraganglioma at age 25 and was found to carry a causal
Predictive Testing for Cancer Predisposition mutation in one of the HPPS genes. It is recommended to start
Syndromes That Present in Childhood screening for HPPS-associated tumors at age 10 in mutation-
In 2012, the American Academy of Pediatrics (AAP) and
positive children.11 Before the HPPS-associated genes were
the American College of Medical Genetics and Genomics
discovered, it was advocated that all children whose par-
(ACMG) published a joint policy statement and technical
ents had manifestations of HPPS begin lifelong biochemical
report on clinical genetic testing in children. 4,5 Earlier
screening for abnormally high levels of catecholamines and
statements made by these organizations objected to the
undergo annual physical examinations, starting at age 10, with
predictive testing in children1,3 but did not distinguish be-
attention to the symptoms associated with paragangliomas
tween testing for conditions that present in childhood and
or pheochromocytomas, such as high blood pressure, heart
those that present in adulthood. The timing of when a
palpitations, anxiety, or headache.12 Today, only those in-
condition is expected to present is crucial, because it affects
dividuals with germline HPPS-associated genetic mutations
the relative importance of the childs role in the decision-
need to undergo surveillance including MRI screening.11,12
making process, whether the child has a right to privacy from
What if the father wanted to test the child for the presence
his or her parents, and whether the parents have a right to
of HPPS mutation when he was an infant? The arguments
make such decisions on behalf of their child.
to support waiting until the child is 10 years old are to avoid
Of the cancer-predisposing conditions that present in child-
demands for earlier initiation of annual surveillance screen-
hood, some will be early-onset (e.g., hereditary retinoblas-
ing; to reduce parental anxiety; and to allow the child to
toma), and others will present in adolescence or young
participate in the decision-making process. However, all three
adulthood (e.g., hereditary paraganglioma/pheochromocytoma
arguments can be refuted. First, the data find that par-
syndrome [HPPS]). Both of these are autosomal dominant,
ents rarely demand inappropriate treatments or initiation
therefore a child whose parent had one of these conditions is at
of therapies earlier than necessary.13 Second, testing is only
done in children with a known family history, so parental
anxiety already exists, and testing would allow half of the
KEY POINTS parents to be reassured.5,8,13 Furthermore, data show that
many individuals find uncertainty worse than a positive test
The guiding principle regarding clinical genetic and/or result.14,15 Third, although ethics supports the inclusion of
genomic testing of children should always be the best children to the extent that they are able to assent, the child in
interest of the child. this scenario does not really have a choice. Although mature
Diagnostic genetic testing of a child with cancer can children can refuse to participate in many types of research,
guide disease management and allow for the initiation
refusal by a child of clinically indicated care is more restricted
of surveillance for second primary cancers.
Predictive genetic testing for an underlying
when diagnosis, surveillance, and treatment can prevent
predisposition syndrome can identify children at serious morbidity or mortality. This is not to ignore the mature
increased cancer risk and allow for patient and family child, but respect can be shown in other ways (e.g., expla-
education, behavioral modification, and cancer nations of what at-risk status means and what the tests can
surveillance. Importantly, such testing can also determine). Of note, given that the child in question is at 50%
eliminate these procedures for children whose genetic risk of inheriting a syndrome in which tumors can be suc-
test results are negative. cessfully treated if diagnosed early, his parents right to re-
Cancer genetic testing of children should take into fuse is also restricted, given the states obligation, as parens
account the availability of evidence-based cancer patriae, to protect children from abuse and neglect. If the
surveillance and risk-reduction strategies and the child and his parents both refuse genetic testing, they could
probability of developing a malignancy during
opt for regular screening (perhaps not the best approach, but
childhood.
Whether to return genetic research results to research
one that can be considered good enough).16
participants is a subject of active debate. A clear plan for The arguments to allow parents to test a young child before
return of genetic research results to research it is clinically indicated are to reduce parental anxiety and to
participants should be formulated prior to recruitment permit them to make other life plans for the child and the
and explained during the informed consent process. family. The AAP/ACMG guidance gives parents wide leeway for
conditions that present at any point in childhood, because

