You are on page 1of 419

Intracranial Tumors

Intracranial Tumors
Diagnosis and Treatment

Lisa M DeAngelis MD
Chairman, Department of Neurology
Memorial Sloan-Kettering Cancer Center
New York, NY 10021
USA

Philip H Gutin MD
Chief, Neurosurgery Service
Department of Surgery
Memorial Sloan-Kettering Cancer Center
New York, NY 10021
USA

Steven A Leibel MD
Chairman, Department of Radiation Oncology
Memorial Sloan-Kettering Cancer Center
New York, NY 10021
USA

Jerome B Posner MD
Attending Neurologist, Department of Neurology
Memorial Sloan-Kettering Cancer Center
New York, NY 10021
USA

MARTIN DUNITZ
2002 Martin Dunitz Ltd, a member of the Taylor & Francis group

First published in the United Kingdom in 2002 by Martin Dunitz Ltd,


The Livery House, 7-9 Pratt Street, London NW1 0AE

Tel: +44 (0) 20 7482 2202


Fax: +44 (0) 20 7267 0159
E-mail: info@dunitz.co.uk
Webiste: http://www.dunitz.co.uk

This edition published in the Taylor & Francis e-Library, 2003.

All rights reserved. No part of this publication may be reproduced, stored in a


retrieval system, or transmitted, in any form or by any means, electronic,
mechanical, photocopying, recording, or otherwise, without the prior permission
of the publisher or in accordance with the provisions of the Copyright, Designs
and Patents Act 1988 or under the terms of any licence permitting limited
copying issued by the Copyright Licensing Agency, 90 Tottenham Court Road,
London, W1P 0LP.

A CIP record for this book is available from the British Library.

ISBN 0-203-21421-8 Master e-book ISBN

ISBN 0-203-27084-3 (Adobe eReader Format)


ISBN 1-90186-537-1

Although every effort has been made to ensure that all owners of copyright
material have been acknowledged in this publication, we would be glad to
acknowledge in subsequent reprints or editions any omissions brought to our
attention.

Distributed in the USA by


Fulfilment Center
Taylor & Francis
7625 Empire Drive
Florence, KY 41042, USA
Toll Free Tel: +1 800 634 7064
E-mail: cserve@routledge_ny.com

Distributed in Canada by
Taylor & Francis
74 Rolark Drive
Scarborough, Ontario M1R 4G2, Canada
Toll Free Tel: +1 877 226 2237
E-mail: tal_fran@istar.ca

Distributed in the rest of the world by


ITPS Limited
Cheriton House
North Way
Andover, Hampshire SP10 5BE, UK
Tel: +44 (0)1264 332424
E-mail: reception@itps.co.uk

Composition by Scribe Design, Gillingham, Kent, UK


Contents

Preface vii 7 Neuronal, mixed and neuronal


glial and embryonal tumors 221
I General principles relevant to
diagnosis and treatment 8 Pineal region tumors 249

1 Classification, incidence and 9 Tumors of cranial nerves and


etiology of intracranial tumors 3 skull base 270

2 Invasion, angiogenesis and the 10 Pituitary and sellar region


bloodbrain barrier 36 tumors 296

3 Principles of diagnosis 65 11 Primary central nervous


system lymphoma and other
4 Principles of therapy 97 hemopoietic tumors 320

12 Miscellaneous central nervous


II Management of specific tumors system neoplasms and tumors 340

5 Glial tumors 149 13 Intracranial metastases 367

6 Meningeal tumors 189 Index 395

v
Preface

This book is intended for clinicians who care have reviewed the best current basic science
for patients with tumors involving the brain. and clinical evidence, compiled up-to-date
These tumors include meningiomas, pituitary, references, and because the book expresses the
pineal and skull base tumors as well as tumors opinions of a highly experienced team of
intrinsic to the brain thus the title neuro-oncologists working together in a cancer
Intracranial Tumors rather than Brain hospital, we hope that even seasoned neuro-
Tumors. The material in the book reflects our oncologists will find the book useful in their
collective experience as: a radiation oncologist own practice.
(Stephen Leibel), a neurosurgeon (Philip Gutin) A word about the front cover. We chose this
and two neurologists (Lisa DeAngelis and design to illustrate advances in the treatment
Jerome Posner), working as a team in a cancer of intracranial tumors. The figure on the left
hospital. The book is divided into two sections. is from a chapter by Walter Dandy in Lewis
The first considers general principles of the (ed) Practice of Surgery, 1944. One is not
biology, diagnosis and treatment of intracranial surprised that in those days, before steroids
tumors whether benign or malignant and and brain imaging, both morbidity and
whether primary or metastatic. The second mortality were high. The figures on the right
section deals with the biology, diagnosis and illustrate the use of frameless stereotaxy in the
management applied to specific tumors. current surgical management of brain tumors,
Physicians from a wide variety of specialties a technique that has substantially decreased
(e.g. family practitioners, internists, neurolo- both mortality and morbidity. Surgical and
gists, psychiatrists) are likely to be the first to other treatments of intracranial tumors are
encounter a patient with an intracranial tumor. discussed in Chapter 4.
Furthermore, neurosurgeons, oncologists, The opinions expressed in this book are
radiation oncologists or neurologists who do shared by many of our colleagues in neurology,
not specialize in intracranial tumors are those radiation oncology and neurosurgery, but we
who treat most of these patients. This book bear the full responsibility for the statements
was written primarily for these physicians and and opinions in this monograph. Our approach
for aspiring neuro-oncologists. Accordingly, to the patients, both diagnostically and thera-
experienced neuro-oncologists may find peutically, has been a team approach, and the
sections of the book involving diagnosis and four of us generally agree on the opinions and
imaging overly simplistic, and medical or radia- approach expressed in this monograph. We
tion oncologists may find the sections on radia- take full responsibility for any omissions,
tion therapy and chemotherapy obvious. We errors or outrageous statements.

vii
PREFACE

We had a lot of technical support in writing Burgess were very patient as we made last
this monograph. Elenita Sambat and Judith minute changes in both the text and the bibli-
Lampron typed the endless drafts, made more ography in an attempt to make the book as
numerous by the availability of the computer. current as possible.
Carol DAnella read the galleys and corrected We would be happy to receive comments,
the more egregious errors of syntax, grammar, corrections and opinions from any of the
and occasionally spelling that Spellcheck did readers who may be so inclined. If this book
not pick up. Amanda May and team at Martin has another edition we will certainly incorpo-
Dunitz were extremely helpful in copyediting rate them. We hope all who read this book will
and putting the book together. They and Alan enjoy it. If not, let us know.

Lisa DeAngelis
Philip Gutin
Stephen Leibel
Jerome Posner

viii
I
General principles relevant to diagnosis and
treatment
1
Classification, incidence and etiology of
intracranial tumors

Introduction Some of the apparent increase in CNS tumors


may be due to better detection with powerful
Central nervous system (CNS) tumors are the imaging techniques such as magnetic resonance
most feared cancers. Although cancers not imaging (MRI) or to the fact that most brain
involving the CNS (systemic cancers, e.g. tumors occur in old age, a cohort whose
breast, lung, colon) can cause pain, substantial numbers are increasing.4 The proponderence of
disability and even death, they attack the body evidence indicates that most or all of the appar-
whereas CNS tumors cause seizures, dementia, ent increase in CNS tumor incidence in both
paralysis and aphasia, symptoms that attack adults and children is related to better diagno-
the self. CNS tumors are also feared because sis.5 Primary CNS lymphoma is the exception
many are intractable to therapies that are effec- (Chapter 11).
tive when applied to systemic tumors. There Despite the fear and pessimism about CNS
are several reasons why many CNS tumors tumors, progress in diagnosis (Chapter 3) and
resist treatment. For example, surgical treatment (Chapter 4) is being made: better
techniques that allow complete removal of a diagnostic techniques, such as MRI and stereo-
breast or colon cancer along with a margin of tactic needle biopsy, have led to earlier diagno-
surrounding normal tissue are not feasible in sis that in some instances improves therapy.
the brain. Radiation therapy that controls New imaging techniques such as functional
tumors elsewhere in the body often fails to do MRI6 (fMRI) and intraoperative MRI7 enable
so in the CNS unless it destroys vital normal the surgeon to excise tumors more radically
CNS tissue along with the tumor. Most CNS because brain structures vital for CNS function
tumors are either intrinsically resistant to (e.g. motor, sensory, visual and language areas)
chemotherapeutic agents or develop resistance can be accurately identified both prior to and
through genetic instability during treatment. during surgery, allowing more extensive and
Adding to the publics concern is evidence safer resection. These techniques are particu-
that the incidence of several brain tumors is larly important because, for most CNS tumors,
increasing. Brain tumors, long the second most surgery is the most effective treatment. New
common childhood cancer after the leukemias,1 techniques of radiation therapy such as three-
are now more common than acute lymphocytic dimensional conformal radiotherapy, radio-
leukemia and may soon surpass all leukemias surgery and fractionated stereotactic
as the most common childhood cancer.2 radiotherapy (FSRT) allow a more potent
Malignant brain tumors also appear to be attack on tumors while sparing normal CNS
increasing in adults, particularly in the elderly.3 tissue. New chemotherapy agents and novel

3
CLASSIFICATION, INCIDENCE AND ETIOLOGY OF INTRACRANIAL TUMORS

Figure 1.1
The general appearance and common
location of several intracranial
tumors, discussed in this book, are
B
I illustrated in this schematic: (A) A
glioma of the anterior corpus
callosum (Chapter 5). (A') A glioma
of the brainstem (Chapter 5). (B) A
A meningioma compressing but not
H G invading the brain (Chapter 6). (C)
D
A medulloblastoma involving the
vermis of the cerebellum (Chapter 7).
F (D) A pinealoma compressing the
tectum of the brainstem (Chapter 8).
A' C
(E) A chordoma of the clivus
E compressing the brainstem (Chapter
9). (F) A pituitary adenoma (Chapter
10). (G) A lymphoma involving the
splenium of the corpus callosum
(Chapter 11). (H) A colloid cyst of
the Foramen of Monro. (I) A
metastasis to the brain (Chapter 13).

combinations of established agents have The term brain tumor, as most physicians use
demonstrated efficacy in certain brain tumors. it, is a misnomer. Most of the time, when we
Preliminary studies of gene therapy and anti- use the term brain tumor we mean, instead,
angiogenesis factors have generated much intracranial tumor (Fig. 1.1). Thus, menin-
excitement, but still no cures. Thus, although giomas that compress but rarely invade the
CNS tumors remain largely intractable, brain are considered to be brain tumors, as are
survival has improved,89 particularly in tumors of the pituitary and pineal glands,
children2 but also in adults. which are not strictly part of the brain,
This book is divided into two major sections. but reside within the intracranial cavity.
Part I (Chapters 14) discusses the general Furthermore, intracranial tumors can be classi-
principles of diagnosis and treatment of CNS fied into two major groups (Table 1.1): newly
tumors. Part II (Chapters 513) discusses diagnosed tumors that arise de novo within the
specific intracranial tumors. The reason for this intracranial contents (primary CNS tumors)
division, despite the inevitable redundancy, is number about 30 000 in the United States each
that many of the principles of diagnosis and year; tumors that spread to the intracranial
treatment and many of the elements of funda- contents from a systemic cancer (metastases)
mental biology of CNS tumors apply to all number about 100 000 in the USA each year.
CNS tumors. Space considerations force us to In this book, the terms CNS and intracranial
concentrate only on major topics. Several much are used interchangeably; the much less
longer and, thus, more comprehensive books common spinal tumors (also CNS) are not
have been published recently.1014 considered.

4
CLASSIFICATION

Table 1.1
Site of intracranial tumors.

Tumor type Site % of Example(s)


Primary
brain tumors
(approximate)b

I Primary intracranial tumorsa (N = 30 000/year)


Neuroepithelial tumors Brain 3645 Glioma, medulloblastoma,
neurocytoma
Meningeal tumors Dura 2640 Meningioma
Pituitary tumors Pituitary 610 Adenoma
Nerve sheath tumors Cranial nerves 47 Acoustic neuroma
Pineal region tumors Pineal, suprasellar cistern 0.3 Germinoma
Lymphocytic tumors Brain 13c Primary CNS lymphoma
Malformative tumors Meninges 2 Dermoid, epidermoid

II Metastatic intracranial tumorsd (N = 100 000/year)


Cells from any organ Brain, dura, meninges etc. Breast, lung, melanoma

aAfter WHO Classification15


bData from Radhakrishnan16 and CBTRUS17
cSee text

dFrom Posner13 and Greenlee et al18

Primary intracranial tumors can arise from making the tumor less than benign. Brain
virtually any cell type (or its precursor)17 found tumors are rarely truly malignant, in the sense
within the intracranial contents (Fig. 1.1); of most systemic cancers, because they seldom
metastases can reach the intracranial contents metastasize to other organs. However, they can
from cancers of any organ or tissue. Although grow rapidly to destroy important surrounding
primary brain tumors are often classified as normal tissues, extensively infiltrate the brain,
benign or malignant, a classification by and may seed virtually the entire neuraxis
histologic grade (low-grade/high-grade) is via cerebrospinal fluid (CSF) pathways. By
preferable: Tumors that arise within the contrast, metastases to the brain are truly
parenchyma of the brain are rarely truly malignant tumors (Chapter 13).
benign, because surgery seldom cures and
many that begin as low-grade tumors become
more biologically aggressive over time. Most
tumors arising outside the parenchyma of the
Classification
brain, such as meningiomas and pituitary The World Health Organization (WHO) classi-
tumors, can be considered benign because they fies CNS tumors by their patterns of differen-
are often cured by surgery, although, at times, tiation and presumed cell of origin15 (Table
complete resection is not technically feasible, 1.2).

5
CLASSIFICATION, INCIDENCE AND ETIOLOGY OF INTRACRANIAL TUMORS

Table 1.2
Histological classification of tumors of the CNS.

Tumors of neuroepithelial tissue Tumors of cranial nerves


Astrocytic tumors Schwannoma
Diffuse astrocytoma Neurofibroma
Anaplastic (malignant) astrocytoma Tumors of meninges
Glioblastoma multiforme Meningioma
Pilocytic astrocytoma Hemangiopericytoma
Pleomorphic xanthoastrocytoma Melanocytic tumor
Subependymal giant cell astrocytoma Hemangioblastoma
Oligodendroglial tumors Primary CNS lymphomas
Oligodendroglioma Germ cell tumors
Anaplastic (malignant) oligodendroglioma Germinoma
Mixed gliomas Embryonal carcinoma
Oligoastrocytoma Yolk sac tumor (endodermal sinus tumor)
Anaplastic oligoastrocytoma Choriocarcinoma
Ependymal tumors Teratoma
Ependymoma Mixed germ cell tumors
Anaplastic (malignant) ependymoma Cysts and tumor-like lesions
Myxopapillary ependymoma (spinal tumor) Rathke cleft cyst
Subependymoma Epidermoid cyst
Choroid plexus Dermoid cyst
Choroid plexus papilloma Colloid cyst of the third ventricle
Choroid plexus carcinoma Tumors of the sellar region
Neuronal and mixed neuronal-glial tumors Pituitary adenoma
Gangliocytoma Pituitary carcinoma
Dysembryoplastic neuroepithelial tumor Craniopharyngioma
Ganglioglioma Granular cell tumor
Anaplastic (malignant) ganglioglioma Metastatic tumors
Central neurocytoma From any primary source
Pineal parenchymal tumors
Pineocytoma
Pineoblastoma
Tumor of intermediate differentiation
Embryonal tumors
Medulloblastoma
Primitive neuroectodermal tumor (PNET)

*Abridged and modified from the WHO classification.15

For clinical purposes, it is often useful to in origin, are discussed together (Chapter 8), as
classify a tumor by intracranial site, as in Fig. are tumors of the pituitary and suprasellar
1.1, as well as by cell of origin. Thus, in this regions (Chapter 10). Tumors considered by
book, tumors of the pineal region, whether the WHO to be embryonal are discussed in
neuroepithelial (e.g. pineocytoma) or germ cell Chapter 7, along with neuronal tumors,

6
INCIDENCE OF INTRACRANIAL TUMORS

because the major embryonal tumor, the be more common than previously believed, an
medulloblastoma, often expresses neuronal important observation because these tumors are
protein markers. chemosensitive and require different treatment
Approximately 70% of symptomatic primary (Chapter 5). Their increase in relative frequency
CNS tumors arise within the parenchyma of the among gliomas probably reflects changing
brain; although their exact lineage is unknown, pathologic criteria19 rather than a true change
they are believed to be of neuroepithelial origin, in incidence. Primary CNS lymphomas are
primarily from glial cells (usually astrocytes) or probably underestimated as a percentage of
their precursors. The remainder arise from brain tumors because of their rapidly increasing
meninges, pituitary or other cell types (e.g. incidence20 (Chapter 11).
meningeal cells, pituicytes, lymphocytes, germ
cells). That neuroepithelial tumors are among
the most common brain tumors is not surpris- Incidence of intracranial
ing because glia (from Greek for glue) consti-
tute 90% of brain cells. Glia include astrocytes
tumors
(from Greek for star and cell), oligodendro- The American Cancer Society (ACS) estimated
cytes (from Greek for few and tree) and the number of new brain and other nervous
ependymal cells (from Greek for to place over system cancers (the term used by the ACS) in
i.e. to line the ventricle). Neurons constitute less 2001 to be 17 200 (9800 males and 7400
than 10% of brain cells; they number 100 females),18 more than twice that of Hodgkins
billion and are mostly postmitotic in the adult disease and over half that of melanoma. These
CNS. They or their precursors are an uncom- figures do not include metastases or benign
mon source of CNS neoplasms. tumors. In 2001, primary CNS cancers killed
Occasionally, CNS tumors arise from cells approximately 13 100 persons. In women, the
not normally found in the nervous system. mortality caused by CNS cancers is about the
These include germ cell tumors, histologically same as that caused by uterine cancer. Brain
identical to those of the testis and ovary. tumors are the second leading cause of cancer
Intracranial germ cell tumors grow in or around deaths in children, the second leading cause of
the pineal gland and the suprasellar area cancer deaths in men aged 2039 and the fifth
(Chapter 8). Primary lymphomas of the nervous in women of that age.
system (Chapter 11) and metastases from Metastases to the CNS from a systemic (i.e.
systemic cancers (Chapter 13) can affect any non-CNS) primary cancer are far more
part of the CNS. Tumors can also arise from common than primary CNS tumors as a cause
faulty migration of embryonic tissues. These of disability and death. Exact data are not
include craniopharyngioma (Chapter 10) and available, but one estimate suggests that over
dermoid and epidermoid tumors (Chapter 12). 100 000 individuals a year will die having
As Table 1.1 indicates, meningeal tumors and suffered from symptomatic intracranial metas-
gliomas account for the majority of intracranial tases.13,21 CNS metastases usually appear late in
tumors. High-grade (malignant) gliomas, such the course of a patients cancer, but in a signif-
as glioblastoma multiforme and anaplastic icant number of patients CNS symptoms are
astrocytoma, make up the majority of gliomas. the presenting complaint. Thus, the physician
Recent evidence suggests that oligodendro- must always consider metastatic disease as a
gliomas and their anaplastic counterparts may possible cause of neurologic symptoms and

7
CLASSIFICATION, INCIDENCE AND ETIOLOGY OF INTRACRANIAL TUMORS

Table 1.3
Primary brain tumors by histological type in Rochester, Minnesota, 1950 to 1989.

Average age- and sex-adjusted incidence rate/100 000/year

All patients Symptomatic patients

Tumor type % of total Rate/100 000/year % of total Rate/100 000/year


All types 100 19.1 100 11.8
Malignant astrocytoma 18 3.6 26 3.3
Low-grade astrocytoma 7 1.3 8 0.9
Meningiomas 40 7.8 16 2.0

Modified from Radhakrishnan et al16

signs in any patient with cancer (Chapter 13), was 19.1/100 000 persons/year for the period
and include metastases in the differential 195089. This includes incidences of 11.8 for
diagnosis of intracranial mass lesions even in symptomatic tumors and 7.3 for asymp-
patients not known to have systemic cancer. tomatic tumors. Gliomas (including oligoden-
A major problem in epidemiologic studies of drogliomas and ependymomas, not included
brain tumors is ascertainment. Incidence in Table 1.3) represent 29% of all brain
figures are affected by the quality of the clini- tumors but 42% of symptomatic tumors
cal evaluation, record-keeping and autopsy (malignant astrocytomas 18% of all tumors
rates. The most complete epidemiologic studies but 26% of symptomatic tumors), menin-
come from the Mayo Clinic16 (Table 1.3). Table giomas 40% and pituitary adenomas 10%.
1.3 includes only astrocytomas and menin- As indicated previously, primary CNS
giomas; other tumors including pituitary lymphomas, which are said to represent 1%,
adenomas (Chapter 10) make up the remain- are probably underestimated with respect to
der of the 100%. current trends, as may be oligodendrogliomas.
This is because Olmstead County, The age-specific incidence rates for malignant
Minnesota where the Mayo Clinic is based, is astrocytomas were highest in the 7584-year
unique in that all medical records containing age groups.
clinical and pathologic diagnosis and surgical More recent but less complete data have
procedures in the community, including those been published from the Central Brain Tumor
kept by private physicians, nursing homes, Registry of the United States (CBTRUS) that
and chronic care facilities, are indexed includes incidence data from 11 state cancer
through a centralized diagnostic registry. registries recording newly diagnosed cases of
Thus, although the population of the county benign and malignant primary brain
is small, the data are complete and include tumors.17 The total number of patients in the
both clinical and autopsy data. Those data registry is over 40 000 (Table 1.4). The overall
indicate that the age- and sex-adjusted incidence rate of 12.73 is similar to that of
incidence rate for primary intracranial tumors symptomatic patients from the Mayo Clinic

8
INCIDENCE OF INTRACRANIAL TUMORS

Table 1.4
Primary brain and CNS tumor incidence rates by major histology groupings and sex, CBTRUS 199297.

Histology Male patients Female patients Total


rate/105/year rate/105/year rate/105/year
(adjusted 2000)

Neuroepithelial tumors 7.50 5.20a 6.25


Cranial and spinal nerve tumors 0.90 0.87 0.89
Meningeal tumors 2.39 4.36a 3.45
Lymphomas and hematopoietic tumors 0.53 0.24a 0.38
Germ cell tumors and cysts 0.11 0.04a 0.07
Sellar region tumors 0.94 0.84 0.87
Skull base tumors 0.03 0.02a 0.03
Unclassified tumors 0.83 0.76 0.79
Total 13.23 12.33a 12.73

ap < 0.05 for difference between male and female patients.


Modified from CBTRUS17

study. This is expected, as state cancer Factors affecting incidence of


registries are unlikely to include patients intracranial tumors
whose diagnosis was made at autopsy. The 14
state cancer registries represent a population The incidence of intracranial tumors in general
of over 60 million people. and of specific histologic types of intracranial
The National Cancer Data Base collects data tumors differs by racial and ethnic group, sex,
from hospital tumor registries for both benign age, geography and even social class.24
and malignant brain tumors. It now contains Differences in brain tumor incidence by race
data from over 60 000 persons with primary and ethnic group include the findings that Jews
brain tumors diagnosed between 198589 and now living in Israel who were born in Europe
199092.22 The data are similar to those of and America have a higher incidence of brain
other databases. tumors than those now living in Israel who
A recent population-based study of all brain were born in Africa or Asia. The overall
scans in two counties in England yielded incidence of brain tumors (especially gliomas)
a primary brain tumor incidence of 21.04/ is greater in whites than blacks. However,
100 000 person years. About 1/5 of patients meningiomas are more frequent in blacks than
were not hospitalized. The incidence of neurep- in whites.24 Pituitary adenomas are also more
ithelial tumors was 9.83 and of meningiomas common in blacks than in whites.
3.99. The sellar tumor incidence was also 3.99; Sex differences are striking. The male to female
the incidence of cranial nerve tumors was ratio is 1.7 for oligodendrogliomas, 1.6 for astro-
2.38.23 Because scans are more easily available cytomas and 1.0 for malignant meningiomas. For
in the USA, such a study here might find a benign meningiomas, the female to male ratio is
higher incidence. 1.5 for intracranial meningiomas but 3.5 for

9
CLASSIFICATION, INCIDENCE AND ETIOLOGY OF INTRACRANIAL TUMORS

spinal meningiomas. Nerve sheath tumors have meningiomas and pituitary adenomas, confirm
an equal sex ratio. Lymphomas and germ cell the SEER findings.
tumors are more common in males. These data CNS tumors can occur at any age. Both the
are from the National Cancers Institutes (NCI) overall incidence and the histologic type of
SEER registry (Surveillance, Epidemiology, and intracranial tumors vary by age. Overall, there
End Results), which collects incidence and is a small peak before age 10 and a steady rise
survival data on malignant tumors from selected from 15 on. Some data indicate that intracra-
cancer registries across the USA (http://www- nial tumor incidence flattens or even falls after
seer.ims.nci.nih.gov/Publications). The CBTRUS age 75 (Fig. 1.2) but this finding may result
data17 that include benign tumors, e.g. from less vigorous evaluation of elderly

Figure 1.2
a) (a) Age-specific incidence
180 All tumors 180 Meningioma A rates of primary
Rate/100 000/year

160 160
140 B intracranial neoplasms in
100
60 Rochester, Minnesota
80
between 1950 and 1989.
60 30 All primary intracranial
40 20 tumors are indicated by
20 10 the red circles. The green
0 0 circles indicate those
Glioma 30 Malignant astrocytoma tumors that were identified
Rate/100 000/year

in clinical evaluation of
20 symptomatic patients.
Note the fall-off in
clinically diagnosed tumors
10
at extreme old age. From
Radhakrishnan et al16 with
0 0 permission. (b) Average
4 4 4 4 4 4 4 4 + 4 4 4 4 4 4 4 4 + annual age-specific
0-1 15-225-335-445-5 55-665-775-8 85 0-1 15-225-335-445-5 55-665-775-8 85
incidence of intracranial
Age groups Age groups
tumors in white men and
b) women in Los Angeles
40
County from 1972 to
Number of cases (per 100 000)

1993. Note in these


20 10 000 patients the
16 leveling in incidence of
12 tumors in old age. From
Preston-Martin25 with
8
permission.
6

4 Male
Female

2
0-14 15-24 25-34 35-44 45-54 55-64 65-74 75+
Age

10
INCIDENCE OF INTRACRANIAL TUMORS

Table 1.5
CNS tumor incidence per 105 person-years by age at diagnosis.

Age at diagnosis (years)

Histology 019 2034 3544 4554 5564 6574 7584 85+

Total tumors 3.69 5.67 9.50 15.78 24.92 36.45 39.81 31.55
Glioblastoma 0.17 0.41 1.21 3.81 8.16 12.34 11.22 5.41
Meningioma 0.10 0.64 2.13 4.35 6.60 11.50 14.70 14.30
Lymphoma 0.01 0.26 0.47 0.41 0.65 1.09 1.22 0.47

Modified from CBTRUS17

patients with neurologic disability. Low-grade Glioblastoma peaks between ages 65 and 74
gliomas, such as astrocytomas, are more and declines slightly after age 75. The menin-
common in the young, and high-grade tumors, gioma rate also increases with age and does not
such as glioblastoma, are more common in decline after age 75 (Table 1.5).
the elderly. Medulloblastomas and germ cell Reported brain tumor incidence also varies
tumors of the pineal region are tumors of child- by geography. The most developed countries
hood. SEER data from 19731991 report only report higher rates of primary brain tumors
208 glioblastomas under age 20, compared to than less developed nations. The age adjusted
3 479 over age 65. Conversely, pilocytic astro- incidence in Scandinavia is reported to be 31.4
cytomas, a low-grade tumor (Chapter 5), was per million, significantly higher than the US
more common under age 20 (n = 252) than rates of 21.7 for blacks and 26.4 for whites. In
over age 65 (n = 7). There were 578 medullo- the USA, Canada, western Europe and
blastomas under age 20 and only 3 over age Australia, the rates are similar and greater than
65. Pineal region tumors numbered 25 under those of eastern Europe. The lowest incidences
age 20 and only 1 over age 65.26 The lifetime among developed countries are in Japan, India
risk of a CNS malignancy is 0.67% for men and Singapore.24 Even though the overall
and 0.52% for women (SEER). incidence is low in Asia, specific tumors are
A recent population study from Japan27 more common. For example, germ cell tumors
compared the incidence of intracranial tumors are much more common in Japanese boys than
in those older and younger than age 70. in any other group in the world (Chapter 8).
Between 1989 and 1995, 1354 new primary Migrant populations usually have higher rates
intracranial tumors were diagnosed in that are closer to those of natives of the
Kumamoto, Japan. The overall age-adjusted adoptive country than those who remain in
incidence was 18.1/105 for those older than 70 their country of origin, suggesting that environ-
and 8.7/105 for those younger. The CBTRUS mental factors are important. How geographi-
data17 show an increasing incidence in all cal differences are affected by diagnostic
intracranial tumors with age; the highest facilities and autopsy rates in individual
rate is in the 7584-year-old age group. countries is not known.

11
CLASSIFICATION, INCIDENCE AND ETIOLOGY OF INTRACRANIAL TUMORS

The reported incidence of intracranial 197592. There was a decrease in astrocytomas


tumors varies by social class (in some studies but a sharp increase in both anaplastic astrocy-
defined by occupation).24 In several studies, the tomas and glioblastomas. Although it is possible
incidence of brain tumors increased with social that the decrease in astrocytomas and increase in
class, this being more evident in men than in anaplastic astrocytomas could be accounted for,
women. The differences between men and in part, by differences in pathologic interpreta-
women would seem to rule out ascertainment tion, it is unlikely that the same could be said
as an explanation, as both sexes within each for glioblastomas, where interpretation is fairly
class should have equal access to medical care. uniform among neuropathologists. A study using
The explanation of social class differences, if data from the French Cancer Registry from 1983
real, is not known. to 1989 indicates that malignant astrocytomas
increased by 5% a year in individuals over age
65.31
Increasing incidence? A study of glial tumors identified in New
An important question is whether the incidence York State between 1976 and 1995 revealed an
of brain tumors in general and certain specific increasing incidence of glioblastomas and
tumors in particular is increasing. Some anaplastic astrocytomas, especially in those
evidence suggests that the incidence is increas- over age 60. The reason for these statistically
ing, making most published data that consist of significant increases is not clear.32
information from previous years unreliable with Not all studies support an increasing
respect to the present situation.23,2830 For incidence.5,16 Furthermore incidence rates that
example, a study using data from the Florida were increasing seem to have stabilized.33
Cancer Registry29 comparing brain tumor Moreover, several problems complicate the
incidence from 198184 with the incidence epidemiologic data that appear to indicate that
from 198689 indicates a consistently signifi- brain tumors are increasing in both adults4 and
cant increase in all primary brain tumors in children.1 In adults, the major problem is
patients over age 65, with the largest increase ascertainment.16 For example, studies of brain
in those over 85. An incidence density ratio tumor incidence based on autopsy data differ
(IDR or risk ratio) was calculated by dividing substantially from those based on clinical
age-adjusted incidence for the period 198689 evaluation. The differences occur in part
by the corresponding age-adjusted incidence for because many brain tumors, particularly small
198184. When specific histologic types were meningiomas and pituitary tumors, are
examined, those patients aged 2064 showed a common but often asymptomatic and, thus,
statistically significant increase with an IDR represent only incidental findings at autopsy.
greater than 3 for both anaplastic astrocytomas Furthermore, diagnosis even of clinically
and primary CNS lymphomas. Those over 65 symptomatic patients, particularly those who
showed a statistically significant increase for are elderly, is often incomplete. Prior to the
anaplastic astrocytomas (IDR = 2.7), glioblast- development of non-invasive brain imaging by
omas (IDR = 1.1) and lymphomas (IDR = 3.4) computed tomography (CT) and MR scanning,
as well as total brain tumors. many elderly patients with neurologic dysfunc-
Olney et al,30 using the NCI SEER data tion were believed to have had strokes or
compared year-by-year incidence of astrocytomas, dementing illnesses and were not further evalu-
anaplastic astrocytomas and glioblastomas from ated. We still encounter elderly patients who

12
INCIDENCE OF INTRACRANIAL TUMORS

Figure 1.3
Failure to diagnose a brain tumor in an elderly woman. A 70-year-old woman presented with new-onset
headache. A non-contrast MR T2 weighted (left image) scan revealed a right temporal lobe lesion (arrow)
interpreted as representing ischemic vascular disease in this elderly patient. The headaches resolved, but when
she developed personality change, confusion and weakness several months later, a contrast-enhanced MR scan
(right image) clearly identified the previously small lesion as a now large glioblastoma (arrow).

suffer a sudden onset of neurologic symptoms as the general attitude toward the diagnosis
and in whom a non-contrast CT scan or, more and treatment of neurologic illness in those of
rarely, an unenhanced MRI is misinterpreted as advanced age has changed, accurate diagnosis
cerebral infarction. Cerebrovascular disease is has become more complete, making it clear
far more common than brain tumors (Fig. 1.3) that the incidence of primary brain tumors
and because a contrast study may have been increases with advancing age. Whether the
omitted either to save costs or to save the increase continues into very advanced age is
patient side-effects, the true nature of the not yet established.
illness may not be discovered, and the patients Another consideration in evaluating the
death attributed to a stroke. If repeat imaging apparent increase in brain tumor incidence is a
with contrast is never performed, the correct change in the age of the population.4 As a
diagnosis is never established. This failure to population ages, its diseases change and the
identify tumors in the elderly has fostered the incidence of a particular disease in the popula-
belief that the incidence of brain tumors peaks tion may rise. Genetic differences in elderly
in late middle age and then declines in the individuals who might in the past have died of
elderly. As diagnostic techniques became less premature cardiac or cerebrovascular disease,
invasive, and therefore more widely used, and which are now treated effectively, may make

13
CLASSIFICATION, INCIDENCE AND ETIOLOGY OF INTRACRANIAL TUMORS

them more susceptible later in life to brain reports, is now a clinical consideration in every
tumors.4 For example, one study found that patient who presents with a mass lesion in the
individuals with a specific genetic variant of a brain. Furthermore, the relative increase of
hepatic metabolic gene were at increased risk primary to metastatic brain lymphoma decreases
for oligodendrogliomas.34 If that same variant the need, in appropriate clinical circumstances,
also conferred protection from heart disease for extensive, repeated searches for an extracra-
and systemic cancers, enabling long-term nial primary lymphoma (Chapter 11). Because
survival, there would be more brain tumors in the diagnostic approach and treatment of
the elderly. Furthermore, when this factor is primary CNS lymphomas differ from those of
combined with the generally better health of other brain tumors, the physician must consider
the elderly population and more vigorous lymphoma, even before the diagnosis has been
evaluation of neurologic complaints in these established. Such patients should be evaluated for
patients, the increase in brain tumors may not HIV infection or other immune-suppressing
reflect an environmental factor. The advent of illnesses, and diagnosed by needle biopsy rather
Medicare, giving many of the elderly better than craniotomy (Chapter 3).
access to medical care, may also play a role. A second clinical implication of the changing
Pediatric brain tumors also are reported to relative incidence of specific types of brain
be increasing in incidence.35 Data from SEER tumors relates to the apparent increase in the
were analyzed for children 14 years of age or relative incidence of oligodendrogliomas (see
younger between 1974 and 1991. There was an Chapter 5). The apparent increase in incidence
average 2% increase in incidence per year for almost certainly represents a change in the crite-
astroglial tumors. These increases were most ria that neuropathologists use for diagnosis:38
apparent among children less than 3 years neuropathologic diagnosis is subjective and
old.36 A recent report from Ontario, Canada based on morphologic interpretation of the
noted a small but significant trend toward tumor tissue. A recently described transcription
increasing incidence in persons under age 20.35 factor (OLIG2) may differentiate oligodendro-
Others interpret the data as showing a one time gliomas from astrocytomas, the tumors most
jump in incidence, suggesting that the appar- likely to be confused with each other.39 However,
ent increase represents either better detection because the treatment of oligodendrogliomas
methods, or more accurate reporting to tumor differs from the more common astrocytoma
registries.1,37 (Chapter 5), the clinician must work closely with
Important for clinicians is a change in the the neuropathologist to determine the correct
relative frequency of specific tumors. For diagnosis, that will dictate therapy to some
example, primary CNS lymphomas (lymphomas degree and predict prognosis. Luckily, genetic
that arise in the nervous system and are not markers may soon solve the problem.40
metastatic from systemic lymphoma) are increas-
ing both in absolute incidence and in relative
frequency when compared to other primary
brain tumors (Chapter 11).20 This increase is
Etiology
apparent in both the immunosuppressed (e.g. Many patients, when informed by their physician
AIDS) and immunocompetent populations. The that they are suffering from a brain tumor, ask
clinical result is that a tumor, once considered a What caused it? For the vast majority of
curiosity and the subject of individual case patients, there is no adequate answer. In a small

14
ETIOLOGY

number of patients one can identify environ- Table 1.6


mental factors that clearly cause or predispose Risk factors related to CNS tumors.3
toward the development of a brain tumor. In an
even smaller number of patients, inherited Definitive
genetic abnormalities are causal. For most Ionizing radiation41,42
patients, the physician cannot identify any factor Immune suppression
HIV infection43,44
that may have caused the tumor. Many patients,
Iatrogenic (e.g. organ transplant)
distressed over the diagnosis and our lack of Possible
knowledge concerning the cause, find this expla- Electromagnetic fields45
nation unsatisfactory and search for potential High tension wires46
causes. Some come up with what they believe to Cellular telephones4749
Diet50
be satisfactory explanations: these include the
N-nitroso compounds
use of cellular telephones, exposure to electro- Aspartame30
magnetic waves from power stations, head Occupation51
trauma, and various occupational or dietary Petroleum industry
exposures. Patients are often supported in their Agricultural pesticides52
belief by anecdotal reports in the medical liter- Household chemicals
Hair dyes and sprays
ature or by a single epidemiologic study.
Household pesticides52,53
Epidemiologic studies of risk factors for brain Head injury54,55
tumors are fraught with difficulty. Even when a Medications
large population is surveyed, the number of brain Vitamins56
tumors is relatively small and statistically sig- Infections
Cysticercosis57
nificant differences may not be reproducible.
Varicella-zoster58
Furthermore, many studies consider brain SV4059,60
tumors as a group and do not stratify by histol-
ogy. Stratification by histologic type is important
because there are undoubtedly different risk
factors for different tumor types, e.g. menin-
giomas compared to gliomas. Thus, two carefully (Table 1.6), only two unequivocal risk factors
done studies may yield conflicting results, one, have been identified.24
for example, suggesting that exposure to high- The first is ionizing radiation; the second is
tension wires increases the number of brain immune suppression. Low-dose irradiation to
tumors, and another showing no such risk. Until the scalp once given for the treatment of tinea
more than one study clearly confirms the risk, capitus, a fungal infection of the scalp, has
the physician should withhold judgment. The been shown to cause meningiomas,42 many of
paragraphs below consider the evidence for both which are anaplastic, and approximately a
environmental and familial risk factors. three-fold increase in the incidence of glial
tumors. Nerve sheath tumors of the head and
neck are increased an astounding 33-fold.61
Environmental risk factors
Higher-dose irradiation for intracranial
Although a large number of studies have tumors, e.g. medulloblastoma or extracranial
examined the relationship between the environ- head cancers, including prophylactic irradia-
ment and the occurrence of brain tumors tion for leukemia, increases the incidence of

15
CLASSIFICATION, INCIDENCE AND ETIOLOGY OF INTRACRANIAL TUMORS

both gliomas and sarcomas seven-fold in those immunosuppressed patient, it is twice as likely
who survive more than 3 years. The cumula- to occur in the brain as elsewhere in the body.
tive relative risk of secondary brain tumors in This is in contradistinction to the situation in
patients treated with cranial irradiation for the immunocompetent patient, where the brain
leukemia is 1.39 at 20 years; approximately is the primary site of lymphoma in only 1% of
two-thirds of the tumors are gliomas and one- patients. The question of increased glial tumors
third meningiomas.62 High-grade gliomas have in HIV-infected patients remains unsettled;
a shorter median latency (9.1 years) from the reports suggested that immune suppression is a
cranial radiotherapy than do meningiomas (19 risk factor for glial tumors44,67 but the incidence
years). The lower dose of prophylactic radia- is not sufficiently high to classify glioma as an
tion now used for leukemia will probably AIDS-defining illness.
decrease, but not eliminate, the incidence of Other studies of environmental risk factors
secondary brain tumors. In one series, the use are substantially less convincing than those of
of antimetabolites during radiation therapy ionizing radiation and immunosuppression.
(RT) may have increased the number of brain Some investigators report that industrial
tumors.63 Prenatal radiographs may predispose exposure to polyvinyl chloride or dietary
to childhood brain tumors. Dental radiogra- exposure to N-nitrosourea compounds could
phy55 and cosmic rays do not appear to be risk be risk factors50 but others have failed to
factors, except for a possible increase in menin- substantiate these findings. Some evidence
giomas in patients given the high-dose whole- suggests that consumption of cured meats,
mouth radiographs used decades ago.64 which contain N-nitroso compounds, may not
Curiously, those dental X-rays appeared to only predispose to brain tumors in the
protect against the later development of consumer, but also in the children of mothers
gliomas. That study also showed a similar who consume the products during pregnancy.68
unexplained decreased risk for glioma associ- Some evidence suggests that vitamins and other
ated with exposure to amalgam fillings. Other antioxidants69 may protect against N-nitroso
studies have not found dental radiographs or compounds. Prenatal or early childhood
amalgam to be either risk or protective vitamin intake may protect children against
factors.65 brain tumors, but the data are equivocal.56
Acquired immune suppression, such as HIV Fruit and vegetable consumption may decrease
infection66 or the use of immunosuppressive risk. Other studies have tentatively identified
agents after organ transplant, increases the high-protein diets and alcohol as risk factors,
incidence of primary lymphomas of the CNS; and a few studies have implicated parental
HIV infection may also increase the frequency exposure to these factors in causation of
of gliomas and intracranial leiomyosarcomas.43 childhood brain tumors; the data are not
Congenital immune-suppressive illnesses such compelling. Thus, the role, if any, of nutrition
as the WiskottAldrich syndrome are also in either causing or protecting from brain
associated with an increased incidence of tumors remains unknown. As a result, the
cerebral lymphomas. physician is unable to give patients evidence-
Primary CNS lymphomas in immunosup- based advice.
pressed patients are driven by pre-existing Head trauma55 has been reported as an
latent EpsteinBarr viral infection of B-lympho- environmental risk factor for the development
cytes. When a lymphoma occurs in an of glial tumors and meningiomas but the

16
ETIOLOGY

evidence is unconvincing. One study relates gliomas. The cytochrome P-450 family of liver
acoustic trauma to the later development of enzymes are required to metabolize drugs and
acoustic neuromas (Chapter 9), but more other toxins. Many of the enzymes are
studies are required to be certain. polymorphic, with some forms metabolizing
There has been recent concern in the lay substances faster than others. There may be
population that exposure to electromagnetic an increased risk of oligodendrogliomas in
radiation, including high tension wires, individuals who carry a poor metabolizer
computer terminals, and cellular telephones CYP2D6 variant allele (allele; from the Greek
may cause brain tumors. Although cellular for reciprocally the same gene location on
telephones cause headache,70 and may cause paired chromosomes) and the GSTT1 null
death by increasing automobile accidents, genotype.34 The role, if any, of individual
evidence suggests that they do not cause brain genetic differences in systemic enzymes in
tumors.47,48 One doubts that we are going to predisposing to brain tumors is not clear.
see an epidemic of left temporal tumors whose In other studies, hair dyes, pesticides,
incidence is related to telephone conversa- formaldehyde, and industrial or occupational
tions. substances have all been implicated as a cause
Olney and colleagues30 have proposed that of brain tumors, but in none has the hypothe-
the recent increase in malignant brain tumors sis been confirmed. If any of these environ-
may be due to ingestion of aspartame, a dipep- mental factors are true risk factors, each can
tide sugar substitute consisting of phenylala- be responsible for only a small proportion of
nine and aspartic acid. Both of these amino brain tumors, because only a small percentage
acids are known to be active in the CNS but of patients have been exposed to such
the evidence that they are a risk factor for substances.
brain tumors remains weak for at least three Prior infection has also been reported to
reasons: (1) brain tumors appear to be increas- play a role in increasing or decreasing brain
ing predominantly in the elderly, the group tumors. A recent report indicates that cysti-
least likely to use sugar substitutes; (2) these cercosis infection increases the incidence of
drugs have only been on the market for a few gliomas.57 Although the study was carefully
years and one would think that if they were a done, the finding needs to be replicated.
risk factor for brain tumors, it would take Cysticercosis is now so common in some parts
decades for this association to become appar- of the USA that one would have expected to
ent; and (3) women use aspartame-containing see almost an epidemic of gliomas if the infec-
products more than men, but gliomas are more tion were an important predisposing factor.
common in men. The first and second reasons Some reports60 find SV40 large T-antigen
apply to cellular telephones as well. sequences at high frequency in gliomas and
A few medical illnesses have been proposed medulloblastomas; others do not.59 The role, if
as risk factors for brain tumors. Breast cancer any, of that virus in the etiology of brain
may predispose to meningioma (Chapter 6). A tumors is unclear. A recent study suggests that
past history of meningitis or epilepsy has also prior varicella-zoster infection, or absence of a
been reported to be associated with an serologic response to varicella-zoster with a
increased incidence of brain tumor. Some history of prior infection, protects against
studies have suggested that diabetes and aller- glioma.58 The mechanism is unclear and this
gic diseases protect against the development of study requires replication.

17
CLASSIFICATION, INCIDENCE AND ETIOLOGY OF INTRACRANIAL TUMORS

Genetic risk factors a specific type or types of brain tumor and


other malignancies. Because each mutation is
Inherited genetic alterations are predisposing found in every cell in the body, having been
risk factors for brain tumors (Table 1.7). These transmitted through parental DNA, the resul-
genetic alterations are usually germline tant brain tumor usually occurs as part of a
mutations that increase an individuals risk for syndrome that includes systemic cancers and

Table 1.7
Some hereditary syndromes associated with brain tumors.