252 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


PEDIATRIC GENETIC TESTING FOR CANCER

parents are the presumptive decision makers for their minor As genomic sequencing becomes cheaper, faster, and
children.5 Genetic counseling can help parents decide when more readily available, questions will continue to emerge
this testing is appropriate for their child, and the decision to about its appropriate use. In 2015, the American Society of
test the child in this scenario at either age should be respected. Human Genetics published updated guidelines for genetic
testing of minors that examined single-gene and genomic
sequencing methodologies for both the clinical and research
Predictive Testing for Cancer Predisposition settings.8 The American Society of Human Genetics con-
Syndromes That Present in Adulthood cluded that targeted tests, or selective sequence analysis,
Some parents seek genetic testing of minors for adult-onset are usually preferable to less-discriminate data acquisition
conditions, although surveillance is not needed until adult- when the clinical challenge can be addressed through a
hood. The 2012 AAP/ACMG statement recommends against targeted approach.8 However, even if the ACMG list of 56
predictive genetic testing for adult-onset disorders, because genes is not intentionally interrogated when a childs blood
the information is not necessary in childhood and because a is sequenced, and even if sequencing is focused on analyzing
delay of testing allows the child to make this decision as an genes that are relevant to the childs condition, testing may
adult for himself or herself (the concept of a childs right to an still identify incidental findings (such as variants in genes that
open future17,18); respects the right to privacy that an adult predispose to other health problems or variants that are only
has, even against his or her own parents; and avoids un- relevant to reproductive decision making [e.g., carrier sta-
necessary anxiety created by the time span between testing tus]). When the sample comes from a child, some seek to
and clinical relevance.5 The AAP/ACMG proscription is not limit disclosure of findings to those conditions that may
absolute but, rather, acknowledges that exceptional cir- manifest in childhood, whereas others seek to limit disclosure
cumstances exist. It permits predictive genetic testing to to childhood conditions for which early surveillance or treat-
resolve disabling parental anxiety or to support life-planning ment can affect outcomes.23,26-30 Some support a policy of
decisions that parents sincerely believe to be in the childs strict nondisclosure of incidental findings to minors and their
best interest particularly when parents and mature ado- parents, but others support pre- and post-test counseling and
lescents jointly express interest in proceeding.5 disclosure of incidental findings as agreed upon by parents,
physicians, and when possible, the child.22,26-30 The clinical,
psychological, and economic costs and benefits of these
Genomic Sequencing for Cancer Predisposition and various approaches on families and on the health care system
Other Conditions remain uncertain and will need to be carefully evaluated.
The 2012 AAP/ACMG statement did not address the use of
genomic sequencing because this test was still considered a
research tool and the guidance focused on clinical care. One CLINICAL FRAMEWORK FOR APPROACHING
month later, however, the ACMG unilaterally proposed that, CANCER GENETIC TESTING IN PEDIATRICS
whenever clinical genomic sequencing was performed, the The germline genetic testing of children with cancer is
sample should be interrogated opportunistically for 56 genes primarily pursued for diagnostic or predictive purposes.
that are highly penetrant in high-risk populations regardless Diagnostic testing can explain tumor formation, provide
of the reason for sequencing, the age of the patient, or the prognostic information, and guide decisions about cancer
consent of the patient and physician.19 treatment. It is recommended that patients with cancer
The Association of Molecular Pathologists objected be- and Li-Fraumeni or Gorlin syndrome for example, who
cause interrogating a sample for these 56 genes requires harbor germline mutations in TP53 or PTCH1 respectively,
additional time, resources, and expertise, and the ACMG not be exposed to ionizing radiation because of the in-
policy required no ones permission, so it was not clear who creased risk for secondary radiation-induced cancers.
was ordering the tests, who was willing to receive and Simiarly, patients with cancer and genetic syndromes
convey the results to patients and families, and who was associated with defective DNA repair, such as Fanconi
going to pay for this analysis.20 Others involved in pediatrics anemia, should not be treated with conventional doses of
and genetics objected to such testing because most of the 56 myelosuppressive chemotherapy and/or radiation because
genes were associated with conditions that would not of the excess toxicity associated with these approaches.
present until adulthood,21-23 and the AAP/ACMG consensus Genetic information can also guide the surgical approach
was that it was not in the childs best interest to routinely to cancer treatment, when no surgery or organ-sparing
test for adult-onset conditions.4,5 The ACMG responded that surgery of paired at risk organs (e.g., the eyes in children
such testing was indeed in a childs best interest because it with hereditary retinoblastoma or the kidneys in children
could identify disease-causing mutations that may be with WT1-associated hereditary Wilms tumor) is preferred
present in their parents.24 One year later, the ACMG revised over removal of an entire organ.
the recommendations to allow patients (and surrogates) to Predictive testing derives its benefit from the tenet that
opt out of the screening of these 56 genes and to allow early detection of tumors will lead to prevention or sur-
health care professionals to limit the amount of information veillance interventions that reduce overall morbidity and
requested.25 mortality. In light of this issue, the American Society of