Hereditary Mode of Type of Involved Gene Reference


condition Inheritance tumor(s) chromosomes

Li-Fraumeni AD Glioma 17p13 TP53 72


Medulloblastoma
Tuberous sclerosis AD Subependymal giant 9q34 TSC1 73
cell astrocytoma, 16p13 TSC2
cortical tubers
Glioma
Neurofibromatosis type 1 AD Glioma (optic nerve) 17q11 NF1 74, 75
(von Recklinghausens Astrocytoma,
disease) Glioblastoma
Neurofibromatosis type 2 AD Meningioma 22q12 NF2 76
Schwannoma (bilateral
acoustic neuroma)
Ependymomas
Multiple endocrine neoplasia AD Pituitary 11q13 Menin 77
type 1
Retinoblastoma AD Retinoblastoma 13q14 RB1 78
Basal cell nevus syndrome AD Medulloblastoma 9q22 PTCH 79
Turcot syndrome AR or AD Brain tumors of 5q21 APC 80
Hereditary nonpolyposis diverse histology, 2p16 HMLH2
colorectal cancer including glioblastoma 3p21 HMLH1
syndrome and medulloblastoma 7p22 h85M2
(HNPCC)
von HippelLindau disease AD Hemangioblastoma 3p2520 VHL 81
Cowdens syndrome AD Dysplastic cerebellar 10p23 PTEN 82
gangliocytoma, (MMAC1)
meningioma,
astrocytoma
Rhabdoid predisposition AD Choroid plexus 22q11 hSNFS/INI1 83
syndrome carcinoma,
medulloblastoma,
primitive neuro-
ectodermal tumors

AD, autosomal dominant; AR, autosomal recessive.

18
BIOLOGY OF INTRACRANIAL TUMORS

other malformations as well as CNS tumors investigations are necessary to clarify the
(Chapter 12). Although these syndromes etiology of brain tumors.
account for a small minority of brain tumors
(about 5% of gliomas are familial and about
1% have a possible autosomal dominant inher- Biology of intracranial
itance),71 the wide range of genetic abnormali-
ties that can lead to CNS neoplasms suggests
tumors
that there may be many genetic avenues that At their core, all tumors have a genetic origin.85
can cause these tumors. Furthermore, the study In familial tumors, the defect is in the germ
of familial syndromes, where genetic abnor- line; thus, the same genetic mutation is trans-
malities are more easily identified, may aid in mitted to all the somatic cells of the carriers
finding similar defects in the more common offspring. Acquired tumors begin when an
sporadic brain tumors. environmental factor alters a gene, usually in a
single somatic cell. However, a single genetic
Interacting genetic and abnormality is insufficient to cause the altered
cell to become a neoplasm. A series of genetic
environmental factors
changes is required before the cell can exhibit
It is likely that for the vast majority of patients unrestrained growth, outstrip the reproductive
no single genetic or environmental factor is rate of its neighboring normal cells and develop
sufficient to explain the development of a brain into a tumor. The large number of genetic
tumor. Interactions among multiple factors mutations in a cancer cell may result from an
may be necessary. For example, N-nitroso early mutation in a gene that is required for the
compounds clearly cause brain tumors in exper- maintenance of genetic stability.86 However,
imental animals. These compounds are found in genes need not be mutated to dysfunction. For
the human diet in bacon, cured meats and other example hypermethylation of a DNA promo-
foods. Their role, if any, in the production of tor region can disrupt gene function.87 Data
human brain tumors may be modified by the from many cancers indicate that every tumor
concomitant ingestion of vitamins and foods expresses a large number of genes not
containing antioxidants that may decrease the expressed in its counterpart normal tissue.
carcinogenic properties of the N-nitroso Although the functions of most of these
compounds. In addition, genetic factors may uniquely expressed genes are not known, most
play a role. For example, the genetically deter- of their protein products appear to be involved
mined presence of the enzyme O6-methylgua- either in regulating the cell cycle or regulating
nine-DNA methyltransferase (MGMT or AGT) how the cell responds to environmental factors
is important in repairing DNA abnormalities that promote differentiation, maturation and
caused by N-nitroso compounds.84 A patient normal cell death, i.e. apoptosis. Apoptosis
with a deficiency of this enzyme and poor (from the Greek apo for off plus ptosis for a
intake of antioxidants may be more susceptible falling) is a normal process of cell death. The
to N-nitroso compounds than another patient process can occur as programmed cell death
with a good intake of antioxidants and a that occurs in embryonic development or it can
relative excess of the enzyme. It will be very be induced by stress signals arising within the
difficult for epidemiologists to dissect all of cell or by signals elicited by binding of death
these multiple interacting factors, but such ligands to the cell. Apoptosis, as opposed to

19
CLASSIFICATION, INCIDENCE AND ETIOLOGY OF INTRACRANIAL TUMORS

necrosis requires energy in order to synthesize


proteins to initiate the process. Cells suffering
damage to their DNA that cannot be repaired a) Wild-type
normally undergo apoptosis. Failure of the
normal apoptotic process allows cells to
accumulate further DNA abnormalities that
can lead to the development of cancers. Several
genes altered in cancer are involved either in
initiating or inhibiting apoptosis. For example,
as indicated below, the p53 tumor suppressor
gene, and possibly the related genes p63 and
p73,88,89 have as one of their functions the
initiation of apoptosis in a cell whose DNA
damage is irreparable. Genetic abnormalities,
such as mutation or deletion of p63 or related
genes, prevent apoptosis and permit survival of b) Amplified wild-type
damaged cells. Apoptosis is also necessary for
radiation and chemotherapy to kill tumor
cells.90 Thus, failure of apoptosis protects the
cancer cells from death by cancer therapies.
Two general kinds of genetic alteration
usually cause CNS and other tumors. The first
is a mutation that inappropriately activates a
proto-oncogene. The second is a mutation that
inactivates a tumor suppressor gene. Proto-
oncogenes are normal cellular genes that
regulate cell proliferation, differentiation and
apoptosis. Proto-oncogenes can encode growth
factors, e.g. basic fibroblastic growth factor
c) Amplified truncation
(bFGF) or growth factor receptors, e.g. epider-
mal growth factor receptor (EGFR), or mediate

Figure 1.4
An example of growth factor receptor abnormalities
in a glioblastoma multiforme. (a) shows the wild-type
epidermal growth factor receptor. The ligand (red
circles) binds to the receptor, and forms a dimer, and
signals to the cells to grow. In (b), when the normal
receptor is amplified, excessive growth occurs. (c)
shows a mutated receptor. The receptor no longer
responds to circulating growth factors, but instead is
constitutively active, signaling unrestrained growth
downstream. Modified from Kleihues and Cavenee15
with permission.

20
BIOLOGY OF INTRACRANIAL TUMORS

signaling pathways, e.g. protein kinase C


(PKC), or regulate gene expression, e.g. c-myc.
Proto-oncogenes become oncogenes by gene Juxtacrine
mutation, gene amplification, gene overexpres- Nucleus Nucleus
sion or chromosomal translocation. Mutations
such as those that occur in the EGFR may lead
to a receptor that is always turned on (consti-
Intracrine
tutively active) and thus not subject to normal
cellular control mechanisms (Fig. 1.4). Gene
amplification refers to the expansion of the
copy number of a gene within the genome. The Autocrine Paracrine Extracellular
process can occur through replication of matrix
genomic DNA or by homogenous staining Endocrine
regions or double minute chromatin bodies
(pieces of extra chromosomal material) leading Blood vessel

to increased expression of genes. Some brain


tumors amplify EGFR; others amplify myc Figure 1.5
genes. Chromosomal translocations may Examples of different modes of action of growth
activate proto-oncogenes. These abnormalities factors. Growth factors secreted by an individual
are often present in hematologic malignancies, tumor cell can stimulate the cell through either its
surface receptor (autocrine cell on left), or an
including primary CNS lymphomas, and are internal receptor (intracrine cell on right). The
found in some glioblastoma cell lines91 but do growth factor can stimulate neighboring cells by
not appear to play an important role in the direct contact (juxtacrine), or by diffusing to nearby
development of brain tumors. Overexpression cells (paracrine), or the growth factor can be secreted
of proto-oncogenes without gene amplification into the systemic circulation to affect cells at a
distance (endocrine).
plays an important role in many brain tumors.
An example is platelet-derived growth factor
(PDGF). Either the ligand, which is homolo-
gous to thymidine phosphorylase and thus stimulated to grow, even in the absence of the
stimulates DNA synthesis, or the receptor receptor ligands, transforming growth factor
(PDGFR), or both, may be overexpressed. alpha (TGF-) and/or epidermal growth factor
(EGF) (Fig. 1.4).
Alterations of growth factors can lead to
Oncogenes
unrestrained growth not only of the cells
Oncogenes function in an autosomal dominant possessing the alteration but of nearby cells
fashion, meaning that mutation of only one and sometimes distant cells by autocrine,
allele is necessary to transform the cell; paracrine or endocrine stimulation (Fig. 1.5).
oncogenes exert their malignant action through One example of autocrine stimulation occurs
a gain of functional activity. One good example when TGF- stimulates EGFRs on the same
is the mutation of EGFR that occurs in 40% cell from which the TGF- is secreted.
of glioblastomas. An in-frame deletion leads to Sometimes the growth factor does not have to
a truncated protein that is constitutively active. be secreted but can stimulate the receptor from
The cells possessing this deletion are constantly within the cell, intracrine stimulation. One

21
CLASSIFICATION, INCIDENCE AND ETIOLOGY OF INTRACRANIAL TUMORS

example of paracrine stimulation is when Many of the growth factors altered in brain
vascular endothelial growth factor (VEGF) tumors exert their function through reversible
secreted by some brain tumor cells stimulates phosphorylation of tyrosine by tyrosine
receptors on nearby endothelial cells, causing kinases.92 Tyrosine kinases play a major role in
the endothelial cells to form new blood vessels both normal and neoplastic growth and cell
(angiogenesis; from Greek for vessel and cycle control as well as in differentiation.
production) (Chapter 2). Endocrine stimula- Growth factor receptors important in brain
tion does not appear to play an important role tumors that function as tyrosine kinases
in many brain tumors. One possible example is include the EGFR, VEGF, PDGFR and fibro-
TGF-, which is secreted by glioblastoma cells blast growth factor receptor (FGFR).93 For
into the bloodstream and may cause the example, overexpression of the -chain of
immune suppression that accompanies many PDGFR can cause brain tumors in mice94 (Fig.
glioblastomas. 1.6).

PDGF

PDGF receptor Plasma membrane

Ras Src
P Y579 Y579 P
P Y581 Y581 P
Sos
Slap
DAG + IP3
p110
G

Erk MAP kinase P P


rb

Y716 Y716
PI3-kinase
2

P Y740 Y740 P
PIP3 PKC
Nck P Y751 Y751 P

Pak GAP P Y771 Y771 P Grb7


PI3,4,5 P2

Fos
Shp2 Y1009 Y1009
Myc Akt
Y1021 Y1021 PLC

Catalytic domain

Figure 1.6
The PDGF receptortyrosine kinase signaling pathway. Both PDGF and its receptor are over-expressed in many
brain tumors. Via autocrine or paracrine stimulation, the excessive activity of the PDGF- receptor triggers a
complicated pathway leading to transcription of genes that increase cellular proliferation. From Hunter92 with
permission.

22
BIOLOGY OF INTRACRANIAL TUMORS

Table 1.8
Some proto-oncogene abnormalities found in brain tumors.

Gene Chromosomal location Mechanism of activation

Oncogene EGFR 7p12 Amplification, rearrangement


PDGFR-A 5q31q32 Rearrangement, amplification
bFGF 4q25 Overexpression
IGF-1/IGF-1R 12q23; 15q26.3 Overexpression
Ros-1 6q22 Overexpression
H-ras, N-ras 11p15; 1p13 Overexpression, point mutation
c-myc, N-myc 8q24; 2p23p24 Amplification
gli 12q13q14 Amplification
met 7q31 Overexpression
gsp 20q12q13.2 Point mutation

Modified from Israel95 with permission.

More than 100 different oncogenes have been suppressor genes produce proteins that are
discovered, most in experimental animal models involved in inhibiting cell growth or promoting
of cancer. Some of these believed to be important cell differentiation. Unlike oncogenes, most
in human brain tumors are detailed in Table 1.8. tumor suppressor genes function in an autoso-
mal recessive manner by a loss of function after
both alleles are disabled. Many tumor suppres-
Tumor suppressor genes sor genes have been identified. Tumor suppres-
Tumor suppressor genes, the opposite of sor genes important in brain tumors include
oncogenes, comprise the second common p5396 (see below), PTEN/MMAC197 and
genetic alteration (Table 1.9). Normal tumor CDKN2 A and B.98

Table 1.9
Gene or locus Chromosomal location Tumor type Tumor suppressor
genes in brain tumors.
P53 17p13.1 Astrocytoma
Glioblastoma
PTEN/MMAC1 10q23 High-grade gliomas
NF1 17q11.2 Pilocytic astrocytoma
RB1 13q14 Glioblastoma
CDKN2 A and B 9p21 Glioblastoma
APC 5q21 ? Medulloblastoma
Gorlin locus 9q31 Medulloblastoma
MEN1 locus 11q13 Pituitary adenoma
NF2 22q12 Meningioma

Modified from Israel95 with permission.

23
CLASSIFICATION, INCIDENCE AND ETIOLOGY OF INTRACRANIAL TUMORS

usually bilateral: one allele is already absent in


every cell because of the germline mutation and
because single hits are relatively common, at least
one cell in each eye is likely to suffer a mutation
of the remaining allele. Non-familial retinoblast-
omas are usually unilateral because two hits in
the same cell are rare.
Not all tumor suppressor genes require
deletions of both alleles in order to cause
tumors. Probably the most common tumor
suppressor gene abnormality in cancer involves
the p53 gene. This gene encodes a 53-kDa
protein that modifies cellular function, includ-
A B C ing the cell cycle, DNA repair after radiation
damage, genomic stability and induction of
Figure 1.7 apoptosis. The protein product of the p53 gene
Knudsons two-hit therapy of oncogenesis due to loss prevents progression of the cell cycle beyond
of tumor suppressor gene function. (A) A normal the G1/S checkpoint when damaged DNA is
somatic cell chromosome pair with two wild type
detected. If the DNA can be repaired, the cell
alleles for a tumor suppressor gene. (B) Typical
somatic cell chromosome pair in a patient with one cycle proceeds; if not, p53 directs the cell to die
wild type allele (black band) and one germ line (apoptosis). If p53 does not function normally,
mutant non-functioning allele (empty band). The cell cells with abnormal DNA can reproduce
has a normal phenotype. (C) Chromosome pair from uncontrollably, causing a neoplasm. Some
a tumor cell arising in a patient with an inherited mutations of the p53 tumor suppressor gene on
mutated non-functioning allele and a partial deletion
of the chromosome on which the tumor suppressor
one allele can cause tumors through a so-called
gene is located. Neither allele of the tumor dominant negative mutation.100 These p53
suppressor gene can function and a tumor develops. mutations lead to the synthesis of a defective
p53 protein tetramer (the active form of the
molecule) in which the mutated protein renders
the entire complex inactive.
Many of the abnormal or deleted genes in PTEN gene mutations are common in high-
familial tumors are tumor suppressor genes.99 The grade gliomas. The protein product of the
loss of a germline tumor suppressor gene on one gene, a protein tyrosine phosphatase, probably
chromosome may not cause a cancer. However, if negatively regulates cell proliferation by
through environmental factors a cell having only inhibiting protein tyrosine kinase activity.97
one normal allele undergoes a genetic mutation Some genetic abnormalities are identified by
of the remaining allele, that cell escapes suppres- examination of the chromosome on which
sion of uncontrolled growth and grows as a the gene resides. Chromosomal abnormalities
tumor (Fig. 1.7). In individuals with a normal are abundant in CNS tumors. Comparative
genetic complement, two hits, one on the tumor genomic hybridization (Fig. 1.8) is a technique
suppressor gene of each chromosome, are neces- to identify regions of chromosomal amplifica-
sary before the cell can become transformed. This tion and deletion by comparing normal DNA
explains why hereditary retinoblastomas are with tumor DNA. A profile is generated that

24
BIOLOGY OF INTRACRANIAL TUMORS

Figure 1.8
a)
Comparative genomic hybridization. (a) Tumor DNA
is labeled with a green flourescent dye and normal
DNA with a red flourescent dye. Equal amounts are
mixed with an excess of unlabeled DNA
Normal DNA Tumor DNA
corresponding to highly repeated sequences and
hybridized to a metaphase spread of chromosomes
from normal human tissue. Areas of chromosome
gain in the tumor appear red and areas of
Hybridization chromosome deletion appear green. (Modified from
Israel95 with permission. (b) A partial profile of an
anaplastic astrocytoma showing loss of chromosomes
1p and 19q, gain of chromosome 1q and loss of
chromosome 9p. (Modified from Bigner et al101 with
permission.)

Normal human metaphase


9p. Many other chromosomal abnormalities
Fluorescence microscopy are also found in anaplastic oligodendro-
gliomas95,101 and other tumors. Furthermore,
Digital image analysis the genetic abnormalities may vary from area
to area within a given tumor (heterogeneity).102
Chromosomal abnormalities found in brain
tumors are usually characterized by a loss of
Amplification in tumor all or a part of one arm of a chromosome
containing a tumor suppressor gene. For
Loss in tumor
example, loss of 22q deletes the tumor suppres-
sor gene NF-2, leading to the development of
Tumor DNA vs normal DNA neurofibromatosis type 2 (Chapter 12). Loss of
chromosome 22 is also common in menin-
b)
giomas and may also occur in anaplastic astro-
cytomas. What specific functional abnormality
this deletion causes in these tumors is unclear.
Chromosomal deletions are not the only
mechanism that can cause genetic dysfunction;
a point mutation in a specific gene may lead to
a similar change in the biology of the cell. For
example, p53 abnormalities can occur either by
deletions on the short arm of chromosome 17,
identifies all of the amplifications and deletions or by point mutations at one or more of several
of chromosomes in a given tumor. The example sites in the gene, particularly at codon 273.
of an anaplastic oligodendroglioma in Fig. 1.9 Attempting to repair p53 abnormalities by
shows deletion of the short arm of chromo- inserting normal p53 into malignant cells is
some 1 (1p), amplification of the long arm of currently undergoing investigation as one form
chromosome 1 (1q), and deletion of 19q, and of gene therapy.

25
CLASSIFICATION, INCIDENCE AND ETIOLOGY OF INTRACRANIAL TUMORS

Figure 1.9
Vit. D, K Cell cycle and its regulator
M Cell differentiation factors. (Modified from
TNF Lupulescu103 with
permission.)
cyclin CDK1
G2 A,B Apoptosis
phase cyclin
G2
cyclin C CDK1
arrest p53+
B
CDK1 BcL-2
CDK4
Rb cyclin C-myc+
cyclin D
E2F TNF
B
CDK2
G1 G1
phase arrest
S CDK2 cyclin
phase E Cell death
cyclin CDK2
A (necrosis)
GFs

Go phase
months
years

Hormones,
vitamins

Gains in chromosomes may lead to amplifi- progression or inhibition of the cell cycle. For
cation of oncogenes. For example, gene ampli- example, cyclin-dependent kinase 4 (CDK-4)
fication can cause an increase in the EGFR. phosphorylates RB protein, which, in turn,
Overactivity of growth factor receptors trans- inhibits binding to transcription factors such as
mits a signal to the cell which, over several E2F, a factor that initiates cell cycle progres-
steps, leads to excessive cellular reproduction. sion. Cyclin-dependent kinase p21 mediates
PKC, also involved in signal transduction, is p53 activity to induce cell cycle arrest; p16 also
expressed in many malignant gliomas. It is inhibits the cell cycle. Mutations in any of these
believed that high-dose tamoxifen, used to treat proteins can lead to uninhibited progression of
malignant gliomas, inhibits that enzyme. cellular reproduction. Abnormalities of cell
As Fig. 1.9 illustrates, the cell cycle is cycle regulatory pathways are common in high-
complicated. Proteins called cyclins and cyclin- grade gliomas. Inactivation of CDKNA2A and
dependent kinases are essential at several steps B genes, inactivation of the RB gene and
in the cell cycle. Alterations in the genes coding overexpression of CDK-4 dysregulate the
for these proteins can cause either uncontrolled normal cell cycle. While a variety of different

26
BIOLOGY OF INTRACRANIAL TUMORS

and sometimes mutually exclusive genetic alter- implies that each individual tumor must be
ations have been documented in malignant examined genetically before therapy directed at
gliomas, many lead to disruption of the same genetic alterations can be applied. To further
pathways that cause uncontrolled cell prolifer- compound the problem, even when a genetic
ation. For example, an inactivating mutation of change is identified in a tumor, not all tumor
p53 inhibits the cells normal mechanisms of cells possess the same genetic change. EGFR
apoptosis in a manner identical to overexpres- overexpression is found in about 40% of all
sion of MDM2. glioblastoma multiforme, but is not present in
The genetic alterations that play important all of the tumor cells of the glioblastoma. The
roles in the pathogenesis of brain tumors, i.e. extreme heterogeneity of these tumors leads to
loss of tumor suppressor gene function, different alterations in different portions of the
increases in growth factors, and mutation or tumor and in different cells in the same portion
amplification of growth factor receptors, repre- of the tumor. The result is that therapy applied
sent not only a challenge but an opportunity. to block growth of tumor cells might be effec-
Each genetic alteration is a potential site for tive in eradicating some tumor cells, but allows
therapeutic intervention. For example, restora- those that are not sensitive to the agent to
tion of wild-type p53 may not only decrease continue to grow. It is likely that successful
growth of malignant cells, but might also cause therapies applied to brain tumors will have to
those cells to undergo apoptosis. Inhibitors of be multiagent rather than single agent and
a growth factor receptor may slow or even stop multimodality rather than single modality.
the growth of tumor cells. A particularly
attractive site for therapeutic intervention is
Other genetic alterations
angiogenesis. Because newly formed tumor
blood vessels are not neoplastic they are not One factor that perhaps plays a role in tumor
genetically unstable as are tumor cells. heterogeneity is microsatellite instability.
Accordingly, unlike tumor cells, they lack the Microsatellites are simple, short, tandem
capacity to develop resistance to a therapeutic repeats of di-, tri- or tetranucleotides occurring
agent. Furthermore, recent experimental 530 times. Normally, the number of repeat
evidence suggests that tumor cells are not only units is highly conserved but when there are
incapable of developing resistance but, after errors in DNA replication, the numbers may
time, they will fail to nourish a tumor even substantially increase or decrease. The term
when the therapeutic agent is withdrawn.104 applied to this disorder is microsatellite insta-
Because of their severe genetic instability, a bility or RER+ (replication error positive) and
major problem in the treatment of brain it implies that the cell is genetically unstable,
tumors, particularly gliomas, is their hetero- making it susceptible to more genetic changes
geneity. The genetic changes shown in Tables that might eventually lead to cancer. The
1.7 and 1.8 are not present in all brain tumors phenomenon was first observed in tumors
of a particular histologic type. For example, associated with hereditary non-polyposis
p53 mutations that are present in a majority of colorectal cancer (HNPCC), where mutations
glioblastoma multiforme are not present in all of DNA were due to dysfunction of mismatch
glioblastomas, indicating that tumors that look repair genes.105 These genes repair DNA altered
identical under the microscope may have by replication errors and environmental insults.
substantially different genetic origins. This Mutation of mismatch repair genes not only

27
CLASSIFICATION, INCIDENCE AND ETIOLOGY OF INTRACRANIAL TUMORS

makes the cell genetically unstable, but also Genetic abnormalities not only promote
reduces the effects of certain chemotherapeutic neoplastic growth and development, but may
agents. Methylating drugs such as streptozocin, affect the sensitivity of tumor cells to therapy.
temozolomide, cisplatin, 6-thioguanine, busul- Some normal genes and some genetic abnor-
fan, etoposide and doxorubicin, all of which malities may lead to the production of proteins
affect DNA at the O6 position of guanine, are that protect tumor cells against cytotoxic
substantially less effective against cells that agents:
have mutations of DNA mismatch repair genes.
The resistance may be a result of failure of 1. The enzyme MGMT repairs DNA damage
degradation of newly synthesized DNA caused by some chemotherapeutic agents,
opposite alkylation damage, or by an inability especially the nitrosoureas (e.g. 1,3-
to detect the damage and cause apoptosis.85,106 bischloro-(2-chloroethyl)-1-nitrosourea
Microsatellite instability is more common in (BCNU)). Some tumors (about 75%)
pediatric than adult CNS tumors.107 express increased amounts of MGMT,
Other genetic alterations also affect cellular conferring resistance to nitrosoureas on the
growth, proliferation and cellular immortality. tumor cells.110 The increased amount of
For example, the enzyme telomerase maintains MGMT does not appear to be related to
the size of the tandem hexamer repetitive proliferation per se, but does heighten the
sequences (TTAGCG)n at the ends of all resistance of brain tumors to therapeutic
chromosomes that are required for cell division. alkylating agents. The presence of the
The number of repetitive sequences progres- enzyme in brain tumors appears to be
sively decreases at approximately 50100 base associated with resistance to temozolomide
pairs per cell division as cells continue to divide. as well as BCNU.111
When the telomere (from the Greek telos = end, 2. P-glycoprotein is a membrane glycoprotein
meros = part) becomes too short, the cell can no that is expressed in a number of brain
longer divide and becomes senescent; the cell tumors112 and also at the luminal
may undergo apoptosis. Thus, telomere short- membrane of the brain capillary endothe-
ening functions as a mitotic clock, inexorably lial cell as a component of the normal
ticking to old age and death. Most normal cells, bloodbrain barrier (Chapter 2). The
with the exception of germ cells, do not contain protein functions as a pump extruding a
the enzyme telomerase and thus possess a built- number of cytotoxic drugs from the cell,
in senescence. Cancers, including many brain including vincristine and doxorubicin, one
tumors contain telomerase, one mechanism that mechanism of resistance to chemotherapy.
ensures cellular immortality. There is also a However, the agents that P-glycoprotein
relationship between telomerase activity and affects are not those usually active against
histologic grade. Telomerase activity is not brain tumors or even in brain tumor cell
present in low-grade CNS tumors, but is lines in tissue culture.
present in more aggressive tumors and is an 3. Metallothioneins (MT) are intracellular
indicator of histologic progression from low to sulfur-containing proteins that have a high
high grade.108 Telomerase may also play a role affinity for heavy metal ions such as zinc,
in resistance of tumor cells to cytotoxic drugs; cadmium, copper and mercury. Their
inhibition of telomerase activity can sensitize normal function involves metabolism of
cells to cisplatin-induced apoptosis.109 trace metals and detoxification of toxic

28
PROGNOSIS

metals. They also bind to cytotoxic agents lacking the p53 protein are chemoresistant,
containing a metal, e.g. cisplatin, and, particularly to paclitaxel and topotecan.
when overexpressed, confer resistance to When the wild-type p53 gene was intro-
tumor cells that express these proteins.113 duced into these cells, the cells regained
The most likely mechanism of resistance to chemosensitivity.114
cisplatin is that the proteins enhance
removal of platinum adducts from DNA,
thus preventing damage by the cytotoxic
agent. MTs are overexpressed in many
Prognosis
brain tumors (66% of astrocytomas) and Several factors are important for predicting the
appear to be inversely related to survival of prognosis of specific intracranial tumors (Table
the patient.113 The proteins are normally 1.10): histopathology, especially mitoses,
expressed at high levels during embryologic vascular proliferation and necrosis, defines an
development and appear to have a role in aggressive tumor with a poor prognosis.
cellular proliferation. MTs are normally The location of the tumor determines
active during the mitotic cell cycle and may symptoms, surgical resectability and, as a result
serve as cellular proliferation markers. of both, prognosis. For example, tumors of the
4. Mutations in p53 also may confer drug non-dominant frontal lobe are often asymp-
resistance. One of the functions of normal tomatic until they reach a large size, can often
p53 is to cause cell cycle arrest in the G1 be completely resected, and have a better
phase when DNA is damaged. If the DNA prognosis than tumors in highly symptomatic
cannot be repaired by the cell, p53 but surgically inaccessible regions such as the
promotes cell death through apoptosis. brainstem or basal ganglia. Tumors arising
Alterations in p53 function may allow cells outside of the brain, such as meningiomas and
with damaged DNA to continue to repro- pituitary tumors, even when histopathologi-
duce, thus conferring resistance to radia- cally aggressive, generally have a better
tion therapy and cytotoxic agents including prognosis than tumors arising within the
topoisomerase inhibitors, nitrosoureas and parenchyma of the brain. The age of the
platinum compounds, all of which have patient is an important prognostic factor. For
DNA as their target. Tumor cell lines certain tumors, such as high-grade gliomas,

Table 1.10
Some factors determining prognosis of CNS tumor.

Factor Tumors in which the factor is particularly important

Histopathology Astrocytoma, pineal tumors


Location Meningioma
Age Astrocytoma, lymphoma, glioblastoma
Extent of surgery Glioblastoma
Performance status Lymphoma, gliomas
Radiation and/or chemotherapeutic sensitivity Oligodendroglioma

29
CLASSIFICATION, INCIDENCE AND ETIOLOGY OF INTRACRANIAL TUMORS

children fare better than adults and young neurologic symptoms. The sensitivities of the
adults fare better than older adults. In adults tumor to radiation and chemotherapy are
with high-grade gliomas, age is the single most important prognostic factors.
important prognostic factor. On the contrary, Histopathologically malignant pineal region
adults with brainstem gliomas usually fare germinomas are cured by radiation, and an
better than children. Race does not appear to increasing number of CNS lymphomas appear
influence survival.115 Although controversial, to be cured by chemotherapy. It is poor sensi-
most evidence suggests that the extent of tivity to radiation and chemotherapy that
surgery is an important prognostic factor.116 In makes the high-grade glioma such an
meningiomas, total surgical resection is intractable tumor. The physician must consider
curative. High-grade gliomas are never cured all of these prognostic factors for each individ-
by surgery, but the less tumor that remains ual patient when prescribing treatment and
after resection, the better the prognosis. The discussing prognosis.
patients neurologic state before and after Data from CBTRUS tabulating 2- and 5-year
surgery (performance status) is also an impor- survivals for specific intracranial tumors are
tant prognostic factor. Relatively asymptomatic tabulated in Table 1.11. Overall, the probability
patients have a better prognosis than those of a patient surviving a high-grade brain tumor
who are paralyzed or suffer other crippling is 36% at 2 years and 27% at 5 years. However,

Table 1.11
Histology % of Cases % 5-year survival Prognosis of brain
tumors from the
Astrocytoma 18.7 30.3 National Cancer Data
Pilocytic astrocytoma 1.4 77.4 Base.
Anaplastic astrocytoma 5.2 21.9
Glioblastoma multiforme 29.6 2.4
Oligodendroglioma 2.4 59.6
Anaplastic oligodendroglioma 0.4 36.5
Ependymoma 1.7 68.1
Anaplastic ependymoma 0.2 54.3
Medulloblastoma 2.4 55.0
Germinoma 0.4 76.3
Pinealoma/pineocytoma 0.1 56.3
Pineoblastoma 0.2 35.6
Choroid plexus papilloma 0.1 70.8
Ganglioglioma 0.3 88.4
Meningioma 14.3 70.1
Malignant meningioma 1.2 54.6
Hemangioblastoma 0.5 83.6
Chordoma 0.2 62.0
Neurilemoma 2.5 84.9
Malignant neurilemoma 0.1 64.6
Lymphoma 4.3 12.9

Table modified from Surawicz et al22 with permission.

30
REFERENCES

for those patients who survive 2 years, the 10. Levin VA. Cancer in the nervous system. 1st
likelihood of surviving 5 years is 76%.117 edn. New York: Churchill Livingstone, 1996:
1474.
Unfortunately, the quality of life for surviving
11. Black PM, Loeffler JS. Cancer of the nervous
patients, particularly children and the elderly is system. 1st edn. Cambridge, MA: Blackwell
generally reduced from their premorbid state Scientific, 1997: 1935.
and not as good as that of most patients success- 12. Kaye AH, Laws ERJ. Brain tumors: an
fully treated for systemic cancer.118 encyclopedic approach. 2nd edn. New York:
Churchill Livingstone, 2001.
13. Posner JB. Neurologic complications of
cancer. Philadelphia: F.A. Davis, 1995.
References 14. Vecht CJ. Neurooncology parts 13. New
York: Elsevier, 1997.
1. Smith MA, Freidlin B, Ries LAG, Simon R.
15. Kleihues P, Cavenee WK. World Health
Trends in reported incidence of primary
Organization classification of tumours:
malignant brain tumors in children in the
tumours of the nervous system-pathology and
United States. J Natl Cancer Inst 1998; 90:
genetics. Lyon: IRAC Press, 2000.
126977.
16. Radhakrishnan K, Mokri B, Parisi JE et al.
2. Bleyer WA. Epidemiologic impact of children
The trends in incidence of primary brain
with brain tumors. Child Nerv Syst 1999; 15:
tumors in the population of Rochester,
75863.
Minnesota. Ann Neurol 1995; 37: 6773.
3. Davis FG, McCarthy BJ. Epidemiology of
17. CBTRUS (2000). Statistical report: primary
brain tumors. Curr Opin Neurol 2000; 13:
brain tumors in the United States, 1992
63540.
1997. Central Brain Tumor Registry of the
4. Riggs JE. The biphasic pattern of age-specific
United States, 2000.
malignant brain tumor mortality rates. 18. Greenlee RT, Hill-Harmon MB, Taylor M,
Neurology 2000; 55: 7503. Thum M. Cancer statistics, 2001. Ca Cancer
5. Gurney JG, Kadan-Lottick N. Brain and J Clin 2001; 51: 1536.
other central nervous system tumors: rates, 19. Coons SW, Johnson PC, Scheithauer BW,
trends, and epidemiology. Curr Opin Oncol Yates AJ, Pearl DK. Improving diagnostic
2001; 13: 1606. accuracy and interobserver concordance in
6. Schulder M, Maldjian JA, Liu WC et al. the classification and grading of primary
Functional image-guided surgery of intra- gliomas. Cancer 1997; 79: 138193.
cranial tumors located in or near the senso- 20. ONeil BP, Janney CA, Olson J et al. The
rimotor cortex. J Neurosurg 1998; 89: continuing increase in primary central
4128. nervous system non-Hodgkins lymphoma
7. Bohinski RJ, Kokkino AK, Warnick RE et al. (PCNSL): a surveillance epidemiology, and
Glioma resection in a shared-resource end results (SEER) analysis. Proc Am Soc Clin
magnetic resonance operating room after Oncol 20, 53. 2001.
optimal image-guided frameless stereotactic 21. Cappuzzo F, Mazzoni F, Maestri A et al.
resection. Neurosurgery 2001; 48: 73142. Medical treatment of brain metastases from
8. Davis FG, Freels S, Grutsch J et al. Survival solid tumours. Forum (Genova) 2000; 10:
rates in patients with primary malignant 13748.
brain tumors stratified by patient age and 22. Surawicz TS, Davis F, Freels S, Laws ER, Jr.,
tumor histological type: an analysis based on Menck HR. Brain tumor survival: Results
surveillance, epidemiology, and end results from the National Cancer Data Base. J
(SEER) data, 19731991. J Neurosurg 1998; Neuroncology 1998; 40: 15160.
88: 110. 23. Pobereskin LH, Chadduck JB. Incidence of
9. Dickman PW, Hakulinen T et al. Survival of brain tumours in two English counties: a
cancer patients in Finland 19551994. Acta population based study. J Neurol Neurosurg
Oncol 1999; 38: 1103. Psychiatry 2000; 69: 46471.

31
CLASSIFICATION, INCIDENCE AND ETIOLOGY OF INTRACRANIAL TUMORS

24. Preston-Martin S. Epidemiology of primary Neurol 1999; 20: 198203.


CNS neoplasms. Neurol Clin 1996; 14: 36. Gurney JG, Davis S, Severson RK et al.
27390. Trends in cancer incidence among children in
25. Preston-Martin S. Descriptive epidemiology the US. Cancer 1996; 78: 53241.
of primary tumors of the brain, cranial nerves 37. Linet MS, Ries LA, Smith MA, Tarone RE,
and cranial meninges in Los Angeles County. Devesa SS. Cancer surveillance series: recent
Neuroepidemiology 1989; 8: 28395. trends in childhood cancer incidence and
26. Davis FG, Freels S, Grutsch J, Barlas S, Brem mortality in the United States. J Natl Cancer
S. Survival rates in patients with primary Inst 1999; 91: 10518.
malignant brain tumors stratified by patient 38. Giannini C, Scheithauer BW, Weaver AL et
age and tumor histological type: an analysis al. Oligodendrogliomas: Reproducibility and
based on surveillance, epidemiology, and end prognostic value of histologic diagnosis and
results (SEER) data, 19741991. J Neurosurg grading. J Neuropathol Exp Neurol 2001; 60:
1998; 88: 110. 24862.
27. Kuratsu J, Ushio Y. Epidemiological study 39. Marie Y, Sanson M, Mokhtari K et al.
of primary intracranial tumours in elderly OLIG2 as a specific marker of oligoden-
people. J Neurol Neurosurg Psychiatry 1997; troglial tumour cells. Lancet 2001; 358:
63: 1168. 298300.
28. Ruiz-Tovar M, Lpez-Abente G, Polln M et 40. Ino Y, Betensky RA, Zlatescu MC et al.
al. Brain cancer incidence in the provinces of Molecular subtypes of anaplastic oligoden-
Zaragoza and Navarre (Spain): effect of age, droglioma: implications for patient manage-
period and birth cohort. J Neurol Sci 1999; ment at diagnosis. Clin Canc Res 2001; 7:
164: 939. 83945.
29. Werner MH, Phuphanich S, Lyman GH. The 41. Strojan P, Popovic M, Jereb B. Secondary
increasing incidence of malignant gliomas and intracranial meningiomas after high-dose
primary central nervous system lymphoma in cranial irradiation: Report of five cases and
the elderly. Cancer 1995; 76: 163442. review of the literature. Int J Radiat Oncol
30. Olney JW, Farber NB, Spitznagel E, Robins Biol Phys 2000; 48: 6573.
LN. Increasing brain tumor rates: Is there a 42. Sadetzki S, Modan B, Chetrit A, Freedman L.
link to aspartame? J Neuropathol Exp Neurol An iatrogenic epidemic of benign menin-
1996; 55: 111523. gioma. Am J Epidemiol 2000; 151: 26672.
31. Fleury A, Menegoz F, Grosclaude P et al. 43. Buttner A, Weis S. Non-lymphomatous brain
Descriptive epidemiology of cerebral gliomas tumors in HIV-1 infection: a review. J
in France. Cancer 1997; 79: 1195202. Neurooncol 1999; 41: 818.
32. McKinley BP, Michalek AM, Fenstermaker 44. Vannemreddy PSSV, Fowler M, Polin RS,
RA, Plunkett RJ. The impact of age and sex Todd JR, Nanda A. Glioblastoma multi-
on the incidence of glial tumors in New York forme in a case of acquired immunodefi-
State from 1976 to 1995. J Neurosurg 2000; ciency syndrome: investigating a possible
93: 9329. oncogenic influence of human immunodefi-
33. Legler JM, Ries LA, Smith MA et al. Brain ciency virus on glial cells Case report and
and other central nervous system cancers: review of the literature. J Neurosurg 2000;
recent trends in incidence and mortality. J 92: 1614.
Natl Cancer Inst 1999; 91: 138290. 45. Salvatore JR, Weitberg AB, Mehta S.
34. Kelsey KT, Wrensch M, Zuo ZF, Miike R, Nonionizing electromagnetic fields and
Wiencke JK. A population-based casecontrol cancer: a review. Oncology 1996; 10: 56374.
study of the CYP2D6 and GSTT1 poly- 46. Wrensch M, Yost M, Miike R, Lee G,
morphisms and malignant brain tumors. Touchstone J. Adult glioma in relation to
Pharmacogenetics 1997; 7: 4638. residential power frequency electromagnetic
35. Keene DL, Hsu E, Ventureyra E. Brain field exposures in the San Francisco Bay area.
tumors in childhood and adolescence. Pediatr Epidemiology 1999; 10: 5237.

32
REFERENCES

47. Muscat JE, Malkin MG, Thompson S et al. 59. Weggen S, Bayer TA, von Deimling A et al.
Handheld cellular telephone use and risk of Low frequency of SV40, JC and BK
brain cancer. JAMA 2000; 284: 30017. polyomavirus sequences in human medullo-
48. Inskip PD, Tarone RE, Hatch EE et al. blastomas, meningiomas and ependymomas.
Cellular-telephone use and brain tumors. N Brain Pathol 2000; 10: 8592.
Engl J Med 2001; 344: 7986. 60. Huang HT, Reis R, Yonekawa Y et al.
49. Zook BC, Simmens SJ. The effects of Identification in human brain tumors of DNA
860 MHz radiofrequency radiation on the sequences specific for SV40 large T antigen.
induction or promotion of brain tumors and Brain Pathol 1999; 9: 3342.
other neoplasms in rats. Radiat Res 2001; 61. Ron E, Modan B, Boice JD, Jr et al. Tumors
155: 57283. of the brain and nervous system after radio-
50. Schwartzbaum JA, Fisher JL, Goodman J, therapy in childhood. New Engl J Med 1988;
Octaviano D, Cornwell DG. Hypotheses 319: 10339.
concerning roles of dietary energy, cured 62. Walter AW, Hancock ML, Pui CH et al.
meat, and serum tocopherols in adult glioma Secondary brain tumors in children treated
development. Neuroepidemiology 1999; 18: for acute lymphoblastic leukemia at St Jude
15666. Childrens Research Hospital. J Clin Oncol
51. Carozza SE, Wrensch M, Miike R et al. 1998; 16: 37617.
Occupation and adult gliomas. Am J 63. Relling MV, Rubnitz JE, Rivera GK et al.
Epidemiol 2000; 152: 83846. High incidence of secondary brain tumours
52. Bohnen NI, Kurland LT. Brain tumor and after radiotherapy and antimetabolites.
exposure to pesticides in humans: a review of Lancet 1999; 354: 349.
the epidemiologic data. J Neurol Sci 1995; 64. Ryan P, Lee MW, North B, McMichael AJ.
132: 11021. Amalgam fillings, diagnostic dental x-rays
53. Pogoda JM, Preston-Martin S. Household and tumours of the brain and meninges.
pesticides and risk of pediatric brain tumors. Eur J Cancer B Oral Oncol 1992; 28B:
Environ Health Perspect 1997; 105: 121420. 915.
54. Inskip PD, Mellemkjaer L, Gridley G, Olsen 65. Rodvall Y, Ahlbom A, Pershagen G,
JH. Incidence of intracranial tumors follow- Nylander M, Spannare B. Dental radiography
ing hospitalization for head injuries after age 25 years, amalgam fillings and
(Denmark). Cancer Causes Control 1998; 9: tumours of the central nervous system. Oral
10916. Oncol 1998; 34: 2659.
55. Wrensch M, Miike R, Lee M, Neuhaus J. Are 66. Camilleri-Broet S, Davi F, Feuillard J et al.
prior head injuries or diagnostic X-rays AIDS-related primary brain lymphomas:
associated with glioma in adults? The effects histopathologic and immunohistochemical
of control selection bias. Neuroepidemiology study of 51 cases. The French Study Group
2000; 19: 23444. for HIV-Associated Tumors. Hum Pathol
56. Preston-Martin S, Pogoda JM, Mueller BA et 1997; 28: 36774.
al. Results from an international case-control 67. Frisch M, Biggar RJ, Engels EA, Goedert JJ.
study of childhood brain tumors: The role of Association of cancer with AIDS-related
prenatal vitamin supplementation. Environ immunosuppression in adults. JAMA 2001;
Health Perspect 1998; 106: 88792. 285: 173645.
57. Del Brutto OH, Castillo PR, Mena IX, Freire 68. Bunin G. What causes childhood brain
AX. Neurocysticercosis among patients with tumors? Limited knowledge, many clues.
cerebral gliomas. Arch Neurol 1997; 54: Pediatr Neurosurg 2000; 32: 3216.
11258. 69. Schwartzbaum JA, Cornwell DG. Oxidant
58. Wrensch M, Weinberg A, Wiencke J et al. stress and glioblastoma multiforme risk:
Does prior infection with varicella-zoster serum antioxidants, gamma-glutamyl
virus influence risk of adult glioma? Am J transpeptidase, and ferritin. Nutr Cancer
Epidemiol 1997; 145: 5947. 2000; 38: 409.

33
CLASSIFICATION, INCIDENCE AND ETIOLOGY OF INTRACRANIAL TUMORS

70. Chia SE, Chia HP, Tan JS. Health hazards of 84. Spiro TP, Gerson SL, Liu L et al. O6-benzyl-
mobile phones Prevalence of headache is guanine: a clinical trial establishing the
increased among users in Singapore. Br Med biochemical modulatory dose in tumor tissue
J 2000; 321: 11556. for alkyltransferase-directed DNA repair.
71. Malmer B, Iselius L, Holmberg E et al. Cancer Res 1999; 59: 240210.
Genetic epidemiology of glioma. Br J Cancer 85. Holland EC. Gliomagenesis: genetic alter-
2001; 84: 42934. ations and mouse models. Nat Rev Genet
72. Tomlinson GE. Familial cancer syndromes 2001; 2: 1209.
and genetic counseling. Cancer Treat Res 86. Loeb LA. A mutator phenotype in cancer.
1997; 92: 6397. Cancer Res 2001; 61: 32309.
73. Johnson MW, Emelin JK, Park SH, Vinters 87. Esteller M, Corn PG, Baylin SB, Herman JG.
HV. Co-localization of TSC1 and TSG2 gene A gene hypermethylation profile of human
products in tubers of patients with tuberous cancer. Cancer Res 2001; 61: 32259.
sclerosis. Brain Pathol 1999; 9: 4554. 88. Simone NL, Bonner RF, Gillespie JW,
74. Gutmann DH, Aylsworth A, Carey JC et al. Emmert-Buck MR, Liotta LA. Laser-capture
The diagnostic evaluation and multidisci- microdissection: opening the microscopic
plinary management of neurofibromatosis 1 frontier to molecular analysis. Trends Genet
and neurofibromatosis 2. JAMA 1997; 278: 1998; 14: 2726.
517. 89. Loiseau H, Arsaut J, Demotes-Mainard J. p73
75. Feldkamp MM, Gutmann DH, Guha A. gene transcripts in human brain tumors:
Neurofibromatosis type 1: Piecing the puzzle overexpression and altered splicing in ependy-
together. Can J Neurol Sci 1998; 25: 18191. momas. Neurosci Lett 1999; 263: 1736.
76. Pollack IF, Mulvihill JJ. Neurofibromatosis 1 90. Villunger A, Strasser A. Does death recep-
and 2. Brain Pathol 1997; 7: 82336. tor signaling play a role in tumorigenesis
77. Asa SL, Somers K, Ezzat S. The MEN-1 gene and cancer therapy? Oncol Res 1998; 10:
is rarely down-regulated in pituitary adeno- 54150.
mas. J Clin Endocrinol Metab 1998; 83: 91. Chernova OB, Somerville RP, Cowell JK. A
32102. novel gene, LGI1, from 10q24 is rearranged
78. Mulligan G, Jacks T. The retinoblastoma and downregulated in malignant brain
gene family: cousins with overlapping inter- tumors. Oncogene 1998; 17: 287381.
ests. Trends Genet 1998; 14: 2239. 92. Hunter T. The Croonian Lecture 1997. The
79. Wicking C, Bale AE. Molecular basis of the phosphorylation of proteins on tyrosine: its
nevoid basal cell carcinoma syndrome. Curr role in cell growth and disease. Philos
Opin Pediatr 1997; 9: 6305. Trans R Soc Lond B Biol Sci 1998; 353:
80. Foulkes WD. A tale of four syndromes: 583605.
familial adenomatous polyposis, Gardner 93. Yamada SM, Yamaguchi F, Brown R, Berger
syndrome, attenuated APC and Turcot MS, Morrison RS. Suppression of glioblas-
syndrome. Q J Med 1995; 88: 85363. toma cell growth following antisense oligo-
81. Decker HJ, Weidt EJ, Brieger J. The von nucleotide-mediated inhibition of fibroblast
HippelLindau tumor suppressor gene. A rare growth factor receptor expression. Glia 1999;
and intriguing disease opening new insight 28: 6676.
into basic mechanisms of carcinogenesis. 94. Uhrbom L, Hesselager G, Nister M,
Cancer Genet Cytogenet 1997; 93: 7483. Westermark B. Induction of brain tumors in
82. Eng C. Genetics of Cowden syndrome: mice using a recombinant platelet-derived
through the looking glass of oncology. Int J growth factor B-chain retrovirus. Cancer Res
Oncol 1998; 12: 70110. 1998; 58: 52759.
83. Svenet N, Sheridan E, Amram D et al. 95. Israel MA. Molecular genetics of brain
Constitutional mutations of the hSNF5/INI1 tumors. In: Martin JB, ed. Scientific American
gene predispose to a variety of cancers. Am J Molecular Neurology. New York: Scientific
Hum Genet 1999; 65: 13428. American, 1999: 95114.