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 253


KESSERWAN ET AL

TABLE 1. Cancer Predisposition Syndromes and Associated Physical Findings


Syndrome Physical Findings
Disorders Associated With Defects in DNA Repair
Ataxia Telangiectasia Ataxia
Telangiectasia
Cafe-au-lait macules
Bloom Syndrome Small size for age
Erythematous skin lesions on the nose and cheeks (butterfly rash)
Underdeveloped malar and lower mandibular areas
Fanconi Anemia Radial ray anomalies: malformations of thumbs, forearms
Short stature
Pigmentary skin changes: cafe-au-lait macules, hypopigmentation
Other anomalies: small eyes, narrow ear canal and abnormal pinna, micropenis
Nijmegen Breakage Syndrome Microcephaly
Growth retardation
Rothmund-Thomson Syndrome Poikyloderma
Dysplastic nails
Rudimentary or hypoplastic teeth
Radial ray anomalies
Acral keratosis
Werner Syndrome Short stature
Premature graying of the hair
Xeroderma Pigmentosum Acute sun sensitivity
Marked freckle-like pigmentation
Severe keratitis
Photophobia
Squamous cell cancer of skin
Acquired microcephaly
Inherited Bone Marrow Failure Syndromes
Diamond-Blackfan Anemia Microcephaly
Growth retardation
Ocular hypertelorism, ptosis
Congenital glaucoma, congenital cataract
Webbed neck, short neck
Klippel-Feil anomaly (short neck, low hair line and/or limited range of motion in the neck due to
fusion of cervical vertebrae)
Sprengel deformity (high scapula)
Flat thenar eminence
Triphalangeal, bifid, hypoplastic, or absent thumb
Dyskeratosis Congenita Dystrophic nails
Dental abnormalities (hypodontia, early tooth loss)
Lacy reticular pigmentation of the upper chest and/or neck
Alopecia
Premature graying of the hair
GATA2 Deficiency Cutaneous warts
Lymphedema of extremities
Shwachman-Diamond Syndrome Short stature
Failure to thrive and poor growth
Continued

254 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


PEDIATRIC GENETIC TESTING FOR CANCER

TABLE 1. Cancer Predisposition Syndromes and Associated Physical Findings (Contd)


Syndrome Physical Findings
Neurocutaneous Syndromes
Neurofibromatosis Type 1 Cafe-au-lait macules
Freckling in the axillary and/or inguinal regions
Hypopigmented macules
Lisch nodules
Macrocephaly
Neurofibromas
Short stature
Tibial pseudarthrosis
Neurofibromatosis Type 2 Cafe-au-lait macules
Cutaneous schwannomas
Tuberous Sclerosis Complex Hypomelanotic macules
Angiofibromas
Shagreen Patch
Confetti skin lesions
Dental enamel pits
Ungual fibromas
Neuroendocrine Syndromes
Carney Complex Spotty skin pigmentation of lips, conjunctiva and inner or outer canthi, vaginal and penile mucosal
surfaces
Blue nevi
Cafe-au-lait macules
Cutaneous and mucosal myxomas
Acromegaly
Multiple Endocrine Neoplasia Type 1 Cutaneous collagenomas
Facial angiofibromas
Multiple Endocrine Neoplasia Type 2B Marfanoid habitus
Neuromas of lips and tongue
Overgrowth Syndromes
AIP-Related Familial Isolated Pituitary Gigantism or acromegaly
Adenomas
Beckwith-Wiedemann Syndrome Macroglossia
Macrosomia
Ear lobe creases, helical pits
Hemihyperplasia
Omphalocele
Umbilical hernia
Diastasis recti
Nevoid Basal Cell Carcinoma Syndrome Coarse facial features, frontal bossing
(Gorlin Syndrome) Macrocephaly
Palmar and plantar pits
Preaxial or postaxial polydactyly
Basal cell carcinomas
PTEN Hamartoma Tumor Syndrome Macrocephaly
Trichilemmomas
Lipomas
Oral mucosal papillomas
Acral keratosis
Hemangiomas
Penile freckling
Continued

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 255


KESSERWAN ET AL

TABLE 1. Cancer Predisposition Syndromes and Associated Physical Findings (Contd)


Syndrome Physical Findings
Simpson-Golabi-Behmel Syndrome Macrocephaly
Macrosomia
Sotos Syndrome Long narrow face with bitemporal narrowing
Long chin
Overgrowth
Sparse frontotemporal hair
Intestinal Polyposis Syndromes
Constitutional Mismatch Repair Defect Cafe-au-lait macules
Hypopigmented macules
Familial Adenomatous Polyposis Syndrome Dental abnormalities: missing or supernumerary teeth
Epidermoid cyst
Osteomas of the skull and/or mandible
Pilomatricomas
Peutz-Jeghers Syndrome Melanotic macules around the mouth, eyes, nostrils, and perianal area
RASopathies
Cardiofaciocutaneous Syndrome Coarse facial features
Bitemporal narrowing
Hypertelorism, downslanting palpebral fissures, and hypoplasia of the supraorbital ridges
Low set ears, ear lobe creases
Macrocephaly relative to body size
Short neck
Short stature
Keratosis pilaris
Lymphedema
Costello Syndrome Coarse facial features
Curly or sparse fine hair
Deep palmar or plantar creases
Friendly, sociable personality
Full lips, large mouth
Papillomata of the face and anal region
Short stature
Ulnar deviation of wrists and fingers
Noonan Syndrome Coarse facial features
Downslanted palpebral fissures
Vivid blue or blue-green irises
Fullness of upper eyelids and ptosis
Low set posteriorly rotated ears with fleshy helices
Broad or webbed neck
Short stature
Pectus deformity of chest
Widely set nipples
Cryptorchidism
Noonan Syndrome With Multiple Lentigines Coarse facial features
Downslanted palpebral fissures
Widely spaced eyes and ptosis
Low set ears, posteriorly rotated with thick helices
Lentigines
Cafe-au-lait macules
Continued