34
REFERENCES

96. Maddalena AS, Hainfellner JA, Hegi ME, 108. Huang F, Kanno H, Yamamoto I, Lin Y,
Glatzel M, Aguzzi A. No complementation Kubota Y. Correlation of clinical features and
between TP53 or RB-1 and v-src in astro- telomerase activity in human gliomas. J
cytomas of GFAP-v-src transgenic mice. Brain Neurooncol 1999; 43: 13742.
Pathol 1999; 9: 62737. 109. Kondo Y, Kondo S, Tanaka Y, Haqqi T,
97. Adachi J, Ohbayashi K, Suzuki T, Sasaki T. Barna BP, Cowell JK. Inhibition of telomerase
Cell cycle arrest and astrocytic differentiation increases the susceptibility of human malig-
resulting from PTEN expression in glioma nant glioblastoma cells to cisplatin-induced
cells. J Neurosurg 1999; 91: 82230. apoptosis. Oncogene 1998; 16: 22438.
98. Simon M, Kster G, Menon AG, Schramm J. 110. Esteller M, Garcia-Foncillas J, Andion E et al.
Functional evidence for a role of combined Inactivation of the DNA-repair gene MGMT
CDKN2A (p16p14ARF)/CDKN2B (p15) gene and the clinical response of gliomas to alkylat-
inactivation in malignant gliomas. Acta ing agents. N Engl J Med 2000; 343: 13504.
Neuropathol (Berl) 1999; 98: 44452. 111. Friedman HS, McLendon RE, Kerby T et al.
99. Bodey B, Bodey B Jr, Siegel SE. Tumor DNA mismatch repair and O6alkylguanine-
suppressor genes in childhood malignancies: DNA alkyltransferase analysis and response
a review. Int J Pediatr Hematol Oncol 1999; to Temodal in newly diagnosed malignant
6: 4764. glioma. J Clin Oncol 1998; 16: 38517.
100. Chne P, Ory K, Redi D, Soussi T, Hegi 112. Ashmore SM, Thomas DG, Darling JL. Does
ME. Functional analyses of a unique p53 P-glycoprotein play a role in clinical resis-
germline mutant (Y236Delta) associated with tance of malignant astrocytoma? Anticancer
a familial brain tumor syndrome. Int J Cancer Drugs 1999; 10: 86172.
1999; 82: 1722. 113. Hiura T, Khalid H, Yamashita H et al.
101. Bigner SH, Rasheed K, Wiltshire RN, Immunohistochemical analysis of metallo-
McLendon R. Morphologic and molecular thionein in astrocytic tumors in relation to
genetic aspects of oligodendroglial neoplasms. tumor grade, proliferative potential, and
Neuroncol 1999; 1: 5260. survival. Cancer 1998; 83: 23619.
102. Jung V, Romeike BFM, Henn W et al. 114. Fueyo J, Gomez-Manzano C, Puduvalli VK
Evidence of focal genetic microhetero- et al. Adenovirus-mediated p16 transfer to
geneity in glioblastoma multiforme by area- glioma cells induces G1 arrest and protects
specific CGH on microdissected tumor from paclitaxel and topotecan: implications
cells. J Neuropathol Exp Neurol 1999; 58: for therapy. Int J Oncol 1998; 12: 6659.
9939. 115. Simpson JR, Scott CB, Rotman M et al. Race
103. Lupulescu A. Cancer cell metabolism: its and prognosis of brain tumor patients enter-
relevance to cancer treatment. Cancer Invest ing multicenter clinical trials. A report from
1999; 17: 42333. the Radiation Therapy Oncology Group. Am
104. Folkman J. Antiangiogenic gene therapy. Proc J Clin Oncol 1996; 19: 11420.
Natl Acad Sci USA 1998; 95: 90646. 116. Black P. Management of malignant glioma:
105. Claij N, Te R. Microsatellite instability in role of surgery in relation to multimodality
human cancer: a prognostic marker for therapy. J Neurovirol 1998; 4: 22736.
chemotherapy? Exp Cell Res 1999; 246: 117. Davis FG, McCarthy BJ, Freels S, Kupelian
110. V, Bondy ML. The conditional probability of
106. Fink D, Aebi S, Howell SB. The role of DNA survival of patients with primary malignant
mismatch repair in drug resistance. Clin brain tumors Surveillance, epidemiology,
Cancer Res 1998; 4: 16. and end results (SEER) data. Cancer 1999;
107. Alonso M, Hamelin R, Kim M et al. 85: 48591.
Microsatellite instability occurs in distinct 118. Foreman NK, Faestel PM, Pearson J et al.
subtypes of pediatric but not adult central Health status in 52 long-term survivors of
nervous system tumors. Cancer Res 2001; 61: pediatric brain tumors. J Neurooncol 1999;
21248. 41: 4753.

35
2
Invasion, angiogenesis and the bloodbrain barrier

Introduction Table 2.1


Sequential steps in growth of brain tumors.
Once the genetic changes that lead to uncon-
trolled cellular reproduction have occurred, 1. Cellular gene mutation (multiple genes)
central nervous system (CNS) tumors must 2. Uncontrolled cell division
grow to a fairly large size (at least 1 cm) before 3. Invasion into normal brain
causing neurologic symptoms. Most brain 4. Formation of a tumor blood supply, i.e.
angiogenesis
tumors grow both by increasing their core mass
5. Bloodbrain barrier disruption
and by invading surrounding normal brain. In 6. Edema
order to grow, a tumor requires nutrients that
are supplied by the vasculature. After a tumor
exceeds a few millimeters in size, it requires the
production of new blood vessels (angiogenesis) mechanisms (Table 2.1) include invasion of
to meet its metabolic demands. The newly tumor cells into normal parenchyma (particu-
formed blood vessels do not possess a normal larly true of glial tumors), angiogenesis
bloodbrain barrier and are thus a source of (required for tumor growth beyond a few
brain edema (see below), which contributes to millimeters) and bloodbrain barrier disruption
the total tumor mass. The combined tumor leading to brain edema (a prominent feature of
mass and its edema not only cause neurologic high-grade gliomas). This chapter describes
dysfunction, but also raise interstitial fluid some of these mechanisms and their clinical
pressure, compromising blood flow and implications.
causing local hypoxia.1,2 The combined size of
a tumor and its edema necessary to cause
symptoms depends, in part, on the location of
the lesion (for example, a 1 cm lesion in the
Invasion
brainstem may be symptomatic, but a 5 cm Some CNS tumors are highly invasive (Fig.
lesion in the frontal lobe may be asymp- 2.1). Gliomas (Chapter 5) and primary CNS
tomatic) and, in part, on the nature of the lymphomas (Chapter 11) are among the most
symptom(s) (e.g. small lesions may cause invasive of cancers. During their migratory
seizures but it requires a large lesion to alter phase, brain tumor cells transiently exit from
cognition or personality). the cell cycle (see Fig. 1.9), making them
Several mechanisms play a role in brain relatively refractory to therapy because quies-
tumor growth and edema formation. These cent cells are less susceptible to radiation and

36
INVASION

Figure 2.1
a)
Invasion of brain by tumor. (a) A glioblastoma with
a relatively sharp interface of the surrounding brain.
A needle passing within millimeters of the neoplasm
may obtain only normal tissue. However, there is
infiltration of tumor beyond the enhancing edge. (b)
and (c) Greater and lesser degrees of differentiation
that can be seen in the same glioma are illustrated in
this untreated glioblastoma multiforme. The areas of
low density marked by small closed and open
triangles represent fibrillary and gemistocytic
LV astrocytes respectively. The markedly cellular regions
(small closed circles) represent small cell glioblastoma
arising in the pre-existing better-differentiated tumor.
A specimen obtained by needle biopsy from areas of
lower density would indicate a well-differentiated
astrocytoma (b), whereas a sample several millimeters
more superiorly (c) would reveal the diagnostic
features of a glioblastoma. (From Burger et al,3 with
permission.)

(a)

LV LV

LV LV

(b) (c)

chemotherapy than are actively reproducing migrate into normal brain, they derive their
cells (Chapter 4). Invasive cells occur at the nourishment from normal cerebral blood
periphery of a tumor mass and can extend vessels. Consequently, even when these cells
many centimeters away from the focal lesion may be intrinsically susceptible to chemical
identifiable on MRI.4 Because individual cells agents, they are sequestered behind a normal

37
INVASION, ANGIOGENESIS AND THE BLOODBRAIN BARRIER

Figure 2.2
Diffuse tumor (gliomatosis cerebri). A 40-year-old man complained of dizziness and unsteadiness on his feet.
Examination revealed nystagmus on left lateral gaze and a slightly ataxic gait. His mental state was normal. An
MR scan (left panel) revealed increased signal in both medial temporal lobes, right greater than left, and
cerebellum (arrow, left panel). Both frontal lobes and the right insula were also involved (arrow, right panel). A
stereotactic needle biopsy of the right temporal lobe revealed increased cellularity and mildly atypical glioma
cells infiltrating the parenchyma, some forming perineuronal satellitosis (arrow), consistent with a low-grade
glioma. Whole-brain radiation therapy to 60 Gy partially relieved the symptoms. He was able to work
effectively for 5 years, when he developed increasing ataxia and new cognitive changes. There was no
significant change in the scan. Chemotherapy was begun.

bloodbrain barrier (see below), making them cerebri), causing a diffuse increase in brain
inaccessible to most water-soluble chemo- mass without evidence of a focal lesion (Fig.
therapeutic agents. Sometimes a glioma may 2.2). Diffuse infiltration may be unaccompa-
invade the entire neuraxis (gliomatosis nied by any change in signal intensity on the

38
INVASION

Table 2.2 invasion and dissemination of tumor into the


Factors required for invasion. CSF.5 Another such molecule, possibly impor-
tant in disaggregation of tumor cells, is N-
Tumor cellcell interactions loosen CAM.
Tumor cells bind to extracellular matrix In order to migrate out from the core of the
Proteases degrade extracellular matrix tumor, tumor cells must adhere to molecules of
Integrins promote cell motility
the extracellular matrix,13 as well as break the
Motility leads to invasion into normal brain
Growth factors promote tumorigenesis adhesion between themselves. Brain extracellu-
lar matrix proteins differ from those of other
tissues4 and also differ from area to area in
brain. Brain tumor cells in vitro adhere to
several structures, including blood vessels,
arachnoid, choroid plexus, ventricular
MRI. Other tumors such as meningiomas ependyma and myelinated fiber tracts. The
rarely invade normal brain but require angio- attachment of neuroepithelial tumor cells to
genesis in order to grow large enough to blood vessels indicates that migration along
compress underlying brain and thus cause blood vessels may be a guiding structure for
symptoms. dissemination of gliomas and perhaps
Several factors are required for tumor lymphomas which grow around vessels. The
invasion of brain5 (Table 2.2). These factors major matrix proteins in small vessels, choroid
are not unique to brain tumors but are a plexus and ependyma are laminin, collagen
general characteristic of most cancers. In the type 4 and fibronectin. Arachnoid fibers
normal state, cells adhere to one another and contain various types of collagen, predomi-
to the underlying basement membrane using a nantly types 1 and 3. The only matrix protein
diverse system of adhesion molecules and their in subependyma is fibronectin. The main ECM
receptors. These include: (1) cadherins,6 (2) proteins of the brain paranchyma are hyaluro-
members of the immunoglobulin superfamily, nan, tenascin and brain enriched hyaluronan
including the neural cell adhesion molecule binding/brevican.5 The last is upregulated in
(N-CAM),7 (3) integrins,8 and (4) selectins.9 gliomas and its cleavage appears important for
Alterations in adhesion molecules play a dual tumor invasion.5
role in tumor invasion. Downregulation of Adhesion molecules, including integrins, are
certain adhesion molecules loosens cellcell responsible for binding to extracellular matrix,
interactions within the tumor.10 Upregulation whereas antibodies to adhesion molecules
of adhesion molecules allows tumor cells to block binding. For example, antibodies to
attach to the extracellular matrix (ECM), a integrin block adhesion to arachnoid tissue,
requirement for cell migration.11 and anti-N-CAM antibodies inhibit attachment
In most tissues, cadherins play a major role to cortex. Antibodies against CD44, adhesion
in cellcell adhesion.12 E-cadherin is expressed molecules that permit binding to the brains
in endothelial cells and N-cadherin in neural extracellular matrix, block intracerebral
cells. E-cadherin is downregulated in most invasion in an experimental glioma model.14
carcinomas. Some data suggest that N-cadherin CD24, another adhesion molecule expressed on
expression is decreased in high-grade neuro- brain tumor cells, promotes invasion by glioma
epithelial tumors and may correlate with tumor cells.15

39
INVASION, ANGIOGENESIS AND THE BLOODBRAIN BARRIER

The extracellular matrix not only serves as a carry tumor cells long distances within the
site of attachment of cells but, when intact, parenchyma of the brain. In addition, tumor
may inhibit migration. Accordingly, tumor cells cells may find their way into the ventricular
produce proteases that break down the extra- fluid or the cortical subarachnoid space and
cellular matrix, allowing for invasion and disseminate along CSF pathways. Particular
migration.16 Extracellular matrix proteins not pathways that appear to be favored by invad-
only inhibit glioma cell proliferation, but also ing brain tumor cells include growth across the
act as a chemoattractant for glioma cells in corpus callosum and subependymal spread
vitro. The inhibition of growth may be a along the lateral ventricles (Fig. 2.3). Growth
requirement for cells to migrate. The situation of a frontal lobe tumor across the anterior
along blood vessels appears somewhat differ- corpus callosum to the other frontal lobe
ent, because cells migrating along blood vessels (butterfly glioma) is a particularly common
appear to remain in the cell cycle and can problem. Tumor growing across the splenium
proliferate, unlike invasive tumor cells of the corpus callosum is somewhat less
elsewhere in the brain. common but is encountered in both gliomas
To break down the extracellular matrix, and lymphomas. Primary CNS lymphomas
tumor cells secrete matrix metalloproteinases have a predilection for invading along blood
(MMPs), zinc-dependent endopeptidases that vessels. This is particularly prominent at the
break down extracellular proteins.17 Tumor microscopic level, where one may see cuffs of
cells not only secrete proteinases to break lymphoma cells surrounding blood vessels,
down the extracellular matrix, but also secrete with intervening parenchyma relatively spared.
proteinase inhibitors.18 TIMP, a metallopro- Inflammatory lymphocytes (T-cells) may also
teinase inhibitor, has been detected in culture be present.22
media from gliomas and meningiomas. The The clinical implication of such widespread
balance between the degrading proteinases and and extensive tumor invasion in the brain is
their inhibitors determines the extent of tumor that surgical cure is rarely possible. Past
growth and invasion. In addition, growth attempts at radical surgery to treat infiltrating
factors can play a role in extracellular matrix tumors have failed. Even hemispherectomy,
degradation. For example, some reports have carried out to resect apparently localized
suggested that transforming growth factor beta tumor, usually results in recurrence of the
(TGF-) can upregulate MMPs (MMP-2 and tumor in the opposite hemisphere within a
MMP-9) while downregulating TIMP-2. Some year. Although it is true that most gliomas that
MMPs also participate in angiogenesis,17 and are treated locally23 recur locally, this is proba-
perhaps in bloodbrain barrier disruption, a bly due to the greater volume of disease
phenomenon inhibited by dexamethasone.19 immediately surrounding a large mass.
Many tumor cells are intrinsically mobile.20 Furthermore, a recent report indicates that
This should not be surprising for CNS cells radiation therapy, when sublethal, may actually
because migration of neurons and glia is neces- promote invasion and migration of glioma
sary for brain development and evidence cells.24
suggests there is continued migration of non- Frequently, lymphomas (Chapter 11) may be
neoplastic stem cells even into adulthood. controlled by local treatment only to recur at
Migratory routes along blood vessels, some distance from the original tumor.25 The
ependyma, and myelinated fiber tracts21 may implication is that tumor cells quiescent behind

40
INVASION

(a)

(b) (c)

(d)

Figure 2.3
Pathways of tumor dissemination. (a) A patient with a butterfly glioma. The tumor presumably originated in
one frontal lobe and grew across the corpus callosum to involve the other frontal lobe in the characteristic
butterfly fashion. Tumors can cross the corpus callosum at any point but most commonly do so anteriorly.
(b) Tumor spreading across the splenium of the corpus callosum. (c) Subependymal spread. A high-grade
glioma is seen growing along the posterior horn of the lateral ventricle just below the ependyma as well as
along the septum (arrow). (d) Distant metastases from a glioblastoma. The patient had a focal glioblastoma of
the right cerebellum (left panel). The tumor infiltrated the leptomeninges of the cerebral hemisphere (middle
panel, arrows) and diffusely infiltrated the lumbar subarachnoid space, forming small nodules as well (right
panel, arrows). Two small metastatic lesions can be seen in the medulla as well as in the meninges surrounding
the medulla (left panel, arrow).

41
INVASION, ANGIOGENESIS AND THE BLOODBRAIN BARRIER

the bloodbrain barrier subsequently enter a


growth phase and become detectable by
imaging when angiogenesis leads to
bloodbrain barrier disruption (see below). A
further clinical implication is that if cell
invasion could be inhibited, local therapy
might be more effective.

Angiogenesis
Brain tumors are among the most highly vascu-
larized tumors. Glioblastomas, meningiomas,
hemangioblastomas and primary CNS
lymphomas demonstrate their impressive
vascularity by the intense contrast enhance-
ment seen on MR scans. Before the days when
contrast-enhanced CT and MR scans were
available, cerebral arteriography often revealed
a blush of tumor vessels that identified the
presence of neovascularity of the underlying
Figure 2.4
brain tumor (Fig. 2.4). A tumor blush identified on magnetic resonance
Despite their vascularity, many areas of a angiography. This elderly man presented with mental
tumor, especially high-grade tumors, are poorly status changes. He was believed to have cerebral
perfused. Poor perfusion results in part from vascular disease and a magnetic resonance angiogram
the tumor growing faster than new blood was performed which showed a blush representing
the breakdown of the bloodbrain barrier in a right
vessels can form, and in part from the fact that temporal lobe tumor (arrow).
the newly formed capillaries are not normal
brain blood vessels (see below) and have an
increased intervascular distance. Furthermore,
the tumor increases interstitial fluid pressure, angiogenesis (see below) and also inhibit
which compresses the blood vessels within the apoptosis, thus promoting tumor survival.26
tumor.2 The result is that many tumors have Several angiogenic factors released by tumor
areas that are ischemic, hypoxic and even cells react with receptors on normal endothe-
necrotic. Even with a normal blood supply, lial cells to allow new vessel formation (Table
tumor cells utilize anaerobic metabolism much 2.3). Perhaps the most important of these
more than normal brain, i.e. they use more factors is vascular endothelial growth factor
glucose and less oxygen, and produce more (VEGF),27 which reacts with FLT-1 and FLK-1
lactate. These metabolic changes allow tumor receptors (Fig. 2.5).28 The VEGF gene family
cells to grow in a nutrient-deprived environ- has several members. Their relative roles are not
ment that also selects for tumor cells of a fully established. VEGF not only promotes
higher grade.26 Furthermore, tumor hypoxia growth of new tumor, but also increases the
and hypoglycemia activate genes that lead to permeability of normal capillaries and thus can

42
ANGIOGENESIS

Table 2.3
Some factors implicated in angiogenesis and anti- a)
(a) Glioma
angiogenesis
Normal blood vessel
Promote angiogenesis Inhibit
angiogenesis

Vascular endothelial growth Thrombospondin


factors (VEGF-A-VEGF-D) Angiostatin
Placenta growth factor (PlGF) Endostatin
Fibroblast growth factors Angiopoietin-2
(aFGF, bFGF)
Angiopoietin-1
Transforming growth factors
(TGF-, TGF-) VEGF Binding sites Tumor cells
Tumor necrosis factor (TNF-) release VEGF
Hepatocyte growth factor/scatter
factor
Gelatinase-B
Interleukin-8 (IL-8)
Platelet endothelial growth (b)
b)
factor-1 (CD31)
Cyclooxygenase-2 (COX-2) Vessels die
Platelet derived growth factor (PDGF)

Tumor shrinks

lead to brain edema;29 in fact, VEGF was first


identified as vascular permeability factor (VPF).
Hypoxia, which is often present in brain
tumors, is an important stimulus to VEGF Dead cells
production.30 The expression of TGF-1, bFGF,
EGF, platelet derived growth factor (PDGF) and
ganglioside in human gliomas can each stimu-
late VEGF, as can COX-2.31
A number of clinical trials are underway that
are attempting to inhibit angiogenesis,32 usually Figure 2.5
by blocking the endothelial cell VEGF receptor. Angiogenesis in a malignant glioma. (a) Tumor cells
These trials are predicated on the assumption produce a variety of angiogenic substances, including
that inhibition would have the effect of VEGF. VEGF binds to the receptors on endothelial
preventing tumor growth by depriving the cells of nearby blood vessels in a paracrine fashion and
promotes the growth of new blood vessels into the
tumor of adequate nutrition.33 Furthermore, tumor to nourish and sustain it. (b) Inhibition of VEGF
because endothelial cells are not neoplastic (a causes the newly formed blood vessels to die. The
controversial point, because endothelial cells in tumor cells deprived of nutrients also shrink or die.

43
INVASION, ANGIOGENESIS AND THE BLOODBRAIN BARRIER

some glioblastomas show increased prolifera- known to inhibit angiogenesis are undergoing
tive activity, suggesting they too may be early clinical trials. These include the drug
neoplastic), and thus not genetically unstable, thalidomide, a small molecule FLK-1 inhibitor,
they should not develop resistance to agents and an anti-VEGF antibody. Antisense VEGF
that inhibit their development.34 has been reported to inhibit brain tumor
Tumor cells secrete both angiogenesis and growth in animal models.37
anti-angiogenesis factors.35 Normal p53 protein Factors other than VEGF are likely to play
promotes release of anti-angiogenesis factors. an important role in angiogenesis as well. A
Mutated p53, a frequent abnormality in brain brain-specific angiogenesis inhibitor called BAI-
tumors, can confer an angiogenic gain of 1 is transcriptionally regulated by p53. This
function. Such factors as angiostatin and inhibitor is downregulated in brain tumors with
endostatin, secreted by systemic tumors, have p53 mutations. Its absence may play a role in
the effect of suppressing growth of tumor brain tumor angiogenesis.38 Fibroblast growth
metastases. Clinically, one occasionally factor (FGF) binding protein mobilizes and
observes a marked increase in growth of metas- activates locally stored FGFs and can serve as
tases once the primary tumor is resected. One an angiogenic switch molecule. The gene is
hypothesis is that the primary tumor secretes upregulated in a number of cancers.39 Its role in
both angiogenesis and anti-angiogenesis brain tumor angiogenesis is unknown, but
factors. The angiogenesis factors promote bFGF, as well as VEGF, is upregulated in many
growth in the primary tumor but are rapidly brain tumors. A recent study indicates that
metabolized in the bloodstream, so they do not placenta growth factor (PlGF) mRNA is
reach any metastasis which may not be synthe- expressed in hypervascular but not hypovascu-
sizing its own angiogenesis factor(s). The anti- lar brain tumors; it is not expressed in
angiogenesis factors are overwhelmed by the metastatic tumors. VEGF and bFGF mRNA are
angiogenesis factors in the primary tumor, so also detected in hypervascular tumors. Making
that the primary tumor continues to invade and cells hypoxic increases PlGF levels, suggesting
grow, but elsewhere in the body the circulating that it may play a role in angiogenesis in
anti-angiogenesis factors dominate and hypoxic brain tumors.40 The extracellular
suppress the growth of metastases. Larger matrix glycoprotein, tenascin-C, is enhanced in
primary tumors provide more inhibition of many brain tumors and correlates with angio-
angiogenesis than the smaller metastases.36 genesis. Expression of that protein may be
When the primary tumor is removed, growth associated with endothelial cell activation and
of the metastases is no longer inhibited and the play a role in angiogenesis.41 Hepatocyte
metastases become symptomatic. The converse growth/scatter factor (SF), which is increased in
has also been reported: rarely, surgical removal high-grade gliomas, has been implicated both in
of the primary tumor leads to regression of motility of tumor cells, causing them to scatter
metastases, presumably because growth or in vitro, and motility of endothelial cells. SF
angiogenic factors secreted by the primary also increases permeability of the bloodglioma
tumor were required to stimulate the metas- barrier in some animal gliomas.42 Thus, the
tases. This distant stimulation may explain the protein may play a role both in tumor cell
occasional finding of a large brain metastasis invasion43 and in migration of endothelial cells
from a small, sometimes undetectable primary necessary for angiogenesis;44 it may also be
tumor (Chapter 13). A number of agents responsible for the microglial infiltration found

44
BLOODCNS BARRIERS

Table 2.4
Steps in angiogenesis.

1. Endothelial cell (EC) and pericyte


activation
2. Basal lamina degradation
3. Migration and proliferation of ECs and
pericytes
4. Formation of new capillary lumen
5. Pericytes infiltrate new capillaries
6. New basal lamina forms
7. Capillary loops form
8. Involution and differentiation of new
Figure 2.6 vessels
Endothelial proliferation in a high-grade brain tumor. 9. Capillary network formation
Two foci of vascular endothelial hyperplasia (arrows) 10. Formation of larger microvessels
are surrounded by neoplastic glial cells.

in some high-grade gliomas.45 Platelet endothe- problem in the therapy of most brain tumors
lial growth factor is expressed in some anaplas- is tumor heterogeneity. Because tumor cells are
tic gliomas and favors angiogenesis.46 genetically unstable, the tumor itself may
The overall result of angiogenesis is that the consist of a large number of cells which differ
tumor develops a vasculature sufficient to from one another in their genetic expression.
sustain growth, even though areas of hypoxia Therefore, some cells in the tumor are likely to
in high-grade tumors are common. be resistant to almost any agent or combina-
Microscopically, the tumors, particularly the tion of agents used for treatment. Even if most
high-grade ones, become hypervascular (Fig. tumor cells are sensitive, the resistant cells
2.6). A study of vascularity in a number of continue to reproduce and repopulate the
brain tumors indicates a relationship between tumor. Although endothelial cells in the tumor
the microvessel density and the biology of the differ from those in normal brain, they are
tumor. Meningiomas had a microvessel count not, like tumor cells, genetically unstable.
(MVC; highest number of microvessels in three Accordingly, if one finds an agent to which
areas of highest vascular density at 200 endothelial cells are sensitive, they should
magnification) of 28, low-grade astrocytomas remain sensitive and not develop genetic
14, anaplastic astrocytomas 42 and glioblas- mutations50 leading to resistance.
tomas 50.47 Furthermore, there are not only Table 2.4 summarizes the steps in angio-
more blood vessels but they are different in genesis.
anatomic structure. The capillaries induced by
VEGF are fenestrated rather than non-fenes-
trated which contributes to permeability of the
bloodbrain barrier within the tumor.48
BloodCNS barriers
Angiogenesis makes a particularly appealing Under normal circumstances, the brain and the
target for brain tumor therapy.33,49 A major CSF do not permit the entry of most

45
INVASION, ANGIOGENESIS AND THE BLOODBRAIN BARRIER

hydrophilic substances that circulate in the


Endothelial
blood and easily enter other organs. Entry of cell
Basal
hydrophilic substances such as peptides and lamina
proteins into brain is restricted by the
bloodbrain barrier, and entry into the CSF by
the bloodCSF barrier. Glucose, some amino
acids and some other vital compounds enter
the brain or CSF by specific transport
systems.51 Water-soluble chemotherapeutic
agents such as cytarabine and methotrexate Lumen
enter the brain poorly,52 although the physician
Tight
can substantially increase the concentration of junction
these substances in the brain by increasing the
Basal
parenteral dose. Most lipid-soluble substances lamina
enter the brain rapidly. Some examples
common in neuro-oncologic practice are
phenytoin, methadone, and 1,3-bischloro-(2-
Pericyte Astrocyte
chloroethyl)-1-nitrosourea (BCNU). footpad

Bloodbrain barrier Figure 2.7


The bloodbrain barrier is formed by capillary
The bloodbrain barrier resides in capillary endothelium and supported by surrounding
endothelial cells.53,54 Endothelial cells require structures, including the basal lamina, astrocytes and
the presence of astrocytes to maintain the pericytes. The pericyte process covers a substantial
bloodbrain barrier,55,56 and astrocytes are portion of the microvascular circumference including
the endothelial tight junctions. (From Blabanov and
present in abundance in normal brain and even
DoreDuffy57 with permission.)
in most brain tumors. The characteristics of
capillary endothelial cells that create the
bloodbrain barrier include (Fig. 2.7):
of most organs. Brain tumor blood vessels
1. Capillary endothelial cells are connected by are fenestrated.48
tight rather than gap junctions.58 The 3. Pinocytotic vesicles transport plasma or
aqueous channels through the tight interstitial fluid bidirectionally across the
junctions have diameters that are no larger endothelium.59 The density of pinocytotic
than 0.60.8 nm, restricting free diffusion vesicles in brain endothelium is about 5%
of most substances including water. Tumor of that in other organs, substantially limit-
capillaries of high-grade gliomas lack tight ing pinocytotic transport. Brain tumor and
junctions; capillaries of low-grade infiltrat- peritumoral capillaries are rich in pino-
ing tumors may retain tight junctions. cytotic vesicles.60
2. Brain capillaries lack fenestrations within 4. Mitochondria that supply the energy for
the endothelial cell, thus preventing the active transport across the endothelium are
entry of large molecules, such as proteins, 10-fold more abundant in brain endothelial
that find their way into the interstitial space cells than in the endothelium of other

46
BLOODCNS BARRIERS

organs. The increased mitochondria in the endothelial cells to form a bloodtissue


brain endothelium supply the energy for barrier. Abnormal glial cells, such as those
transport of water-soluble substances that lacking glial fibrillary acidic protein
cannot otherwise cross the bloodbrain (GFAP), or neoplastic glial cells, are
barrier and promote the expulsion of incapable of forming a normal barrier.
potentially toxic substances that accumu- 7. The extracellular matrix (ECM) also serves
late as a result of brain activity (e.g. potas- to support the bloodbrain barrier.
sium, glutamate). Brain tumor vessels have Disruption of ECM components may
a low density of mitochondria.61 disrupt the bloodbrain barrier.66,67
5. P-glycoprotein, a unique membrane glyco- 8. Peritumoral blood vessels also show
protein with a molecular mass of morphologic changes that suggest to differ-
170180 kDa, is encoded by a multi-drug ent investigators either increased perme-
resistance gene (MDR).62 P-glycoprotein is ability68 or a role in edema resolution.69
expressed either at the luminal membrane of 9. Aquaporins, a family of water channels,
cerebral endothelial cells or at astrocytic foot regulate the brains water content. They
processes62 and can be measured by PET may play a role in formation of
scanning in rats.63 Increased expression of P- bloodCNS barriers and in development
glycoprotein has been associated with resis- and restoration of brain edema.70,71
tance of some systemic tumors to several
chemotherapeutic agents. The protein
BloodCSF barrier
pumps out certain chemotherapeutic drugs,
e.g. vincristine and doxorubicin. Unlike The bloodCSF barrier differs from the blood
systemic tumors, brain tumor capillaries are brain barrier in several respects.72 It has a
characterized by the absence of P-glyco- 5000-fold smaller surface area than that of the
protein, which contributes to the impaired bloodbrain barrier. The major sites of trans-
bloodbrain barrier seen in brain tumors.64 port across the bloodCSF barrier are at the
6. Capillary endothelial cells rest on a contin- choroid plexus73 (where a substantial portion
uous basement membrane, which in turn is of the spinal fluid is secreted and the composi-
surrounded by astrocytic processes. At one tion of newly formed CSF is determined), and
time, this glial sheath was considered to be at the arachnoid villi (and, to a lesser extent,
the site of the bloodbrain barrier, but along nerve root sheaths), where spinal fluid is
electronmicroscopy has demonstrated spaces reabsorbed. Certain trace elements essential for
of 200 angstroms between astrocytic brain nutrition, such as folic acid and vitamin
processes, allowing for diffusion. Studies B12, that do not cross the bloodbrain barrier
with horseradish peroxidase, a protein may be transported into the nervous system via
marker, have shown that the barrier resides the choroid plexus and reach the brain by
at the tight junctions of endothelial cells. diffusion.74,75 Certain acidic substances such as
However, it is clear that normal astrocytes penicillin and methotrexate are transported out
are essential for capillary endothelia to of the CSF to the blood by the choroid plexus,
maintain an intact bloodbrain barrier. Co- particularly in the fourth ventricle.
culture of non-brain endothelial cells with CSF is reabsorbed by bulk flow through the
normal astrocytes or with glial cell line- arachnoid villi and, especially when intracranial
derived neurotrophic factor65 induces the pressure is increased, through lymphatics that

47
INVASION, ANGIOGENESIS AND THE BLOODBRAIN BARRIER

drain from the basal subarachnoid space into barrier from the vasculature to enter brain
cervical lymph nodes.76 The rate of CSF forma- interstitial space, or are secreted or excreted by
tion and absorption is about 0.35 ml/min. The neurons and glia, leave the nervous system by
CSF volume in the adult is approximately diffusing into the CSF and then being absorbed
150 ml, a volume achieved in children at about by bulk flow. In addition to diffusion, hydro-
age 4 years. Therefore, the total CSF volume static pressure may drive substances either
turns over approximately four times a day. A toward or away from the CSF. A brain tumor
barrier does not exist between the CSF and the and its surrounding plasma-derived edema
brain, because the ependyma lining the ventri- cause increased tissue pressure; the pressure
cles and the pia-arachnoid surrounding brain drives substances from the tumor through
and spinal cord allow free diffusion of edematous brain and normal brain, and then
substances, including proteins such as albumin. to the subarachnoid space. Conversely, when
However, the diffusion rate into and through subarachnoid pathways are blocked, pressure
brain parenchyma is considerably slower than may drive substances from CSF into brain.
that of bulk CSF flow. Furthermore, many
substances are reabsorbed by the brain capillary
bed into the systemic circulation once they have
diffused a short distance into the parenchyma. Disruption of bloodCNS
Thus, most substances introduced into the CSF barriers
do not penetrate very far into the brain or
spinal cord. The result is that drugs injected
Angiogenesis
intrathecally, while exposing leptomeningeal
tumor to high concentrations, usually fail to Although angiogenesis leads to blood vessels
reach parenchymal lesions such as brain tumors that do not have a normal bloodCNS barrier,
in significant concentration. Consequently, the degree of disruption in the barrier varies
administration of chemotherapeutic agents substantially from tumor to tumor, as well
directly into CSF is unreliable for treating intra- as within an individual tumor. Clinically,
parenchymal lesions.77 In experimental animals, bloodbrain barrier disruption is visualized by
an appreciable concentration of methotrexate the passage of intravenously injected contrast
has been shown to reach about 40% of the material into the extracellular space of the
brain within an hour after intraventricular tumor. In general, high-grade intrinsic tumors
administration, but an adequate concentration of the brain are associated with substantial
cannot be achieved in the much larger human disruption of the bloodbrain barrier and
brain. In both experimental animals and lower-grade tumors with lesser or absent
humans, white matter adjacent to CSF contains disruption (Fig. 2.8). There are, however,
the highest drug concentration, possibly notable exceptions. Low-grade tumors, such as
explaining the tendency for drug-induced pilocytic astrocytomas, are often associated
leukoencephalopathy to be periventricular. with significant bloodbrain barrier break-
The CSF also serves as the brains lymphatic down and contrast enhancement. Meningio-
system although there are lymph channels mas, benign tumors, characteristically intensely
around cranial nerves and perhaps around large contrast enhance because their blood supply
arteries at the base of the brain as well. comes from the external carotid circulation,
Substances that either cross the bloodbrain and they would not be expected to possess a

48
DISRUPTION OF BLOODCNS BARRIERS

A B C D E F G
Liquid
soluble Transport
solute solute
L-Dopa
Na
K

Second Altered cell


K Na messenger function
ATP-
ase
Na K
Dopamine
Tight junctions

H I J
Astrocyte
Neuron

Vesicular transport
Disrupted tight Diffusion through
junctions cellular fenestration

Figure 2.8
A schematic representation of the normal (top) and disrupted (bottom) bloodbrain barrier. The endothelial cell
lipid membrane (A) and tight junctions connecting endothelial cells (B) prevent the movement of polar
molecules between the blood and the brain. Lipid-soluble molecules (C) and solutes for which the endothelial
cell had transporters, such as glucose, some amino acids, and some micronutrients such as thiamine (D), cross
from blood to brain by two-way transport systems. Other substances, such as potassium and sodium, are
transported asymmetrically from either the blood to the brain or the brain to the blood (E). Some molecules
that cross the luminal endothelial membrane, such as L-DOPA (F), are metabolized within the endothelial cell
so that a metabolic product such as dopamine reaches the brain. Receptors on the endothelial surface, including
selectrins and integrins, react with circulating messengers that affect the brain via a second messenger system
(G). In patients with brain tumors or other lesions, the bloodbrain barrier is disrupted. This occurs in part via
increased vesicular transport (H), in part because the tight junctions are disrupted, allowing polar solutes to
cross from the blood to the brain (I), and in part because fenestrations in the endothelial cell allow increased
diffusion of polar solutes through the cell (J).

49
INVASION, ANGIOGENESIS AND THE BLOODBRAIN BARRIER

bloodbrain barrier. Conversely, some high- significantly reduced extension of pericytic and
grade tumors have little or no contrast glial investments. The capillaries are rich in
enhancement, particularly early in the course of pinocytotic vesicles, which suggests increased
their development. Medulloblastomas, among capillary permeability although fenestrations
the most high-grade of brain tumors, may and disruption of tight junctions have not been
sometimes be associated with minimal or observed.68 The increased permeability proba-
absent contrast enhancement. Even when the bly results from substances secreted by tumor
tumor does contrast enhance, disruption of the cells that diffuse into surrounding brain; these
barrier is rarely complete. Substantial variabil- substances can result from an inflammatory
ity is found in the degree of bloodbrain response to the tumor, or entry into the tumor
barrier breakdown in primary brain tumors via breakdown of the bloodbrain barrier.
implanted into rats. Nitrosourea (ENU)- VEGF is secreted by a variety of tumors and
induced oligodendrogliomas have an almost increases permeability of normal vessels.37
normal bloodbrain barrier, for instance, Other substances, such as free radicals,
whereas avian sarcoma virus (ASV)-induced bradykinins and arachidonic acid may be
tumors are substantially leaky.78 Although the secreted by microglia or other cells in reaction
clinician can use the degree of contrast to tumor formation. Vasoactive mediators such
enhancement as a clue to the histologic grade as glutamate, histamine and hormones may
of the tumor, it is not sufficiently dependable enter the tumor from the serum and diffuse
to eliminate the need for biopsy to establish a into normal brain, increasing permeability of
definitive histology (Chapter 3). otherwise normal brain capillaries.

Other sources of disruption


Tumor growth and its associated angiogenesis
Brain edema
are not the only mechanisms that can disrupt There is a close but incomplete correlation
the bloodbrain barrier. An increase in hydro- among the factors that cause angiogenesis and
static pressure, such as that caused by severe those that lead to disruption of the bloodbrain
arterial hypertension, can disrupt the barrier in barrier and production of brain edema.80 The
tissue compressed by tumor.79 Compression can best example is the most important angiogene-
also disrupt the barrier, interfering with venous sis factor, VEGF. This factor was originally
drainage and increasing capillary hydrostatic designated vascular permeability factor (VPF)
pressure; this is possibly one mechanism of because it increased the permeability of capil-
bloodbrain barrier disruption caused by laries, both within the brain and elsewhere in
tumors such as meningiomas that compress the the body. The increase in permeability may be
brain but do not directly invade it. mediated by nitric oxide.81 In most circum-
stances, brain tumors that cause angiogenesis
have a disrupted bloodbrain barrier, producing
Peritumoral blood vessels
contrast enhancement on MRI and substantial
Vessels surrounding brain tumors may also brain edema. However, the correlation among
show increased permeability. Peritumoral capil- these three characteristics is not complete. For
laries show an increase in endothelial surface- example, in some high-grade gliomas, angio-
connected vesicles, a thicker basal lamina and genesis, disruption of the bloodbrain barrier

50
BRAIN EDEMA

Figure 2.9
Edema versus infiltrating tumor. The panel on the left shows a tumor surrounded by edema, whereas the panel
on the right shows a non-edematous low-grade tumor. The hyperintensity on the T2-weighted image seen on
the left panel spares the cerebral cortex and the deep gray structures (arrows). The hyperintensity on the T2-
weighted image on the right involves both the subcortical structures and the cerebral cortex.

and contrast enhancement may be present but surrounding normal brain by increased hydro-
edema may be minimal or absent (Fig. 2.9). static pressure within the tumor. They may be
Similarly, a meningioma, which also requires eliminated by filtering through the white
substantial angiogenesis, usually causes little or matter and then into the cerebral ventricles or
no edema of the underlying brain. When a subarachnoid space, to be reabsorbed by bulk
meningioma does cause brain edema, it may flow of CSF.
result from secretion of VEGF by the tumor.82 The edema caused by brain tumors develops
When the bloodbrain barrier is focally primarily in white matter rather than the more
disrupted, water-soluble substances and large tightly packed gray matter with its interdigi-
molecules such as proteins can enter the brain tating dendritic processes. The edema is extra-
more easily. Because the brain has limited cellular, and follows low resistance white
lymphatic drainage, these substances are not matter fiber tracts rather than diffusing in a
easily eliminated but are often driven into spherical pattern.83 Thus, a small occipital

51
INVASION, ANGIOGENESIS AND THE BLOODBRAIN BARRIER

tumor may cause edema infiltrating along fiber the transcapillary movement of plasma-derived
tracts all the way to the tip of the temporal fluid from blood to tumor and from tumor into
lobe. Edema fluid has more water and is less surrounding brain; and (4) hydrostatic pressure
dense than normal brain, so it is relatively gradients within the extracellular tissue driving
easy to image by MR or CT scans. fluid movement from areas of bloodbrain
Characteristically, edema fluid is hypodense on barrier breakdown into surrounding normal
CT scan and T1 MRI and hyperintense on the brain. The tissue hydrostatic pressure gradients
T2-weighted MRI scan; typically, it extends and movement of edema fluid through the
like fingers into the white matter outlining the brain depend on arterial blood pressure and the
cerebral cortex (Chapter 3). Edema fluid does hydraulic conductivity of the tissue itself (white
not contrast enhance, making it easy to differ- matter to gray matter).
entiate from bulky tumor (Fig. 2.9); however, The formation within tumors of new vessels
because some glial tumors are highly invasive, that do not have a bloodbrain barrier is
isolated tumor cells are often found in edema- certainly important to the pathogenesis of
tous brain areas, several centimeters from the brain edema, but new vessel formation (i.e.
main tumor mass. Edema has an elevated angiogenesis) may not be the only way that
protein content, including albumin, and most
studies suggest that vasogenic edema, no
Table 2.5
matter how produced, is similar in composi-
Some mediators of brain edema.
tion. However, one study of human tumors
suggests that the mean serum protein content
in tissues adjacent to tumor varies consider- Mediator Proposed mechanism
ably, depending on the nature of the tumor; the
Oxygen-free radicals Peroxidation of lipid
serum protein content is high in peritumoral
membranes
edema surrounding glioblastomas and low in DNA strand breaks
edema surrounding a metastasis.84 Depletion of cellular
The mechanism by which brain tumors cause energy stores
vasogenic edema is complex and multifactorial. ? Reaction with nitric
oxide to form toxic
Increased vascular permeability to plasma
peroxynitrite
constituents, including albumin, results in: (1) Bradykinin Dilation of cerebral
an increase in conductance of osmotically vessels
active solutes and a decrease in their reflection Arachidonic acid Dilation of cerebral
coefficients across the capillary wall; (2) an vessels
alteration of the normal concentration gradient ? Production of oxygen
free radicals
of osmotically active solutes between plasma
Glutamate Cellular energy depletion
and brain extracellular fluid, by an increase in Dilation of cerebral
brain solutes, creating an imbalance between vessels
osmotic and hydrostatic pressures across brain Hormones
and tumor capillaries, favoring fluid movement Arginine vasopressin Disruption of water and
Atrial natriuretic electrolyte homeostasis
from blood to brain; (3) an increase in
peptide
hydraulic conductivity across tumor vessels and Nitric oxide? Vasodilatation
through an expanded extracellular space Histamine
between white matter fiber tracts, augmenting

52
BRAIN EDEMA

Figure 2.10
Not all intrinsic brain tumors cause significant edema nor are all tumors causing edema necessarily within the
brain. The two panels on the top show the enhanced T1- and T2-weighted images of a patient with a large
glioblastoma multiforme with no surrounding edema. Two panels (below) show the enhanced T1- and T2-weighted
images of a patient with a meningioma (arrows) surrounded by substantial edema (arrow head) (Chapter 6).

53
INVASION, ANGIOGENESIS AND THE BLOODBRAIN BARRIER

edema is formed in brain tumors. Substantial particularly vulnerable to the effects of tumors
evidence indicates that tumors secrete growing within the intracranial cavity:
substances that not only promote angiogenesis
of vessels lacking a bloodbrain barrier, but 1. The brain is enclosed in bone (the skull).
also promote leakiness of normal brain capil- As tumors grow, they distort and compress
laries in the brain immediately surrounding nearby normal brain and they raise tissue
tumor (Table 2.5).37,85,86 pressure both in the immediate area and
The neurologic symptoms that result from at a distance (i.e. overall intracranial
brain edema are often more severe than those pressure), thus interfering with neural
resulting from the tumor itself. However, not all function remote from the tumor and
intracranial tumors are accompanied by edema sometimes giving rise to false localizing
(Fig. 2.10); for example, most meningiomas signs (Chapter 3). Furthermore, tissue
(Chapter 6) do not cause edema. However, pressure is not equally distributed.
whether edema fluid in and of itself is toxic to Regional brain tissue pressure gradients
the brain or whether all of its symptoms are a develop, with the pressure highest near
consequence of mass effect compressing and the lesion. These pressure gradients may
distorting normal tissue is not clear. No clinical further serve to shift tissue from its normal
or experimental evidence has defined whether compartment.87 Mechanical compression
or not symptoms are caused by the edema fluid can initiate glial proliferation, possibly via
itself. Edema differs from normal brain extra- a tyrosine kinase pathway. This may
cellular space because it contains substances explain gliosis surrounding many brain
normally excluded from the brain. For example, tumors.88 Increased tissue pressure in the
potassium, calcium and glutamate, all tumor may retard entry of drugs into the
substances that affect neuronal function, are tumor and may promote their diffusion
found at much lower concentrations in brain away from the tumor.
extracellular space and in CSF than in edema 2. The brain lacks lymphatics. Lymph
fluid. The normal CSF/plasma ratios are 0.62, channels positioned along cranial nerves,
0.49 and 0.40, respectively. Disruption of the especially the olfactory nerve, and spinal
bloodbrain barrier increases those ratios in the roots may absorb some CSF, and macro-
brains extracellular space and could contribute molecules draining in this manner localize
to altered neural function. Positron emission in cervical lymph nodes.89 The amount,
tomography (PET) studies consistently show however, is not sufficient to prevent brain
relative hypometabolism of edematous brain edema and the resultant increased intra-
surrounding tumors that cannot be completely cranial pressure. The paucity of lymphatics
explained by a lesser cell density. The pragmatic may also play a role in preventing an
issue is that there is no question that cerebral immune response to some brain tumors.
edema is instrumental in causing symptoms in However, the lymphatic drainage into
patients with intracranial tumors; amelioration cervical lymph nodes appears sufficient in
of the edema with steroids (see below) often experimental animals to allow immune
substantially improves symptoms even before reactions. Because of the few lymphatics,
the tumor itself is treated. most of the absorption of brain edema
Two additional aspects of the anatomy and fluid can only occur by the long, tortuous
physiology of the nervous system make it route of convection or bulk flow through

54
TREATMENT OF CEREBRAL HERNIATION

the brain substance to the ventricular clinical consequences of these herniations are
system or subarachnoid space, where it is described in Chapter 3. Focally increased
eventually absorbed along with the CSF. tissue pressure can also cause symptoms by
Unfortunately, the tumor, with its increased interfering with the local blood supply. Both
intracranial pressure and elevated CSF mechanisms are probably responsible for
proteins, often reduces the capacity of many of the symptoms caused by brain
normal CSF absorptive pathways, thus tumors.
compounding the problem of brain edema. Some patients suffer intermittent episodes of
neurologic dysfunction that result from sudden
rises in intracranial pressure called plateau
Consequences of barrier waves.9193 These episodes last 520 min and
then cease. The waves, first described by
disruption and brain edema Lundberg92 in 1960 (Fig. 3.6), appear to result
from an increase in cerebral blood volume due
Increased intracranial pressure, to a sudden decrease in cerebral vascular resis-
plateau waves and cerebral tance;94,95 blood flow may actually decrease as
the blood volume rises. Patients with an
herniation
intracranial mass and an elevated intracranial
In those patients with mass lesions involving baseline pressure resulting in diminished CSF
or compressing nervous system structures, the absorption are more likely to develop plateau
breakdown of bloodCNS barriers with the waves than those with normal CSF absorp-
production of brain edema often leads to tion.96
increased intracranial pressure. The pressure Plateau waves can either occur sponta-
may be distributed evenly throughout the neously or be precipitated by certain activities.
brain, such as when obstruction of the Common precipitating causes include tracheal
superior vena cava or sagittal sinus raises suctioning, coughing or sneezing, and, particu-
venous pressure and thus intracranial larly, rising from a lying or sitting position,97
pressure uniformly, or pressure may be especially in the early morning after a nights
distributed unevenly, as exemplified in sleep. Plateau waves are often asymptomatic
patients with focal mass lesions.90 Evenly even when they cause a dramatic increase in
distributed increased intracranial pressure intracranial pressure. However, at times the
probably causes no symptoms until the symptoms can be dramatic and varied (Chapter
intracranial pressure approaches the arterial 3). Plateau waves respond rapidly to relief of
blood pressure, resulting in cerebral ischemia. raised intracranial pressure either by steroids or
Focally increased tissue pressure causes herni- other mechanisms.
ation of portions of normal brain into areas
of lower pressure. The most common sites of
herniation, as illustrated in Fig. 3.2, include Treatment of cerebral
herniation of the medial frontal lobe under
the falx cerebri, herniation of the medial
herniation
temporal lobe through the tentorium Immediate treatment of increased intracranial
cerebelli, and herniation of the cerebellar pressure is required to reverse or prevent
tonsils through the foramen magnum. The cerebral herniation and to prevent death. The

55
INVASION, ANGIOGENESIS AND THE BLOODBRAIN BARRIER

treatment of increased intracranial pressure solution at a dose of 0.52 g/kg.99 This drug
and cerebral edema includes hyperventilation, may also decrease CSF formation and
hyperosmolar agents, and adrenocorticos- volume.100 In a few patients with severe and
teroids. sustained increased intracranial pressure,
pressure can be monitored and the dose
Hyperventilation tailored to maintain decreased pressure.99 In
patients with normal intracranial pressure,
Hyperventilation is the most rapid technique mannitol may actually increase the pressure
for lowering intracranial pressure and revers- briefly, probably due to a transient increase in
ing herniation, but its effect is short-lived. cerebral blood flow and volume. This does
Hyperventilation lowers pCO2, causing not appear to occur in patients with increased
cerebral vasoconstriction and decreasing intracranial pressure.99,101 The mannitol effects
cerebral blood volume. If the patient is uncon- are rapid and last several hours. Mannitol
scious, an endotracheal tube should be injections may be repeated in smaller doses if
inserted and the patient ventilated to a pCO2 the patient first responds and subsequently
level between 2530 mm Hg, which lowers relapses. Glycerol, urea and hypertonic
intracranial pressure rapidly in most patients saline102,103 are less widely used. Repeated
but only transiently, with a return to baseline doses of mannitol can also cause a reverse
blood volume in about an hour. Many osmotic effect as the compound leaks into
patients with cerebral herniation sponta- edematous brain. The addition of a diuretic
neously hyperventilate to these levels and (furosemide) enhances and prolongs the
require no additional respiratory intervention mannitol effect.104
from the physician. However, intubation is
advisable to protect the airway. Respiratory Corticosteroids
function must be monitored carefully, because
brain herniation can cause respiratory failure. Corticosteroids decrease the transfer across
Mechanical ventilation can raise as well as the disrupted bloodbrain barrier of serum
lower intracranial pressure, and ventilated substances into brain. They lower intracra-
patients with brain lesions are at risk,98 nial pressure, diminish plateau waves and
particularly when intrathoracic pressure is eventually decrease edema (see below).
kept high, diminishing venous return. The Dexamethasone (100 mg IV) is given
increased intrathoracic pressure is transmitted immediately, followed by doses of
to the superior vena cava and to intracranial 40100 mg/24 h, depending on the patient's
vessels. response to treatment. Some physicians add
furosemide (40120 mg IV) to the steroids
and believe that the combination is better
Osmolar therapy
than steroids alone;102 however, intravascu-
Hyperosmolar agents decrease the water lar volume depletion must be closely
content of the brain by creating an osmolar watched. With such vigorous treatment,
gradient between the blood and that portion most patients who herniate from brain
of the brain with an intact bloodbrain tumors can be stabilized, and many have
barrier. The agent of choice is mannitol given complete amelioration of their symptoms
intravenously over 1020 min as a 20% within a few days.

56
TREATMENT OF BRAIN EDEMA

Treatment of brain edema is also less likely than other synthetic steroids
to cause cognitive and behavioral dysfunc-
Several drugs, including glutamate inhibitors, tion.101
non-steroidal anti-inflammatory agents (e.g. Only a few experiments address a dose-
indomethacin) and lazeroids (21-aminosteroids response curve for CNS effects of corticos-
that have antioxidant but not glucocorticoid teroids. One such experiment in an animal
activity) have also been proposed for the model of spinal cord compression suggests that
treatment of brain and spinal cord edema,105 doses equivalent to 100 mg/24 h of dexa-
but their clinical efficacy is unproved. methasone may be superior to lesser doses both
Corticotropin-releasing factor (CRF) has in decreasing edema and in ameliorating clini-
proved effective in ameliorating tumor-induced cal symptomatology.112 Because reports thus far
brain edema in experimental animals; the give no indication that these higher doses given
mechanism of action is not known. The edema- for a few days are deleterious, our practice is
ameliorating effects are not a result of CRF- to use high doses in seriously symptomatic or
induced cortisone release, because CRF is deteriorating patients, tapering the dose after a
effective in adrenalectomized brain tumor- few days to the lowest dose consistent with
bearing animals.106,107 An arginine vasopressin symptomatic control.113
receptor antagonist may also prove useful.86 Steroid treatment begins with the physician
However, at present, corticosteroids are the selecting a dose appropriate for the neurologic
drug of first choice to treat tumor-induced disorder and its severity (see below). Because
brain edema. of its long half-life, dexamethasone can be
given twice daily, although most physicians
give four divided doses. The drug is well
Corticosteroids absorbed from the gastrointestinal tract, but
The most widely used drugs in neuro-oncology first-pass hepatic metabolism may decrease the
are the synthetic glucocorticoids, commonly effectiveness of an oral dose, especially in
referred to simply as steroids.108 They are the patients taking phenytoin. Because side-effects
mainstay of treatment for nervous system are numerous and often serious, patients
edema and increased intracranial pressure. The should be maintained on the lowest dose of
optimum dose and best preparation of corti- steroid that affords relief of symptoms. Thus,
costeroids are not established.109 The dosage once symptomatic control is established and
probably should differ both with the nature of more definitive therapy (e.g. surgery, radiation,
the problem and its severity.110 and chemotherapy) is underway, the steroid
The steroid most widely used remains should be tapered to the lowest possible dose
dexamethasone, largely because it was the (see below).
drug whose usage was established by Galicich
and French111 to treat brain tumors.
Steroid taper
Dexamethasone may be preferred to other
synthetic glucocorticoids for several theoretical Because of the deleterious effects of corticos-
reasons. First, it has no mineralocorticoid teroids (Chapter 4), patients should be treated
effect, and therefore is the least likely steroid with the smallest effective dose for the short-
to cause salt retention and systemic edema est time possible. Virtually all patients begun
formation. Second, some investigators believe it on corticosteroid therapy for brain tumors are

57
INVASION, ANGIOGENESIS AND THE BLOODBRAIN BARRIER

treated subsequently with either radiation Mechanisms of steroid action


therapy (RT) or chemotherapy. Patients with
brain tumors usually continue steroids during The mechanisms by which steroids stabilize
the course of RT, but the dose can be bloodbrain, bloodspinal cord, and probably
decreased after the first week and gradually bloodCSF barriers are not entirely known,
tapered thereafter. Most patients can be off although several have been proposed.
steroids by the completion of the RT, and the Glucocorticoids inhibit the production or release
remainder should be weaned from the steroids of a number of biochemical substances shown
entirely if possible. The steroid taper begins to increase vascular permeability and induce
34 days after surgery or during week 2 of vasodilatation (an effect that, by increased
RT and should be tapered gradually enough hydrostatic pressure, also increases permeabil-
to prevent the development of steroid ity). Glucocorticoids induce formation of
withdrawal symptoms, but rapidly enough so lipocortins, which inhibit phosphorylase A2,
that the patient is not taking the drugs for an thus preventing the release of arachidonic
extended period. For patients receiving 16 mg acid.115 Arachidonic acid and its metabolites
dexamethasone, the drug should be tapered increase vascular permeability; thus, the reduc-
by 24 mg every fifth day. If at any time tion of arachidonic acid by steroids might
during the taper the patient develops reduce brain edema. Nonetheless, other
symptoms of either brain tumor or steroid inhibitors of this pathway, such as
withdrawal, the drug is increased to the next indomethacin, do not stabilize the bloodbrain
higher dose for 48 days before tapering barrier or ameliorate the symptoms of brain
again. If, after the patient is withdrawn, brain tumors as effectively as corticosteroids. Steroids
tumor symptoms develop, it is probably wise inhibit the release of IL-1; whether the inter-
to start the full regimen of 16 mg/24 h dexa- leukins play a role in bloodbrain barrier break-
methasone. down is not known. Steroids also appear to have
For patients who have been on steroids for a direct effect on endothelial cells, increasing
many months, who fail the usual taper sched- resistance to transendothelial fluid flow and
ule, or who have large amounts of residual increasing the number of tight junctions.116 In
tumor, the drug is tapered more slowly (e.g. several organisms, steroids inhibit the increased
12 mg/week) to the lowest dose tolerable. For permeability that results from endothelial cell
patients taking large doses of steroids (e.g. interaction with a number of chemical agents.117
100 mg/24 h dexamethasone) who have stabi- Experimental evidence suggests that steroids can
lized and are receiving more definitive treat- induce the synthesis of a protein that inhibits
ment, the dose can be halved every 45 days microvascular permeability, a direct action on
depending on the patients clinical state. The the endothelial cell. It appears that the inhibitory
dose should be raised again if the clinical protein is distinct from lipocortin and, thus, the
condition deteriorates. effect is independent from the inhibition of
Weissman et al114 propose a more rapid taper phosphorylase A2. Whatever their mechanisms,
schedule, beginning at 16 mg/day (8 mg bid) steroids appear to be unique in their ability to
for 4 days followed by 8 mg/24 h for 4 days ameliorate clinical symptoms and stabilize the
and 4 mg/24 h until completion of RT. This bloodbrain barrier. Clinically, steroid-induced
procedure was well tolerated by their patients reconstitution of the bloodbrain barrier can
who had brain metastases. occasionally be visualized on neuroimaging.

58
REFERENCES

Contrast enhancement can diminish in tumors and its Coverings, 3rd edn. Churchill
after initiation of dexamethasone but before the Livingstone, New York, 1991.
4. Kelly PJ, Daumas-Duport C, Scheithauer BW.
start of definitive treatment. Sometimes, this is
Stereotactic histologic correlations of computed
misinterpreted as a response to treatment if the tomography and magnetic resonance imaging-
steroid-induced diminution of enhancement is defined abnormalities in patients with glial
not appreciated or recognized. neoplasms. Mayo Clin Proc 1987; 62: 45059.
In addition to restoring capillary imperme- 5. Gary SC, Hockfield S. BEHAB/brevican: an
ability,118 dexamethasone also decreases extracellular matrix component associated
with invasive glioma. Clin Neurosurg 2000;
cerebral blood flow and volume119,120 and 47: 7282.
increases the fractional extraction of oxygen 6. Asano K, Kubo O, Tajika Y, Takakura K,
throughout the brain without affecting oxygen Suzuki S. Expression of cadherin and CSF
utilization. Reports suggest that the drug dissemination in malignant astrocytic tumors.
probably has a direct vasoconstrictive effect on Neurosurg Rev 2000; 23: 3944.
7. Tews DS. Adhesive and invasive features in
cerebral blood vessels.121 Dexamethasone treat- gliomas. Pathol Res Pract 2000; 196: 70111.
ment reduces or eliminates the filtration of 8. Tonn JC, Wunderlich S, Kerkau S, Klein CE,
plasma-derived fluid across tumor capillaries Roosen K. Invasive behaviour of human
and the movement of albumin through the gliomas is mediated by interindividually
extracellular space by solvent drag. These different integrin patterns. Anticancer Res
1998; 18: 2599605.
effects may be mediated by reducing the size of 9. Hallahan DE, Staba-Hogan MJ, Virudachalam
the extracellular space or decreasing the pore S, Kolchinsky A. X-ray-induced P-selectin
size of tumor capillaries, which probably repre- localization to the lumen of tumor blood
sents an important mechanism for cortico- vessels. Cancer Res 1998; 58: 521620.
steroid control of peritumoral brain edema.122 10. Sasaki H, Yoshida K, Ikeda E, et al. Expression
of the neural cell adhesion molecule in astro-
Although most studies have not demonstrated
cytic tumors an inverse correlation with
an effect of dexamethasone on the bloodbrain malignancy. Cancer 1998; 82: 192131.
barrier of normal brain, at least one study123 11. Sehgal A, Boynton AL, Young RF et al. Cell
suggests that dexamethasone may decrease the adhesion molecule Nr-CAM is over-expressed
permeability to macromolecules of normal in human brain tumors. Int J Cancer 1998;
76: 4518.
cerebral vasculature, possibly by interfering
12. Yap AS. The morphogenetic role of cadherin
with vesicular transport.123 cell adhesion molecules in human cancer: a
thematic review. Cancer Invest 1998; 16:
25261.
References 13. Thorsen F, Tysnes BB. Brain tumor cell
invasion, anatomical and biological consider-
1. Milosevic MF, Fyles AW, Hill RP. The ations. Anticancer Res 1997; 17: 41216.
relationship between elevated interstitial fluid 14. Gunia S, Hussein S, Radu DL et al. CD44s-
pressure and blood flow in tumors: a bioengi- targeted treatment with monoclonal antibody
neering analysis. Int J Radiat Oncol Biol Phys blocks intracerebral invasion and growth of
1999; 43: 111123. 9L gliosarcoma. Clin Exp Metastasis 1999;
2. Boucher Y, Salehi H, Witwer B, Harsh GR, 17: 22130.
Jain RK. Interstitial fluid pressure in intra- 15. Senner V, Sturm A, Baur I et al. CD24
cranial tumours in patients and in rodents. promotes invasion of glioma cells in vivo. J
Br J Cancer 1997; 75: 82936. Neuropathol Exp Neurol 1999; 58: 795802.
3. Burger PC, Scheithauer BW, Vogel FS. 16. Nakada M, Kita D, Futami K et al. Roles of
Surgical Pathology of the Nervous System membrane type 1 matrix metalloproteinase and

59
INVASION, ANGIOGENESIS AND THE BLOODBRAIN BARRIER

tissue inhibitor of metalloproteinases 2 in factor in human brain tumors. Acta


invasion and dissemination of human malignant Neuropathol (Berl) 1998; 96: 45362.
glioma. J Neurosurg 2001; 94: 46473. 28. Carroll RS, Zhang JP, Bello L et al. KDR
17. Forsyth PA, Wong H, Laing TD et al. activation in astrocytic neoplasms. Cancer
Gelatinase-A (MMP-2), gelatinase-B (MMP- 1999; 86: 133541.
9) and membrane type matrix metallopro- 29. Machein MR, Kullmer J, Fiebich BL et al.
teinase-1 (MT1-MMP) are involved in Vascular endothelial growth factor expres-
different aspects of the pathophysiology of sion, vascular volume, and capillary perme-
malignant gliomas. Br J Cancer 1999; 79: ability in human brain tumors. Neurosurgery
182835. 1999; 44: 73240.
18. Kachra Z, Beaulieu E, Delbecchi L et al. 30. Zagzag D, Zhong H, Scalzitti JM et al.
Expression of matrix metalloproteinases and Expression of hypoxia-inducible factor 1 in
their inhibitors in human brain tumors. Clin brain tumors association with angiogensis,
Exp Metastasis 1999; 17: 55566. invasion and progression. Cancer 2000; 88:
19. Harkness KA, Adamson P, Sussman JD et al. 260618.
Dexamethasone regulation of matrix metal- 31. Joki T, Heese O, Nikas DC et al. Expression
loproteinase expression in CNS vascular of cyclooxygenase 2 (COX-2) in human
endothelium. Brain 2000; 123: 698709. glioma and in vitro inhibition by a specific
20. Fillmore HL, Shurm J, Furqueron P et al. An COX-2 inhibitor, NS-398. Cancer Res 19.
in vivo rat model for visualizing glioma 2000; 60: 492631.
tumor cell invasion using stable persistent 32. Schirner M. Antiangiogenic chemothera-
expression of the green fluorescent protein. peutic agents. Cancer Metastasis Rev 2000;
Cancer Lett 1999; 141: 919. 19: 6773.
21. Bayer TA, Thier M, Roeb E et al. Migratory 33. Folkman J. Antiangiogenic gene therapy. Proc
potential of transplantable neural tumor cell Natl Acad Sci USA 1998; 95: 906466.
lines. Acta Neuropathol (Berl) 1999; 97: 34. Lund EL, Spang-Thomsen M, Skovgaard-
60712. Poulsen H, Kristjansen PE. Tumor angio-
22. Molnar PP, O'Neil BP, Scheithauer BW, genesis a new therapeutic target in gliomas.
Groothuis DR. The bloodbrain barrier in Acta Neurol Scand 1998; 97: 5262.
primary CNS lymphomas: ultrastructural 35. Strik HM, Schluesener HJ, Seid K,
evidence of endothelial cell death. Meyermann R, Deininger MH. Localization
Neurooncol 1999; 1: 89100. of endostatin in rat and human gliomas.
23. Hochberg FH, Pruitt A. Assumptions in the Cancer 2001; 91: 101319.
radiotherapy of glioblastoma. Neurology 36. Sckell A, Safabakhsh N, Dellian M, Jain RK.
1980; 30: 90711. Primary tumor size-dependent inhibition of
24. Wild-Bode C, Weller M, Rimner A, Dichgans angiogenesis at a secondary site: an intravital
J, Wick W. Sublethal irradiation promotes microscopic study in mice. Cancer Res 1998;
migration and invasiveness of glioma cells: 58: 58669.
implications for radiotherapy of human 37. Oku T, Tjuvajev JG, Miyagawa T et al.
glioblastoma. Cancer Res 2001; 61: 274450. Tumor growth modulation by sense and
25. Loeffler JS, Alexander E, Hochberg FH et al. antisense vascular endothelial growth factor
Clinical patterns of failure following stereo- gene expression: effects on angiogenesis,
tactic interstitial irradiation for malignant vascular permeability, blood volume, blood
gliomas. Int J Radiat Oncol Biol Phys 1990; flow, fluorodeoxyglucose uptake, and pro-
19: 145562. liferation of human melanoma intracerebral
26. Dang CV, Semenza GL. Oncogenic alter- xenografts. Cancer Res 1998; 58: 418592.
ations of metabolism. Trends Biochem Sci 38. Shiratsuchi T, Nishimori H, Ichise H,
1999; 24: 6872. Nakamura Y, Tokino T. Cloning and charac-
27. Nishikawa R, Cheng SY, Nagashima R et al. terization of BAI2 and BAI3, novel genes
Expression of vascular endothelial growth homologous to brain-specific angiogenesis

60
REFERENCES

inhibitor 1 (BAI1). Cytogenet Cell Genet 50. Hanahan D. A flanking attack on cancer. Nat
1997; 79: 1038. Med 1998; 4: 1314.
39. Czubayko F, Liaudet-Coopman ED, Aigner 51. Tamai I, Tsuji A. Transporter-mediated
A et al. A secreted FGF-binding protein can permeation of drugs across the bloodbrain
serve as the angiogenic switch in human barrier. J Pharm Sci 2000; 89: 137188.
cancer. Nat Med 1997; 3: 113740. 52. Habgood MD, Begley DJ, Abbott NJ.
40. Nomura M, Yamagishi S, Harada S et al. Determinants of passive drug entry into the
Placenta growth factor (PlGF) mRNA expres- central nervous system. Cell Mol Neurobiol
sion in brain tumors. J Neurooncol 1998; 40: 2000; 20: 23153.
12330. 53. Drewes LR. What is the bloodbrain barrier?
41. Jallo GI, Friedlander DR, Kelly PJ et al. A molecular perspective Cerebral vascular
Tenascin-C expression in the cyst wall and biology. Adv Exp Med Biol 1999; 474:
fluid of human brain tumors correlates with 11122.
angiogenesis. Neurosurgery 1997; 41: 54. Li JY, Boado RJ, Pardridge WM. Bloodbrain
10529. barrier genomics. J Cereb Blood Flow Metab
42. Book AA, Ranganathan S, Abounader R, 2001; 21: 618.
Rosen E, Laterra J. Scatter factor hepatocyte 55. Schroeter ML, Mertsch K, Giese H et al.
growth factor gene transfer increases rat Astrocytes enhance radical defence in capil-
bloodglioma barrier permeability. Brain Res lary endothelial cells constituting the
1999; 833: 17380. blood brain barrier. FEBS Lett 1999; 449:
43. Welch WC, Kornblith PL, Michalopoulos GK 2414.
et al. Hepatocyte growth factor (HGF) and 56. Kuchler-Bopp S, Delaunoy JP, Artault JC,
receptor (c-met) in normal and malignant Zaepfel M, Dietrich JB. Astrocytes induce
astrocytic cells. Anticancer Res 1999; 19: several bloodbrain barrier properties in non-
163540. neural endothelial cells. Neuroreport 1999;
44. Lamszus K, Schmidt NO, Jin L et al. Scatter 10: 134753.
factor promotes motility of human glioma 57. Balabanov R, Dore-Duffy P. Role of the CNS
and neuromicrovascular endothelial cells. Int microvascular pericyte in the blood brain
J Cancer 1998; 75: 1928. barrier. J Neurosci Res 1998; 53: 63744.
45. Badie B, Schartner J, Klaver J, Vorpahl J. In 58. Kniesel U, Wolburg H. Tight junctions of the
vitro modulation of microglia motility by bloodbrain barrier. Cell Mol Neurobiol
glioma cells is mediated by hepatocyte growth 2000; 20: 5776.
factor scatter factor. Neurosurgery 1999; 44: 59. Stewart PA. Endothelial vesicles in the
107782. blood brain barrier: are they related to
46. Aroca F, Renaud W, Bartoli C, Bouvier- permeability? Cell Mol Neurobiol 2000; 20:
Labit C, Figarella-Branger D. Expression 14963.
of PECAM-1/CD31 isoforms in human 60. Vaz R, Borges N, Cruz C, Azevedo I.
brain gliomas. J Neurooncol 1999; 43: Cerebral edema associated with menin-
1925. giomas: the role of peritumoral brain tissue.
47. Assimakopoulou M, Sotiropoulou-Bonikou J Neurooncol 1998; 36: 28591.
G, Maraziotis T, Papadakis N, Varakis I. 61. Coomber BL, Stewart PA, Hayakawa EM,
Microvessel density in brain tumors. Farrell CL, Del Maestro RF. A quantitative
Anticancer Res 1997; 17: 474753. assessment of microvessel ultrastructure in C6
48. Roberts WG, Palade GE. Neovasculature astrocytoma spheroids transplanted to brain
induced by vascular endothelial growth factor and to muscle. J Neuropathol Exp Neurol
is fenestrated. Cancer Res 1997; 57: 76572. 1988; 47: 2940.
49. Malonne H, Langer I, Kiss R, Atassi G. 62. Golden PL, Pardridge WM. Brain microvascu-
Mechanisms of tumor angiogenesis and thera- lar P-glycoprotein and a revised model of
peutic implications: angiogenesis inhibitors. multidrug resistance in brain. Cell Mol
Clin Exp Metastasis 1999; 17: 114. Neurobiol 2000; 20: 16581.

61
INVASION, ANGIOGENESIS AND THE BLOODBRAIN BARRIER

63. Hendrikse NH, De Vries EGE, Eriks-Fluks L plexus epithelial cells in culture: regulation of
et al. A new in vivo method to study P-glyco- barrier properties and transport processes.
protein transport in tumors and the blood Microsc Res Tech 2001; 52: 137152.
brain barrier. Cancer Res 1999; 59: 241116. 76. Boulton M, Armstrong D, Flessner M, Hay J,
64. Jolliet-Riant P, Tillement JP. Drug transfer Szalai JP, Johnston M. Raised intracranial
across the bloodbrain barrier and pressure increases CSF drainage through
improvement of brain delivery. Fundam arachnoid villi and extracranial lymphatics.
Clin Pharmacol 1999; 13: 1626. Am J Physiol Regul Integr Comp Physiol
65. Igarashi Y, Utsumi H, Chiba H et al. Glial 1998; 275: R889R896.
cell line-derived neurotrophic factor induces 77. Grossman SA, Reinhard CS, Loats HL. The
barrier function of endothelial cells forming intracerebral penetration of intraventricularly
the bloodbrain barrier. Biochem Biophys administered methotrexate: A quantitative
Res Commun 1999; 261: 10812. autoradiographic study. J Neurooncol 1989;
66. Robert AM, Robert L. Extracellular matrix 7: 31928.
and bloodbrain barrier function. Pathol Biol 78. Molnar P, Blasberg RG, Groothuis D, et al.
(Paris) 1998; 46: 53542. Regional blood-to-tissue transport in avian
67. Yong VW, Krekoski CA, Forsyth PA, Bell R, sarcoma virus (ASV)-induced brain tumors.
Edwards DR. Matrix metalloproteinases and Neurology 1983; 33: 70211.
diseases of the CNS. Trends Neurosci 1998; 79. Suzuki N, Sako K, Yonemasu Y. Effects of
21: 7580. induced hypertension on blood flow and
68. Vaz R, Borges N, Cruz C, Azevedo I. capillary permeability in rats with experi-
Cerebral edema associated with menin- mental brain tumors. J Neurooncol 1991; 10:
giomas: the role of peritumoral brain tissue. 21318.
J Neurooncol 1998; 36: 28591. 80. Schilling L, Wahl M. Mediators of cerebral
69. Bertossi M, Virgintino D, Maiorano E, edema. Adv Exp Med Biol 1999; 474:
Occhiogrosso M, Roncali L. Ultrastructural 12341.
and morphometric investigation of human 81. Mayhan WG. VEGF increases permeability of
brain capillaries in normal and peritumoral the bloodbrain barrier via a nitric oxide
tissues. Ultrastruct Pathol 1997; 21: 419. synthase/cGMP-dependent pathway. Am J
70. Venero JL, Vizuete ML, Machado A, Cano J. Physiol Cell Physiol 1999; 276: C114853.
Aquaporins in the central nervous system. 82. Goldman CK, Bharara S, Palmer CA et al.
Prog Neurobiol 2001; 63: 32136. Brain edema in meningiomas is associated
71. Nico B, Frigeri A, Nicchia GP et al. Role of with increased vascular endothelial growth
aquaporin-4 water channel in the develop- factor expression. Neurosurgery 1997; 40:
ment and integrity of the bloodbrain barrier. 126977.
J Cell Sci 2001; 114: 12971307. 83. Geer CP, Grossman SA. Interstitial fluid flow
72. Saunders NR, Habgood MD, Dziegielewska along white matter tracts: A potentially
KM. Barrier mechanisms in the brain, I. important mechanism for the dissemination
Adult brain. Clin Exp Pharmacol Physiol of primary brain tumors. J Neurooncol 1997;
1999; 26: 1119. 32: 193201.
73. Segal MB. The choroid plexuses and the 84. Bodsch W, Rommel T, Ophoff BG, et al.
barriers between the blood and the Factors responsible for the retention of fluid
cerebrospinal fluid. Cell Mol Neurobiol in human tumor edema and the effect of
2000; 20: 18396. dexamethasone. J Neurosurg 1987; 67:
74. Ghersi-Egea JF, Strazielle N. Brain drug deliv- 2507.
ery, drug metabolism, and multidrug resis- 85. Strugar J, Rothbart D, Harrington W,
tance at the choroid plexus. Microsc Res Criscuolo GR. Vascular permeability factor in
Tech 2001; 52: 838. brain metastases: correlation with vasogenic
75. Haselbach M, Wegener J, Decker S, brain edema and tumor angiogenesis. J
Engelbertz C, Galla HJ. Porcine choroid Neurosurg 1994; 81: 5606.

62
REFERENCES

86. Bemana I, Nagao S. Treatment of brain 98. Colice GL. How to ventilate patients when
edema with a nonpeptide arginine vasopressin ICP elevation is a risk: Monitor pressure,
V1 receptor antagonist OPC-21268 in rats. consider hyperventilation therapy. J Crit
Neurosurgery 1999; 44: 14854. Illness 1993; 8: 100320.
87. Wolfla CE, Luerssen TG. Brain tissue 99. Ravussin P, Abou-Madi M, Archer D, et al.
pressure gradients are dependent upon a Changes in CSF pressure after mannitol in
normal spinal subarachnoid space. Acta patients with and without elevated CSF
Neurochir Suppl (Wien) 1998; 71: 31012. pressure. J Neurosurg 1988; 69: 86976.
88. Oishi Y, Uezono Y, Yanagihara N et al. 100. Donato T, Shapria Y, Artru A, et al. Effect
Transmural compression-induced prolifera- of mannitol on cerebrospinal fluid dynamics
tion and DNA synthesis through activation of and brain tissue edema. Anesth Analg 1994;
a tyrosine kinase pathway in rat astrocytoma 78: 5866.
RCR-1 cells. Brain Res 1998; 781: 15966. 101. Fishman RA. Cerebrospinal Fluid in Diseases
89. Weller RO, Engelhardt B, Phillips MJ. of the Nervous System, 2nd Edn. W.B.
Lymphocyte targeting of the central nervous Saunders Co., Philadelphia, 1992.
system: a review of afferent and efferent 102. Rottenberg DA, Hurwitz BJ, Posner JB. The
CNS-immune pathways. Brain Pathol 1996; effect of oral glycerol on intraventricular
6: 27588. pressure in man. Neurology 1977; 27: 6008.
90. Weaver DD, Winn HR, Jane JA. Differential 103. Qureshi AI, Suarez JI. Use of hypertonic
intracranial pressure in patients with unilateral saline solutions in treatment of cerebral
mass lesions. J Neurosurg 1982; 56: 6605. edema and intracranial hypertension. Crit
91. Hayashi M, Handa Y, Kobayashi H, et al. Care Med 2000; 28: 330113.
Plateau-wave phenomenon (I). Correlation 104. Pollay M, Fullenwider C, Roberts PA, et al.
between the appearance of plateau waves and Effect of mannitol and furosemide on blood
CSF circulation in patients with intracranial brain osmotic gradient and intracranial
hypertension. Brain 1992; 114: 268191. pressure. J Neurosurg 1983; 59: 94550.
92. Lundberg N. Continuous recording and 105. Kondziolka D, Mori Y, Martinez AJ et al.
control of ventricular fluid pressure in neuro- Beneficial effects of the radioprotectant 21-
surgical practice. Acta Neurol Scand 1960; aminosteroid U-74389G in a radiosurgery rat
Supp 149: 1193. malignant glioma model. Int J Radiat Oncol
93. Rottenberg DA, Posner JB. Intracranial Biol Phys 1999; 44: 17984.
pressure control. In: Cottrell JE, Turndorf H. 106. Tjuvajev J, Uehara H, Desai R et al.
(eds) Anesthesia and Neurosurgery. C.V. Corticotropin releasing factor as an alterna-
Mosby Co., St. Louis, 1980. tive treatment of peritumoral brain edema. In
94. Hayashi M, Kobayashi H, Handa Y, et al. Ito S (ed). Intracranial Pressure IX, Springer
Brain blood volume and blood flow in Verlag, Tokyo, 1994.
patients with plateau waves. J Neurosurg 107. Villalona-Calero MA, Eckardt J, Burris H et
1985; 63: 55661. al. A phase I trial of human corticotropin-
95. Matsuda M, Yoneda S, Handa H, et al. releasing factor (hCRF) in patients with peritu-
Cerebral hemodynamic changes during moral brain edema. Ann Oncol 1998; 9: 717.
plateau waves in brain-tumor patients. J 108. Weissman DE. Glucocorticoid treatment for
Neurosurg 1979; 50: 4838. brain metastases and epidural spinal cord
96. Hayashi M, Kobayashi H, Handa Y, et al. compression: A review. J Clin Oncol 1988; 6:
Plateau-wave phenomenon (II). Occurrence of 54351.
brain herniation in patients with and without 109. Oka K, Shimodaira H. Telepharmacodynamics
plateau waves. Brain 1991; 114: 26939. to predict therapeutic effects of glucocorti-
97. Magnaes B. Body position and cerebrospinal coids. Letter to the Editor. Lancet 1991; 338:
fluid pressure. Part I: Clinical studies on the 385.
effect of rapid postural changes. J Neurosurg 110. Renaudin J, Fewer D, Wilson CB, et al. Dose
1976; 44: 687697. dependency of decadron in patients with

63
INVASION, ANGIOGENESIS AND THE BLOODBRAIN BARRIER

partially excised brain tumors. J Neurosurg 117. Weissman DE, Stewart C. Experimental drug
1973; 39: 3025. therapy of peritumoral brain edema. J Neuro
111. Galicich JH, French LA. Use of dexametha- oncol 1988; 6: 33942.
sone in the treatment of cerebral edema result- 118. Williams TJ, Yarwood H. Effect of glucocor-
ing from brain tumors and brain surgery. Am ticosteroids on microvascular permeability.
Practit-Dig Treat 1961; 12: 16974. Am Rev Respir Dis 1990; 141: S39S43.
112. Delattre J-Y, Arbit E, Thaler HT, Rosenblum 119. Jarden JO, Dhawan V, Moeller JR, et al. The
MK, Posner JB. A dose-response study of time course of steroid action on blood-to-
dexamethasone in a model of spinal cord brain and blood-to-tumor transport of 82Rb:
compression caused by epidural tumor. J a positron emission tomographic study. Ann
Neurosurg 1989; 70: 9205. Neurol 1989; 25: 23945.
113. Lieberman A, LeBrun Y, Glass P, et al. Use 120. Tajima A, Yen M-H, Nakata H, et al. Effects
of high-dose corticosteroids in patients with of dexamethasone on blood flow and volume
inoperable brain tumors. J Neurol Neurosurg of perfused microvessels in traumatic brain
Psychiatry 1977; 40: 67882. edema. Adv Neurol 1990; 52: 34350.
114. Weissman DE, Janjan NA, Erickson B, et al. 121. Leenders KL, Beaney RP, Brooks DJ, et al.
Twice-daily tapering dexamethasone treat- Dexamethasone treatment of brain tumor
ment during cranial radiation for newly patients: Effects on regional cerebral blood
diagnosed brain metastases. J Neurooncol flow, blood volume and oxygen utilization.
1991; 11: 2359. Neurology 1985; 35: 161016.
115. Chan PH, Fishman RA. The role of arachi- 122. Nakagawa H, Groothuis DR, Owens ES, et
donic acid in vasogenic brain edema. Fed Proc al. Dexamethasone effects on [125I] albumin
1984; 43: 21013. distribution in experimental RG-2 gliomas
116. Underwood JL, Murphy CG, Chen J et al. and adjacent brain. J Cereb Blood Flow
Glucocorticoids regulate transendothelial Metab 1987; 7: 687701.
fluid flow resistance and formation of inter- 123. Hedley-Whyte ET, Hsu DW. Effect of dexa-
cellular junctions. Am J Physiol Cell Physiol methasone on bloodbrain barrier in the
1999; 277: C33042. normal mouse. Ann Neurol 1986; 19: 3737.

64
3
Principles of diagnosis

Introduction History
The diagnosis of central nervous system (CNS) The duration of symptoms may be helpful in
tumors differs quantitatively but not qualita- distinguishing brain tumors from other disor-
tively from the diagnosis of other neurologic ders and in distinguishing among histologic
diseases. The qualitative elements of history, grades of brain tumors. Although seizures are
general physical examination, neurologic sometimes present for many years before a
examination and laboratory findings, especially brain tumor is identified1 (this was much truer
imaging, are the same for all neurologic before current imaging techniques were avail-
diseases. However, in most neurologic disor- able), a long duration of seizures usually
ders, such as migraine, epilepsy and even, early indicates a diagnosis other than brain tumor.
in its course, multiple sclerosis, the history is Neurologic symptoms other than seizures in
by far the most important element, represent- patients with brain tumors usually evolve
ing more than 80% of the information neces- subacutely over weeks or months. A sudden
sary for diagnosis. This is because most onset of neurologic symptoms, followed by
patients who present for neurologic evaluation stability suggests cerebral infarction rather than
have symptoms unaccompanied by neurologic brain tumor; a very slow onset over years
signs or abnormal images. In patients with suggests degenerative disease rather than brain
CNS tumors, the situation is different. This tumor. More aggressive brain tumors, such as
history is quantitatively far less important than glioblastoma, metastases and primary CNS
the laboratory evaluation. Unless the patient lymphoma, usually evolve rapidly over weeks,
presents with a focal seizure, symptoms may be whereas lower-grade tumors, such as astro-
vague and subtle and resemble those of many cytoma and oligodendroglioma, may evolve
less serious neurologic syndromes. For over months or a few years.
example, one cannot easily distinguish, by
history, the headache of brain tumor from that Neurologic examination
of migraine or tension-type headache (see
below). Similarly, it is often hard to distinguish The general physical and neurologic examina-
personality change associated with brain tion is often not helpful, because many patients
tumors from depression or other psychological have no neurologic signs, and when signs are
disorders. present they do not differ substantially from

65
PRINCIPLES OF DIAGNOSIS

those of patients with cerebrovascular disease Clinical findings


or other much more common neurologic
disorders. Thus, unlike most neurologic Introduction
disorders, the physician evaluating a patient
Several factors determine the symptoms and
with a possible intracranial tumor must depend
signs in a patient with a CNS tumor (Table 3.1
on the laboratory, especially brain imaging.
and Fig. 3.1.)

Laboratory examination Table 3.1


Factors determining symptoms and signs of CNS
By far the most important element in the evalu-
tumors.
ation of patients suspected of harboring an
intracranial tumor is the magnetic resonance
image (MRI). Often, MRI performed before Location
Supratentorial versus infratentorial
and after the injection of contrast material Cortical versus subcortical
(gadolinium) is the only laboratory test required Intraparenchymal versus extraparenchymal
prior to initiating therapy. At other times, more Eloquent versus non-eloquent
extensive evaluation is required, as indicated in Growth (histology)
Rapid versus slow
the paragraphs below. With very few excep- Infiltrative versus discrete
tions, a negative MRI in a patient suspected of Size
harboring an intracranial mass lesion effectively Large versus small
Secretions
excludes that diagnosis. This, of course, Angiogenic factors
assumes that the scan is of high quality, that it Pituitary hormones
has been read correctly and that it has been Immunosuppressive factors
?Melatonin
performed with injection of contrast. The clini- Cytokines
cian should review each MRI with the neuro- Neuropeptides
radiologist, paying careful attention to the area,
if any, of clinical interest suggested by the
history or examination. Early in their course,
Figure 3.1
tumors may be either missed or confused with
The impact of size, growth rate and location on clinical
ischemic changes, particularly in the elderly. We symptomatology. (a) The MR scan shows an enlarging
do not suggest that every patient with a right frontal tumor in 1995 (left) and 1999 (right) in a
headache receive an MRI2 or that the history patient who had occasional focal seizures over 6 years
and physical findings not be meticulously characterized by sudden brief panic attacks. Despite the
reviewed before ordering an image, but we growth of the lesion, she has not had other neurologic
symptoms and her clinical examination was normal.
believe that, for most patients suspected of The diagnosis was oligodendroglioma. (b) The contrast-
harboring an intracranial mass, MRI is the first enhanced MRI of a 65-year-old man with progressive
and often the only step required for diagnosis. weakness of the left side over a 2-day period. A small
This chapter considers symptoms, signs and enhancing tumor can be identified in the brainstem
laboratory tests necessary for the diagnosis of (arrow), probably a brainstem glioma. (c) The MRI of
a 25-year-old woman with facial myokymia and slight
intracranial tumors. Specific laboratory tests
facial weakness but no other neurologic
necessary for the diagnosis of specific intra- symptomatology. A large mass infiltrated an enlarged
cranial tumors are considered in the chapters pons, completely changing the signal in that structure
on individual tumors. without major neurologic symptoms.