256 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


PEDIATRIC GENETIC TESTING FOR CANCER

TABLE 1. Cancer Predisposition Syndromes and Associated Physical Findings (Contd)


Syndrome Physical Findings
Short stature
Cryptorchidism
Disorders of Sex Chromosomes

Klinefelter Syndrome Gynecomastia and/or small genitalia


Tall stature
Turner Syndrome Short stature
Webbed neck
Low hair line
Shield-like chest
Wide-spaced nipples
Lymphedema of hands and feet
WT1-Related Disorders
Denys-Drash Syndrome Ambiguous genitalia
Frasier Syndrome Ambiguous genitalia
Wilms Tumor, Aniridia, and Growth Aniridia
Retardation Syndrome Ambiguous genitalia
Other
Rubinstein Taybi Syndrome Convex nasal bridge with low hanging columella
Downslanting palpebral fissures
Broad radially deviated thumbs
Broad terminal phalanges
Grimacing smile
Pilomatricomas
Talon cusps (extra cusp-like projection of the primary or permanent anterior teeth)
Undescended testis

Clinical Oncology (ASCO) advises that cancer genetic test- Factors to Consider When Pursuing Clinical Cancer
ing of children take into account the availability of evidence- Genetic Testing in Children
based risk-reduction strategies and the probability of The factors most commonly used to prompt referral of a
developing a malignancy during childhood. 31 Currently, child to a cancer genetics specialist include a personal
there are only a limited number of conditions for which effective medical or family history consistent with, or suggestive of,
preventive measures are known to exist. One such example is an underlying hereditary cancer syndrome. The personal
multiple endocrine neoplasia type 2 (MEN2), which is caused by features that most support an underlying cancer predis-
germline RET gene mutations. Predictive RET gene testing is the position in a child include the presence of: a clinical or
clinical standard of care for all individuals with a positive family molecular diagnosis of a known predisposition syndrome;
history of MEN2 because of the very high risk to develop early- very early onset or adult-type tumors (e.g., epithelial breast
onset medullary thyroid cancer in affected individuals, including or colon cancer); multifocal tumors; bilateral involvement of
paired organs (e.g., eyes, kidneys, adrenal glands, breasts);
children. Prophylactic thyroidectomy during childhood is
specific cancer types and histologic features (e.g., medullary
therefore indicated for anyone who harbors a pathogenic gain-
thyroid cancer and MEN2); second primary neoplasms; and
of-function RET gene mutation because this procedure greatly
specific physical findings (examples of these physical find-
reduces or even eliminates the risk of developing thyroid ings are provided as a reference and listed in Table 1).
cancer. Risk stratification that is based on RET genotype The family history is also a key factor that helps guide
can inform decisions about the age to perform the prophylactic a cancer genetics evaluation. ASCO has recognized the
thyroidectomy.32 The identification of a predisposing germline importance of an adequate family history in the identifi-
mutation also guides strategies for cancer monitoring. Although cation of patients whose cancers may be associated with
surveillance protocols are under investigation for many con- inherited genetic factors. For each relative of a child with
ditions, effective approaches do exist for certain conditions, cancer, it is important to record whether that relative de-
such as familial ademonatous polyposis, Beckwith Wiedeman veloped cancer and, if so, to also record the type of cancer
syndrome, Li-Fraumeni syndrome, and the WT1-associated and its histologic features; the age at cancer diagnosis; laterality;
Wilms tumor syndromes, among others.33-38 maternal or paternal lineage relationships of affected relatives