66
CLINICAL FINDINGS

(a)

(b) (c)

67
PRINCIPLES OF DIAGNOSIS

Location whereas rapidly growing tumors (e.g. glioblas-


A major factor determining symptomatology tomas) often present with focal neurologic signs,
is brain location. Supratentorial tumors often including paralysis and aphasia. Before modern
present with seizures, whereas infratentorial imaging techniques, low-grade tumors often
tumors are more likely to cause headache and caused seizures for many years (20 years in one
vomiting. Tumors involving the cortex (e.g. of our patients with an oligodendroglioma)
meningioma) are more likely to cause seizures before other symptoms developed.
as the only sign, whereas deep-lying tumors
(e.g. lymphomas) are more likely to cause Size
personality or cognitive changes. Tumors The size of the tumor also helps determine the
located in eloquent areas (the term eloquent neurologic symptoms. False localizing signs
comes from the Latin for to speak and is used (see below) were first described in patients
by neurologists to indicate those vital cerebral with meningiomas, where the large tumors in
structures necessary for language, e.g. Brocas relatively silent areas caused major shifts of
and Wernickes areas, or for movement or normal brain structures before the tumors were
somatic sensory perception, e.g. sensorimotor detected. Such false localizing signs are uncom-
cortex) usually present with either easily identi- mon now, because other minor symptoms often
fiable focal seizures or focal signs (e.g. speech lead to early neuroimaging and diagnosis.
arrest from frontal lobe tumors, hemiparesis
from tumors involving the motor strip). Secretions
Tumors in non-eloquent areas may present Intracranial tumors can cause symptoms not
with what appear to be generalized seizures only by their size and location, but also by their
(actually seizures of focal origin in which the secretions. Small tumors of the pituitary gland
focal origin is unrecognized), or behavioral and (Chapter 10) may secrete growth hormone and
cognitive changes. Tumors of the brainstem cause acromegaly associated with headache.
usually begin with cranial nerve palsies causing Other tumors secrete prolactin, causing galact-
symptoms such as diplopia or facial weakness. orrhea and amenorrhea in women or impotence
Other posterior fossa or base of brain tumors in men. Adrenocorticotropic hormone (ACTH)
often have headache as an early symptom, secreting tumors cause Cushings disease, also
because they either involve pain-sensitive struc- associated with behavioral change, usually
tures at the base of the brain, or cause early depression. Larger tumors (macroadenomas)
hydrocephalus (4th ventricular tumors). may cause pituitary failure with associated
Infiltrating tumors are more likely to cause cognitive dysfunction. Pineal region tumors
generalized symptoms of headache and person- have been reported to interfere with normal
ality change, whereas focal tumors are more melatonin secretion, leading to insomnia,
likely to cause seizures, hemiparesis and other sometimes with personality changes. Precocious
focal neurologic signs. puberty with behavioral changes may also
complicate pineal region tumors (Chapter 8).
Growth rate Some tumors, e.g. high-grade gliomas and some
The growth rate of tumors also determines clini- meningiomas, secrete factors such as vascular
cal findings. Slowly growing tumors (e.g. astro- endothelial growth factor (VEGF) and fibrob-
cytomas) usually present with seizures without last growth factor (FGF) that cause angiogene-
other evidence of neurologic dysfunction, sis and peritumoral edema (Chapter 2). The

68
CLINICAL FINDINGS

edema may cause more symptoms than the diagnosis and treatment. However, all too
tumor itself. Some tumors secrete cytokines often, they reveal a small meningioma that
such as interleukins and tumor necrosis factor does not require treatment or show a structure
(TNF) and may alter brain neuropeptides,3 misinterpreted as a tumor, e.g. a pineal cyst
which may affect cognitive function and behav- or enlarged VirchowRobin space, leading to
ior, e.g. anorexia. much patient anxiety.

Imaging Pathophysiology of symptoms and


CT and MR imaging have substantially
signs
changed the symptoms at diagnosis of intra-
cranial mass lesions. In the past, lesions often CNS tumors cause neurologic symptoms by
grew to considerable size and caused obvious one or more of four mechanisms (Table 3.2,
signs and symptoms before the physician was Fig. 3.2). These mechanisms include invasion,
willing to suggest such invasive and painful compression, CSF obstruction and herniation.
diagnostic tests as angiography or pneumo- Invasion and compression cause symptoms and
encephalography. Now patients present with signs suggestive of focal brain disease, i.e. signs
more subtle symptoms that tax the clinical referable to a particular region of the brain.
skills of the physician. The diagnosis of brain CSF obstruction and herniation cause more
tumor is now often made long before intra- generalized brain symptoms, such as diffuse
cranial pressure is substantially increased and headache and alterations of consciousness not
sometimes made when the tumor is completely ascribable to a specific brain area. When focal
asymptomatic, the imaging having been masses cause herniation, signs and symptoms
procured for reasons unrelated to the tumor, may originate from portions of the brain
such as minor head trauma or a syncopal distant from the tumor, so-called false localiz-
attack. In one series of 1000 healthy, mostly ing signs.
young (average age 30), asymptomatic volun-
teers, 18% had some abnormal finding on MRI Invasion
and 2 (0.2%) had a primary brain tumor.4 The The tumor invades and replaces or displaces
numbers would undoubtedly be greater if older normal brain. The underlying normal brain
volunteers were selected. Such unindicated tissue may or may not be destroyed.
scans may reveal a tumor, leading to early Widespread brain invasion is particularly

Table 3.2
Pathophysiology of symptoms and signs of brain tumors.

Mechanism Signs and symptoms Example

Invasion Focal Hemiparesis, focal seizures


Compression Focal Hemiparesis
Cerebrospinal fluid obstruction Generalized Headache
Herniation Generalized, false localizing Confusion, stupor, abducens nerve
paralysis

69
PRINCIPLES OF DIAGNOSIS

Figure 3.2
Pathophysiology of signs and symptoms in patients
with brain tumors. This schematic coronal section of
the brain enclosed in the skull illustrates the changes
caused by a large brain tumor. The lesion itself (1)
(green sphere) underlies the arm area of the motor
9
strip and causes weakness of the contralateral arm. 2
The edema (2) surrounding the mass involves most of 3
the motor area and is likely to cause a contralateral
1
hemiplegia. Because of the size of the brain tumor
and the surrounding edema, the normal brain shifts, 4
compressing itself and other normal brain areas.
Herniation of the cingulate gyrus (3) under the falx 5
6
cerebri compresses not only the contralateral frontal
7
lobe but also the anterior cerebral arteries. Such
herniation can cause bilateral frontal ischemia, with
weak legs, urinary incontinence, and mental changes.
The diencephalon (4) shifts toward the contralateral
side, compressing itself, the third ventricle, and the
8
opposite diencephalon. The resulting diencephalic
dysfunction causes diminished consciousness.
Herniation of the uncus and hippocampal gyrus (5)
of the temporal lobe into the tentorial notch
compresses the posterior cerebral artery, leading to midbrain and pontine hemorrhages (Duret
infarction in the ipsilateral occipital lobe (Fig. 3.3). hemorrhages). Herniation of the cerebellar tonsils (8)
Uncal herniation also stretches the ipsilateral through the foramen magnum compresses the lower
oculomotor nerve and compresses the brainstem and brainstem and may cause respiratory arrest.
diencephalon, causing changes in the state of Hydrocephalus (9) occurs in the contralateral lateral
consciousness. Compression of the opposite cerebral ventricle as a result of obstruction of the third
peduncle (6) against the tentorium causes a ventricle and Sylvian aqueduct by compression. Any
hemiparesis that is ipsilateral to the side of the lesion combination of the mechanisms illustrated in this
(a false localizing sign). Downward displacement of figure may play a role in producing the symptoms
the brainstem (7) alters consciousness and may cause caused by a brain tumor.

characteristic of infiltrating gliomas but Cerebrospinal fluid obstruction


sometimes occurs with meningiomas, pineal The tumor obstructs cerebrospinal fluid (CSF)
region tumors or metastases. pathways, causing hydrocephalus. In some
instances, the only symptoms of the tumor may
Compression be those of hydrocephalus. Examples include
The tumor and surrounding edema compress colloid cysts at the foramen of Monro, choroid
normal tissue and its blood vessels, causing plexus papillomas, intraventricular menin-
distortion and ischemia. Compression is a giomas, pineal region tumors, fourth ventricu-
common mechanism in focal tumors such as lar ependymomas and occasionally intraspinal
meningiomas and metastases. Blood flow is tumors.6 In other instances, hydrocephalus may
reduced in peritumoral areas,5 and capillaries be a late complication of tumors that have
may collapse, leading to brain ischemia. caused preceding symptoms. These include

70
CLINICAL FINDINGS

large meningiomas at the base of the brain,


intrinsic tumors of the hemisphere, such as
glioblastomas and metastases, and brainstem
gliomas.

Herniation
Large brain tumors with their peritumoral
edema and sometimes hydrocephalus can
herniate normal cerebral structures under the
falx cerebri, through the tentorium cerebelli, or
through the foramen magnum, often causing
acute neurologic decompensation and at other
times leading to false localizing signs (see
below) (Fig. 3.3).

Symptoms and signs


As a result of these four mechanisms, neuro-
logic symptoms and signs may be divided into
one (or more) of three categories (Table 3.3).
Patients may show: (1) generalized symptoms
caused by raised intracranial pressure; (2) focal
symptoms caused by invasion, ischemia and
compression, or (3) false localizing symptoms Figure 3.3
caused by shifts of cerebral structures. Cortical blindness caused by herniation. This 32-year-
Generalized or false localizing symptoms are old woman was 9 months pregnant with a history of
particularly likely to be caused by slowly 6 weeks of progressive headache. An MR scan
revealed a large right frontal tumor. She was
growing tumors that reach a large size in the
admitted to the hospital for elective cesarean section
relatively silent frontal lobe, whereas focal and tumor removal. She became comatose shortly
symptoms occur with even small tumors in following admission, and an emergency cesarean
more functionally important areas of the brain, section and craniotomy were performed. When she
such as the motor strip and brainstem. recovered from the surgery she was cortically blind.
An MRI 2 weeks after the ictus revealed enhancing
infarction in both occipital lobes (arrow) and the
Generalized symptoms and signs residual right frontal tumor. She eventually recovered
Headache, the most common symptom of macular vision, but peripheral fields remained
increased intracranial pressure, is the first abnormal and she had severe prosopagnosia.
symptom in about 3040% of patients with a
brain tumor.7,8 Although headache may result
from increased intracranial pressure, it occurs developing papilledema, do not have headache.
with almost equal frequency in patients To cause headache, a tumor must stimulate
without raised intracranial pressure. pain-sensitive structures.9 The brain itself is not
Conversely, many patients with elevated pain sensitive. The dura over the cerebral
intracranial pressure, even to the point of convexity and the floor of the middle fossa are

71
PRINCIPLES OF DIAGNOSIS

Table 3.3
Category Symptoms Signs Some symptoms of signs
of intracranial tumors.
Generalized Headache Confusion
Vomiting Papilledema
Drowsiness Apathy, abulia
Visual obscurations
Personality change
Focal Seizures Postictal paralysis
Hemiparesis Focal weakness
Paresthesias Sensory loss
Cognitive changes Aphasia, agnosia, ataxia
Incoordination Apraxia, ataxia
Diplopia Ocular muscle paralysis
Dysphagia Aspiration
False localizing Diplopia III, VI nerve paralysis
Tinnitus Hearing loss
Visual loss Cortical blindness

also insensitive to pain, except immediately may explain the frequent similarity between
along or within a few millimeters of the brain tumor and migraine headaches.
meningeal arteries or dural sinuses. By Headache associated with brain tumor has
contrast, the dura over the floor of the anterior several mechanisms: (1) traction on venous
fossa is sensitive over its entire surface. The sinuses or their tributaries; (2) traction on
superior surface of the tentorium, transverse meningeal arteries; (3) traction on large arteries
and straight sinuses as well as the posterior at the base of the brain; (4) pressure on cranial
portion of the superior and inferior sagittal and cervical pain-sensitive structures; (5) dila-
sinuses are all pain sensitive, and like all of the tation of intracranial arteries; and (6) inflam-
supratentorial structures, are supplied by the mation of pain-sensitive structures. Interestingly,
first division of the trigeminal nerve, thus often headache is more frequent in those patients
referring the pain of a supratentorial brain with a prior history of non-tumoral headaches,
tumor, no matter where above the tentorium it suggesting an individual predilection for
is located (e.g. occipital lobe), to the eye and headache.
frontal area of the head. Posterior fossa struc- Most brain tumor headache is non-specific;
tures are supplied by lower cranial and upper however, a brain tumor should be suspected:
cervical nerves, referring pain to the occipital (1) when mild to moderate headache is present
area. The trigeminal nerve, in addition to being on the patients awakening from sleep but
the sole source of supratentorial, dural and disappears within 1 h (severe headaches that
meningeal vessel sensory afferents, is also the awaken one from sleep are more probably due
source of sensory fibers to the vessels of the to non-tumoral causes); (2) when headaches
anterior circulation via the first or ophthalmic begin in a middle-aged or older person who has
division of the trigeminal nerve. The common not previously experienced them; (3) when the
nerve supply to dura and vascular structures character or severity of headache in a chronic

72
CLINICAL FINDINGS

headache sufferer suddenly changes; (4) when language, should be carefully sought. A careful
a headache becomes progressively more severe neurologic examination may reveal sensory
over time (more often the result of brain edema or motor abnormalities unrecognized by the
rather than increasing tumor size8). patient.
Localized headache is a reliable indicator of
laterality but does not mark the precise Vomiting with or without preceding nausea,
location of the tumor. For example, a right particularly on awakening, and often described
frontal headache indicates that the tumor is on as explosive or projectile, is a common
the right but does not indicate that the tumor symptom of brain tumor in children but is less
is frontal; the tumor could be occipital or even common in adults. Vomiting with increased
cerebellar, the pain probably resulting from intracranial pressure is particularly frequent in
traction on the tentorium, the superior surface children with medulloblastoma, ependymomas
of which is supplied by the trigeminal nerve. of the fourth ventricle and other posterior fossa
Because intracranial tumors are progressive, tumors, probably because they directly
headache is usually mild and intermittent at compress the brainstem emetic center. These
onset, and often relieved by over-the-counter tumors are common in children but rare in
analgesics. The headache becomes progres- adults, explaining in part the high frequency of
sively more severe and more intractable as time vomiting in children. However, that is not the
passes. The headache can be either steady or entire explanation, because even with the same
throbbing, but is characteristically exacerbated tumor, children are more likely to vomit than
by alterations of intracranial pressure, e.g. adults. Vomiting as an ictal event can occur in
coughing, sneezing, bending, head-shaking or patients with tumors involving the insula (Latin
sexual activity. However, most cough or for island), an island of cortex deep within the
exertional headaches are not due to brain Sylvian fissure.
tumors.10 Sudden short-lived episodic Acute headache followed immediately by
headaches may be associated with plateau vomiting is characteristic of a brain tumor and
waves in patients with raised intracranial indicates increased intracranial pressure; by
pressure from a brain tumor (see below). As in contrast, a more prolonged headache followed
patients with increased intracranial pressure several hours later by vomiting is characteris-
from trauma, plateau waves are associated tic of migraine. Vomiting in patients with brain
with acute vascular dilatation. tumors usually results from irritation of the
Because headache is such a common vomiting center in the floor of the 4th ventri-
symptom and intracranial tumors are uncom- cle. The emetic center can be activated either
mon, even those headaches having the charac- by increased intracranial pressure or by direct
teristics listed above are more likely to be involvement by tumor. Accordingly, tumors of
caused by disorders other than brain tumors. the posterior fossa and brainstem are most
Nevertheless, patients with these characteristics likely to cause vomiting, usually but not always
require careful evaluation. Most patients with preceded by headache. Vomiting in the absence
headache due to brain tumors have other of increased intracranial pressure has been
symptoms or neurologic signs suggesting brain reported in patients with brainstem tumors.12
tumor by the time they seek medical help.11
Thus other symptoms, in particular, changes in Vertigo and dizziness. Vertigo occurs
memory, personality, behavior (see below) or occasionally in patients with vestibular

73
PRINCIPLES OF DIAGNOSIS

schwannomas (Chapter 9) because these suffer significant visual abnormalities, includ-


tumors originate in the vestibular portion of ing central scotomata.13 More commonly in
the VIIIth cranial nerve. However, because the severe papilledema, there may be episodes of
tumors grow so slowly, the vestibular system transient visual obscuration ranging from
usually compensates, so that patients may graying out of the visual field to complete
have either no abnormal sensation or only a blindness. These episodes are commonly associ-
vague sensation of dizziness. Vertigo also ated with a plateau wave (see Chapter 2 and
accompanies tumors of the cerebellum and below). Visual obscurations associated with
brainstem. In these cases, it is likely to be papilledema are not dangerous in themselves,
more severe and intractable. Many patients as they do not presage a permanent visual
with brain tumors and increased intracranial abnormality.
pressure complain of a sensation of dizziness
or light-headedness, possibly resulting from Mental and cognitive abnormalities. Changes
decreased blood flow to the brain causing in mental and cognitive function are of two
mild brain hypoxia. The symptoms are non- types. Specific cognitive abnormalities such as
specific. Patients who experience dizziness aphasia, alexia, agnosias and apraxias are a
may have a great deal of difficulty in precisely result of focal lesions in eloquent areas of
describing the sensation. brain. Less specific changes in behavior are
often the presenting symptom of relatively
Papilledema occurs frequently in children and large tumors in more silent areas of brain.
young adults but is less common in older These abnormalities vary from patient to
patients and infants. (In older patients, brain patient, but often mislead the family and physi-
atrophy gives a tumor more room to expand cian into believing the patient is suffering
without raising intracranial pressure, and from a psychological rather than a neurologic
fibrosis of the optic nerve sheath may prevent disorder.
pressure from being transmitted to the optic Non-specific mental changes begin with
disk; in infants, the unfused skull bones irritability and progress to apathy. Patients
can expand to accommodate the mass.) sleep longer at night, may fall asleep easily
Papilledema itself is usually asymptomatic, during the day, seem preoccupied when awake,
often found by an ophthalmologist on a and often fail to initiate activity, including
routine examination or when a patient conversation; however, if they are spoken to,
complains of headache and consults an they usually respond appropriately. In adults,
ophthalmologist for eye strain. Papilledema psychiatric consultation for the treatment of
always causes enlargement of the blind spot, what is thought to be depression is frequently
because the optic disk produces the blind spot obtained before a brain tumor is suspected.
in the visual field, and when swollen the blind The astute psychiatrist will recognize the
spot is enlarged; the patient is unaware of this symptoms as being those of structural disease
change. In some patients, papilledema may of the nervous system rather than depression.
cause constriction of the visual field or even Although the patient usually first becomes
loss of central vision. Many patients with irritable, in two of our patients with large,
pseudotumor cerebri (increased intracranial non-dominant frontal lobe tumors, the first
pressure without a tumor), but fewer patients alteration in behavior was a change from estab-
with brain tumors (even with papilledema), lished irascibility to placidness. As their

74
CLINICAL FINDINGS

spouses put it, the patients suddenly became Table 3.4


nice and easy to get along with. Paroxysmal symptoms from plateau waves in patients
with intracranial space-occupying lesions.14

Episodic symptoms that include headache,


visual loss, altered consciousness and Impairment of consciousness
sometimes transient weakness of the extremi- Trance-like state
Unreality/warmth
ties are often precipitated by rising from a
Confusion, disorientation
recumbent position, coughing, or sneezing.
Restlessness, agitation
They are caused by plateau waves, abrupt Disorganized motor activity
increases in an already elevated intracranial Sense of suffocation, air hunger
pressure that last for 520 min (Table 3.4). Cardiovascular/respiratory disturbances
Plateau waves are not seizures; they respond to Headache
Pain in the neck and shoulders
Nasal itch
Blurring of vision, amaurosis
Mydriasis, pupillary areflexia
Nystagmus
Oculomotor/abducens paresis
Conjugate deviation of the eyes
External ophthalmoplegia
Dysphagia, dysarthria
Nuchal rigidity
Retroflexion of the neck
Opisthotonus, trismus
Rigidity and tonic extension/flexion of the
arms and legs
Bilateral extensor plantar responses
Sluggish/absent deep tendon reflexes
Generalized muscular weakness
Loss of muscle tone in the eyes
Facial twitching
Clonic movements of the arms and legs
Facial/limb paresthesias
Rise in temperature
Nausea, vomiting
Facial flushing
Pallor, cyanosis
Figure 3.4 Sweating
The MR of a patient with callosal amnesia. This man Shivering and goose flesh
presented complaining of loss of recent memory. He Thirst
had severe loss of recent memory but other cognitive Salivation
functions were relatively intact and his segmental Yawning, hiccoughing
neurologic examination was entirely normal. The Urinary and fecal urgency/incontinence
initial impression was that he was suffering from
Alzheimers disease, but an MR scan of the brain Bold text indicates the more common symptoms
revealed a tumor that proved to be a glioblastoma.

75
PRINCIPLES OF DIAGNOSIS

corticosteroids or a decrease in the intracranial Other focal symptoms and signs of a brain
pressure but do not respond to anticonvulsant tumor depend on the site of the lesion. These
therapy. focal symptoms and signs are the same as those
of CNS infection, stroke or other structural
Focal symptoms and signs diseases of the brain. Table 3.5 lists some focal
symptoms and signs of intracranial tumors in
Seizures are the most common focal sign of a relation to their location.
brain tumor.1 They affect at least 1/3 of brain Signs such as contralateral hemiparesis with
tumor patients and are often the first and only basal ganglia or motor cortex tumors or ataxia
symptom. They are more common in patients and nystagmus with cerebellar tumors are
with low-grade gliomas than those with high- obvious. A few other less obvious signs deserve
grade tumors.15 Focal seizures are particularly comment. Tumors that arise posterior to the
common in patients who have tumors, such as motor strip in the parietal and posterior tempo-
meningiomas, that compress the cortex or arise ral lobe may present with focal cognitive and
in or near the motor strip or the temporal lobe. behavioral changes that confuse both the
Focal seizures caused by frontal or temporal patient and the physician. We have encoun-
foci often cause episodic behavioral or tered several patients whose first symptom of a
emotional symptoms that are sometimes non-dominant parietal lobe lesion was dressing
confused with panic attacks or psychological apraxia. The patients noticed that it took
disorders. Episodic symptoms such as a longer to get dressed because the visualspatial
hemiparesis or aphasia without clear seizure relationship between the clothing and the
activity can last for hours to days, and then patients body seemed unclear. The family often
resolve. These episodes may respond to noticed that the patient would wear clothes
anticonvulsants. Generalized convulsions, inside out or backwards.
without the patient experiencing an aura (Latin A second perplexing problem is that of a
for breeze denoting the focal onset of a visual field defect from an occipital tumor. The
seizure as recognized by the patient), when patient is usually unaware of the loss of periph-
caused by brain tumors, are not truly general- eral vision but he or she suffers several minor
ized at onset, but represent a focal seizure, car accidents, usually damaging the same side
arising from an asymptomatic focal discharge of the car in each accident. If the visual field
and then secondarily generalizing; the focal defect is non-dominant, the patient may also
signature is not apparent either to the patient complain of difficulty in reading, because as his
or an observer. Depending on the growth rate eyes return to the beginning of the next line,
of the tumor, seizures may be present for he loses his place. With dominant visual field
months to years before other symptoms defects (right side), there is less difficulty in
develop. Any patient with focal or generalized reading unless the lesion involves more anterior
seizures that begin in adulthood should association areas, in which case the patient
undergo diagnostic evaluation for a brain may develop alexia with or without agraphia.
tumor (see below). However, only a minority Lesions of the posterior corpus callosum
of patients with adult-onset seizures have brain (splenium), probably due to invasion of
tumor as the cause. The figures vary from hippocampal structures lying immediately
2%22% depending on the series.16 The higher below them, often present with memory loss18
figures are probably the more accurate. in the absence of other cognitive abnormalities.

76
CLINICAL FINDINGS

Table 3.5
Focal symptoms and signs of intracranial tumors.

Frontal lobe Thalamus


Generalized seizures Sensory loss (contralateral)
Focal motor seizures (contralateral) Behavioral changes (posterior)
Expressive aphasia (dominant side) Language disorder (dominant side)
Behavioral changes
Dementia Midbrain/pineal
Gait disorders, incontinence Paresis of vertical eye movements
Hemiparesis Pupillary abnormalities
Precocious puberty (boys)
Basal ganglia
Hemiparesis (contralateral) Sella/optic nerve/pituitary
Movement disorders (rare) Endocrinopathy
Bitemporal hemianopia
Parietal lobe Monocular visual defects
Receptive aphasia (dominant side) Ophthalmoplegia (cavernous sinus)
Spatial disorientation (non-dominant side)
Cortical sensory dysfunction (contralateral) Pons/medulla
Agnosias Cranial nerve dysfunction
Ataxia, nystagmus
Occipital lobe Weakness, sensory loss
Hemianopsia (contralateral) Spasticity
Visual disturbances (unformed)
Cerebello-pontine angle
Temporal lobe Deafness (ipsilateral)
Complex partial (psychomotor) seizures Loss of facial sensation (ipsilateral)
Generalized seizures Facial weakness (ipsilateral)
Behavioral changes Ataxia
Olfactory and complex seizures
Visual auras Cerebellum
Visual field defect Ataxia (ipsilateral)
Nystagmus
Corpus callosum
Dementia (anterior)
Behavioral changes
Memory loss (posterior)
Asymptomatic (mid)

Modified from Vick.17

One of our patients with a lymphoma involv- vermis. Characteristically, the patient
ing the posterior corpus callosum was unaware complains of ataxia and is found to be ataxic
of the day, month, or year, but was able to when walking, but point-to-point tests of both
determine the season by noting that he was upper and lower extremities are performed
wearing a summer-weight suit. well, sometimes leading the physician to
Truncal ataxia, sparing the limbs, is a believe that the ataxic gait is hysterical.
common finding in lesions of the cerebellar Hydrocephalus can cause gait abnormalities,

77
PRINCIPLES OF DIAGNOSIS

Figure 3.5
A patient with a large base of skull meningioma and secondary hydrocephalus. She presented with gait
instability and ataxia, which were first believed to be secondary to compression of the brainstem by the tumor.
However, despite the large tumor, the entire ventricular system was open, and communicating hydrocephalus
(left) was evident. A ventriculoperitoneal shunt relieved her symptoms and the hydrocephalus (right). Note the
decreased ventricular size after surgery. The tumor did not change in size.

so-called frontal ataxia, sometimes mistaken amnesia has been the presenting event of a
for cerebellar dysfunction (Fig. 3.5).19 brain tumor.20 Most patients who have
As indicated above, prolonged episodic transient global amnesia caused by a brain
symptoms of a behavioral and cognitive nature tumor have some persistent abnormality of
may be the presenting complaint of a brain memory on careful neurologic evaluation.
tumor. One of our patients suffering a
dominant parietal lobe glioblastoma suddenly False localizing symptoms and signs
became unable to decide how to turn off the False localizing symptoms and signs are caused
bath water that he had turned on a few by shifts of cerebral structures. Diplopia may
minutes before. He then attempted to call his result from displacement or compression of
daughter, and although he remembered the the abducens nerve at the base of the brain.
phone number, could not dial it. There was no Hemianopsia or even cortical blindness may be
weakness, sensory loss, or other form of confu- caused by tentorial herniation that compresses
sion. He eventually dialed the number. By the the posterior cerebral artery. A number of other
time he got to the emergency room, his neuro- cranial nerve palsies associated with shifts of
logic examination was entirely normal. brainstem structures may also occur. Table 3.6
On a few occasions, episodic transient global lists some false localizing signs associated with

78
CLINICAL FINDINGS

brain tumors. Table 3.6


Most false localizing signs occur in patients Some false localizing signs associated with brain
tumors.
with large, slowly growing brain tumors, e.g.
meningiomas, but they have been reported in
other situations as well. A few deserve Cranial nerves
comment. Anosmia
Diplopia, ptosis, anisocoria
Face pain, numbness, and weakness
Tinnitus, with or without hearing loss, is a Tinnitus, hearing loss
common complaint in patients suffering
Parenchymal signs
increased intracranial pressure of any cause. At
Ipsilateral hemiparesis
least one form of tinnitus appears to be due to Ipsilateral gaze palsy
turbulent flow through the transverse sinuses, Visual field defect, cortical blindness
which pass near the middle ear. The noise is Ataxia
heard as a venous hum.21 Increasing cerebral Other signs
venous pressure, either by a Valsalva maneuver Nuchal rigidity
or by gently compressing the jugular vein in the
neck, will often alter the tinnitus or make it
disappear. In other patients, tumor compres-
sion of the acoustic nerve leads to hearing loss
and more high-pitched tinnitus. (usually extracranial, such as a meningioma)
shifts the brainstem laterally, compressing the
Diplopia. Increased intracranial pressure leads contralateral cerebral peduncle against the
to compression of the abducens nerve, the tentorium cerebelli (Kernohans notch). The
cranial nerve that runs the longest intracranial patient may have either ipsilateral hemiparesis
course, causing horizontal diplopia on lateral only or bilateral hemiparesis.
gaze. The patient may complain only of
diplopia when he looks into the distance and Anosmia resulting from compression of the
not notice it for near vision, when the abducens olfactory nerves by the overlying frontal lobe
nerve is at rest. Abducens paralysis may be is a common false localizing sign, but is rarely
unilateral or bilateral. Less common, but more noted by the patient or identified or tested for
ominous, is involvement of the ipsilateral third by the physician.
nerve, compressed by the hippocampal gyrus as
it herniates through the tentorium cerebelli Cortical blindness and prosopagnosia. Cortical
(uncal herniation) (see Fig. 3.2). The first sign blindness (Fig. 3.3) results from compression of
is ipsilateral pupillary dilatation followed by posterior cerebral arteries, a result of herniation
vertical and horizontal diplopia and sometimes of the bilateral hippocampus gyri through the
ptosis. The patient may be fully conscious tentorial notch. One of our patients with a large
when the third nerve symptoms begin. frontal oligodendroglioma herniated just prior
to surgery and awoke cortically blind. Because
Ipsilateral hemiparesis. Unlike the contralateral the middle cerebral artery supplies macular
hemiparesis that usually accompanies a supra- vision, that vision returned, leaving her with a
tentorial mass lesion, ipsilateral hemiparesis small area of central vision. Involvement of
occurs when a supratentorial mass lesion nearby association areas led to some memory

79
PRINCIPLES OF DIAGNOSIS

loss and prosopagnosia (from Greek prosopon weighted image was normal. As his symptoms
face and gnosis recognition) so profound that progressed over the next month, a repeat MRI
she was unable to recognize her husbands face was obtained and a large enhancing glioblas-
or her own face in the mirror. When looking toma was seen in the right temporal lobe.
into a mirror, she would protrude her tongue to MRI identifies tumors that CT misses,
be sure the image was hers. particularly in the posterior fossa, and distin-
guishes tumors from arteriovenous malforma-
Radicular pain can be a false localizing sign of tions. MRI often gives signal characteristics
increased intracranial pressure.22 that suggest the histology (Table 3.7).23 Except
for biopsy (see below), other laboratory tests
are usually unnecessary. MR scans can be
performed relatively easily and repeatedly in
Laboratory diagnosis most patients. Even in patients who are
somewhat confused and therefore restless, fast
Imaging
scans often suffice to give information that may
All adults with new-onset seizures and all not be easily detected on a CT scan. About
patients with papilledema or new focal motor 30% of patients are uncomfortable in the
or sensory signs require an MRI23,24 with the scanner because of claustrophobia. Most toler-
injection of contrast material (gadolinium ate the procedure with encouragement from the
DPTA). Other presenting symptoms listed in technician or a companion sitting in the room
Table 3.7 and discussed above should also with them. About 510% of patients require
prompt consideration of imaging. Once a sedation. We use lorazepam 12 mg prior to
tumor is identified, other imaging techniques, the scan. Probably fewer than 1% of patients
such as diffusion tensor MRI (to identify cannot tolerate the scan even with sedation. A
distortion of white matter near a tumor that cardiac pacemaker is an absolute contraindica-
appears normal on routine MRI25,26), positron tion, as are ferromagnetic foreign bodies (e.g.
emission tomography (PET) to measure glucose shrapnel) in the patient. For the claustrophobic
metabolism or amino acid uptake in the tumor, patient who cannot tolerate a standard scan,
single photon emission computed tomography imaging in an open scanner, although yielding
(SPECT),24 magnetic resonance spectroscopy an inferior scan, is preferable to CT.
(MRS),26,27 and functional MRI (fMRI),24 may Certain signal characteristics suggest a brain
each prove useful in ascertaining histologic tumor (Table 3.7). The increased water content
grade and in guiding surgical procedures. of brain tumors and their surrounding edema
yield a hypointense (darker than normal brain)
MRI T1 image and a hyperintense (lighter than
MRI visualizes the entire intracranial contents normal brain) T2 image. T1 and T2 refer to
clearly, unlike CT scans, where bone and teeth the proton relaxation time for the acquisition
can obscure lesions, particularly in the poste- of MRI data. Disruption of the bloodbrain
rior fossa and middle cranial fossa. A negative barrier characteristically occurs in certain brain
MRI almost always excludes a tumor as the tumors (Chapter 2). A contrast agent (e.g.
cause of the patients symptoms or signs. gadolinium) leaks across and enters the brain
However, we had one patient who presented tumors extracellular space, causing hyper-
with confusion and whose MRI including a T2- intensity (enhancement) on T1 images.

80
LABORATORY DIAGNOSIS

Table 3.7
Imaging appearance of brain tumors.

CT (MR)* MR

Non-Contrast (T1) Contrast (T1) T2

Malignant glioma Density Irregular peripheral


enhancement
Low-grade astrocytoma Density enhances
Oligodendroglioma to iso, Ca2+ in 91% < 50% + enhances
with blood Heterogeneous if bleed
Lymphoma Density Solid portion
enhances
Ganglioglioma Density, 33% Ca2+ 50% enhances
Ependymoma Density, Ca2+ > 50% Irregular, enhances
(often peripheral)
PNET Iso to density Enhances Mixed
Choroid plexus papilloma Iso and Ca2+ + Mixed
Choroid plexus carcinoma Iso and Ca2+ + Mixed
Germinoma Iso to + Iso to
Embryonal carcinoma Iso to +
Teratoma Mixed density Mixed
Hemangioblastoma +
Brainstem glioma
Cerebellar astrocytomas + Largely cystic
Meningioma Iso to + Iso to
Schwannoma to iso + Iso to
Metastases to + Variable to
CSF disseminated tumor + Iso
Neurocytoma Mixed cystic, Ca2+ 60% + Cystic inhomogeneous
Hemangiopericytoma or +

*CT and T1-weighted MRI show similar changes


Modified from Zimmerman.29

Hemorrhages are best identified on gradi- becomes hyperintense after about 3 days and
ent echo MRI.30 They usually appear as may remain so for weeks to months. The T2
hyperintense on non-contrast T1 images and image becomes hyperintense, sometimes with
hypointense on T2. However, these radio- a rim of low intensity after about a week and
graphic features vary depending on the age of develops uniformly low intensity after
the hemorrhage. Acute hemorrhage can be months. Enhancement is absent acutely but
detected on MRI within 2 h.31 In general, the hemorrhage may develop a rim of
hemorrhage demonstrates low intensity to enhancement after about a week. Earlier
isointensity on T1 and low intensity on T2 enhancement suggests hemorrhage into a pre-
during the first 3 days. The T1 image existing tumor.

81
PRINCIPLES OF DIAGNOSIS

Hemorrhage is hyperdense on CT scan. have multiple lesions, whereas only 5% of


Calcifications are difficult to identify on MR, patients with gliomas have multifocal disease.
but are easily seen on CT scan as more dense Of patients with primary CNS lymphoma
than hemorrhage. On CT scan, calcification is (PCNSL), 2040% have multiple lesions that
a positive prognostic factor,32 and enhancement are located periventricularly, usually exhibit
a negative factor within each histologic grade diffuse contrast enhancement, have poorly
of glioma. circumscribed margins compared with gliomas
Although both brain edema and the tumor and metastases, and are usually surrounded by
appear hyperintense on T2, edema is usually less edema than these other tumors. On T2,
more hyperintense (i.e. appears whiter). PCNSL has relatively low signal intensity
Sometimes, tumor and edema can be distin- compared with the high signal intensity of the
guished by the fact that edema spares the surrounding edema. The MR scans of immuno-
cortex, producing finger-like projections of T2 compromised patients with PCNSL may not
hyperintensity between normal-appearing corti- demonstrate enhancement or may show ring
cal gyri. By contrast, infiltrating tumor involves enhancement. Cortical and basal ganglia
the cortex, changing the cortical signal and involvement is more common in these patients.
often expanding gyri. Infiltrating tumor may A separate technique, diffusion-weighted
have the appearance of a well-defined border MR imaging, permits assessment of the mobil-
on T2 MRI images, whereas edema typically ity of water molecules, and may distinguish
has blurred and indistinct radiographic among: (1) ischemia; (2) cytotoxic edema
borders. within parts of the tumor; (3) radiation-
In low-grade gliomas, edema is often absent induced gliosis; (4) vasogenic edema. However,
and there is usually no contrast enhancement. the differences are relatively slight and have not
High-grade gliomas usually exhibit contrast been very clinically useful to date. Perfusion
enhancement on the T1-weighted image, images measured during the first pass of
showing an enhanced rim of irregular shape contrast through the brain reflect blood volume
and thickness that surrounds a hypointense and, thus, vascularity. The relative cerebral
center; the T2 image shows only hyperintensity. blood volume measured by the perfusion scan
Although the contrast enhancement does not is a good indicator of tumor grade and corre-
encompass the entire infiltrating margin, it lates strongly with PET F18-deoxyglucose
represents a clinically useful approximation of studies (see next section) of tumor grade.34
tumor volume. Fast fluid-attenuated inversion Serial MR scans can also be used to deter-
recovery (FLAIR) sequences provide prominent mine the effectiveness of treatment. The
distinction between normal brain and brain efficacy of treatment with radiation or
tumor or edema and give high tumor-to- chemotherapy is generally divided into four
background contrast ratios;33 however, tumor categories: a complete response (CR) indicates
cannot be differentiated from edema on FLAIR disappearance of the tumor; a partial response
images. Metastases have a regular and spheri- (PR) indicates a decrease in the size of the
cal rim because they grow by expansion and contrast-enhancing tumor to 50% or less of its
compression of adjacent brain tissue rather former size; stable disease (SD) indicates no
than by invasion and infiltration as gliomas do. decrease, but also no increase, in the tumor
Metastases are much more likely than gliomas size; and progressive disease (PD) indicates
to be multiple; 50% of patients with metastases growth of the tumor. Some investigators

82
LABORATORY DIAGNOSIS

include the term minor response (MR) to Positron emission tomography (PET)
indicate tumor shrinkage by more than 25% This procedure is performed by injecting
but less than 50%. Although extremely useful substances such as glucose, an amino acid, or
in large series, these criteria are less useful in even a nucleotide labeled with a positron-
an individual patient. Because the patients emitting isotope (Fig. 3.6). These substances
head is not fixed in the scanner, the orientation are taken up by cells and metabolized through
differs from one scan to the next, making exact normal biochemical pathways. Positron-
measurements difficult. Furthermore, patients emitting isotopes include O15, C11, N13, and F18.
are often scanned on different machines using These isotopes have short half-lives of minutes
different computer settings and at different to hours and disintegrate by emitting a
time intervals after contrast injection. Finally, positron, a positively charged electron, from
unless quantitative measurements are made in the nucleus of the atom. The positron travels a
all three dimensions and a volume calculated, very short distance before it encounters an
differences of as much as 20% between scans electron. The two annihilate each other and
may be missed. Even with quantitative convert a small amount of mass into a pair of
measurements, 10% differences cannot be photons that travel in opposite directions. The
easily detected. The 50% difference required of photons are recorded when they strike a crystal
a PR, however, is usually easily detectable. detection system (with a large circular array)

Figure 3.6
Comparison of MR and PET scan in a patient with a low-grade glioma. The MR scan (left) shows a lesion
hypointense on the T1-weighted image (arrow). A coregistered PET scan reveals that the lesion is
hypometabolic (arrow). A biopsy revealed a low-grade tumor.

83
PRINCIPLES OF DIAGNOSIS

and their location is identified by simultaneous because treatment is determined by the highest
detection electronics built into the tomograph. grade in a heterogeneous tumor.
The number of reconstructed images reflects FDG PET scans are also useful, after radia-
the emissions detected, which are proportional tion therapy, in helping to distinguish radiation
to the amount of isotope found in a particular necrosis from recurrent tumor. Both may
volume of tumor or brain. If the arterial appear similar on MRI, but radiation necrosis
concentration of F18-deoxyglucose (FDG) is usually hypometabolic and recurrent tumor
radioactivity is measured to correct for isotope is usually normometabolic or hypermetabolic.
in the vascular compartment, the absolute Unfortunately, this test is not completely
metabolic rate of glucose utilization in specific reliable. Necrotic areas within recurrent tumor
brain tumor regions can be calculated. At may result in a hypometabolic image and
present, FDG is the most common isotopically inflammation associated with radiation necro-
labeled compound used for evaluating brain sis may result in a hypermetabolic image. Some
tumors,35 although C11-methionine can also be reports suggest that FDG PET may predict the
used, as can other amino acids.36,37 The differ- tumor response to irradiation or chemotherapy.
ential accumulation of a positron-emitting Acute increases in glucose metabolism were
metabolite in normal brain compared with associated with response to radiosurgery.40
brain tumor tissue can provide information
about the grade of the tumor.38 For example, Single photon emission spectroscopy (SPECT)
FDG imaging from glucose roughly defines the This procedure is less technically demanding,
metabolic rate of the area being examined. and is sometimes used to define blood flow and
Hypermetabolism (high FDG uptake) is blood volume of tumors when compared with
common in high-grade tumors, and hypo- normal tissue. This method uses gamma
metabolism (low FDG uptake) is seen in low- cameras and computers. SPECT is widely
grade tumors. Amino acid uptake is increased available, whereas PET has limited availability.
in high-grade tumors as well. The injection of SPECT generally provides information similar
nucleotides, such as iododeoxyuridine (IUDR), to PET and has been called the poor mans
may help define DNA metabolism and thus the PET scan. The most commonly used agent is
proportion of tumor cells progressing through technetium-99m-labeled hexamethylpropylene-
the cell cycle. amine oxime (HMPAO), which is lipophilic
PET scans are performed with FDG in some and crosses the bloodbrain barrier in propor-
patients with putative low-grade diffuse astro- tion to blood flow. This agent identifies high-
cytomas prior to biopsy.39 In a patient with a grade tumors by their increased blood volume
non-enhancing lesion who suffers only seizures due to extensive neovascularity. No increased
controllable by anticonvulsants and has no uptake of technetium-99m-labeled-HMPAO is
other neurologic symptoms or signs, if the PET observed in low-grade, infiltrating tumors. One
scan is hypometabolic (e.g. glucose metabolism recent report,41 using dual isotope SPECT with
less than normal white matter) we may elect to thallium-201 ion and technetium-99m-labeled
follow that patient clinically rather than biopsy HMPAO, accurately predicted histopathologic
or treat (Chapter 5). If an FDG PET scan that findings and survival after re-resection in
is generally hypometabolic shows an area of patients treated with radiotherapy for glioblas-
hypermetabolism, the neurosurgeon can direct toma. Thallium-201, a potassium analog, does
the stereotactic needle biopsy to that area not cross the intact bloodbrain barrier but

84
LABORATORY DIAGNOSIS

does cross an impaired bloodbrain barrier,


NAA
where it is taken up in proportion to the activ-
ity of the sodiumpotassium ATP pump. Thus, 500
necrotic tissue, which has a disrupted barrier,
400
but does not take up much thallium, can be
distinguished from viable contrast-enhancing 300 CHO
tumor tissue. If the thallium uptake is more CR
than 312 times that of the scalp it suggests 200
tumor recurrence, whereas if it is less than 2
100
times the scalp uptake, it represents necrosis.
The technique has proved useful in differenti- 0
ating radiation necrosis from recurrent tumor
in patients who have been previously treated. 4 3 2 1 0
It is also useful for following pediatric brain Frequency (ppm)
tumors after treatment.42 Thallium can differ-
entiate toxoplasmosis infection from tumor in
Figure 3.7
patients with AIDS. Toxoplasmosis is largely Magnetic resonance spectroscopy of normal brain.
necrotic and has a low thallium uptake, The choline (CHO) and creatine (CR) peaks are
whereas tumor, usually a lymphoma or glioma, substantially the same size. The largest peak is made
has a higher uptake. A recent report suggests by N-acetyl aspartate (NAA), a component of normal
that methyltyrosine SPECT is superior to neurons.
glucose PET in grading tumor recurrence in
patients treated for glioma43,44 Technetium-
99m-labeled sestamibi has been reported to
detect recurrent tumor after radiation
therapy.45 Sestamibi imaging can also identify dent systems and choline marks cell membrane
activity of the MDR gene and P-glycoprotein.46 turnover. Choline levels are elevated with
increased cell turnover. Lactate marks anaero-
Magnetic resonance spectroscopy (MRS) bic metabolism and is never measurable in
This procedure is performed in a fashion normal brain. Lipids may accumulate in areas
similar to a standard MRI except that of necrosis. The pattern of chemicals can help
biochemical spectra from areas of interest are distinguish the grade of glioma, and sometimes
measured (Fig. 3.7). The major spectra assayed the histologic type of glioma, and distinguish
by proton MRS are N-acetyl aspartate (NAA), tumors from infections and demyelinating
phosphocreatine, creatine, choline-containing lesions.47 However, these data are preliminary
compounds, myoinositol, several amino acids, and the technique should still be considered
including gamma-aminobutyric acid, lipids, experimental.
lactate and glucose. NAA is a marker of Current evidence suggests that NAA and
neuronal and axonal density. Its presence is gamma-aminobutyric acid are decreased in
decreased with neuronal loss from tumor, brain tumors, whereas choline is increased and
ischemia, necrosis, infection and other lactate levels correlate with histologic grade
processes that damage neurons. Creatine and (Fig. 3.8). Brain abscesses generally show
phosphocreatine are markers of energy-depen- absent creatine, choline and NAA with a large

85
PRINCIPLES OF DIAGNOSIS

CHO
Functional MRI (fMRI)
Areas of brain function can be identified. The
fMRI technique takes advantage of the fact
that when an area of the brain is activated,
CR
blood flow increases, and therefore propor-
tionate oxygen extraction decreases in the
activated region. The change in oxyhemoglobin
NAA
LAC to deoxyhemoglobin ratio results in an
increased signal intensity on the MR scan.48
This change can be imaged on a standard MRI
without using contrast, although greater sensi-
4 3 2 1 0 PPM tivity is achieved by scanners with echoplanar
capability. The technique is particularly useful
for identifying language and motor areas. fMRI
Figure 3.8 is clinically useful for presurgical planning,
Magnetic resonance spectrogram of a high-grade allowing the surgeon to maximally resect a
glioma. Note that the choline peak (CHO) is higher tumor while avoiding areas that perform criti-
than the creatinine peak (CR) and substantially
higher than the N-acetyl aspartate (NAA) peak. NAA
cal neurologic functions (Fig. 4.2).
represents neurons and axons. The low peak
indicates replacement of these structures by tumor.
Lactate (LAC), not identifiable in normal brain, is
Biopsy
also present, suggesting anaerobic metabolism. Although MRI may suggest the histologic
diagnosis (for example most low-grade tumors
do not contrast enhance, whereas most high-
grade tumors do), only biopsy is definitive.49
An exception to this rule occurs in primary
lactate peak; amino acids are also elevated CNS lymphoma, in which lumbar puncture or,
in an abscess. Demyelinating disorders are if the eyes are involved, vitrectomy, may yield
marked by normal or decreased NAA and malignant cells, obviating the necessity for
increased choline. Meningiomas have no brain biopsy (Chapter 11). In other tumors,
creatine and no NAA but show an increased lumbar puncture poses a risk of cerebral herni-
choline and increased alanine peak. ation and rarely contributes to diagnosis.
Phosphorus MRS allows the measurement Stereotactic (from the Greek word stereo for
of varied phosphate-containing compounds, three dimensions and the Latin term tactic for
including phosphodiesters, phosphomono- touch) needle biopsy is a technique used to
esters, phosphocreatine, ATP, ADP and obtain tissue in patients whose tumor location,
inorganic phosphate. Phosphorus MRS has extent or multiplicity precludes craniotomy and
been studied less, and its role in identifying operative removal. The procedure can be done
brain tumors is uncertain. Preliminary evidence either under local or general anesthesia. The
suggests that the phosphocreatine/inorganic technique is usually performed by bolting a
phosphate ratio is reduced because the pH of frame to the patients head under local anesthe-
brain tumors is alkaline, a counterintuitive sia and performing a CT or MR scan to
observation. localize the lesion. The frame defines the

86
LABORATORY DIAGNOSIS

coordinates. Recently, frameless systems have The second complication is that of neurologic
become available which provide the same local- worsening without bleeding. There is little in
izing information without requiring a frame to the literature on this complication, but we have
be secured to the skull. The needle is passed found a significant number of patients (partic-
through a burr-hole to the appropriate spot ularly those with widespread and diffuse
and a core of tissue is removed. Several passes tumors) whose neurologic condition worsens
with the needle may be necessary to procure substantially after stereotactic needle biopsy in
sufficient tissue to establish a diagnosis.50 In the absence of hemorrhage. Most recover over
experienced hands (both the surgeon and the several days to a few weeks, but some do not.
neuropathologist), a diagnosis can be estab- The neurologic worsening is of two types. One
lished in over 90% of patients (Fig. 3.9). is an increase in focal signs (e.g. hemiparesis).
Molecular genetic analysis can be performed The second is an alteration of mental status
on most samples,51 although sampling error is without focal signs. Some of our patients who
a potential problem. demonstrate memory loss or personality change
Stereotactic needle biopsy can cause may, following a stereotactic needle biopsy,
problems. Although it is easily performed, the become and remain lethargic or confused
small sample retrieved may make it difficult for without making a substantial recovery.
the neuropathologist to establish a diagnosis.
Even when the pathologist can establish a
diagnosis, he may not be able to accurately
grade the tumor. The neurosurgeon performing
a stereotactic needle biopsy should direct the
needle to what appears to be the highest grade
of the tumor. This can be established either by
areas of contrast enhancement on the MR scan,
or by areas of hypermetabolism on the PET
scan.
Stereotactic needle biopsy can also cause two
major complications. The first is bleeding.
Although the overall rate of symptomatic bleed-
ing in patients who undergo stereotactic needle
biopsy is about 1%, the rate is substantially
higher in patients with diffuse astrocytoma,
particularly those with high-grade lesions. In
one study, as many as 6% of patients with
glioblastoma multiforme bled after stereotactic
needle biopsy.52 Complications are fewer in
lower-grade tumors, but still greater than 1%.
Asymptomatic small hemorrhages identified on
Figure 3.9
post-biopsy scan may affect as many as 60% of Stereotactic needle biopsy of a patient with a
patients.53 In patients with AIDS, post-biopsy progressive hemiparesis. The tissue core obtained
mortality (30 days) was 2.9% and morbidity revealed increased cellularity with atypical cells,
was 8.4%.54 suggesting astrocytoma.