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 257


KESSERWAN ET AL

TABLE 2. Family History Features Suggestive of an guidelines for cancer surveillance of many predisposition
Underlying Cancer Predisposition syndromes that affect children do not yet exist. Multicenter,
prospective studies to assess the clinical utility of surveillance
Feature Findings protocols are critically needed. Third, there remains concern
Pattern of Three generations affected by cancer (on the same that the testing of children for cancer predisposition will cause
Cancer side of the family) adverse psychological outcomes for the child, the parents, and
Occurrence
Three or more family members with the same type of other family members. Although it has been reported that
cancer (on the same side of the family)
some children experience distress, discrimination, guilt, or
One or more first-degree relatives with cancer (parent, regret, most of the published literature indicates that there is
child, sibling)
no increase in mean anxiety, depression, and distress scores
Age of Cancer Earlier than expected age of onset for cancer type
Onset
as a result of testing.41,42 Fourth, the increasing use of mul-
tigene panel and genomic testing has increased the difficulty
Cancer Type/ Adult type cancers in children (e.g., epithelial cancers
Presentation such as adenocarcinoma of the breast, ovary, and level and time required to adequately counsel patients and
colon) parents. A 2015 ASCO policy statement highlights the el-
Constellation of tumors consistent with a familial ements of pretest counseling recommended for multigene
syndrome (e.g., breast and ovarian cancer in panel testing. In addition to including the general compo-
hereditary breast and ovarian cancer syndrome;
uterine and colon cancer in Lynch syndrome) nents of traditional counseling, the possibility of detecting
Multiple primary cancers
predisposing mutations not suggested by the personal or
family history factors, and the implications of positive results
Bilateral or multifocal cancers
in lesser-penetrance genes are key points to address.43 The
Ethnic Ashkenazi Jewish background
Background
counseling process should include a discussion of variants of
Consanguinity unknown significance, which remain a source of confusion
Other Features Clinical or molecular diagnosis of a predisposition for health care providers and patients. Providers should
syndrome in a close relative
ensure that families comprehend the concept of variants of
unknown significance and recognize that the presence of
with cancer; ethnicity; and results of prior genetic testing.39 A
such variants does not generally change clinical manage-
minimum adequate family history should include cancer in first-
ment. They should consult the literature and seek the as-
and second-degree relatives of the proband and no less than a
sistance of experts for additional opinions, when necessary.
three-generation pedigree. Table 2 lists the family history
Given the challenges associated with genetic and genomic
features suggestive of an underlying predisposition.
testing, interpretation of test results, and the potential
impacts on clinical management, it is recommended that
Guidelines for the Referral of Children for Clinical children who are candidates for cancer genetic or genomic
Cancer Genetic Testing testing be referred to physicians and genetic specialists with
The ACMG and National Society of Genetic Counselors clinical expertise and research interests in these subject
(NSGC) have issued practice guidelines to facilitate id- areas.
entification and referral of adults with cancer and at-risk
individuals to a cancer genetics specialist. 40 Currently,
guidelines dedicated to the evaluation of children are GUIDELINES AND CHALLENGES RELATED TO
lacking. Tables 3, 4, and 5 provide practical information to aid RESEARCH GENETIC TESTING AMONG CHILDREN
in the genetics referral of children with liquid, central As large, prospective, cohort studies with banked DNA in-
nervous system (CNS), and non-CNS solid tumors. creasingly have been used to evaluate the effects of genes,
the environment, and lifestyle, there has been increasing
Challenges Associated With Cancer Genetic Testing in debate about the obligations, if any, to share individual
Children genetic research results with research participants.44 The
Despite the benefits of childhood cancer genetic testing, traditional approach has been to not share individual re-
several challenges remain. First, providers often lack the search results with research participants or, in the setting of
time, tools, or expertise to collect and meaningfully interpret pediatrics, with their parents or surrogates.45,46 This tradi-
family history information. Furthermore, it is important to tional approach is rooted in distinctions between the goals of
recognize that family histories may appear negative because research and clinical care.27,45,47,48 Classically, the primary
of inaccurate or inadequate medical information and be- goal of research is to create generalizable knowledge to
cause of incomplete penetrance or presence of de novo ultimately benefit a population as a whole. In contrast, the
germline mutations. Providers must not overlook the pos- goal of clinical care is to benefit an individual patient and
sibility of an underlying syndrome, even if the family history improve health outcomes. Arguments against returning
appears negative. This is particularly true for children who individual research results include blurring this important
have cancers that are known to have a heritable basis in a distinction between research and clinical care,27,47-51 the
clinically relevant proportion of cases (e.g., retinoblastoma, potential for misunderstanding or over-interpretation of the
adrenocortical carcinoma, paraganglioma). Second, consensus clinical significance or limitations of genetic results identified