87
PRINCIPLES OF DIAGNOSIS

Differential diagnosis have often encountered a similar finding in


brain tumors. Subacute infarcts that promi-
Table 3.8 lists some lesions that must be nently enhance can also be difficult to differ-
considered in the differential diagnosis of entiate from a tumor. Often, the clinical
intracranial lesions. history, pattern of enhancement (i.e. ribbon-
Three main non-neoplastic lesions should be like following cortical gyri) and surrounding
considered in the differential diagnosis. The edema (little in subacute infarct, much in brain
first is ischemic infarction. Older patients with tumor) help clarify the diagnosis.
intracranial tumors often present with transient A second important distinction occurs in
symptoms presumably due to seizures. Unlike patients with large contrast-enhancing lesions
the short-lived focal seizures occurring with caused by demyelinating disease that have the
other epileptogenic lesions, these episodes may appearance of a high-grade diffuse astro-
last many minutes to hours, are often associ- cytoma. One distinction between demyelinating
ated with abnormalities of cognition and lesions and glioblastoma is that the contrast-
behavior, and typically cause negative signs, enhancing ring is usually incomplete in
e.g. hemiparesis, but not typical tonicclonic demyelinating disease but complete in glio-
seizure activity. Because of their length, they blastoma.56 However, usually one cannot easily
may be confused with transient ischemic distinguish these lesions on MR scan, and
attacks or even small resolving cerebral biopsy may be necessary. It is the obligation of
infarcts. A CT scan without contrast may show the neurologist and neurosurgeon to call to the
an area of hypodensity that is interpreted as attention of the neuropathologist the possibil-
cerebral infarction. However, CT hypodensity ity of demyelinating disease, because the
associated with intrinsic brain tumors is usually macrophages in a demyelinating lesion may be
restricted to white matter and does not involve misinterpreted as tumor cells and patients
cortex. On the contrary, cerebral infarction is subjected to major resection and radiation
generally triangular-shaped with the base at the therapy. Radiation therapy appears to be
cortical surface. A contrast-enhanced MR scan particularly damaging in patients with demyeli-
often, but not always, establishes the diagno- nating disease, and the distinction is therefore
sis. Hyperintensity on a diffusion-weighted vital.57
MRI is characteristic of acute ischemia, but we Cerebral infection can mimic brain tumors.
The diagnosis is often clinically obvious when
the infection occurs in immune-suppressed
Table 3.8 patients or patients suffering from a severe
Differential diagnosis of brain tumors.17 systemic infection. Furthermore, certain MR
characteristics (as indicated in a previous
Hematoma section) suggest infection rather than neoplasm,
Abscess55 but often the diagnosis cannot be made clini-
Granuloma cally.55 In those cases, biopsy either as part of
Parasitic infections, e.g. Cysticercosis, resection of a single surgically accessible lesion,
histoplasmosis
or by needle biopsy in patients who have
Vascular malformations
Multiple sclerosis inaccessible and multiple lesions, will establish
Cerebral infarcts the diagnosis. The clinician should indicate to
the surgeon the possibility of an infection, so

88
LABORATORY DIAGNOSIS

that appropriate cultures are performed on the and bizarre-appearing tumor cells often
surgical material. mistaken for glioblastoma multiforme.
The individual histologic characteristics of
each tumor are discussed in the chapters on
Pathology
individual tumors in Part 2 of this book. A few
A correct pathologic diagnosis is essential to general principles that apply to most intra-
prescribe appropriate treatment and to predict cranial tumors are listed here:
prognosis. Examples abound. Meningiomas
that can be completely resected need no further 1. All tumor specimens should be reviewed by
treatment; they do not metastasize. Hemangio- a neuropathologist. Neuropathology is as
pericytomas, once believed to be a variant of different from general pathology as neurol-
meningioma, have a much worse prognosis and ogy is from internal medicine, and exper-
often metastasize. Postoperative radiation tise in the specialty is essential for making
therapy is required. Atypical or anaplastic an appropriate diagnosis. That review
meningiomas require focal radiation therapy should be completed before any postoper-
even when completely resected. Current ative treatment is undertaken.
evidence suggests that anaplastic astrocytomas 2. The responsible physician should review the
are best treated with radiation therapy slides with the neuropathologist. Often, vital
followed by chemotherapy, whereas anaplastic information, not available on the standard
oligodendrogliomas are best treated by requisition form, is exchanged during a
chemotherapy, perhaps even without radiation. personal review of the slides with the
The prognosis for intracranial germinomas and neuropathologist. One example will suffice.
mature teratomas is excellent, whereas the One of our patients with a large contrast-
prognosis for other intracranial germ cell enhancing mass that the neuropathologist
tumors is poor. was content to read as a glioblastoma had
Unfortunately, the histologic diagnosis of a history of a melanoma removed 2 years
intracranial tumors is sometimes difficult for earlier. The patient was told that it was
several reasons: (1) often the surgeon can cured and did not report it to the surgeon.
supply only a very small biopsy sample because Only during the course of a postoperative
the location of the tumor prohibits a larger physical examination, when a small scar
resection; (2) many intracranial tumors are was found on the patients leg, was the
quite heterogeneous with substantial variability history elicited. Once the neuropathologist
in the histology of the tumor from area to area; was informed of this, he performed appro-
(3) some relatively benign-behaving tumors can priate special stains to establish the diagno-
appear malignant under the microscope sis of a metastatic melanoma. It is often
whereas some relatively clinically aggressive helpful to review the imaging studies with
tumors can appear low-grade under the micro- the neuropathologist, particularly when
scope. A few examples will suffice. A few determining tumor grade or identifying rare
mitotic figures in an otherwise benign menin- variants of glial tumors (Chapter 5).
gioma do not alter the excellent clinical 3. The surgeon should submit as large a
prognosis or the treatment. The relatively sample as possible. This not only allows
benign pleomorphic xanthoastrocytomas may the neuropathologist sufficient material for
contain a few mitoses, lymphocytic infiltration making a diagnosis, but also allows an

89
PRINCIPLES OF DIAGNOSIS

estimate of the degree of heterogeneity. If a stereotactic needle biopsy, a frozen


the patient is not a candidate for resection, section or a smear preparation can tell the
but only a stereotactic needle biopsy, the surgeon that he has successfully biopsied a
biopsy should include the area that appears tumor, even if the exact nature of the
most aggressive on preoperative imaging, tumor must await identification on perma-
such as any area of contrast enhancement nent sections. During the course of open
on gadolinium MR scan or a region of surgery, identification of the tumor may
hypermetabolism on FDG PET. determine the course of surgery. For
4. Utilize the most modern diagnostic example, a lymphoma does not require
techniques to identify the tumor. Several extirpative surgery but is best treated by
techniques beyond routine staining of chemotherapy, whereas gliomas benefit
pathologic specimens now aid in diagnosis. from removal of as much tumor as is feasi-
These include electron microscopy, ble. Often these two tumors are not distin-
immunohistochemical techniques and guished preoperatively but only at the time
identification of genetic abnormalities. of frozen section. The pathologist may also
Molecular genetic analysis can be be able to tell on frozen section whether
performed on needle biopsy specimens.51 some tissue should be fixed for future
Electron microscopy of appropriately electron microscopy to assist in definitive
prepared tissue can distinguish central diagnosis. Finally, during the course of
neurocytomas (neuronal tumors) with their surgery the surgeon may want to sample
synaptic vesicles from their almost identi- the margins of his resection to determine
cally appearing oligodendrogliomas, which whether tumor remains or he has reached
lack synaptic vesicles. In similar fashion, normal white matter. Immediate histology
microvilli and cilia identify an ependy- can also be procured by smear or touch
moma that otherwise appears to be an preparations.62 In these preparations, cellu-
astrocytoma. B-cell markers can distinguish lar processes and nuclei are much better
primary CNS lymphoma from inflamma- preserved than in frozen section although
tory diseases. Synaptophysin helps identify the tumor pattern is lost.63
tumors of neuronal origin, and two 6. CSF is sometimes useful to identify tumor
recently described antibodies, anti-Hu58 cells, particularly if the leptomeninges have
and Neu-N,59 may prove to be even more been seeded. In most instances, however, it
effective in distinguishing neuronal and is of no use in the evaluation of a brain
mixed glioneuronal tumors. Although there tumor, and the risks of lumbar puncture
is no clear marker yet for identifying an far outweigh the benefits. In the case of
oligodendroglioma, deletions of chromo- suspected primary CNS lymphoma, how-
somes 1p and 19q are common in oligo- ever, examination of CSF can be fruitful in
dendrogliomas and not in astrocytomas.60 identifying lymphoma cells, obviating the
Other genetic markers may prove to be necessity for brain biopsy. Immuno-
equally valuable in the future.61 histochemistry for B-cells is particularly
5. It is often wise to request a frozen section helpful, since most primary CNS
during the course of surgery. Immediate lymphomas are B-cell lymphomas, whereas
identification of histologic material can T-cells in the spinal fluid are generally
have several purposes. During the course of associated with inflammatory states. It is

90
LABORATORY DIAGNOSIS

extremely rare for tumors arising outside of 1), distinguish cells in all phases of the cell
the substance of the brain, such as menin- cycle save for Go. The number of positive
giomas, Schwannomas and neurofibromas, nuclei correlates well with the tumor grade
to exude cells into the CSF unless malig- and biological behavior in most studies.65
nant variants of these tumors have seeded Tumors with MIB indices over 5%, even
the leptomeninges. with otherwise benign histology, should be
7. Routine histology hematoxylin and eosin viewed with suspicion by the clinician.
staining of formalin-fixed paraffin-embed- Different isoforms of S100 protein are
ded tissue still remains the most common expressed in different tumors; recent
histologic test for intracranial tumors. A reports indicate that modifications of S100
variety of other stains are used for identi- A3 protein expression distinguished
fication of particular abnormalities in pilocytic from diffuse astrocytoma.65 The
individual tumors. Examples include retic- presence of p53 protein by immunohisto-
ulin stains, which are often positive in chemical staining occurs in many astro-
ganglioglioma, Massons trichome stain to cytomas, particularly of higher grade, but
identify gliosarcoma, and a myelin stain to is rare in oligodendrogliomas.
differentiate tumor from multiple sclerosis.
8. Immunohistochemistry tumors of glial
Approach to the patient
origin are often positive for glial fibrillary
acidic protein (GFAP). This stain is often Figures 3.10 and 3.11 describe the general
positive in astrocytomas, particularly of approach to a patient who presents with
higher grade, and in some oligodendro- seizures or other neurologic symptoms or signs.
gliomas. Reactive astrocytosis also gives If after a history and physical examination the
positive reactions. A recent report describes physician suspects a structural lesion, he should
a specific marker for tumors of oligo- proceed to MR scanning both with and without
dendroglial origin.64 Microglia are visual- contrast. If the scan suggests an intracranial
ized by macrophage markers, such as tumor, further decisions will depend on whether
CD68, which helps to distinguish demyeli- the mass enhances or not. The algorithm does
nating disorders from tumor. Antibodies not apply to every patient with a brain tumor
against cytokeratins are helpful in distin- or every type of brain tumor, but describes a
guishing metastatic epithelial tumors from general approach which is most applicable to
primary tumors. The HMB-45 stain identi- intrinsic intracranial tumors such as gliomas.
fies melanoma and differentiates it from Appropriate therapy depends on the clinical
primary gliomas. Immunohistochemistry state of the patient and the type of tumor. The
can also help in grading tumors. Cell diagnosis and treatment of specific tumors are
proliferation markers, such as Ki-67 (MIB- detailed in Chapters 513.

91
PRINCIPLES OF DIAGNOSIS

Seizure or other neurologic


symptom(s) or sign(s)

Suspect structural lesion Do not suspect


structural lesion

MRI with contrast


Appropriate care

Enhancing
Non-enhancing mass mass
No mass

PET + MRS Steroids (if indicated)


Anticonvulsants (if indicated)

Low grade High grade


Multifocal, unifocal Unifocal operable
but inoperable or
suspect PCNSL

Seizures only Neurologic Resection with frozen


symptoms section analysis
or signs
Stereotactic biopsy

Treat seizures

Appropriate therapy depending


on tumor type
Success Failure

Follow with MRI

Figure 3.10
An algorithm describing the approach to a patient who presents with seizures or other neurologic symptoms or
signs. The approach is a general one and does not apply to every patient or to every type of tumor. The
indications for steroids and anticonvulsants are described in detail in Chapter 4.

92
LABORATORY DIAGNOSIS

Figure 3.11
Evaluation of a patient with a suspected primary brain tumor. This 50-year-old woman presented with mild
progressive weakness of the right leg. An MR scan (upper left panel) revealed a ring-enhancing mass in or near
the motor strip. A PET scan (middle panel) revealed the ring enhancement to be hypermetabolic (arrow) and
the center of the lesion to be iso- or hypometabolic, perhaps representing necrosis. An MRS (upper right panel)
revealed a high choline peak (left arrow) with a smaller N-acetyl aspartate (NAA) peak (middle arrow). There
was a high lipid peak (right arrow) compatible with necrosis. The patient was advised that the tumor was
inoperable because of its proximity to the motor strip. However, functional MR (lower left) revealed that a
posterior approach might successfully avoid both the laterally replaced sensorimotor areas (arrow) and the
anteriorly placed supplementary motor area (double arrow). About 90% of the tumor was removed. The
patient had a modest but transient increase in her weakness after the operation. The tumor proved to be a
glioblastoma multiforme (lower right).

93
PRINCIPLES OF DIAGNOSIS

References 14. Lundberg N. Continuous recording and control


of ventricular fluid pressure in neurosurgical
1. Liigant A, Haldre S, Oun A, Linnamgi , Saar practice. Acta Neurol Scand 1960; 149: 1193.
A, Asser T, Kaasik AE. Seizure disorders in 15. Lote K, Stenwig AE, Skullerud K, Hirschberg
patients with brain tumors. Eur Neurol 2001; H. Prevalence and prognostic significance of
45: 4651. epilepsy in patients with gliomas. Eur J Cancer
2. Frishberg BM. Neuroimaging in presumed 1998; 34: 98102.
primary headache disorders. Semin Neurol 16. Holt-Seitz A, Wirrell EC, Sundaram MB.
1997; 17: 37382. Seizures in the elderly: etiology and prognosis.
3. Ilyin SE, Gayle D, Gonzlez-Gmez I, Miele Can J Neurol Sci 1999; 26: 1104.
ME, Plata-Salamn CR. Brain tumor develop- 17. Vick N. Intracranial tumors. In: Bennett JC,
ment in rats is associated with changes in Plum F, eds. Cecil textbook of medicine.
central nervous system cytokine and neuropep- Philadelphia: W.B. Saunders Co., 1996:
tide systems. Brain Res Bull 1999; 48: 36373. 212530.
4. Katzman GL, Dagher AP, Patronas NJ. 18. Rudge P, Warrington EK. Selective impairment
Incidental findings on brain magnetic resonance of memory and visual perception in splenial
imaging from 1000 asymptomatic volunteers. tumours. Brain 1991; 114: 34960.
JAMA 1999; 282: 369. 19. Terry JB, Rosenberg RN. Frontal lobe ataxia.
5. Behrens PF, Ostertag CB, Warnke PC. Regional Surg Neurol 1995; 44: 5838.
cerebral blood flow in peritumoral brain edema 20. Shuping JR, Rollinson RD, Toole JF. Transient
during dexamethasone treatment: a xenon- global amnesia. Ann Neurol 1980; 7: 2815.
enhanced computed tomographic study. 21. Meador KJ, Swift TR. Tinnitus from intra-
Neurosurgery 1998; 43: 23540. cranial hypertension. Neurology 1984; 34:
6. Kordas M, Czirjak S, Doczi T. The spinal 125861.
tumour related hydrocephalus. Acta Neurochir 22. Groves MD, McCutcheon IE, Ginsberg LE,
(Wien) 1997; 139: 104954. Kyritsis AP. Radicular pain can be a symptom
7. Forsyth PA, Posner JB. Headaches in patients of elevated intracranial pressure. Neurology
with brain tumors: a study of 111 patients. 1999; 52: 10935.
Neurology 1993; 43, 167883. 23. Gilman S. Imaging the brain. N Engl J Med
8. Pfund Z, Szapary L, Jaszberenyi O, Nagy F, 1998; 338: 88996.
Czopf J. Headache in intracranial tumors. 24. Hoffman JM. New advances in brain tumor
Cephalalgia 1999; 19: 78790. imaging. Curr Opin Oncol 2001; 13: 14853.
9. Fricke B, Andres KH, Von Dring M. Nerve 25. Wieshmann UC, Symms MR, Parker GJM et al.
fibers innervating the cranial and spinal Diffusion tensor imaging demonstrates devia-
meninges: morphology of nerve fiber terminals tion of fibres in normal appearing white matter
and their structural integration. Microsc Res adjacent to a brain tumour. J Neurol Neurosurg
Tech 2001; 53: 96105. Psychiatry 2000; 68: 5013.
10. Pascual J, Iglesias F, Oterino A, Vazquez- 26. Pomper MG, Port JD. New techniques in MR
Barquero A, Berciano J. Cough, exertional, and imaging of brain tumors. Magn Reson Imaging
sexual headaches: an analysis of 72 benign and Clin N Am 2000; 8: 691713.
symptomatic cases. Neurology 1996; 46: 15204. 27. Lee PL, Gonzalez RG. Magnetic resonance
11. Edmeads JG. Headache as a symptom of organic spectroscopy of brain tumors. Curr Opin Oncol
diseases. Curr Opin Neurol 1995; 8: 2336. 2000; 12: 199204.
12. Mann SD, Danesh BJ, Kamm MA. Intractable 28. Snyder H, Robinson K, Shah D, Brennan R,
vomiting due to a brainstem lesion in the Handrigan M. Signs and symptoms of patients
absence of neurological signs or raised intra- with brain tumors presenting to the emergency
cranial pressure. Gut 1998; 42: 8757. department. J Emerg Med 1993; 11: 2538.
13. Rowe FJ, Sarkies NJ. Assessment of visual 29. Zimmerman RA. Brain tumor imaging. In:
function in idiopathic intracranial hypertension: Vecht C, ed. Neuro-oncology, Part 1.
a prospective study. Eye 1998; 12: 1118. Amsterdam: Elsevier, 1997: 16792.

94
REFERENCES

30. Mitchell P, Wilkinson ID, Hoggard N et al. 41. Schwartz RB, Holman BL, Polak JF et al. Dual-
Detection of subarachnoid haemorrhage with isotope single-photon emission computerized
magnetic resonance imaging. J Neurol tomography scanning in patients with glio-
Neurosurg Psychiatry 2001; 70: 20511. blastoma multiforme: association with patient
31. Linfante I, Llinas RH, Caplan LR, Warach S. survival and histopathological characteristics
MRI features of intracerebral hemorrhage of tumor after high-dose radiotherapy. J
within 2 hours from symptom onset. Stroke Neurosurg 1998; 89: 608.
1999; 30: 22637. 42. Maria BL, Drane WE, Mastin ST, Jimenez LA.
32. Lote K, Egeland T, Hager B, Skullerud K, Comparative value of thallium and glucose
Hirschberg H. Prognostic significance of CT SPECT imaging in childhood brain tumors.
contrast enhancement within histological Pediatr Neurol 1998; 19: 3517.
subgroups of intracranial glioma. J Neurooncol 43. Bader JB, Samnick S, Moringlane JR et al.
1998; 40: 16170. Evaluation of L-3-[123I]iodo--methyltyrosine
33. Essig M, Knopp MV, Schoenberg SO et al. SPET and [18F]fluorodeoxyglucose PET in the
Cerebral gliomas and metastases: assessment detection and grading of recurrences in patients
with contrast-enhanced fast fluid-attenuated pretreated for gliomas at follow-up: a compar-
inversion-recovery MR imaging. Radiology ative study with stereotactic biopsy. Eur J Nucl
1999; 210: 5517. Med 1999; 26: 14451.
34. Siegal T, Rubinstein R, Tzuk-Shina T, Gomori 44. Matheja P, Schober O. 123I-IMT SPET: intro-
JM. Utility of relative cerebral blood volume ducing another research tool into clinical
mapping derived from perfusion magnetic neurooncology? Eur J Nucl Med 2001; 28: 14.
resonance imaging in the routine follow up of 45. Soler C, Beauchesne P, Maatougui K et al.
brain tumors. J Neurosurg 1997; 86: 227. Technetium-99m sestamibi brain single-photon
35. Delbeke D. Oncological applications of FDG emission tomography for detection of recurrent
PET imaging: brain tumors, colorectal cancer gliomas after radiation therapy. Eur J Nucl Med
lymphoma and melanoma. J Nucl Med 1999; 1998; 25: 164957.
40: 591603. 46. Hendrikse NH, Franssen EJ, van der Graaf WT,
36. Shoup TM, Olson J, Hoffman JM et al. Synthesis Vaalburg W, de Vries E.G. Visualization of
and evaluation of [18F]1-amino-3-fluorocyclo- multidrug resistance in vivo. Eur J Nucl Med
butane-1-carboxylic acid to image brain tumors. 1999; 26: 28393.
J Nucl Med 1999; 40: 3318. 47. De Stefano N, Caramanos Z, Preul MC,
37. Inoue T, Shibasaki T, Oriuchi N et al. 18F Francis G, Antel JP, Arnold DL. In vivo differ-
-methyl tyrosine PET studies in patients entiation of astrocytic brain tumors and
with brain tumors. J Nucl Med 1999; 40: isolated demyelinating lesions of the type seen
399405. in multiple sclerosis using 1H magnetic
38. Mineura K, Shioya H, Kowada M et al. Blood resonance spectroscopic imaging. Ann Neurol
flow and metabolism of oligodendrogliomas: 1998; 44: 2738.
a positron emission tomography study with 48. Forster BB, MacKay AL, Whittall KP et al.
kinetic analysis of 18F-fluorodeoxyglucose. J Functional magnetic resonance imaging: the
Neurooncol 1999; 43: 4957. basics of blood-oxygen-level dependent (BOLD)
39. Sasaki M, Kuwabara Y, Yoshida T et al. A imaging. Can Assoc Radiol J 1998; 49: 3209.
comparative study of thallium-201 SPET, 49. Hall WA. The safety and efficacy of stereotac-
carbon-11 methionine PET and fluorine-18 tic biopsy for intracranial lesions. Cancer 1998;
fluorodeoxyglucose PET for the differentiation 82: 174955.
of astrocytic tumours. Eur J Nucl Med 1998; 50. Soo TM, Bernstein M, Provias J, Tasker R,
25: 12619. Lozano A, Guha A. Failed stereotactic biopsy
40. Maruyama I, Sadato N, Waki A et al. in a series of 518 cases. Stereotact Funct
Hyperacute changes in glucose metabolism of Neurosurg 1995; 64: 18396.
brain tumors after stereotactic radiosurgery: a 51. Mller MB, Schmidt MC, Schmidt O et al.
PET study. J Nucl Med 1999; 40: 108590. Molecular genetic analysis as a tool for evaluating

95
PRINCIPLES OF DIAGNOSIS

stereotactic biopsies of glioma specimens. J 59. Wolf HK, Buslei R, Schmidt-Kastner R et al.
Neuropathol Exp Neurol 1999; 58: 405. NeuN: a useful neuronal marker for diagnostic
52. Bernstein M, Parrent AG. Complications of CT- histopathology. J Histochem Cytochem 1996;
guided stereotactic biopsy of intra-axial brain 44: 116771.
lesions. J Neurosurg 1994; 81: 1658. 60. Cairncross JG, Ueki K, Zlatescu MC et al.
53. Kulkarni AV, Guha A, Lozano A, Bernstein M. Specific genetic predictors of chemotherapeutic
Incidence of silent hemorrhage and delayed response and survival in patients with anaplas-
deterioration after stereotactic brain biopsy. J tic oligodendrogliomas. J Natl Cancer Inst
Neurosurg 1998; 89: 315. 1998; 90: 14739.
54. Skolasky RL, Dal Pan GJ, Olivi A, Lenz FA, 61. Firlik KS, Martinez AJ, Lunsford LD. Use of
Abrams RA, McArthur JC. HIV-associated cytological preparations for the intraoperative
primary CNS morbidity and utility of brain diagnosis of stereotactically obtained brain
biopsy. J Neurol Sci 1999; 163: 328. biopsies: a 19-year experience and survey of
55. Klein CJ, DiNapoli RP, Temesgen Z, Meyer FB. neuropathologists. J Neurosurg 1999; 91: 4548.
Central nervous system histoplasmosis mimick- 62. Slowinski J, Harabin-Slowinska M, Mrwka R.
ing a brain tumor: difficulties in diagnosis and Smear technique in the intra-operative brain
treatment. Mayo Clin Proc 1999; 74: 8037. tumor diagnosis: its advantages and limitations.
56. Masdeu JC, Quinto C, Olivera C et al. Open- Neurol Res 1999; 21: 1214.
ring imaging sign highly specific for atypical 63. Giannini C, Scheithauer BW, Burger PC et al.
brain demyelination. Neurology 2000; 54: Cellular proliferation in pilocytic and diffuse
142733. astrocytomas. J Neuropathol Exp Neurol 1999;
57. Peterson K, Rosenblum MK, Powers JM et al. 58: 4653.
effect of brain irradiation on demyelinating 64. Marie Y, Sanson M, Mokhtari K et al. OLIG2
lesions. Neurology 1993; 43: 210512. as a specific marker of oligodendroglial tumour
58. Gultekin SH, Dalmau J, Graus Y, Posner JB, cells. Lancet 2001; 358: 298300.
Rosenblum MK. Anti-Hu immunolabeling as 65. Camby I, Nagy N, Lopes MB et al. Supratentorial
an index of neuronal differentiation in human pilocytic astrocytomas, astrocytomas, anaplastic
brain tumors-a study of 112 central neuro- astrocytomas and glioblastomas are characterized
epithelial neoplasms. Am J Surg Pathol 1998; by a differential expression of S100 proteins.
22: 195200. Brain Pathol 1999; 9: 119.

96
4
Principles of therapy

Introduction three conventional therapeutic approaches


surgery, radiation and chemotherapy as well as
Intracranial tumor therapy can be directed at several forms of experimental therapy.
the tumor (definitive therapy) or at symptom Supportive therapy encompasses management
control (supportive therapy) or, preferably, at of tumor symptoms, e.g. seizures, and of
both (Table 4.1). Definitive therapy includes the complications of the tumor or its treatment, e.g.
deep vein thrombosis, radiation necrosis. This
Table 4.1
chapter considers the principles of both defini-
Therapy of intracranial tumors. tive and supportive therapy. Therapy directed at
individual tumors is considered in later
chapters.
Definitive therapy
Surgery
Biopsy
Resection
Radiation
Principles of surgery
External beam
Radiosurgery or stereotactic radiotherapy Introduction
Heavy particles
Surgery is the most important single modality
Brachytherapy
Chemotherapy in the treatment of intracranial tumors.1,2
Parenteral Truly benign tumors, such as pituitary adeno-
Local mas and meningiomas, are often cured by
Experimental modalities surgery. For those tumors that cannot be
Angiogenesis inhibitors
cured, almost all observers believe that surgery
Growth factor inhibitors
Differentiating agents substantially enhances both the duration and
Immunotherapy quality of survival. The goals of surgery are
Gene therapy several (Table 4.2).
Antisense oligonucleotides The first goal of surgery is to establish a
Supportive therapy diagnosis, either by biopsy or more definitive
Anticonvulsants
tumor resection. Stereotactic needle biopsy is
Corticosteroids
Anti-thrombosis agents usually reserved for: (1) surgically inaccessi-
Psychotropic agents ble tumors such as those in the basal ganglia,
Physical therapy corpus callosum or brainstem; (2) multifocal
tumors; (3) diffuse gliomatosis (Chapter 5);

97
PRINCIPLES OF THERAPY

Table 4.2 A second aim of surgery is cure. Meticulous


Goals of surgery. attempts to remove the entire tumor sometimes
result in cure without additional therapy. When
Establish the diagnosis cure is not possible, the goal is to remove as
Cure the patient much tumor as possible without compromising
Decrease tumor burden neurologic function. Removal of substantial
Relieve symptoms
amounts of even infiltrative tumors has several
Improve neurologic function
Extend duration and quality of life salutary effects:
1. It reduces the number of tumor cells that
must subsequently be eliminated by radia-
tion therapy and/or chemotherapy. RT and
(4) putative primary CNS lymphomas, a
chemotherapy exert their effects by killing
tumor better treated by chemotherapy than
a percentage of tumor cells regardless of
surgery (Chapter 11). With these exceptions,
tumor volume (see below). Thus, these
surgery should remove as much tumor as
modalities are more likely to improve
feasible.
survival when a smaller rather than a larger
Stereotactic needle biopsy is performed under
number of cells remain after resection.
either CT or MR guidance.3 Fiducials (Latin for
2. Reducing tumor size decreases areas of
trust, thus a fixed point of reference) or a frame
hypoxia, thus increasing radiation sensitivity.
is fixed to the head; a small incision is made in
3. Because small tumors often grow exponen-
the scalp and a burr-hole drilled through the
tially, but larger tumors grow more slowly,
skull. A needle is inserted through the burr-hole,
with many tumor cells in a quiescent phase,
guided by the scan, into what appears to be
surgical cytoreduction puts more cells into
the most malignant area of the tumor, and a
the cell cycle, making them susceptible to
core taken for histologic evaluation. In some
RT and chemotherapy. Therefore, even with
instances, several passes are made to ensure an
high-grade tumors such as glioblastoma,
adequate sample. Stereotactic needle biopsy and
most evidence suggests a direct correlation
its complications are discussed in Chapter 3.
between extent of surgery and improved
Needle biopsies have several limitations. The
duration and quality of survival.46 Extent of
limited tissue sample procured often makes
surgery is now recognized by most observers
diagnosis difficult, especially when attempting
to be an important prognostic factor.
to grade tumors such as gliomas. For example,
samples taken from the tumor edge may Because virtually no tissue is removed by needle
suggest low-grade glioma or even gliosis, biopsy in patients with large tumors, postopera-
whereas deeper samples may demonstrate a tive brain swelling often worsens neurologic
higher-grade histology. Stereotactic needle symptoms; however, a substantial resection of
biopsy is preferred for deep-seated lesions, such tumor provides space for any postoperative
as those in the brainstem or basal ganglia, or cerebral swelling to occur safely without causing
those where direct inspection will not assist the clinical deterioration. Several studies indicate that
surgeon. A biopsy performed under direct patients undergoing gross total resection of intrin-
inspection allows the surgeon to procure a sic brain tumors have less postoperative morbid-
larger and thus more easily diagnosed specimen ity and better neurologic function than do those
and to control bleeding. undergoing only biopsy or subtotal resection.7

98
PRINCIPLES OF SURGERY

(a) Figure 4.1


Old and new techniques for brain
tumor surgery. (a) A figure redrawn
from an illustration by neurosurgeon
Walter Dandy, indicating the surgical
approach used for brain tumors in
the first half of last century.
Corticosteroids were not available.
The trauma of the finger removing
the tumor engendered severe brain
edema. Mortality and morbidity
were high. (b) Illustration of modern
computerized imaged-guided therapy.
The viewing wand is a fully
computerized, frameless MR-guided
Finger in system that directs the surgeon
ventricle cleaning toward the target by continually
brain to base of providing the location of the
skull
operating site. A mechanical arm is
freely mobile and is attached to a
computer that contains the patients
(b)
preoperative image data.

Because of improved ability to identify function- As Fig. 4.1 illustrates, the surgery of
ally important areas of brain, e.g. by fMRI, the intracranial tumors has improved substantially
neurosurgeon is now able to remove more tumors because of technical improvements over the
safely than in the past, without compromising past several decades.9 The result has been
neurologic function.8 dramatic reduction in both mortality and

99
PRINCIPLES OF THERAPY

morbidity resulting from neurosurgical proce- bone and determine if the bone is eroded or
dures. In the days of the early neurosurgical invaded by tumor. An angiogram may be
giants, Dandy and Cushing, surgical mortalities required of some tumors both to evaluate the
of 2530% were common, and many patients vascularity of the tumor and to identify normal
who were operated on for brain tumors arterial and venous structures that may help
suffered substantial neurologic disability after dictate the neurosurgeons approach to the
the procedure. Of course, because of limited tumor. Sometimes MR angiography or MR
diagnostic techniques, the tumors were usually venography will suffice to identify the anatomy,
larger than those operated on today. obviating the need for more invasive arteriog-
Improvements in diagnostic, neuroanesthetic raphy. For a few highly vascular tumors, includ-
and surgical techniques have lowered operative ing some meningiomas, hemangioblastomas
mortality in the best centers to 3% or less and and metastases, preoperative embolization may
postoperative neurologic worsening to less than allow a better and safer resection.
10% (see below). If the tumor is in or close to a functionally
important area, a functional MRI will reliably
identify language, motor and sensory areas
Preoperative evaluation
(Fig. 4.2) and allow the surgeon to plan his
As indicated in Chapter 3, an MRI is usually approach to avoid those regions.10,11
sufficient for the neurosurgeon to plan the Identification of language areas is particularly
procedure. The day prior to surgery, a contrast- important when operating on dominant tempo-
enhanced MR scan is performed and fiducial ral lobe lesions, since they are quite variable.12
markers placed upon the scalp to allow use of Some surgeons prefer awake craniotomy to
frameless stereotactic guidance apparatus map language areas during surgery.13 For
during the procedure. In some instances, other patients undergoing needle biopsy, an FDG
imaging techniques give the surgeon additional PET scan will identify any areas of hyper-
information (Table 4.3). metabolism and perfusion MRI will identify
A CT scan of a skull base tumor may help areas of increased blood flow, both of which
the neurosurgeon evaluate the anatomy of the presumably represent the most rapidly growing
tumor, allowing the neurosurgeon to target that
area for biopsy.
The preoperative workup should also
Table 4.3 include, depending on the patients age and
Preoperative evaluation. general medical status, an evaluation of cardiac
and pulmonary function. Aspirin or aspirin-
All patients Some patients containing compounds (there are about 400 of
them) should be discontinued 710 days prior
Localizing MRI CT scan to surgery. Non-steroidal anti-inflammatory
Discontinue aspirin Functional MRI drugs (NSAIDs) also inhibit platelet function
Give anticonvulsants Angiogram
but do so reversibly and can be discontinued
Start steroids PET
Evaluate general Tumor embolization 48 h prior to surgery. Except in the case of
physical and primary central nervous system lymphomas
neurologic condition (PCNSL), corticosteroids in a dose equivalent
to 16 mg dexamethasone a day should be given

100
PRINCIPLES OF SURGERY

(a) Figure 4.2


The usefulness of functional
imaging in planning surgery. (a)
This 16-year old girl had
intractable seizures and a non-
enhancing mass in the right
posterior frontal lobe. Several
neurosurgeons refused surgery
because they believed that the
tumor was too close to the
sensorimotor strip. Functional
MR scan revealed sensory and
motor activity posteriorly and
medially to the tumor. A gross
total resection was carried out
without postoperative weakness.
The seizures ceased. (b) At the
(b)
time of surgery, areas close to
the sensorimotor strip are
marked with numbered tags.
Motor areas are identified by
stimulation of the cortex and
sensory areas by peripheral
stimulation.

for at least 48 h prior to the operative proce- (15 mg/kg) or fosphenytoin just prior to
dure. Other medications, except aspirin, surgery. The anticonvulsants can be continued
should be continued. Controversy exists for 2 months, and then if the patient has not
concerning the use of prophylactic anticonvul- had prior seizures, tapered or discontinued
sants in those patients who have not previ- (see below). For standard, clean neurosurgical
ously had a seizure. Two studies disagree. One procedures, we give a broad-spectrum antibi-
shows protection during the first postoperative otic such as cefazolin 1 g every 8 h for three
week;14 the other does not.15 We give glioma doses, the first just before the skin is incised.
patients a loading dose of phenytoin Anticoagulants are discussed on page 137.

101
PRINCIPLES OF THERAPY

Table 4.4
Operative procedures Operative tools.
Specific operative procedures differ depending
on the location and size of the tumor and the Frameless stereotaxy
goal of the surgeon (e.g. biopsy, subtotal resec- Ultrasound
tion, total resection). These factors are consid- Microscope
Cortical mapping
ered in the chapters on individual tumors.
Ultrasonic aspirator
For all patients undergoing craniotomy, Laser
during induction of anesthesia an arterial Endoscope
catheter is placed in the radial artery for Intraoperative MRI
continuous monitoring of blood pressure.
Some measure jugular bulb oxygen saturation
to ensure adequate cerebral perfusion.16
Pneumatic compression boots are placed on by tumor, it may have to be discarded and later
each calf. The patient is given 11.5 g/kg of replaced by an acrylic prosthesis.
a 20% solution of mannitol over 2030 min. Modern neurosurgical techniques include
This hyperosmolar substance extracts water several tools that allow better access to and
from normal, non-edematous brain. In some identification of a tumor once the skull has
instances, a lumbar drain is placed to drain been opened (Table 4.4).
cerebrospinal fluid (CSF). Both mannitol and
CSF drainage serve to slacken the brain for Frameless stereotaxy, ultrasound and
easier and safer retraction. If the patient is microscope
being operated on in the sitting position, a Frameless stereotactic systems, based on preop-
central venous line and a cortical Doppler are erative imaging, allow precise placement of the
used to monitor for air emboli. bony opening, accurate planning of the trajec-
Once the patient is positioned and prepared, tory to the tumor, and feedback about the
an incision is made in the scalp as defined by extent of resection. Some surgeons prefer,
the location of the tumor on MR scan and instead or in addition to frameless stereotaxy,
dictated by frameless stereotactic localization. to localize and approach the tumor using intra-
The stereotactic apparatus links the preopera- operative ultrasound applied directly to the
tive scan to landmarks on the surface of the cortical surface. This modality also provides
head, providing continuous information on the real time feedback about the presence of resid-
relationship of the surgeons instruments to the ual tumor and helps the surgeon to gain more
intracranial tumor. Both CT and MR stereotac- complete resections. The operating microscope
tic systems are accurate to within a few millime- will sufficiently magnify to allow good visual-
ters.17 Unfortunately, as the surgery proceeds, ization of blood vessels and other critical struc-
shifts of intracranial structures decrease the tures and, when combined with microdissection
reliability of the information that was based using micro-instruments, improves the safety of
on the preoperative scan. Intraoperative MRI surgical resections.
remedies this problem.18 The incision should
provide optimal exposure of the tumor but Cortical mapping
maintain an adequate blood supply to the scalp. If the tumor is in or near functionally impor-
The overlying skull is removed. If it is involved tant areas, such as language, primary motor or

102
PRINCIPLES OF SURGERY

sensory cortex, cortical mapping may comple- been secured (in some instances by raising the
ment fMRI (Fig. 4.2).19 Cortical mapping of systolic blood pressure to 140 mm Hg to
language can be carried out only by direct ensure there is no bleeding), the wound is
cortical stimulation in an awake patient being closed and the patient monitored in a recovery
operated on under local anesthesia.20 Motor room for several hours before being returned
and sensory cortex can be mapped by cortical to the neurosurgical unit.
stimulation or by somatosensory evoked poten-
tials in patients being operated on under
general anesthesia.19 Somatosensory evoked Postoperative care
potentials or nerve stimulation are also very At Memorial SloanKettering, we return
useful in assessing cranial nerve function patients from the recovery room to a small,
during surgery on the skull base. constant-observation unit, where they remain
for 2448 h (Table 4.5).
Ultrasonic aspirator, laser and The monitoring includes careful neurologic
neuroendoscope evaluation with particular attention to state of
An ultrasonic aspirator utilizes high-frequency consciousness, blood pressure via the arterial
vibrations to shatter tumor cells with sound catheter until the morning postsurgery (i.e.
waves before suctioning the debris away. 1214 h), and careful measurement of urine
The technique inflicts minimal damage on output via Foley catheter placed at the time of
surrounding normal structures and blood surgery. Depending on the extent of surgery
vessels and is especially effective in firm tumors and degree of preoperative edema, corticos-
such as meningiomas and acoustic neuromas. teroids in doses of 1640 mg a day are
Additional operative tools include an operating prescribed for several days following the
laser, sometimes useful in removing tumor in surgery and then tapered to the patients toler-
tight working places, and a neuroendoscope, a ance. Pneumatic pressure boots are maintained
tool that can be used to explore the ventricu- until the patient is fully ambulatory. Most
lar system, remove intraventricular masses, and patients are able to eat and are mobilized the
even explore the posterior fossa to aid acous- morning after surgery, first to a chair and then
tic neuroma surgery.21 to ambulation.
Intraoperative pathology
Intraoperative consultation with a skilled Table 4.5
neuropathologist establishes the diagnosis in Postoperative care.
most instances.22 Frozen sections help guaran-
tee that appropriate tissue has been obtained State of consciousness
for diagnosis. It may also help the surgeon Pupillary reactions
decide whether to proceed with further tumor Segmental neurologic exam
removal. For example, if a lymphoma is seen Cerebral perfusion pressure = blood
pressurevenous pressure
on a frozen section, there is no need for a gross
Intravenous fluids
total excision, which could lead to neurologic Corticosteroids
deficits. Anticonvulsants
After the tumor has been resected to the MRI
maximum extent possible and hemostasis has

103
PRINCIPLES OF THERAPY

For most neurosurgical procedures, one can hyperintense so early after the surgery is proba-
anticipate the patients neurologic examination bly because the exposure of the tumor bed to
will be the same or improved over the preop- room air, i.e. oxygen, has promoted the early
erative examination in the immediate postoper- development of paramagnetic methemoglobin),
ative period. Because the brain swells 2448 h but contrast enhancement indicates residual
after the surgical manipulation, some patients tumor. After about 72 hours, normal brain
become drowsy or lethargic, or have worsening structures surrounding the tumor bed begin to
focal signs after having initially appeared wide enhance and may remain enhanced for weeks to
awake, with improved neurologic function. In months following the surgery, thus precluding
most instances, this disappears after 2436 h accurate identification of residual tumor.
and, if it does not, can usually be obviated by For certain tumors, particularly those at the
increasing the dose of corticosteroids. Rarely, skull base or tumors operated on after radia-
the swelling can cause herniation and/or perma- tion therapy or prolonged use of steroids, a
nent neurologic disability. CSF leak may develop in the postoperative
A postoperative gadolinium-enhanced MR period. Bedrest and placement of a lumbar
scan is performed 2472 h after surgery to drain to lower CSF pressure are usually effec-
determine the extent of residual tumor, if any. tive treatments. Other complications that may
The pre-contrast T1 image is compared with the occur include hyponatremia and postoperative
post-contrast image (Fig. 4.3). Blood appears seizures. Cerebral infarction may be caused
hyperintense on both (the reason blood appears either by arterial occlusion or spasm or by

Figure 4.3
The value of immediate postoperative scans. This patient had a non-enhancing left temporal lobe tumor (left
panel). His physicians did not order an immediate postoperative scan. When the scan was repeated some weeks
later, the physicians became concerned that the enhancement represented a change in the biology of residual
tumor (middle panel). However, a PET scan (right panel) shows that the area was hypo- not hypermetabolic,
and after several months the enhancement disappeared. The tumor was a low-grade glioma.