258 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


PEDIATRIC GENETIC TESTING FOR CANCER

TABLE 3. Indications for Consideration of Genetics Evaluation in Children With Hematopoietic Malignancies
Type of
Hematopoietic Consider Referring for Genetics Evaluation Cancer Predisposition
Malignancy When Any of the Following Is Present Syndrome(s) to Consider Gene(s)
ALL Family history of LFS-associated tumors LFS TP53
Hypodiploid ALL LFS TP53
Cafe -au-lait macules CMMRD MLH1, MSH2, MSH6, PMS2, EPCAM
Family history of Lynch syndromeassociated tumors
Consanguinity
Pure red cell aplasia DBA RPL11, RPL26, RPL35A, RPL5, RPS7,
RPS10, RPS17, RPS19, RPS24,
RPS26, RPS29, GATA1
Pancreatic insufficiency SDS SBDS
Growth failure
Bone marrow failure
Family history of ALL Familial ALL caused by PAX5 PAX5
mutations
Family history of ALL Familial ALL caused by ETV6 ETV6
mutations
Thrombocytopenia
Family history of ALL Familial ALL caused by SH2B3 SH2B3
mutations
AML/Myelodysplastic Family history of LFS-associated tumors LFS TP53
Syndrome
Cafe -au-lait macules CMMRD MLH1, MSH2, MSH6, PMS2, EPCAM
Family history of Lynch syndromes-associated tumors
Consanguinity
Cafe -au-lait macules FA BRCA2, BRIP1, ERCC4, FANCA, FANCB,
FANCC, FANCD2, FANCE, FANCF,
Radial ray defect
FANCG, FANCI, FANCL, PALB2,
Growth and developmental delay RAD51C, SLX4, XRCC2
Microcephaly
Skeletal anomalies
Pure red cell aplasia DBA RPL11, RPL26, RPL35A, RPL5, RPS7,
RPS10, RPS17, RPS19, RPS24,
RPS26, RPS29, GATA1
Pancreatic insufficiency SDS SBDS
Growth failure
Bone marrow failure
Neutropenia Severe congenital neutropenia/ CSF3R, ELANE, G6PC3, GF11, HAX1,
Kostmann syndrome JAGN1
Pigmentary changes (neck/chest) Dyskeratosis congenita ACD, C16orf57, CTC1, DKC1, NHP2,
NOLA3, PARN, RTEL1, TERT, TERC,
Pulmonary fibrosis
TINF2, WRAP53
Bone marrow failure
Dysplastic nails
Oral leukoplakia
Family history of AML Familial AML caused by CEBPA CEBPA
mutations
Family history of AML Familial platelet disorder with RUNX1
predisposition to acute
Thrombocytopenia
myelogenous leukemia
Cytopenias MonoMac syndrome/Emberger GATA2
syndrome
Immunodeficiency and increased risk for M. avium
complex infection
Lymphedema
Radial ray defect Rothmund-Thomson syndrome RECQL4
Poikiloderma
Sparse hair/nail abnormalities
Continued
asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 259
KESSERWAN ET AL

TABLE 3. Indications for Consideration of Genetics Evaluation in Children With Hematopoietic Malignancies
(Contd)
Type of
Hematopoietic Consider Referring for Genetics Evaluation Cancer Predisposition
Malignancy When Any of the Following Is Present Syndrome(s) to Consider Gene(s)
JMML Family history and/or features of RASopathies or NF1 Noonan syndrome and related BRAF, CBL, HRAS, KRAS, MAP2K1,
RASopathies MAP2K2, NRAS, PTPN11, RAF1,
RIT1, SHOC2, SOS1
NF1 NF1
Mosaic Monosomy 7 Family history of monosomy 7 Familial mosaic monosomy 7 Unknown
syndrome
Non-Hodgkin Males with B-cell lymphoma and a history of EBV X-linked lymphoproliferative SH2D1A
Lymphoma infection and/or family history of XLP features disease
Cafe -au-lait macules CMMRD MLH1, MSH2, MSH6, PMS2, EPCAM
Family history of Lynch syndromeassociated cancers
Consanguinity
Gait abnormalities Ataxia-telangiectasia ATM
Cutaneous and/or ocular telangiectasia
Males with lymphoma, immunodeficiency, and/or Wiskott-Aldrich syndrome WAS
eczema
Thrombocytopenia, small platelets
Failure to thrive
Males with humoral immunodeficiency X-linked agammaglobulinemia BTK
Abbreviations: ALL, acute lymphoblastic leukemia; AML, acute myeloid leukemia; CMMRD, constitutional mismatch repair deficiency; DBA, Diamond-Blackfan anemia; EBV, Epstein-
Barr virus; FA, Fanconi anemia; JMML, juvenile myelomonocytic anemia; LFS, Li-Fraumeni syndrome; NF1, neurofibromatosis type 1; NF2, neurofibromatosis type 2; SDS, Shwachman-
Diamond syndrome.

in the research setting,47,48,50 maintenance of privacy and interest. Nonetheless, other potentially actionable muta-
the right of the participant to not know,52 the prohibitive tions (e.g., the 56 highly penetrant genes outlined by the
costs for research team and biobanks,53-55 and the potential ACMG) may have been sequenced, but the interpretation
negative impact on research progress if costs are shifted to and variant calling required to return individual results to
cover return of results at the expense of discovery.27,53,56 research participants would require additional resources
Yet, this traditional framework has been questioned over beyond the scope of the scientific aims. Third, somatic tumor
time, given an increasing appreciation that some genetic profiling (e.g., sequencing of tumors to identify acquired
findings identified in the research setting could significantly genomic aberrations that can be used to identify targeted
affect the health of a research participant.27,45,46,47,57-59 therapy) can indirectly or directly reveal inherited variants of
Others have argued that returning individual genetic re- clinical significance.66,67 Many somatic sequencing plat-
search results should be considered on the basis of the forms use a normal sample for subtraction analysis to clarify
principles of beneficence, autonomy, reciprocity, and respect which variants are tumor specific. Dedicated analysis of the
for persons, and, in pediatrics, that the return of individual normal or germline sample may identify inherited muta-
genetic research results is in the best interest of the child.57-64 tions. Tumor-only sequencing (those that do not include a
This debate has become more and more important with normal or germline sample) may also identify potential
the advent of next-generation sequencing, which allows germline mutations, because any variant found in the cancer
multiple genes or the entire genome to be sequenced rapidly could, in theory, reflect variation in the normal DNA. Thus, a
and at relatively accessible costs.48,65-67 Now, there are germline susceptibility to cancer may be incidentally iden-
numerous ways that potentially clinically significant genetic tified in the process of cataloging somatic mutations. Given
findings can be identified in the research setting. First, many the increasing range of scenarios in which research can
studies designed to evaluate the genetic underpinnings of identify potentially clinically significant genetic variants,
disease evaluate a number of genes suspected to be related there is an increasing need to clarify what obligations re-
to a specific condition. Even if these genes are not known to search teams have to return genetic research results to
have any relationship to the health of research participants participants.
at the time they enroll in the study, the level of evidence may
accumulate over time, such that they become clinically
actionable in the future. Second, a research study may se- Guidelines Regarding Returning Research Results
quence large portions of the genome but only invest in the In 1999, the National Bioethics Advisory Commission advised
interpretation of genetic variants related to the disease of against return of incidental research findings, except in rare

260 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


PEDIATRIC GENETIC TESTING FOR CANCER

TABLE 4. Indications for Consideration of Genetics Evaluation for Children With Central Nervous System, Spinal, or
Intracranial Tumors
Consider Referring for Genetics Evaluation When Any Cancer Predisposition
Type of Tumor of the Following Is Present Syndrome to Consider Gene(s)
Astrocytoma Anaplastic/high grade and family history of LFS TP53
LFS-associated tumors
Subependymal giant cell astrocytoma Tuberous sclerosis complex TSC1, TSC2
Cortical tuber
NF1 features NF1 NF1
Family history of NF1
Cafe-au-lait macules CMMRD MLH1, MSH2, MSH6, PMS2,
EPCAM
Family history of Lynch syndrome-associated cancers
Consanguinity
Atypical Teratoid/ Refer all RTPS SMARCB1, SMARCA4
Rhabdoid Tumor
Cerebellar Refer all VHL VHL
Hemangioblastoma
Choroid Plexus Refer all LFS TP53
Carcinoma
Endolymphatic Sac Refer all VHL VHL
Tumor
Ependymoma Spinal ependymoma NF2 NF2
Glioblastoma Cafe-au-lait macules CMMRD MLH1, MSH2, MSH6, PMS2,
EPCAM
Family history of Lynch syndrome-associated cancers
Consanguinity
Family history of LFS-associated tumors LFS TP53
Medulloblastoma Age , 3 NBCCS/Gorlin syndrome PTCH1, SUFU
Desmoplastic/extensive nodularity
Macrocephaly
Basal cell carcinoma
Palmar/plantar pitting
Jaw keratocysts
Calcification of the falx
Sonic Hedgehog subtype LFS TP53
Family history of LFS-associated tumors
Cafe-au-lait macules CMMRD MLH1, MSH2, MSH6, PMS2,
EPCAM
Family history of Lynch syndromeassociated cancers
Consanguinity
Family history of polyps, early-onset colon cancer FAP APC
Cafe-au-lait macules Fanconi anemia BRCA2, BRIP1, ERCC4, FANCA,
FANCB, FANCC, FANCD2,
Radial ray defect
FANCE, FANCF, FANCG, FANCI,
Developmental delays FANCL, PALB2, RAD51C, SLX4,
Microcephaly XRCC2

Skeletal anomalies
Multiple congenital anomalies
Meningioma Clear cell subtype Predisposition to clear cell SMARCE1
meningiomas
Macrocephaly NBCCS (Gorlin syndrome) PTCH1, SUFU
Basal cell carcinoma
Palmar/plantar pitting
Jaw keratocysts
Calcification of the falx
Personal/family history of Schwannomas NF2 NF2
Continued