104
PRINCIPLES OF SURGERY

Figure 4.4
A postoperative infarct causing weakness of the left face and arm. This 62-year-
old man with a long history of hypertension had a single generalized seizure. The
MR scan (not shown) showed a tumor involving the right temporal lobe and the
insula. Some of the tumor was in the sylvian fissure. His neurologic examination
was normal. A partial resection was carried out, and in the postoperative period
his face and arm were paralyzed, with the leg being relatively normal. The
diffusion-weighted image (left) showed evidence of an acute infarct (arrow). The
T2-weighted image (right) showed the recent infarct plus multiple hyperintensities
in the white matter related to hypertensive small vessel disease.

venous occlusion. Arterial occlusion or spasm Infection following clean craniotomy is rare,
causes bland infarction (Fig. 4.4). Tumors especially when prophylactic antibiotics are
involving the sylvian fissure, through which the used. Infection is more likely after second
middle cerebral artery passes, or meningiomas surgery in a patient who has been treated with
abutting veins that enter the sagittal sinus, are steroids and who has received RT, as both
at particularly high risk for postoperative treatments interfere with wound healing.
vascular complications. Interestingly, long-term remissions of high-
Another cause of neurologic deterioration grade tumor have been reported in several
after surgery may be the development of a patients after intracranial infection,23 perhaps a
hematoma in the operative bed. In any patient result of inhibition of angiogenesis.24
whose neurologic condition worsens signifi-
cantly in the postoperative period, an urgent
Future developments
unenhanced CT scan should be performed to
look for a hematoma. Hematomas usually As indicated above, surgical technology has
develop within the first 24 h postoperatively. improved dramatically. New techniques
Those of sufficient size to cause neurologic promise to improve the neurosurgical approach
symptoms require evacuation. to tumors even more in succeeding years.

105
PRINCIPLES OF THERAPY

Intraoperative MRI, robotic equipment and cases after surgical debulking or gross total resec-
endoscopic surgery tion. The goal of the radiation oncologist is to
Intraoperative MRI18 allows the surgeon to deliver radiation in a precise fashion to the
stand in an MR machine and, using ceramic tumor, usually defined by the area of contrast
rather than metal tools, to monitor the tumor enhancement on MR scan with a margin of
in real time as it is resected.25 This eliminates 23 cm, sparing as much as possible the
the errors that occur because of slight shifts of surrounding normal tissue.
the brain as the procedure is being carried out
that change the position of the tumor from the
Physics of radiation
preoperative MR scan. This technique is
already in place in some centers. Robotic X-rays and -rays represent two forms of
equipment and virtual reality techniques may, electromagnetic radiation used conventionally
in the future, replace the hands of the neuro- in clinical practice to treat cancer.30 X-rays are
surgeon. Endoscopic surgery has already generated by an electrical device known as a
proved useful in the treatment of vestibular linear accelerator, which accelerates electrons
schwannoma (Chapter 9)26 and colloid cysts to high energies so that they emit X-rays
(Chapter 12)27 for some neurosurgeons. when they encounter a metal target. -Rays,
produced by cobalt-60 teletherapy units, arise
through the decay of the radioactive isotope.
Electromagnetic radiation can be considered
Principles of radiation either as a wave or a quantum of energy, called
therapy a photon. When X-rays of sufficient energy are
absorbed in biological material, a photon may
Introduction
encounter an atom within a target molecule,
For most CNS tumors, RT is the second most leading to the release of an energetic electron
important treatment modality. For some tumors, from its orbit around the nucleus and a
such as germinomas, RT is curative; for others it secondary energy-attenuated photon. The
substantially prolongs survival or at least retards atom, having lost an electron, is now an ion.
progression. Techniques of radiation have Hence, X-rays are also referred to as ionizing
improved over the years, allowing higher doses radiation. The ejected electrons and the
to the tumor and sparing normal brain (Fig. 4.5). secondary photons interact with other cellular
For example, in the original Brain Tumor Study molecules, leading to a chain of reactions
Group prospective randomized trial of high- which dissipates the original photon energy
grade gliomas,28 the median survival of patients within the cell and produces a variety of short-
treated by surgery alone was 14 weeks, whereas lived, chemically unstable free-radical species.
those receiving postoperative whole-brain RT of Other atomic particles used to deliver radiation
5060 Gy had a median survival of 36 weeks. A therapy include electrons, produced by linear
direct relationship between dose and survival accelerators, and neutrons, protons, -particles
was also demonstrated. Patients receiving 50 Gy and heavy charged particles such as helium
had a median survival of 28 weeks, and those ions, which are generated by cyclotrons.
treated with 60 Gy had a median survival of 42 When radiation is absorbed, ionizing events
weeks.29 RT is usually delivered after the histo- are usually localized along tracks of individual
logic diagnosis has been established and in most charged particles with a pattern that is dependent

106
PRINCIPLES OF RADIATION THERAPY

(a) Figure 4.5


The evolution of radiation therapy for
brain tumors. Up to early 1980, most
patients with primary and metastatic
tumors of the brain received whole-
brain irradiation to lateral opposing
ports (a). In the early 1980s, when it
was discovered for most primary brain
tumors that recurrences were local
rather than elsewhere in the brain, the
treatment was modified to give 40 Gy
to the whole brain and an additional
20 Gy to the tumor (b). This was
further modified to deliver only partial
brain irradiation through bilaterally
opposing ports. This treatment
nevertheless irradiated a substantial
(b) portion of normal brain. The
development of conformal field
radiation (c) now allows one to treat
the tumor and a surround that is
probably infiltrated by microscopic
disease, but spares most of the normal
brain. The color bar indicates the dose
that that area received.

(c)

107
PRINCIPLES OF THERAPY

on the type of radiation involved. The energy more sensitive. Cells in G0 are resistant.
deposited per unit length of the track is referred Although these rules hold in general, there are
to as the linear energy transfer (LET). Densely a number of exceptions.
ionizing radiation with higher LET (e.g. neutron Ionizing radiation damages DNA either
radiation) causes more biological damage directly or indirectly. When a photon strikes the
than sparsely ionizing, low-LET radiation (e.g. DNA itself, ionizing that molecule, the damage
X-rays). is direct. This type of interaction is relatively
rare with X-rays, but it represents the dominant
Mechanisms of radiation damage process of high-LET radiation such as neutrons.
The predominant mechanism by which radia- Alternatively, if the radiation interacts with
tion kills mammalian cells is the reproductive other atoms or molecules within the cell,
(also known as clonogenic) pathway of cell especially water, to produce highly reactive free
death. The target is DNA, and double-strand radicals that secondarily damage the DNA, the
breaks are regarded as the specific lesion that action is indirect. Indirect action is the predom-
initiates the lethal response. Most radiation- inant mechanism in conventional external beam
induced DNA single-strand and some double- photon irradiation. The presence of molecular
strand breaks, particularly if both strands are oxygen prolongs the life of the reactive species
broken at the same site, are rapidly repaired that attack DNA, making oxygenated cells more
by constitutively expressed repair mecha- susceptible to radiation damage than hypoxic
nisms (known as sublethal damage repair). cells. On the other hand, sulfhydryl compounds
Residual unrepaired or misrepaired breaks neutralize and reduce the lifespan of free
result in genetic instability, increased radicals, and analogs of these agents are being
frequency of mutations and chromosomal investigated as radioprotectors. Radiation also
aberrations, and, thus, carcinogenesis. Lethal causes apoptosis, although significantly less
mutations or dysfunctional chromosomal frequently than clonogenic cell death. Ionizing
aberrations eventually lead to cell death at radiation also attacks lipids, proteins and RNA,
the first or a subsequent attempt of the causing cellular damage and probably cell death.
damaged cell to reproduce. When radiation is
directed at the brain, cellular damage may Radiation dose and fractionation schedules
become apparent only months or years later, The unit of measurement that describes the
because glial and Schwann cells reproduce amount of radiation energy absorbed per unit
slowly. mass or the absorbed dose was previously
Because DNA uncoiled during the reproduc- called a rad (for radiation absorbed dose). A
tive cycle is more susceptible to radiation rad is equal to 100 ergs of energy absorbed per
damage than DNA coiled in the more quiescent gram of tissue. The currently used unit of
phases of the cycle, cells are more radio- absorbed dose is the Gray (after Louis Gray, a
sensitive at particular times in the cell cycle British radiologist). One Gray is equal to
(Chapter 1). Cells are most sensitive in the 100 rad. The term centiGray (cGy) is
mitotic (M) stage of the cell cycle and most frequently used to equate the dose with the rad.
resistant in the latter part of the S phase. G1 For example, 6000 cGy is equal to 6000 rad.
and G2 show intermediate sensitivity, although Radiotherapy is usually given in a series of
G2 is more sensitive than G1. If G1 is long, the equal-sized fractions, either daily or more than
early phase is resistant and the latter phase once a day for several weeks.31 Each fraction

108
PRINCIPLES OF RADIATION THERAPY

kills a similar proportion of tumor cells, result- fractionation schedule. The ability to cure a
ing in a logarithmic decline in the number of tumor without excessive complications depends
surviving cells as the number of fractions on a complex interplay of these factors. Late
increases. Thus, smaller tumors are more complications of radiotherapy are influenced
susceptible than larger ones at any given dose. by the total dose, the size of the dose per
The biological basis for fractionation is that fraction, and the time interval between
dividing a dose into a number of fractions fractions when they are closely spaced. Acute
spares normal tissues, because sublethal reactions are most affected by the fraction size
damage of normal cells is repaired between and the overall time taken to deliver the treat-
dose fractions, whereas tumor cells are not ment. In acute-reacting tissues such as skin,
repaired. Cellular division also allows repopu- mucous membranes, bone marrow and,
lation of normal cells if the overall treatment hopefully, tumor cells, injury occurs early but
time is sufficiently long. At the same time, regeneration occurs quickly. In late-reacting
dividing a dose into a number of fractions tissue such as the CNS and probably many
increases damage to the tumor by allowing CNS tumors, injury may not occur for months
time for cells that were quiescent and thus or years, and recovery, if it occurs at all, occurs
resistant to radiation to re-enter the cell cycle late. However, prolonging the overall treatment
and become more sensitive. In addition, each time excessively allows surviving tumor cells to
fraction selectively kills cells in the more divide more quickly, and some tumors may
radiosensitive phases of the cell cycle. The actually proliferate during the treatment.
remaining cells progress into the more Most brain tumors are treated with doses
radiosensitive phases of the cycle, where they ranging from 4560 Gy in daily fractions of
can be killed by the next fraction (known as 1.82.0 Gy. Higher doses per fraction have
reassortment). Furthermore, tumors contain been associated with an increased incidence of
hypoxic cells. Oxygenated mammalian cells are late radiation injury to the CNS (see below).
2.53 times more sensitive to radiation than The dose used for each tumor type is discussed
hypoxic cells. Fractionation permits the shrink- in the chapters reviewing the management of
ing tumor to reoxygenate, thus making the specific tumors.
tumor cells in a previously hypoxic area more Changing the conventional fractionation
sensitive to radiotherapy. These phenomena parameters of time and dose can lead to differ-
also apply to normal cells, but they can repair ential effects on the tumor and normal tissues.
themselves. This has stimulated the design of altered
A course of external beam irradiation is fractionation schedules that deliver more than
described in terms of the total dose delivered, one dose fraction per day. These schemes seek
the amount of radiation given at each treat- to reduce the size of the dose per fraction or
ment session (known as fraction size), the reduce the overall time of treatment.
frequency of dose fractions, and the overall
time in days over which the course of radiation Hyperfractionated irradiation differs from
is given. For example, in the treatment of conventionally fractionated irradiation in that
malignant gliomas, a typical treatment course two or more treatments are given daily with
is 59.4 Gy delivered in 33 daily fractions of fraction sizes that are smaller than the
1.8 Gy each, five days per week, given over standard fractions of 1.82.0 Gy. The goal is
45 days. Together, these factors constitute a to deliver a higher total dose (1015%) in the

109
PRINCIPLES OF THERAPY

same overall time as a conventional treatment Accelerated fractionation attempts to improve


schedule (66.5 weeks) without increasing radiation-induced tumor cell killing in rapidly
toxicity, by exploiting differences in the capac- proliferating tumors by reducing the length of
ities of normal and tumor tissues to repair time needed to complete the treatment course.33
radiation damage. The ability of normal Conventional dose fractions (1.62.0 Gy) are
neural tissues to repair sublethal radiation given two or three times daily. This treatment
injury increases if the dose per fraction is schedule may further improve the therapeutic
small.32 Hence, the risk of late complications ratio by overcoming the effects of accelerated
can be reduced by using smaller individual tumor cell repopulation during treatment,
fractions. Because rapidly proliferating tumor thereby increasing the probability of tumor
cells are less efficient at repairing damage from control for a given dose level.31 However, trials
reduced dose fractions, and because tumor cell using different accelerated fractionation
death is influenced more by total dose than by regimens for brain tumors have not shown a
fraction size, there should be a therapeutic survival benefit over conventional irradia-
advantage to using a larger number of smaller tion.36,37
fractions. It is necessary to administer more
than one fraction per day to prevent tumor
Radiation therapy treatment
repopulation from negating the potential gain
in tumor control.33 The 68 h interval between planning and delivery approaches
doses allows sufficient time for normal tissues Conventional treatment planning
to repair sublethal radiation damage. Ionizing radiation can be delivered to a brain
However, the repair process may require more tumor in several ways (Table 4.6).
than 8 h in the CNS, making hyperfractiona- The most common technique, external beam
tion less safe than some dosage formulas radiotherapy, aims several radiation beams
suggest. Proliferating tumor cells progress into at the tumor to maximize the dose while
more radiosensitive phases of the cell cycle minimizing the dose to normal tissue outside
during the interval between fractions, increas- of the target. Recent advances in external
ing the opportunity for tumor cell killing. beam radiation therapy techniques have
Target cells for late sequelae proliferate slowly,
so for these tissues little cell cycle reassortment
or self-sensitization occurs during irradiation. Table 4.6
Studies using white matter changes on MR Radiation therapy of brain tumors.
imaging as an endpoint demonstrate that
morbidity is decreased in patients receiving Conventional Experimental
doses in excess of 70 Gy with hyperfraction-
ated schedules, compared to conventional Conformal fields
fractionation regimens. However, studies of Intensity-modulated Charged particles39
radiotherapy38 (Fig 4.6)
RT administered in multiple doses per fraction
Radiosurgery Boron neutron
to treat brain tumors have failed to demon- capture40
strate superior efficacy over single daily dose Stereotactic radiotherapy Radioprotectors
RT.34 In vitro studies indicate that hyperfrac- Interstitial radiation41
tionation may prove useful for tumors express- Radiosensitizers
ing wild-type p53.35

110
PRINCIPLES OF RADIATION THERAPY

Figure 4.6
Three-dimensional dose
display for a four-field
treatment plan designed
using intensity
modulation. The dose
cloud, which represents
the prescription isodose
level, conforms closely
to the shape of the
planning target volume
(PTV), while avoiding
nearby optical
structures, including the
chiasm (orange),
bilateral optic nerves
(pink), retinas (green)
and brainstem (blue).

improved the efficacy and lowered the toxic- the beam, the more energy it carries to greater
ity associated with the treatment of brain depths within the brain. Beams with energies
tumors. To plan and carry out a course of of up to 10 MV are typically used in the treat-
radiation therapy, the radiation oncologist ment of brain tumors. Cobalt-60 -rays, used
must determine the volume of the brain to less often to deliver brain irradiation, have an
treat based on neuroimaging studies, the average energy of 1.25 MV. The total radia-
operative findings and the patterns of tumor tion dose to be administered and dose given
spread. The target volume is made up of at each daily treatment are based on the
three components: (1) the tumor visible histology of the tumor.
on neuroimaging studies; (2) a margin of In the past, especially for gliomas, radiation
surrounding tissue considered to be at risk was directed to the entire brain (Fig. 4.5). This
for microscopic tumor spread; and (3) an was in part because of the recognition that
additional 0.51.0 cm margin to account for many brain tumors were diffusely infiltrative,
daily variations in positioning the patient on but was mainly because imaging techniques
the treatment machine and patient motion. which clearly defined the location of the tumor
The radiation beam energy and field arrange- were not available. Whole-brain irradiation at
ments are selected after consideration of the doses that affect most brain tumors causes
location of the target volume within the brain substantial toxicity. In addition, most primary
and its geometry. Beam energies produced by brain tumors are unifocal at the time of initial
linear accelerators range from 4 MV (million diagnosis and they relapse at their original
electron-volts) to 25 MV. The more energetic location after either whole-brain or limited-field

111
PRINCIPLES OF THERAPY

irradiation. Accordingly, as imaging techniques Three-dimensional conformal radiation


improved, radiation was delivered only to the therapy. The development of modern three-
part of the brain encompassing the tumor and dimensional conformal radiation therapy (3D-
its surrounding microscopic extensions. An CRT) treatment planning techniques permits
exception is the treatment of tumors such as the design of treatment plans which match the
medulloblastomas (Chapter 7), which often high dose of radiation to the three-dimensional
disseminate via CSF pathways. Such tumors are contour of the target volume while the dose to
treated with a shrinking field approach, in surrounding tissues is reduced to a minimum
which the entire craniospinal axis is treated and (Fig. 4.5). Conformal treatment planning not
a smaller field, encompassing the site of the only decreases the risk of normal tissue injury,
primary tumor, receives a higher dose. but also allows higher than traditional doses to
Treatment planning begins on a simulator. be safely administered to selected patients with
Simulators are X-ray machines designed to malignant gliomas42 and other intracranial
duplicate the geometry and configuration of a tumors.
treatment machine, but they produce standard
radiographs, rather than therapeutic X-rays. Intensity-modulated radiation therapy (IMRT)
Specialized CT simulations are now available recently has been introduced as an advanced
that permit three-dimensional reconstruction of form of 3D-CRT43 (Fig. 4.6) to produce an
tumors and normal tissues. During the simula- additional refinement of dose configuration.
tion, patient positioning (supine or prone) is Dose distributions generated in this fashion
determined and an individualized custom- exquisitely conform to the shape of the target
designed immobilization device is constructed. volume and sculpt around adjacent critical
Such devices allow daily reproduction of normal tissues. As with all focal RT techniques,
patient position on the treatment couch and great care and expertise are required to ensure
prevent patient movement during each radio- accuracy of field alignment. Errors in treatment
therapy session. planning or delivery may reduce the efficacy or
For conventional treatment planning, lateral increase the toxicity to the normal brain.44
or 90 orthogonal simulator radiographs are
obtained. The target volume is reconstructed Stereotactic radiosurgery is a technique for
directly on the simulation film from the CT delivering highly focal, external irradiation to
and/or MR images. For more complex field a clearly defined small target45 (Fig. 4.7). It
arrangements, an outline of the target volume requires the use of a stereotactic frame, identi-
is drawn on a contour of the head, taken cal to that used for neurosurgery, to precisely
through the central axis of the proposed localize the tumor within the intracranial
treatment portals. The contour is digitally cavity. Image-guided robotic radiosurgery may
transferred to a treatment planning computer, eliminate the need for fixing the head in a
radiation beams are defined, and radiation stereotactic frame.46 Multiple radiation beams
dose distributions (known as isodose distribu- intersect at one (or sometimes more) point(s),
tions) are generated. The goal of treatment known as isocenters, within the skull after
planning is to cover the target in its entirety entering through numerous points or arcs
while minimizing the dose to normal brain distributed over the head. The technique deliv-
tissue, especially the retina, optic nerve, and ers a high radiation dose to an intracranial
optic chiasm. target in a single session without delivering

112
PRINCIPLES OF RADIATION THERAPY

(Fig. 4.7) requires rotation of the gantry, the


couch, or both. In either approach, a single large
radiation fraction is delivered to the pathologic
tissue. For larger tumors, micro-Multileaf
Collimators provide superior dose distributions
for the linear accelerator compared to the
Gamma Knife, eliminating the need for multiple
overlapping isocenters, which leads to dose
inhomogeneity and an increased risk of morbid-
ity. The amount of radiation that most normal
tissue receives is minimal, except to the area
immediately surrounding the tumor. This is a
particular problem when the tumor abuts the
brainstem or optic nerve. Calculations of late
effects on normal tissue relative to fraction size
are available.49

Figure 4.7 Stereotactic radiotherapy. The terms stereotactic


The concept of radiosurgery. Multiple beams are aimed
radiotherapy and fractionated radiosurgery
at the tumor from several angles and the normal brain
receives only a small amount of radiation. Maximal refer to several fractions delivered stereotacti-
radiation can be delivered to the tumor. cally, allowing for a larger total dose to the
pathologic tissue with minimal exposure to
surrounding brain.50 Stereotactic radiotherapy
significant radiation to adjacent normal tissues. combines the focal advantages of radiosurgery
To maintain a steep dose gradient at the edges with the radiobiological advantage of fractiona-
of the field, the target volume must be small. tion. Fractionation improves the therapeutic
Furthermore, because the volume is limited, the ratio, allowing larger tumors and those located
application of radiation in effective single doses within or near critical intracranial structures to
is restricted to lesions 4 cm or less in diameter. be treated. Fractionated stereotactic radiotherapy
Stereotactic radiosurgery originally found its is accomplished using head frames that can be
greatest usefulness in treating small vascular relocalized daily in a non-invasive, reproducible
malformations in the brain, but it is now being fashion. This approach is being applied to the
widely used to treat metastatic tumors, menin- treatment of malignant gliomas, craniopharyn-
giomas, acoustic neuromas, pituitary adenomas giomas, pituitary adenomas, and small tumors of
and other relatively small tumors (see appro- the optic tracts, and as a boost for medullo-
priate chapters). blastomas.51 Recent reports have suggested that
Stereotactic radiosurgery by Gamma Knife47 fractionated radiosurgery is superior in some
uses 201 cobalt-60 sources positioned such that instances to single-dose radiosurgery.
the long axis of each source is oriented along the
radius of a sphere and aimed precisely at the Interstitial brachytherapy. The surgical implan-
central point of the unit. Some sources may be tation of radiation isotopes into a brain tumor
selectively blocked to alter the shape of the dose is called brachytherapy (from Greek for
distribution. Radiosurgery by linear accelerator48 short).52 This conformal technique allows

113
PRINCIPLES OF THERAPY

maximal radiation to the tumor, while Proton beams interact with the nuclei of
surrounding tissues receive considerably lower atoms rather than their orbital electrons. Like
doses. This approach has been applied to the conventional radiation, protons are sparsely
treatment of newly diagnosed and recurrent ionizing as they enter tissue. However, they
malignant gliomas in the USA and has been become more densely ionizing as they deceler-
used in Europe for low-grade gliomas arising ate in tissue, whereupon the dose deposited
in functionally critical areas of the brain.52 reaches a sharp maximum, after which it falls
Because of the requirement for high-activity to zero. The region of the sharp rise and fall
sources for malignant brain tumors, stereotac- in dose is called the Bragg peak, allowing large
tic techniques have been devised to place after- doses of radiation to be deposited in one area
loading catheters that are removed after the while dramatically sparing adjacent tissues.
prescribed dose has been delivered. Controlled Because of this feature, protons and heavy
trials using high-activity iodine-125 sources charged particles such as helium ions, which
plus external beam irradiation for high-grade behave in a similar fashion, have been used for
gliomas have failed to show efficacy over radiosurgery as well as to deliver fractionated
external beam irradiation alone.53 Other treatments. The depth of penetration can be
radioisotope sources such as iridium-192 have controlled by varying beam energy or by
also been used but less frequently. In some placing brass or plastic absorbing material in
instances, interstitial radiation has been the beam path. Charged particle beams can be
coupled with hyperthermia for the treatment made to stop in front of a critical structure,
of gliomas.54 Low-activity iodine-125 sources and in combination with other lateral or
have been implanted directly into the tumor oblique beams, can be wrapped around a criti-
bed as an intraoperative boost in patients with cal structure.56 This form of radiation is ideally
meningiomas and gliomas and as retreatment suited for the treatment of skull-base tumors
for patients with previously resected skull base such as meningiomas, chordomas and low-
tumors. grade chondrosarcomas that are located
adjacent to critical structures such as the optic
Experimental approaches in radiation nerves, chiasm, and brainstem. Pituitary adeno-
therapy mas and choroidal melanomas are also treated
As with surgery, recent developments in radia- with this modality. Note that the biological
tion therapy have made that treatment modal- properties of protons are similar to those of
ity both more effective for some tumors and X-rays and that it is only the physical dose
less toxic to the brain (Table 4.6). distribution that provides an advantage over
conventional photon radiotherapy.
Heavy particle irradiation. Protons, neutrons
and heavy charged particles have been used to Boron neutron capture therapy (BNCT). This
treat CNS tumors.55 For malignant gliomas, technique has been utilized off and on for over
neutrons successfully eradicated tumor tissue, 50 years. It has gained renewed prominence, in
but caused diffuse white matter degeneration, part because it is theoretically so satisfying and
probably because the relative biological effec- in part because new techniques for delivery of
tiveness of neutrons was underestimated. No boronated agents have been developed. The
known dose of neutrons eradicates tumor patient is injected with a boron-10-containing
without unacceptable injury to normal tissue. compound that, because of bloodbrain barrier

114
PRINCIPLES OF RADIATION THERAPY

disruption, finds its way to the tumor but not Halogenated pyrimidine analogs are radiosen-
to normal brain. The tumor is then irradiated sitizers that become incorporated into the DNA
with low-energy epithermal neutrons. The of dividing cells because of their similarity to
boronated compound captures the neutron, the DNA precursor, thymidine;60 they enhance
releasing an -particle, which travels only a the number of DNA strand breaks by dissoci-
short distance, approximately the diameter of ating hydrogen atoms from adjacent deoxy-
a cell. In theory, if the boron is within the cell, ribose moieties.
the -particle should destroy no more than the Radiosensitization appears to be directly
cell itself. The procedure is being used to treat related to an increased production of
gliomas and other brain tumors but is still unrepaired double-strand breaks in drug-
experimental.57,58 This approach cannot reach exposed cells. Sensitizer enhancement ratios of
microscopic disease that resides behind the 1.53 have been observed, and the degree of
bloodbrain barrier. The problem of getting enhancement is dependent on the percentage of
sufficient quantity of the boronated compound thymidine replaced. Two analogs, bromod-
into the tumor59 may be helped by the use of eoxyuridine (BrdU) and iododeoxyuridine
bloodbrain barrier opening, but this will also (IdU), have been tested in patients with malig-
expose normal brain. nant gliomas and in brain metastases. These
studies have not demonstrated an improvement
Chemical modifiers of the radiation response in outcome.37 Further, tumor biopsies obtained
Radiosensitizers. Oxygen assists in the DNA in some patients showed relatively low levels of
damage induced by radiation. Hypoxia drug incorporation.
protects cells from the effects of low-LET
radiation (X-rays). The radiation dose must be Motexafin gadolinium may enhance radiation
increased by a factor of three to obtain in therapy of brain metastases.61Trials in primary
hypoxic cells the same effect achieved in cells brain tumors are underway.
that are fully oxygenated. Even when only
23% of such resistant cells are present, a two- The Herpes thymidine kinase gene and a
fold increment in radiation dose may be dominant-negative epidermal growth factor
required to completely eradicate a tumor. The receptor may also be radiosensitizers.62,63
radioresistance of glioblastoma multiforme
may, in part, be due to the presence of radio- Radioprotectors. Because the limiting factor in
biologically hypoxic but viable tumor cells radiating brain tumors is toxicity to normal
within necrotic areas. brain, a number of attempts have been made
to find substances that protect normal brain
Nitroimidazoles are electron-affinic compounds against the effects of radiation but do not affect
that mimic oxygen in damaging DNA by free tumor tissue. Agents containing sulfhydryl
radicals. They are ineffective in the presence of groups will help return a free radical to its
oxygen. Unfortunately, no survival improve- normal state. Omega fatty acids and
ment was observed in malignant glioma bioflavonoids may be protective.64 Some
patients receiving the most extensively tested thiophosphate compounds such as amifostine
compound, misonidazole, along with radiation (WR2721), when transformed by enzymes
in any of the dose-fractionation or drug sched- inside the living cell, do give some protection
ules tested. but have not yet been shown to be clinically

115
PRINCIPLES OF THERAPY

effective. In cell culture experiments, neurons from radiation depends not only on total radia-
are protected against damaging effects of radia- tion dose, dose per fraction and, to a lesser
tion by the additional presence of astrocytes as extent, overall treatment time, but also on
well as by free-radical scavengers.65 However, additional factors, most of which depend on
clinical efficacy has not been demonstrated for the host (Table 4.7).
any putative radioprotectors.66 The volume of tissue irradiated is important.
Whole-brain radiation is considerably more
likely to cause toxicity than partial brain
Radiation toxicity
radiation or conformal field radiation. Host
The goal of RT is to cause as much damage to factors also play a role. Both the developing
the tumor with as little damage to the nervous (under age 3) and the elderly (over age 60)
system as possible (Fig. 4.8). Several isoeffect brain are more susceptible to radiation than
formulas exist to calculate the sensitivity of the brain of a young adult. For example, in
normal brain to different radiation timedose- some studies, prophylactic whole-brain RT for
fractionation schemes.67 These formulas leukemia affects the IQ of children under 5,
consider total radiation dose, dose per fraction but not those older. Thus, special attempts
and, to a lesser extent, total treatment time; should be made to avoid RT in infants and
they indicate that normal brain tissue can children, using chemotherapy to delay radia-
tolerate about 60 Gy of radiation delivered in tion as long as possible. Genetic factors may
1.82.0 Gy fractions. However, CNS damage also play a role. Certain familial disorders,

Figure 4.8
Three-dimensionally reconstructed anterior view of a patient showing the placement of seven lateral and
superior arcs used for stereotactic radiosurgery of a small midline lesion (left). The brainstem (yellow) and eyes
(green) have been delineated for geometrical perspective and to allow the determination of dose to these critical
structures. A coronal CT image (right) shows the dose distribution through the center of the target volume for
the radiosurgery plan. The prescription level is the 80% isodose line (shown in yellow).

116
PRINCIPLES OF RADIATION THERAPY

Table 4.7
Radiation factors Host and other factors Risk factors for CNS
radiation damage.
Dose per fraction Age
Total dose Sex
Total duration of therapy Genetic predisposition
Volume of tissue irradiated Pre-existing nervous system disease
Energy of radiation Infection
Systemic disease (e.g. hypertension,
diabetes, hypoxia)
Chemotherapy
Environment (e.g. smoking)

particularly those associated with defects in oligodendroglial, and Schwann cells have been
DNA repair, cause marked photosensitivity proposed as the most vulnerable. Also, because
leading to skin tumors. These and other most neurons are post-mitotic, they are less
genetic defects may also cause increased sensi- susceptible to RT damage, although either
tivity to RT. Patients with pre-existing nervous apoptosis or DNA damage may lead to neuronal
system disease are often more sensitive to dysfunction or death. Radiation damage directed
radiation. Patients mistakenly given RT for at white matter causes a leukoencephalopathy.
demyelinating lesions appear to be inordi- Typical is the diffuse white matter destruction
nately sensitive.68 Hypertension and cerebral seen after whole-brain radiation. Some
atherosclerosis may also be risk factors. chemotherapeutic agents appear to exacerbate
Hypoxia promotes radiation-induced blood this type of damage in patients who have received
brain barrier disruption.69 RT and chemother- doses of radiation not usually sufficient to cause
apy may be synergistic or at least additive in such damage.
causing CNS toxicity. This appears to be The second mechanism proposed is that RT
particularly important in patients treated with damages vascular endothelium.70 Hopewell and
chemotherapy and RT for primary CNS Wright71 postulate that after RT has killed
lymphoma (Chapter 11). Timing may also be many capillary endothelial cells, a few localized
important; some studies suggest that clones survive at irregular locations along the
chemotherapy given before RT is safer than vessel. These clones multiply, leading to focal
that given with or after RT, perhaps because luminal constrictions and eventually to vascu-
RT disrupts the bloodbrain barrier, allowing lar occlusion. The result is ischemia and necro-
neurotoxic amounts of chemotherapy to enter. sis of neural tissue. In this process, luminal
The mechanism(s) by which the brain is occlusion is due to endothelial proliferation;
damaged is controversial. Three hypotheses, not intraluminal clotting plays no role. This
necessarily mutually exclusive, have been hypothesis suffers from the fact that, although
proposed. The first is that ionizing radiation vascular occlusions occur in areas of radiation
directly damages nervous system cells. Because necrosis, the degree of tissue damage is usually
RT-induced nervous system damage predomi- greater than can be explained by the degree of
nantly affects white matter, glial cells, particularly vascular insufficiency.

117
PRINCIPLES OF THERAPY

Table 4.8
Cerebral radiation injury.

Designation Time after RT Clinical findings Pathogenesis Outcome

Acute Immediate Headache, vomiting, Increased Recovery (usually) with


(minutes to neurologic signs intracranial steroids
hours) pressure
Early-delayed 416 weeks Somnolence, Demyelination, Recovery, responds to
increased focal possibly cerebral steroids
signs, worsening edema
MR scan
Late-delayed Months to years Focal signs Brain necrosis Responds to steroids
Necrosis Months to years MR enhancement, Possibly vascular Surgical removal and
memory loss steroids
Atrophy, Months to years Dementia, gait Cellular loss, Modest response to
hydrocephalus ataxia, incontinence, demyelination, shunting in some cases
MR atrophy, possibly
hydrocephalus, spongiosis of
leukoencephalopathy white matter
Hemorrhage Years Focal signs Telangiectasia Partial recovery

Infarction Years Focal signs Cerebral/carotid Variable


atherosclerosis
Encephalopathy Years Confusion, Hypothyroidism Recovery with hormone
disorientation replacement
Neoplasm Years Focal signs RT-induced Usually poor
neoplasm

The third mechanism, proposed by Lampert Whatever the mechanism, when radiation
et al,72 suggests that damaged glial cells release injury follows focal radiation for treatment of
antigens identified as foreign by the immune a brain tumor, the necrosis is focal and the signs
system. An immune response leads to both the and symptoms often mimic those of the origi-
necrosis and vascular changes of a hypersensi- nal brain tumor. The MR scan may also appear
tivity reaction. Almost no direct evidence exists similar, making it difficult to distinguish recur-
for this third hypothesis, although in one rent tumor from radiation necrosis. The differ-
patient a focal vasculitis in an irradiated area ential diagnosis was discussed in Chapter 3.
of brain was associated with circulating Radiation injury can occur at almost any
immune complexes. Thus, the exact cause of time, from seconds to years after the therapy is
radiation damage to the peripheral and central delivered. Side-effects can be divided into those
nervous systems is still unknown, although, at that are acute, usually observed within the first
least in experimental animals, lower total RT few days, early-delayed, seen within 4 weeks to
doses appear to cause neurotoxicity via the 4 months following RT, or late-delayed, many
endothelial cells, and higher doses via glial months to many years after RT is completed.
cells. Late-delayed RT-induced brain dysfunction can

118
PRINCIPLES OF RADIATION THERAPY

take several forms (Table 4.8) and is the most larger than 2.0 Gy. Second, these patients
important form of RT injury, because it is should be protected with corticosteroids such
irreversible and often progressive. as dexamethasone, 816 mg/24 h, or more, if
increased intracranial pressure is sympto-
matic. The drugs should be administered for
Acute encephalopathy at least 4872 h prior to initiating RT.
This disorder usually follows large fractions Steroids are not required if the RT portal and
(more than 300 cGy) delivered to a large the tumor are small.
volume of brain in patients with increased
intracranial pressure from a brain tumor.
Early-delayed encephalopathy
Because such large doses are rarely used, acute
RT toxicity is uncommon; however, absence This disorder usually begins 4 weeks to 4
of corticosteroid treatment increases the risk. months after RT. Early-delayed symptoms do
Acute effects, usually mild, also follow stereo- not predict or predispose to later complications.
tactic radiotherapy and radiosurgery. The symptoms of early-delayed encephalopathy
Immediately or within a few hours following simulate tumor progression. The patient devel-
treatment, susceptible patients develop ops headache, lethargy, and worsening or
headache, nausea, vomiting, somnolence, fever, reappearance of the original neurologic
and worsening of pre-existing neurologic symptoms. Alternatively, patients may develop
symptoms. In rare instances, the disorder memory deficits.74 The disorder usually peaks 2
culminates in cerebral herniation and death. months after RT and resolves within 6 months.
Acute encephalopathy usually follows the first The symptoms suggest tumor progression but
radiation dose and becomes progressively less improve spontaneously over the ensuing 4
severe with each ensuing fraction. A mild form months. MR or CT scans reveal an increase in
of the disorder is common and consists of the contrast-enhancing area and surrounding
headache and nausea immediately following edema. In some instances, new areas of contrast
radiation. The pathogenesis of acute enhancement appear. Both clinical and radio-
encephalopathy is probably disruption of the logic changes resolve spontaneously over the
bloodbrain barrier by ionizing radiation, ensuing months, hastened by corticosteroids.
causing increased cerebral edema and a rise in Radiosurgery may also cause an early-delayed
intracranial pressure. Evidence indicates that a encephalopathy, usually characterized by MR
single dose of 300 Gy to the brain in an exper- scans showing an increased enhancing area,
imental animal causes substantial disruption of suggesting an increase in tumor volume and
the bloodbrain barrier if measured 2 h after more hyperintensity around the tumor, suggest-
the radiation. The P-glycoprotein level, a ing brain edema. These changes usually appear
marker of bloodbrain barrier function, 312 months after treatment and resolve in 57
decreased to 60% of control within 5 days of months. They may be accompanied by loss of
one dose of 25 Gy to an experimental animal.73 enhancement in the center of the tumor,
These observations carry two clinical suggesting necrosis.
messages. First, patients harboring large The clinical and scan findings of early-
brain tumors, particularly tumors causing delayed radiation encephalopathy can follow
signs of increased intracranial pressure, conventional external beam RT or radio-
should probably be treated with fractions no surgery. The clinical implication is that failure

119
PRINCIPLES OF THERAPY

of the patient or the scan to improve, or even Radiation necrosis


a worsening within 2 or 3 months of RT, does Radiation necrosis usually begins 12 years
not mean that the therapy has failed. The after RT is completed but can start as early as
usual sequence is that the scan appears 3 months after treatment or be delayed for
somewhat improved immediately at the end of several years.75,76 The symptoms generally
RT and then worsens in the several months recapitulate those of the brain tumor, leading
following radiation, only to improve once the physician to suspect tumor recurrence.
again. The patient should be supported with Rarely, new focal signs indicate a lesion distant
corticosteroids, with close follow-up examina- from the original lesion. The MRI may show
tions and scanning. Because an MRI cannot increased contrast enhancement at the original
distinguish either early or late changes due to tumor site or, less frequently, at a remote site,77
radiation from those due to residual or recur- but within the radiation portal. Neither the
rent tumor, a number of non-invasive tests clinical symptoms nor CT or MR scans distin-
have been used in an attempt to make that guish tumor recurrence from necrosis. PET
distinction (Chapter 3). These have included scan measuring glucose consumption shows
PET and SPECT scanning as well as MRS. hypometabolism in areas of necrosis and
However, it is not clear that these tests do hypermetabolism in areas containing tumor,
make this distinction in early-delayed radia- but is not sufficiently sensitive to unequivocally
tion encephalopathy. establish a diagnosis.78 SPECT and MRS may
The pathogenesis of early-delayed be somewhat better than glucose PET, although
encephalopathy is believed to be demyelina- they are not infallible. Our experience has been
tion resulting from transient damage to that many patients, particularly those treated
oligodendroglia with the subsequent break- for glioma, have a mixture of radiation necro-
down of myelin sheaths. The best evidence sis and tumor.
supporting this hypothesis comes from Histologically, the typical radiation necrosis
pathologic studies in patients with early- lesion consists of coagulative necrosis in the
delayed brainstem encephalopathy, in whom white matter with relative sparing of the
confluent areas of demyelination with overlying cortex and deep gray matter.
varying degrees of axonal loss were found in Microscopically, striking abnormalities are
areas receiving the radiation. Associated with found in blood vessels, with hyalinized thick-
this is the loss of oligodendrocytes and the ening and fibrinoid necrosis often associated
occurrence of abnormal, often multinucle- with vascular thrombosis, hemorrhage, and
ated, giant astrocytes. The timing of the accumulated perivascular fibrinoid material. In
appearance and resolution of symptoms less severely affected areas, demyelination is
tends to parallel the known turnover of noted with a loss of oligodendrocytes, variable
myelin. Other pathologic changes include axonal loss, axonal swellings, dystrophic calci-
perivenous inflammatory infiltrates. Necrosis fications, fibrillary gliosis, and scattered
and vascular changes are absent. perivascular infiltrates and mononuclear cells.
Telangiectatic vessels also form there and may
be responsible for late hemorrhages.
Late-delayed encephalopathy
Radiation necrosis is usually treated by
Late-delayed radiation damage to the brain can surgical resection. Most patients respond
take several forms (Fig. 4.9) (see Table 4.8). transiently to steroids but relapse when steroids

120
PRINCIPLES OF RADIATION THERAPY

Atrophy, hydrocephalus
A more frequent consequence of brain radiation
than radiation necrosis is brain atrophy. If
substantial volumes of brain tissue are irradi-
ated, most patients develop MRI-documented
evidence of enlarged cerebral sulci and ventricu-
lomegaly. In addition, MR scans may reveal
hyperintensity, most marked in cerebral white
matter around the ventricles on the T2-
weighted or fast fluid-attenuated inversion
recovers (FLAIR) images; hypodensity is noted
on the CT scan. These changes may worsen
over time. Both atrophy and white matter
abnormalities are usually clear-cut by 1 year
following RT, but occasionally they may
become obvious within 2 or 3 months after RT
is completed. They can persist or progress there-
after. The symptoms are those of a subcortical
dementia79 with memory loss, apathy and
Figure 4.9 slowed cognitive responses. Some patients also
A new lesion appearing 14 years post-radiation have gait abnormalities suggesting gait apraxia,
therapy. This man developed symptoms of a right and urinary urgency followed by incontinence.
frontal brain tumor, which was treated surgically and
with radiation therapy. The radiation included 40 Gy
The triad of gait difficulties, incontinence and
of whole-brain radiation with a 20 Gy boost to the dementia suggest the syndrome of normal
right frontal area. Sixteen years later, the patient is pressure hydrocephalus.80 If the scan also
asymptomatic, working full-time as a physician. suggests hydrocephalus, i.e. if ventricular dilata-
Fourteen years after the radiation therapy, a small
tion is greater than cortical atrophy, the patient
contrast-enhancing asymptomatic lesion appeared in
the posterior frontal area on the left (arrow). This may respond at least temporarily to ventricular
lesion, probably representing a radiation-induced shunting. The gait disturbance and incontinence
vascular abnormality, is asymptomatic and has not respond best to shunting, whereas the memory
progressed over the ensuing 2 years. impairment usually does not respond. Cerebral
atrophy is untreatable.
Virtually all patients in whom a substantial
portion of the brain is irradiated to treat
brain tumors complain of memory loss
resembling that experienced by most older
are discontinued, although reports have people (e.g., forgetting names, telephone
described prolonged responses after steroid numbers, appointments, and recent events
therapy without surgery. Other suggested treat- but remembering remote events well). The
ments presupposing a vascular mechanism have memory loss may even prevent the individual
included aspirin and anticoagulation, but we from returning to gainful employment.
have not found them to be effective. Recent analysis of long-term survivors of RT

121
PRINCIPLES OF THERAPY

for glioma indicate that 60% were employed growth often follow RT, especially when deliv-
at jobs comparable to those they held before ered to the entire neuraxis. Growth hormone
receiving RT.81 In a minority of patients, failure is a particular problem in children with
memory loss progresses and affects other brain tumors.84
cognitive functions, leading to a more severe
dementia.
In our experience, many of our patients
complain of two additional symptoms. The first
is a feeling of being cold. Characteristically, the
Principles of chemotherapy
patients require more blankets than the spouse, Introduction
often a reversal of the pre-illness situation. The
patients also require more clothing when Chemotherapeutic agents have been disap-
outdoors than previously. Endocrinological pointing in the treatment of most brain
workup does not reveal abnormalities in thyroid tumors.85,86 Exceptions include cerebral germi-
function, and the symptoms do not respond to nomas, medulloblastomas, primary lymphomas
treatment with thyroid hormones. Another of the nervous system and some oligoden-
frequent symptom is loss of libido or impotence. drogliomas. The role of chemotherapy in the
Patients rarely complain of this symptom but management of astrocytomas is controversial,
spouses are often quite distressed. Some male although most believe it has a small role to
patients are not impotent but simply indifferent play in improving survival in some patients. No
to sexual activity. It is useful to probe patients adequate chemotherapeutic agents have been
and their spouses for the above symptoms, developed for meningiomas, acoustic neuromas
because simply knowing they are common often and most pituitary adenomas.
relieves anxiety. In addition, sildenafil can be Certain principles of chemotherapy are
useful for some patients with impotence. believed to apply to all cancers. These are listed
in Table 4.9 and discussed in the paragraphs
below.
Many of these concepts have been developed
Vasculopathy by treating tumors in experimental animals and
Radiation therapy decreases microvascular then testing the concepts in humans with systemic
density in tumor and in surrounding normal cancers, such as carcinoma of the breast. How
brain.82 Telangiectasias sometimes develop that
can result in cerebral hemorrhage.70 Occlusive
vasculopathy with subsequent Moya-Moya
vessel formation may follow RT to optic gliomas Table 4.9
Basic concepts in cancer chemotherapy.
in patients with neurofibromatosis type 1.80

Chemotherapeutic agents kill only a


percentage of cancer cells
Endocrine dysfunction Some drugs kill only dividing cells
Multiple drugs with differing sites of action
Hypothyroidism, either primary from cervical are better than a single drug
irradiation or secondary from cranial irradia- High doses are better than low doses
tion, pituitary failure and retarded bone

122
PRINCIPLES OF CHEMOTHERAPY

they apply to intracranial tumors is uncertain. vincristine and 1-(2-chloroethyl)-3-cyclohexyl-1-