asco.org/edbook | 2016 ASCO EDUCATIONAL BOOK 261


KESSERWAN ET AL

TABLE 4. Indications for Consideration of Genetics Evaluation for Children With Central Nervous System, Spinal,
or Intracranial Tumors (Contd)
Consider Referring for Genetics Evaluation When Any Cancer Predisposition
Type of Tumor of the Following Is Present Syndrome to Consider Gene(s)
Neurofibroma Refer all NF1 NF1
Personal/family history of Schwannomas NF2 NF2
Optic Pathway Glioma Family history or features of NF1 NF1 NF1
Pineoblastoma Family history of DICER1-associated tumors DICER1 syndrome DICER1
Family history of retinoblastoma Hereditary retinoblastoma RB1
Pituitary Blastoma Refer all DICER1 syndrome DICER1
Schwannoma Refer all NF2 NF2
Familial schwannomatosis SMARCB1
Abbreviations: CMMRD, constitutional mismatch repair deficiency; FAP, familial adenomatous polyposis; LFS, Li-Fraumeni syndrome; NBCCS, nevoid basal cell carcinoma syndrome;
NF1, neurofibromatosis type 1; NF2, neurofibromatosis type 2; RTPS, rhabdoid tumor predisposition syndrome; VHL, Von Hippel-Lindau; yo, years old.

circumstances. Since this initial guidance, various organi- Remaining Areas of Controversy and Unique
zations, advisory commissions, and scholars have provided Considerations for Children
additional guidance statements, which reflect a growing Despite this emerging consensus, there remain many areas
consensus that there may be some cases in which re- of continued debate. There is no consensus on criteria or
search participants should be offered the option of learning settings in which there is an obligation to return action-
about research results that could significantly affect their able research findings to pediatric research participants,
health.45,68-72 Although the 2013 ACMG statement proposes their parents, or their surrogates45,51,52,59,86,87 Although the
that laboratories have a fiduciary responsibility to seek and clinical utility or actionability of results is often used as a
report mutations in a minimum list of 56 genes (23 of which deciding factor, defining actionability is challenging, par-
are cancer related) regardless of test indication or age, ticularly as commercial tests become available prior to ev-
these recommendations have been controversial and were idence of clinical utility.88 There are additional complexities
designed specifically for sequencing in clinical care as op- in pediatrics, given that some conditions (e.g., adult-onset
posed to research.21,22,66,73-81 There currently is no con- conditions such as adult cancer or Alzheimer disease) may
sensus list of actionable genes for obligatory disclosure in not be immediately relevant to the child but could be im-
the research setting.19,82-84 Later in 2013, the Presidential mediately relevant to his or her relatives.46 Although some
Commission for Bioethical Issues published Anticipate and have argued that this is sufficient rationale to return results
Communicate: Ethical Management of Incidental and Sec- in the pediatric setting, others have rejected this rationale
ondary Findings in the Clinical, Research and Direct-to- for justifying the return.19,27,46 Second, the duration of
Consumer Contexts, which provides guiding principles obligations has been debated. Can research teams be held
and general recommendations for the return of incidental responsible for returning results that may not become ac-
findings in the research setting.85 Key recommendations tionable until many years after the research was completed
included the following: (1) anticipating the potential for and when they may not have remaining funding or resources
incidental genetic findings; (2) communicating the poten- to support the return of results? This is particularly relevant in
tial and plans for returning individual results in advance pediatric research, because a child may not have the ability to
(e.g., at the time of consent to the study); and (3) honoring make an informed decision about receiving research results
the right of participants to refuse receipt of incidental at the time of enrollment but may be able to do so in the
genetic research findings. Importantly, the Presidential future.46 Third, there is debate about whether genetic re-
Commission rejected an obligation to opportunistically search results must be conducted or confirmed in a Clinical
seek actionable genetic variants (e.g., secondary findings) Laboratory Improvement Amendments (CLIA)certified lab-
in the research setting, highlighting the negative impact oratory prior to return of results to research participants.
this could have on the research enterprise, in which society Some groups argue that only select results (e.g., those that are
has a significant interest.85 In addition, the Commission clearly clinically actionable and have been confirmed in a CLIA
emphasized the need for additional research, deliberation, laboratory) should be returned.51,87,89 Yet, many research
and context-specific guidelines from professional orga- investigations occur in non-CLIA certified laboratories, and
nizations. These recommendations are consistent with such requirements would significantly increase costs, detract
recommendations published by other expert groups and from the research at hand, and create barriers to returning
represent a growing consensus that some analytically potentially actionable genetic research findings. Thus, others
valid genetic research findings should be considered for argue that genetic research results should be offered to
return to participants and, in pediatrics, to parents and participants, and, in pediatrics, to their parents or surrogates,
surrogates.27,45,46,52 to let them make decisions about which results they would

262 2016 ASCO EDUCATIONAL BOOK | asco.org/edbook


PEDIATRIC GENETIC TESTING FOR CANCER

TABLE 5. Indications for Consideration of Genetics Evaluation for Children With Solid Tumors
Type of Solid Tumor by Consider Referring When Any Cancer Predisposition
Anatomic Location of the Following Is Present Syndrome to Consider Gene(s)
Breast

You might also like