The underlying concept is that most tumors are nitrosourea (CCNU)] may be superior to BCNU
monoclonal, i.e. they arise from a single cell. for anaplastic astrocytomas but not glioblastoma,
However, because of genetic instability, as the but this occurred in a subgroup analysis of an
tumor grows it becomes progressively more essentially negative study. A recent review of
heterogeneous, some of the cells having developed RTOG protocols did not show a survival benefit
intrinsic resistance to some chemotherapeutic of PCV over BCNU for patients with anaplastic
agents. Furthermore, although small tumors tend astrocytoma.89 A randomized trial from England
to grow exponentially with virtually all of the failed to demonstrate an advantage in survival by
cells dividing, as the tumor gets larger, a number using PCV with RT over RT alone,90 although
of cells enter a quiescent phase, making them less both randomized trials and a meta-analysis from
susceptible to many chemotherapeutic agents. In the US demonstrate that BCNU increases survival
addition, rapidly growing tumors may outstrip in high-grade glioma patients (Chapter 5).91 Even
their blood supply, leaving cells in relatively in chemosensitive tumors such as oligoden-
ischemic areas less exposed to chemotherapeutic droglioma and primary CNS lymphoma, there is
agents and also putting the ischemic cells into a no evidence to demonstrate the superiority of
more quiescent phase. When a cancer is exposed multiagent over single agent chemotherapy
to sublethal doses of a chemotherapeutic agent, (BCNU in the former, methotrexate in the
resistance to further doses of that drug develops. latter).87 Germinomas (Chapter 8) may be an
These cells, along with the intrinsically resistant exception, where multiagent therapy does appear
cells, lead to rapid regrowth and restoration of to be efficacious.
the tumor, now more resistant to treatment than Table 4.10 classifies antineoplastic drugs and
it was initially. Thus, for most cancer therapy, gives some examples of drugs that have been used
oncologists use the largest possible doses of multi- to treat brain tumors. Individual drugs are consid-
ple chemotherapeutic agents. (This is the rationale ered in the paragraphs below. Recent reviews
for high-dose chemotherapy with bone marrow describe the neurotoxicity of these and a number
or stem cell rescue, so-called bone marrow trans- of other drugs not included in this section.92
plant protocols.) Each agent should have a
different mechanism of action, including cell Problems unique to intracranial
cycle-specific drugs (drugs which only kill divid- tumors
ing cells) and cell cycle-non-specific (drugs that
directly damage DNA and are toxic to resting Table 4.11 lists some of the problems specific
cells as well as dividing cells). Unfortunately, for to the chemotherapy of intracranial tumors.
most brain tumors, there is no compelling Many oncologists overrate the bloodbrain
evidence that multiagent chemotherapy is barrier as a problem for chemotherapy of brain
superior to single agent chemotherapy or that tumors. Many intracranial tumors, including
dose intensity improves outcome.87 Controlled meningiomas, schwannomas, most pineal
trials by the Brain Tumor Study Group (BTSG) region tumors and anterior pituitary tumors, do
failed to demonstrate superiority of multi-agent not possess a bloodbrain barrier. In other
chemotherapy over 1,3-bischloro-(2-chloroethyl)- tumors such as metastases and high-grade
7-nitrosourea (BCNU) alone.88 In the treatment gliomas, disruption of the bloodbrain barrier
of malignant glioma, one study suggests that is often sufficient for water-soluble chemother-
multiagent chemotherapy, PCV [procarbazine, apeutic agents to reach tumors in substantial

123
PRINCIPLES OF THERAPY

Table 4.10
Class Examples Classification of
antineoplastic
Alkylating agents Platinum-based, e.g. cisplatin, carboplatin drugs.
Nitrosoureas, e.g. carmustine (BCNU), lomustine (CCNU)
Temozolomide
Antibiotics Doxorubicin
Bleomycin
Anthracyclines e.g. doxorubricin
Antimetabolites Cytarabine
5-Fluorouracil
Methotrexate
Plant alkaloids Vincristine
Epipodophyllotoxins, e.g. etoposide, teniposide
Taxanes, e.g. paclitaxel
Miscellaneous Suramin
Hydroxyurea
Procarbazine

Table 4.11 usually presents with one or multiple contrast-


Chemotherapy of intracranial tumors: problems enhancing lesions, but there is also widespread
specific to location.
infiltration of tumor that does not contrast
enhance. Standard doses of water-soluble agents
Bloodbrain barrier may eliminate the area(s) of contrast enhance-
Paucity of lymphatics ment, only to have the tumor rapidly appear
Heterogeneity of gliomas
Intrinsic resistance
elsewhere in the brain, where it had been
Low therapeutic/toxic ratio sequestered behind the bloodbrain barrier.
A number of attempts have been made to
circumvent the bloodbrain barrier in the treat-
ment of brain tumors. These have included
opening the bloodbrain barrier using a hyper-
concentrations. However, for certain tumors, osmolar agent such as intra-arterial mannitol or
the bloodbrain barrier does prevent entry of Cereport, a bradykinin analog.93 The latter is
water-soluble chemotherapeutic agents. said to open the bloodbrain barrier more in
Examples include low-grade gliomas in which tumors than in normal brain. Opening of the
the bloodbrain barrier is intact, and higher- bloodbrain barrier has been used in the treat-
grade gliomas that disrupt the bloodbrain ment of primary CNS lymphomas (Chapter 11);
barrier in the bulk of the tumor, but not the there is as yet no evidence that barrier opening
edge where tumor infiltrates normal tissue. The is superior to chemotherapy without barrier
bloodbrain barrier is relatively or completely opening. Furthermore, opening the bloodbrain
intact at the infiltrating margins of the tumor. barrier, at least with hyperosmolar agents,
Perhaps the most striking example is primary results in a greater proportionate increase of
lymphoma of the nervous system. The patient drug entry into normal brain than into brain

124
PRINCIPLES OF CHEMOTHERAPY

tumor. Osmotic bloodbrain barrier opening is within an acceptable range of toxicity is more
influenced by the choice of anesthesia; propo- effective than standard doses.
fol/N2O with hypercarbia is optimal.94 Other problems unique to intracranial
Other approaches have attempted to deliver tumors include the fact that a deficient
more chemotherapy to the tumor and less to lymphatic system prevents the easy removal of
normal tissue. These include intra-arterial detritus caused by effective chemotherapy from
infusions,95 intratumoral injections using the brain. Necrotic tissue is not cleared easily
catheters, implanting drug-impregnated from the brain and may form a nidus for
wafers or microspheres, and drugs altered to
96 97 further edema and worsening of neurologic
cross the bloodbrain barrier.98,99 None has yet symptoms. This is one reason why radiation
proved more effective or less toxic than necrosis causes symptoms.
conventional routes of administration, and Even the time-honored concept that a small
wafer implantation has been reported to result tumor responds better than a big tumor does not
in postoperative complications, including infec- appear to apply to many primary brain tumors,
tion and poor wound healing.100 perhaps because of their intrinsic resistance. In
The best drugs for the treatment of infiltrat- the treatment of gliomas, current evidence
ing gliomas have proved to be the nitrosoureas suggests that there is little or no advantage to
(and perhaps temozolomide), which are lipid- treating patients in the adjuvant setting with
soluble and thus not affected by the chemotherapy over waiting for a recurrence and
bloodbrain barrier. Nevertheless, despite their then treating. This probably reflects the intrinsic
ability to reach the entire tumor, they have not resistance of these tumors to the chemothera-
proved dramatically effective. On the other peutic agents that are currently available.
hand, methotrexate, which does not easily Chemotherapy directed at intracranial tumors
cross the bloodbrain barrier, has been shown, can be given by a number of routes of adminis-
when given in very high doses, to be effective tration. The most common is systemic adminis-
for primary lymphomas of the nervous system, tration, either oral or intravenous. The
even though it is not a major agent for the advantage of systemic administration is that the
treatment of comparable systemic lymphomas. agents are easy to administer. The disadvantages
Major problems with chemotherapy for most include the fact that some drugs are unable to
brain tumors include: (1) intrinsic resistance to cross the bloodbrain barrier easily and that
most chemotherapeutic agents,101,102 probably much of the agent reaches and damages tissues
because of heterogeneity of the higher grade other than the tumor. The second route of
tumors; (2) rapid development of resistance to administration is local. Most intrinsic brain
chemotherapeutic agents; and (3) susceptibility tumors, unlike systemic tumors, do not metas-
only to doses of chemotherapeutic agents that are tasize, but the vast majority recur at the origi-
unacceptably toxic both to the normal brain and nal site.42 Accordingly, gliomas should be ideal
the tissues outside the CNS. Thus, extremely for focal therapy. Focal therapy has the advan-
high-dose BCNU with bone marrow rescue given tage not only of preventing systemic toxicity, but
for the treatment of malignant gliomas is success- also of circumventing the bloodbrain barrier
ful in eradicating the tumor, but causes severe and getting higher doses of a chemotherapeutic
encephalopathy that eventually kills the patient. agent to the tumor than one could get by
Furthermore, there is little evidence that increas- systemic administration. Techniques of local
ing the dose of standard chemotherapeutic agents therapy include intra-arterial injection,103 usually

125
PRINCIPLES OF THERAPY

in the carotid artery or one of its branches, may have an advantage in that the vascular
intrathecal injection, into the subarachnoid supply to the tumor is better than it is after
space by lumbar puncture or via an intraven- radiation. In addition, by giving chemotherapy
tricular cannula, or intratumoral injection. Intra- first, one can determine if the agents are effec-
arterial injection has been compared in a tive and, if so, continue using them after RT.
controlled trial with intravenous injection of If they are ineffective, they can be abandoned.
carmustine and not found to be superior but to There is also some evidence, at least for
increase neurotoxicity.104 Intrathecal injection methotrexate, that chemotherapy given prior
may treat leptomeningeal tumor but cannot to radiation is less neurotoxic than when it
reach intrinsic tumors of the brain in sufficient follows radiation. Alternatively, chemotherapy
concentrations to be effective. Several studies are following radiation may encounter a
now investigating the efficacy of intratumoral bloodbrain barrier that has been partially
injection either by the implantation of wafers disrupted by the radiation. This may allow
that slowly release chemotherapeutic agents or more water-soluble drug to enter both the
by single or repeated injections through an tumor and, unfortunately, the normal brain.
implanted cannula. Wafers impregnated with Given all of these considerations, it is not yet
BCNU implanted in the tumor bed appear to clear what the optimal sequence of radiation
give longer survival than placebo implantation and chemotherapy should be. We generally
for recurrent gliomas, but the increased survival prefer to use RT, the more effective modality,
was only 2 months, so that the advantage, if first and follow with chemotherapy.
any, is modest.96 The advantage of local therapy
is that it saves normal tissues from the side- Specific chemotherapeutic agents
effects of the chemotherapeutic agent. The
disadvantage is that it often requires a surgical Table 4.12 lists some chemotherapeutic agents
procedure. Combinations of systemic and local used for the treatment of brain tumors. Some
therapy include opening the bloodbrain barrier of these are considered in the paragraphs below.
by intra-arterial injection of various substances,
followed by either intra-arterial or systemic Alkylating agents
chemotherapy.
The timing of chemotherapy with respect to The alkylating agents, including the nitro-
radiation is important but controversial. On soureas, platinum compounds and procar-
the one hand, evidence suggests that many bazine, are among the most widely used drugs
chemotherapeutic agents act as radiation sensi- for the treatment of brain tumors. Although
tizers. Theoretically, if such agents are given cell cycle non-specific, they appear to be more
along with radiation therapy, the effect will be active against rapidly dividing cells, probably
better than if they are given independently. because of the lesser time these cells have to
However, there is no evidence demonstrating repair DNA damage. By alkylating DNA bases,
enhanced efficacy against brain tumors when they cross-link DNA, resulting in single and
chemotherapy and RT are given together than double-strand breaks. The most widely used of
when they are used sequentially. Furthermore, the alkylating agents for the treatment of brain
there is some evidence that chemotherapeutic tumors are the nitrosoureas. Several nitro-
agents such as cisplatin may induce radiation soureas treat gliomas, including BCNU
resistance.105 Chemotherapy given prior to RT (carmustine), CCNU (lomustine), PCNU,

126
PRINCIPLES OF CHEMOTHERAPY

MeCCNU and ACNU [3-((4-amino-2-methyl- being used experimentally to try to increase


5-pyrimidinyl-methyl)-12(2-chloroethyl)-1- tumor sensitivity to nitrosoureas and other
nitrosourea] and HeCCNU The drugs have chemotherapeutic agents.106
been used in single agent and multiagent The major toxicities of nitrosoureas are bone
chemotherapeutic regimens, either adjuvantly marrow suppression and pulmonary fibrosis.
after surgery and radiation, at relapse, or The drugs are relatively non-neurotoxic.
during radiation as radiation sensitizers. The ACNU has been safely given intrathecally.
nitrosoureas alkylate O-6-guanine in DNA. However, high doses of nitrosoureas can
Substantial evidence indicates that patients cause encephalopathy. Retinal toxicity and
whose tumors intrinsically express the repair encephalopathy were side-effects of an experi-
enzyme guanine-6-methyltransferase are more mental program in which the drug was given
resistant to the chemotherapeutic effects of intra-arterially to increase the dose delivered to
nitrosoureas.101 Inhibitors of the enzyme are the tumor.

Procarbazine rapidly crosses the bloodbrain


Table 4.12 barrier. Its mechanism of action is not known,
Specific chemotherapeutic agents used for intracranial although it is believed to interfere with DNA
tumors. synthesis. The drug has been widely used as part
of conventional treatment of malignant gliomas.
Cytosine arabinoside (Ara-C) Encephalopathy and peripheral neuropathy have
Arazidinylbenzoquinone (AZQ) been reported, but rash and bone marrow suppres-
1,3-bis(2-Chloroethyl)-1-nitrosourea (BCNU)
sion are the main side-effects. Because it is a weak
1-(2-Chloroethyl)-3-cyclohexyl-1-nitrosourea
(CCNU) monoamine oxidase inhibitor, patients are advised
Cisplatin to avoid foods containing tyramine. Although the
Carboplatin importance of this precaution is doubtful, mono-
Dianhydrogalacticol amine oxidase inhibitors should be discontinued
Dibromodulcitol 2 weeks before starting procarbazine.
Dacarbazine (DTIC)
Temozolomide
5-Fluorouracil The platinum compounds, cisplatin and particu-
Hydroxyurea larly carboplatin, have been used to treat a
Methyl-1-(2-chloroethyl)-3-cyclohexyl-1- number of brain tumors. Cisplatin is neurotoxic
nitrosourea (MeCCNU) (Table 4.13), but carboplatin is not. These drugs
Misonidazole
are efficacious against medulloblastomas and
Methotrexate
1-(2-Chloroethyl)-3-(2,6-dioxo-3-piper-idyl)-1- germinomas and although widely used in the
nitrosourea (PCNU) treatment of gliomas, their role is not established.
Procarbazine A preliminary report suggests that carboplatin in
Vincristine combination with an opening of the bloodbrain
Teniposide barrier might be effective in glioma.107
Etoposide (VP16)
Paclitaxel (Taxol)
Bromodeoxyuridine (BrdU) Cyclophosphamide is a prodrug that must be
Eflornithine metabolized in the liver to 4-hydroxyperoxycyclo-
Irinotecan (CPT-11) phosphamide to be active. The active agent has
safely been given intrathecally for the treatment

127
PRINCIPLES OF THERAPY

Table 4.13 treatment of CNS tumors. DTIC has been used to


Neurotoxicity of cisplatin. treat malignant meningiomas. Temozolomide is
approved for treatment of malignant gliomas.
Common Early enthusiastic reports suggesting a 50%
Peripheral neuropathy (large fiber, sensory) response rate have not been borne out by more
Lhermittes sign current data. However, temozolomide appears to
Hearing loss (high frequency) be active in about 20% of patients with malignant
Tinnitus
Uncommon
glioma, about the same as the nitrosoureas. The
Encephalopathy fact that it is administered orally and is relatively
Visual loss nontoxic makes it appealing.108 These drugs are
Retinal toxicity marrow suppressive and occasionally cause
Optic neuropathy encephalopathy but are basically non-neurotoxic.
Cortical blindness
Hepatic veno-occlusive disease and necrosis have
Seizures
Cerebral herniation (hydration related) been reported.
Electrolyte imbalance (Ca++, Mg++, Na+,
SIADH) Estramustine is an alkylating agent that crosses
Vestibular toxicity the bloodbrain barrier poorly but has been
Autonomic neuropathy reported to have some effect on malignant
gliomas. It is not neurotoxic.
SIADH = Syndrome of Inappropriate Antidiuretic Hormone
Secretion
Diazaquine (AZQ) crosses the bloodbrain
barrier. The drug has been used to treat malig-
nant gliomas and has been given intrathecally
to treat leptomeningeal tumor. Its main toxici-
of leptomeningeal tumor. Cyclophosphamide has ties are myelosuppression, nausea, anorexia,
been used as part of multiagent chemotherapy for and skin discoloration.
a number of intracranial tumors. It is relatively Busulfan is a bifunctional alkylating agent that
non-neurotoxic except at high doses, when it can easily crosses the bloodbrain barrier. It has
cause lethargy, seizures or both. Ifosfamide, a been used to treat some malignant brain tumors,
related compound that also requires hydroxylation but does not appear to be effective. With high-
by liver, more commonly causes encephalopathy. dose therapy used to prepare children for bone
Thiotepa crosses the bloodbrain barrier easily and marrow transplantation, seizures occur. At
has been used both intravenously and intrathecally standard doses the drug is not neurotoxic.
for the treatment of brain tumors. High-dose
thiotepa is used in some bone marrow rescue Plant alkaloids
protocols, particularly for the treatment of anaplas-
tic oligodendroglioma. The drug is not neurotoxic, The plant alkaloids include: the vinca
although myelopathy has been reported in a few alkaloids, vincristine, vindesine, and vinblas-
patients after intrathecal injection. tine; podophyllins, including etoposide and
teniposide; and the taxanes, paclitaxel and
Dacarbazine (DTIC) and a related compound docetaxel. The drugs all bind to tubulin, the
temozolomide are both cell cycle-non-specific microtubular protein, but the antineoplastic
alkylating agents that have been used for the mechanism of each is different.

128
PRINCIPLES OF CHEMOTHERAPY

The vinca alkaloids, because of their size, without toxicity but can cause mild peripheral
cross the bloodbrain barrier poorly. They neuropathy when given systemically. Teni-
cause metaphase arrest by binding to tubulin poside causes peripheral neuropathy less
during S-phase and thus are cell cycle commonly.
specific. Vincristine is part of a common
regimen for the treatment of malignant The taxanes bind to tubulin, but instead of
gliomas (PCV). The major side effect is a inhibiting polymerization, as do the vinca
peripheral neuropathy, but myopathies, alkaloids, they stabilize and promote micro-
autonomic neuropathies, cranial neuropathies tubular assembly. How this interaction causes
and central toxicity, including seizures and cytotoxicity is not certain. The drugs have been
inappropriate secretion of antidiuretic used for the treatment of a number of brain
hormone with hyponatremia, have been tumors, particularly malignant gliomas, but
reported (Table 4.14). their role is not yet established.110 The taxanes
can cause peripheral neuropathy and proximal
The podophyllins are lipid-soluble compounds weakness which is probably neuropathic as
but do not cross the bloodbrain barrier well, well.111
because of their size. They bind to tubulin and
inhibit microtubular assembly, and thus are cell
Antimetabolites
cycle specific, causing arrest in G2. Etoposide is
widely used for the treatment of gliomas and The antimetabolites are all cell cycle specific
forms part of multiagent therapy for other and are subdivided into the antifolates,
brain tumors.109 It has been given intrathecally including methotrexate, the cytidine analogs,

Table 4.14
Spectrum of vincristine neurotoxicity.

Toxic effect Subacute Intermediate Chronic


(1 day to 2 weeks) (1 to 4 weeks) (> 4 weeks)

Peripheral neuropathy Depressed Achilles Other tendon reflexes Sensory loss, extensor
reflex, paresthesias depressed, paresthesias weakness, especially
foot-drop
Myopathy Muscle pain, tenderness
(especially quadriceps);
jaw pain
Autonomic neuropathy Ileus with abdominal Constipation, urinary
cramping, pain hesitancy, impotence,
orthostatic hypotension
Cranial neuropathy Optic atrophy; ptosis;
(uncommon) VIth, VIIth, and VIIIth
cranial nerve
dysfunction;
hoarseness; dysphagia
Central toxicity Seizures, SIADH

129
PRINCIPLES OF THERAPY

including cytarabine (Ara-C), the fluorinated reversibly to dihydrofolate reductase, thus


pyrimidines, including 5-fluorouracil, the blocking synthesis of tetrahydrofolate, reducing
purine analogs, including 6-thioguanine, the folate pool and preventing thymidine and
6-mercaptopurine and cladribine, and a variety purine biosynthesis. The primary side-effects of
of other agents, including hydroxyurea. the drugs are bone marrow suppression and
gastrointestinal toxicity, but neurotoxicity is a
Hydroxyurea inhibits ribonucleotide diphos- significant problem (Table 4.15). The drug
phate reductase, an enzyme required in DNA appears to be synergistic with brain irradiation
synthesis. The drug crosses the bloodbrain in producing leukoencephalopathy. High doses
barrier readily and has been used as a radiosen- of the drug occasionally cause a transient
sitizer for the treatment of malignant gliomas, encephalopathy, and intrathecal injection can
without substantial effect. It has also been used cause a myelopathy. Gene amplification of
as a single agent in the treatment of recurrent dihydrofolate reductase can result in over-
meningiomas, also without significant effect. production of the enzyme, leading to tumor
The drug causes mild nausea, anorexia, skin resistance to methotrexate.
discoloration and myelosuppression but is not
neurotoxic. Cytarabine (Ara-C) is an S-phase cell cycle-
specific drug that inhibits DNA polymerase-,
Methotrexate is a drug widely used in cancer is incorporated into DNA and terminates DNA
chemotherapy but has limited use in the treat- chain elongation. The drug does not cross the
ment of brain tumors, with the exception of bloodbrain barrier unless given in very high
primary CNS lymphoma. The drug is water- doses. However, because the enzyme that
soluble and does not cross the bloodbrain metabolizes the drug is present at very low
barrier well, but intravenous injection of very levels in the CNS, the drug persists for an
high doses (with folinic acid rescue) forces extended period. Cytarabine is not widely used
enough drug across the bloodbrain and blood- for the treatment of brain tumors, with the
CSF barriers to produce therapeutic levels exception of primary CNS lymphoma (Chapter
within the CNS (Chapter 11). The drug binds 11). At very high doses, the drug can cause

Table 4.15
Methotrexate neurotoxicity.

Route of Dose Toxic effect


administration

Oral or intravenous Conventional Leukoencephalopathy (if prior brain irradiation)


Intravenous High-dose Acute transient encephalopathy, chronic
leukoencephalopathy
Intra-arterial Conventional or high-dose Hemorrhagic cerebral infarction, seizures
Intrathecal Conventional Acute aseptic meningitis, paraplegia, seizures,
chronic leukoencephalopathy, cerebral atrophy,
calcification

130
PRINCIPLES OF MISCELLANEOUS THERAPIES

cerebellar degeneration, a peripheral neuropa- have some efficacy in the treatment of intracra-
thy or an encephalopathy. A depo-preparation nial tumors,113 especially metastases.114
has been used to treat leptomeningeal tumors Camptothecins cause marrow suppression but
(Chapter 11). are not neurotoxic.

5-Fluorouracil is a pyrimidine analog that Eflornithine (DFMO) inhibits ornithine decar-


interferes with DNA synthesis. It is not effica- boxylase and depletes cells of putrescine and
cious in the treatment of brain tumors, even spermidine. It has been used in brain tumors as
though it crosses the bloodbrain barrier well. a radiation enhancer. The drug does not readily
The drug is generally non-neurotoxic, although cross the bloodbrain barrier. It can cause
high doses can cause encephalopathy or ototoxicity, but other kinds of neurotoxicity
cerebellar degeneration. The major side-effects are not reported; its primary toxic effect is on
are stomatitis and diarrhea. bone marrow.

6-Mercaptopurine and 6-thioguanine are Tamoxifen is a synthetic anti-estrogen that


purine analogs that inhibit purine biosynthesis. crosses the bloodbrain barrier and appears to
They have been used to treat gliomas but are concentrate in the brain. It has been reported to
not widely used anymore. They cause myelo- have some effect against malignant gliomas115
suppression but are not neurotoxic. They do and has been used without much effect for the
not cross the bloodbrain barrier easily. treatment of meningiomas. Its mechanism of
action on malignant gliomas, if any, is probably
not as an anti-estrogen but as an inhibitor of
Antineoplastic antibiotics
protein kinase C (PKC); it requires high doses
The antineoplastic antibiotics consist of the to achieve this effect. The drug at high doses
anthracyclines such as doxorubicin, and other causes dizziness and ataxia,115 and reversible
agents such as mitomycin and bleomycin. Most effects on the retina have been described: the
are not used for the treatment of brain tumors, drug may promote thrombophlebitis.
although a recent experimental protocol calls
for direct injection of bleomycin into the tumor
bed. The drugs are generally not neurotoxic,
although bleomycin in combination with
Principles of miscellaneous
cisplatin has been reported to cause strokes and therapies
myocardial infarction. Doxorubicin is not
Experimental therapies
neurotoxic when given by conventional routes,
but accidental injection into the CSF is lethal. Table 4.16 lists a number of approaches to
therapy now being tried experimentally either
in animals or early clinical trials.
Miscellaneous agents
Camptothecins are topoisomerase inhibitors Angiogenesis inhibitors
used experimentally for the treatment of Many brain tumors are highly vascular, and
malignant gliomas. Topotecan crosses the angiogenesis is necessary for their growth.
bloodbrain barrier112 and encouraging prelim- Accordingly, drugs that interfere with angio-
inary reports suggest that camptothecins may genesis might retard the growth of tumors. In

131
PRINCIPLES OF THERAPY

Table 4.16 a bystander effect.123 Although clever, this


Experimental therapies.
approach has not yet proved efficacious in the
treatment of malignant gliomas. The poor
Angiogenesis inhibitors116,117 outcome may be due to technical limitations
Gene therapy118 that further research may resolve.
Herpes simplex thymidine kinase, p53 gene An even more attractive therapeutic approach
Immunotherapy119
Vaccines
would be to try to normalize mutated genes in
T-cells tumor cells. Therapeutic attempts using the
Tumor cells wild-type p53 gene, which should cause tumor
Antibodies cells that lack this gene to stop reproducing, are
Dendritic cells underway. Other forms of gene therapy are still
Growth factor inhibitors
in early experimental stages.
Toxins120,121
Differentiating agents Antisense oligonucleotides that probably
inhibit the function of the normal messenger
RNA are being used in experimental systems.
For example, antisense fibroblast growth factor
(FGF) and antisense IGF receptors inhibit
experimental animals, anti-angiogenesis factors growth of glioblastoma cells.
not only retard the growth of tumor, but have
been shown eventually to cure tumors. Immunotherapy
Furthermore, because blood vessels are not Immunotherapy has proved a disappointment.
neoplastic, they do not possess the genetic insta- A number of human trials involving the injec-
bility of tumor cells and do not develop resis- tion of T-cells with interleukin-2 (IL-2) into the
tance to the anti-angiogenesis factors. A number tumor bed are ongoing, but none have yet
of drugs are now in clinical trials, including proved efficacious. Vaccination of patients with
thalidomide efficacy122 and analogs of fumag- their own radiation-killed tumor cells, or with
illin. These drugs are being used either alone or tumor cells and T-cells incubated with IL-2, has
in conjunction with chemotherapeutic agents been undergoing therapeutic trials, but, again,
(i.e. thalidomide plus carboplatin). The future efficacy has not been shown.
roles, if any, of these drugs are unknown.
Growth factor inhibitors
Gene therapy Growth factor inhibitors are also undergoing
Gene therapy is another attractive experimen- clinical trials. Most brain tumors are dependent
tal approach to the treatment of brain tumors. on growth factors such as epidermal growth
Most studies have focused on the insertion of factor (EGF), platelet-derived growth factor
the herpes simplex thymidine kinase gene into (PDGF) and vascular endothelial growth factor
tumor cells; the patient is then treated with (VEGF), as well as FGF, for both angiogenesis
ganciclovir, a drug that causes a lethal reaction and tumor growth. Monoclonal antibodies
with the herpes simplex but not the human against EGF receptor have been used to treat
thymidine kinase gene, killing the tumor cell. brain tumors but effectiveness has not yet been
Some evidence suggests that not only are tumor established. A number of drugs that inhibit
cells possessing the gene destroyed, but growth factors are currently undergoing clini-
surrounding tumor cells are also destroyed by cal trials, but again their efficacy is not known.

132
PRINCIPLES OF MISCELLANEOUS THERAPIES

Table 4.17
Supportive therapy Brain tumors: anticonvulsants.
Anticonvulsants
Table 14.17 details the principles of using Prophylaxis
anticonvulsants. Does not prevent first seizure
Seizures comprise one of the common Probably useful in perioperative period
Treatment
symptoms in patients with intracranial tumors.
Efficacy unclear
They usually occur as the presenting symptom Side-effects more common
but may in some instances be late complica- Interactions common
tions following surgery or radiation therapy. Hard to control levels
The late onset of seizures does not necessarily Best drug unknown
presage recurrence of a tumor. It may simply
result from damage to surrounding normal
brain caused by the tumor or as a result of
treatment. All seizures due to intracranial due to brain tumors, either those that occur
tumor have a focal onset. If the focus of epilep- before or those that occur after treatment.
tic discharge is in a silent area, the seizure may Furthermore, untoward side-effects of anticon-
secondarily generalize without the patient vulsants are more common in patients with
being aware of any initial symptoms. Focal brain tumors than in the general population.
seizures generally resolve spontaneously and do Especially dangerous are skin reactions includ-
not cause permanent damage. Focal seizures ing the StevensJohnson syndrome, which can
may vary in frequency from rare (a few a be fatal124 (Fig. 4.10). Most (but not all)
month to a few a year) to several times a day. anticonvulsants have interactions with drugs
Although occasional focal seizures do not cause used to treat patients with brain tumors. The
significant damage or interfere with activities interactions may affect the metabolism of
of daily living, they are distressing and chemotherapeutic agents. Interactions may also
attempts should be made to control them. affect the metabolism of anticonvulsants, thus
Generalized seizures, although usually not making therapeutic levels hard to maintain.
dangerous, can be lethal if the patient injures Although some of the newer anticonvulsants
himself during the course of a seizure or if (e.g. gabapentin) do not have known interac-
repetitive seizures lead to status epilepticus. tions, presently unknown interactions may
Thus, as a general principle, attempts should eventually surface.
be made to control seizures. We recommend anticonvulsants for all
Unfortunately, as Table 4.17 indicates, patients who have either focal or generalized
attempts to control seizures are not without seizures. Table 4.18 lists the anticonvulsants
problems. Both focal and generalized seizures generally used to treat patients with brain
may be extremely difficult to control in tumors. One anticonvulsant does not appear to
patients with a brain tumor. In patients with be superior to the others.125
low-grade glioma, there is some evidence that The first-line anticonvulsants include
treating the tumor with RT decreases the phenytoin, carbamazepine, valproate and
incidence of intractable seizures. However, phenobarbital. All of these have potentially
there is no good evidence that anticonvulsants deleterious effects, including disorders of
themselves are effective in controlling seizures cognitive function and interactions with

133
PRINCIPLES OF THERAPY

Figure 4.10
StevensJohnson syndrome in a man with a brain
tumor. Although this patient did not have seizures,
he was begun on corticosteroids and phenytoin prior
to surgery. The surgery was without incident, and
radiation therapy was started about 3 weeks after the
operation. A week after radiation was started, he
broke out with a total body rash, including
involvement of the oral mucosa and the conjunctiva.
He was hospitalized for about a week before the rash
began to clear. This is a typical StevensJohnson
syndrome that occurs in patients with a combination
of recent onset of anticonvulsant drugs (usually
phenytoin or carbamazepine), radiation therapy to
the head and tapering of corticosteroids.

Table 4.18
Some antiepileptic drugs for brain tumors.

Drug Usual daily dose Dose intervals

Standard agents
Phenytoin 200500 mg Once daily (qd) or twice daily (bid)
Carbamazepine 6001600 mg Three times a day (tid) or four times a day (qid) (bid
for extended release tablets)
Valproate 1560 mg/kg tid or qid
Phenobarbital 14 mg/kg qid

Newer agents
Gabapentin 9006000 mg tid or qid
Lamotrigine 200800 mg bid
Topiramate 2001600 mg bid
Tiagabine 3256 mg bid qid

chemotherapeutic agents. The newer anti- dysfunction; gabapentin and lamotrigine


convulsant drugs appear to have fewer side- rarely do so.126 However, it is not clear how
effects and many have fewer interactions. In effective these newer agents are in the treat-
healthy adults, topiramate can cause cognitive ment of patients with brain tumors, or

134
PRINCIPLES OF MISCELLANEOUS THERAPIES

whether more side-effects will emerge with Corticosteroids dramatically relieve the
longer experience. Accordingly, we recommend symptoms of brain tumors, reducing edema
the following approach. An active seizure and thus decreasing intracranial pressure.
should be stopped, first with intravenous Symptomatic improvement may begin within
lorazepam (0.52 mg over 12 min), followed minutes and patients often become asympto-
by 1000 mg (15 mg/kg) of phenytoin or prefer- matic within 2448 h. The mechanisms by
ably fosphenytoin by intravenous injection (the which corticosteroids exert their effects are
latter drug does not cause cardiovascular poorly understood. One mechanism may be
problems and may be given more rapidly than a decrease in the flux of water-soluble
phenytoin itself). For patients not actively compounds across the disrupted bloodbrain
having seizures, we begin either with phenytoin barrier (Chapter 2), thus effectively reconsti-
or carbamazepine, increasing the dose until tuting the bloodbrain barrier. Unintended
either seizures are stopped or side-effects effects include reduction of chemotherapeutic
prevent further increases. Measurement of drug entry into the tumor. Furthermore, restor-
blood levels will indicate to the physician the ing the bloodbrain barrier reduces contrast
likelihood that a further dose increase may be enhancement on MRI, which can sometimes be
toxic or safe, but it is wise to treat the seizures mistaken for a response to treatment.
and not the blood levels. Some patients are well Corticosteroids are indicated in all
controlled on doses that are considered symptomatic patients with intracranial tumors,
subtherapeutic. Conversely, many patients with the exception of suspected but undiag-
tolerate supratherapeutic blood levels without nosed lymphoma. In a patient with lymphoma,
evidence of toxicity. If one drug is ineffective, corticosteroids may cause tumor necrosis due
one can switch to another or add a second to their lympholytic effect, precluding definitive
drug. We generally add gabapentin to either diagnosis (Chapter 11). Corticosteroids are not
phenytoin or carbamazepine in an attempt to given to asymptomatic patients, except those
control seizures. In some patients with non- undergoing surgery or RT.
disabling focal seizures, control with anticon-
vulsants may not be possible without toxicity
and it may be wise to allow the patient to have
occasional focal seizures.
There is no evidence that anticonvulsant drugs Table 4.19
prevent seizures in patients with brain tumors Advantages and disadvantages of corticosteroids.
who have not had prior seizures. Controlled trials
indicate that with either metastatic or primary Advantages
brain tumors, prophylactic anticonvulsants are Controls neurologic symptoms by
not helpful.127 One exception may be in the reducing edema
immediate perioperative period, when the use of Decreases acute RT toxicity
Relieves emesis from chemotherapy
prophylactic anticonvulsants for several weeks
Oncolytic (lymphoma)
may be indicated. Disadvantages
Side-effects common (see Table 4.20)
Corticosteroids Decreases chemotherapy entry
Table 4.19 lists some of the advantages and Oncolytic (lymphoma)
disadvantages of using corticosteroids.

135
PRINCIPLES OF THERAPY

The corticosteroid usually administered is good neurologic function. It can often be


dexamethasone at 16 mg a day. The drug is completely discontinued after surgery or radia-
long-acting and need not be given more than tion. There is no upper limit on the use of
twice daily, but is often administered more corticosteroids to control brain edema. If
frequently to reduce acute gastrointestinal 16 mg a day does not relieve symptoms, it is
toxicity and insomnia. We do not routinely use often wise to double the dose for a few days
gastric protection for patients receiving corti- to see if symptoms improve. Some investigators
costeroids. In patients expected to be on corti- have indicated that doses of 100 mg a day for
costeroids for a prolonged period, e.g. more several days may be required to fully relieve
than 6 weeks, prophylactic antibiotics to symptoms of a brain tumor. If one doubles or
prevent Pneumocystis carinii infection are quadruples the dose of corticosteroids for 48 h
indicated. Unfortunately, these drugs often and does not see an effect, one can rapidly
cause side-effects, particularly platelet suppres- taper the drug back to baseline. Recent
sion and rash. Once begun, the administration evidence suggests that doses of prednisone less
of a corticosteroid is continued until the than 5 mg daily can be abruptly withdrawn
patients symptoms are relieved and intracra- without causing adrenal suppression.128
nial pressure is diminished. The drug is then Despite the necessity for their use, the side-
tapered to the lowest dose commensurate with effects of corticosteroids are many (Table 4.20),

Table 4.20
Side effects of corticosteroids.

Common but Non-neurologic Neurologic Neurologic


usually mild but serious (common) (uncommon)

Insomnia Gastrointestinal bleeding Myopathy133 Psychosis


Sensation of Bowel perforation Behavioral alterations Paraparesis
abdominal Osteoporosis Hallucinations (high dose)
bloating Avascular osteonecrosis Hiccoughs
Increased appetite (usually hip) Tremor
Visual blurring Glaucoma Visual blurring
Urinary frequency Opportunistic infections
and nocturia Hyperglycemia
Acne Kaposis sarcoma
Edema Pancreatitis
Lipomatosis
Genital burning
(IV push)
Oral candidiasis
Memory loss132

IV, intravenous
For further discussion of supportive care of brain tumor patients, see Posner.67

136
PRINCIPLES OF MISCELLANEOUS THERAPIES

and not all are dose-related. Because of the side- may increase the risk of symptomatic intracranial
effects of steroids, investigators have made hemorrhage.139
efforts to find substitutes with fewer Treatment of an established thrombosis in
side-effects. These have included 21-amino neurosurgical patients is even more contro-
steroids,129 corticotropin-releasing factor,130 and versial than prophylaxis. Therapeutic options
boswellic acid.131 All of these treatments remain include the placement of a vena cava filter or
experimental. the use of an anticoagulant such as heparin or
low molecular weight heparin. Many neuro-
Anticoagulants surgeons remain reluctant to prescribe antico-
Deep vein thromboses commonly complicate agulation during the immediate postoperative
brain tumors and their therapy. They may be period. There is disagreement about when
present when the patient is first evaluated or anticoagulation can safely be started after a
occur after treatment is underway.134 There are craniotomy140 once a clinically apparent
multiple factors which contribute to deep vein thrombosis develops. The brain appears to
thrombosis, including immobility, neurosurgery, become less susceptible to hemorrhage after
the release of thromboplastins from the brain about 48 h. Some neurosurgeons begin
and hypercoagulability related to systemic cancer heparin treatment at 24 h, while others wait
and/or chemotherapy. Most episodes occur in for 57 days. When making the decision to
close proximity to the patients initial neurosur- begin anticoagulants, the physician must
gical procedure, often beginning during anesthe- weigh the risk that the patient will suffer a
sia. Pneumatic compression boots decrease the thromboembolic episode against the risk of an
incidence of deep vein thrombosis, especially if intracranial hemorrhage. In patients with
applied preoperatively and continued in the thromboembolic disease, anticoagulation,
postoperative period until the patient is fully avoiding supratherapeutic levels, is probably
ambulatory;135 low molecular weight heparin safe within 2 or 3 days of surgery. Because the
combined with the boots is even better.136 placement of vena cava filters has complica-
However, deep vein thromboses occur even in tions of its own, we prefer anticoagulants to
patients who ambulate the day after craniotomy vena cava filters whenever possible;141 many of
and are protected by pneumatic boots during and our patients who receive filters subsequently
after surgery. A recent study addressed the safety require anticoagulation for recurrent throm-
of perioperative subcutaneous heparin for bosis and chronic venous insufficiency of the
prophylaxis of venous thromboembolism during legs. We usually start low molecular weight
craniotomy. Treatment was begun at the induc- heparin with concurrent coumadin and treat
tion of anesthesia and continued for 7 days. The patients for the standard 36 months that
anticoagulants did not increase intraoperative is recommended for thromboembolism.
blood loss, transfusion requirements or postop- Occasionally, we have found that daily aspirin
erative platelet counts. No patient with a brain can help reduce repeated thromboembolic
tumor bled into the tumor bed after surgery. The episodes in patients with chronic venous stasis.
authors concluded that perioperative heparin at
a dose of 5000 units every 12 h is safe.137,138 Low Alternative therapy
molecular weight heparin given prophylactically One study suggests that about one-quarter of
24 h after surgery also appears to be safe,136 but brain tumor patients try alternative therapy.
one study indicates that, given preoperatively, it Although no major side-effects have been noted,

137
PRINCIPLES OF THERAPY

one cannot be sure that the agents did not inter- References
fere with conventional therapy. For example,
some antioxidants, a favorite alternative 1. Gutin PH, Posner JB. Neuro-oncology:
Diagnosis and management of cerebral
therapy, inhibit the cytotoxicity of cisplatin on gliomas past, present, and future.
glioma cells.142 As in breast cancer patients on Neurosurgery 2000; 47: 18.
alternative therapies, the brain tumor patient 2. Black PM. Surgery for cerebral gliomas: past,
using alternative therapies may have a lower present, and future. Clin Neurosurg 2000; 47:
quality of life.143 2145.
3. Kondziolka D, Lunsford LD. The role of
stereotactic biopsy in the management of
Nutrition gliomas. J Neurooncol 1999; 42: 20513.
Nutrition is often poor in patients with brain 4. Mohan DS, Suh JH, Phan JL et al. Outcome
tumors. Both the tumor and its treatment may in elderly patients undergoing definitive
surgery and radiation therapy for supratento-
cause nausea or anorexia. Dysphagia and rial glioblastoma multiforme at a tertiary care
obtundation may also decrease food intake. institution. Int J Radiat Oncol Biol Phys
The physician must be scrupulous in ensuring 1998; 42: 9817.
adequate nutrition. Patients with brain tumors 5. Simpson JR, Horton J, Scott C et al. Influence
should not suffer the additional and clinically of location and extent of surgical resection on
survival of patients with glioblastoma multi-
confusing insult of Wernickes encephalo-
forme: results of three consecutive Radiation
pathy.144 Therapy Oncology Group (RTOG) clinical
trials. Int J Radiat Oncol Biol Phys 1993; 26:
Quality of life 23944.
6. Toms SA, Ferson DZ, Sawaya R. Basic surgi-
The goal of all of the above treatments is not cal techniques in the resection of malignant
only to promote survival either by curing the gliomas. J Neurooncol 1999; 42: 21526.
tumor or by retarding its growth, but also to 7. Fadul C, Wood J, Thaler H et al. Morbidity
ensure that the patient sustains the best quality and mortality of craniotomy for excision of
of life possible. A number of recent studies supratentorial gliomas. Neurology 1988; 38:
13749.
have addressed quality of life in patients being
8. Sawaya R, Hammoud M, Schoppa D et al.
treated for high-grade brain tumors.145147 Neurosurgical outcomes in a modern series of
Patients with brain tumors can suffer other 400 craniotomies for treatment of parenchy-
symptoms such as depression or anxiety, mal tumors. Neurosurgery 1998; 42:
which should be vigorously treated and do 104455.
respond to therapy. Physical therapy can help 9. Gumprecht HK, Widenka DC, Lumenta CB.
BrainLab VectorVision neuronavigation
patients to maximize their functional capacity. system: technology and clinical experiences in
Occasionally, stimulants such as methyl- 131 cases. Neurosurgery 1999; 44: 97104.
phenidate can improve a patients level of 10. Pujol J, Conesa G, Deus J et al. Clinical appli-
alertness and decrease lethargy.148 Although cation of functional magnetic resonance
cognitive dysfunction and mood alterations imaging in presurgical identification of the
central sulcus. J Neurosurg 1998; 88: 8639.
are common, the quality of life does not
11. Roux FE, Boulanouar K, Ranjeva JP et al.
appear worse than in patients with other Cortical intraoperative stimulation in brain
chronic neurologic diseases. Most factors that tumors as a tool to evaluate spatial data from
affect the patients quality of life are related to motor functional MRI. Invest Radiol 1999;
the tumor rather than to the treatment. 34: 2259.

138
REFERENCES

12. Schwartz TH, Devinsky O, Doyle W, Perrine infection: a report of four cases. Neuro-
K. Preoperative predictors of anterior tempo- surgery 1999; 44: 63642.
ral language areas. J Neurosurg 1998; 89: 24. Hunter CA, Yu D, Gee M et al. Cutting edge:
96270. systemic inhibition of angiogenesis underlies
13. Taylor MD, Bernstein M. Awake craniotomy resistance to tumors during acute toxoplas-
with brain mapping as the routine surgical mosis. J Immunol 2001; 166: 587881.
approach to treating patients with supraten- 25. Hall WA, Martin AJ, Liu HY et al. Brain
torial intraaxial tumors: a prospective trial of biopsy using high-field strength interventional
200 cases. J Neurosurg 1999; 90: 3541. magnetic resonance imaging. Neurosurgery
14. North JB, Penhall RK, Hanieh A, Frewin DB, 1999; 44: 80713.
Taylor WB. Phenytoin and postoperative 26. King WA, Wackym PA. Endoscope-assisted
epilepsy. A double-blind study. J Neurosurg surgery for acoustic neuromas (vestibular
1983; 58: 6727. schwannomas): early experience using the
15. Shaw MD, Foy PM. Epilepsy after rigid Hopkins telescope. Neurosurgery 1999;
craniotomy and the place of prophylactic 44: 1095100.
anticonvulsant drugs: discussion paper. J R 27. King WA, Ullman AS, Frazee LG, Post KD,
Soc Med 1991; 84: 2213. Bergsneider M. Endoscopic resection of
16. Jansen GFA, Van Praagh BH, Kedaria MB, colloid cysts: surgical considerations using the
Odoom JA. Jugular bulb oxygen saturation rigid endoscope. Neurosurgery 1999; 44:
during propofol and isoflurane/nitrous oxide 11039.
anesthesia in patients undergoing brain tumor 28. Walker MD, Alexander EJ, Hunt WE et al.
surgery. Anesth Analg 1999; 89: 35863. Evaluation of BCNU and/or radiotherapy in
17. Dorward NL, Alberti O, Palmer JD, Kitchen the treatment of anaplastic gliomas. A
ND, Thomas DGT. Accuracy of true frame- cooperative clinical trial. J Neurosurg 1978;
less stereotaxy: in vivo measurement and 49: 33343.
laboratory phantom studies technical note. 29. Walker MD, Strike TA, Sheline GE. An
J Neurosurg 1999; 90: 1608. analysis of doseeffect relationship in the
18. Nimsky C, Ganslandt O, Kober H, radiotherapy of malignant gliomas. Int J
Buchfelder M, Fahlbusch R. Intraoperative Radiat Oncol Biol Phys 1979; 5: 172531.
magnetic resonance imaging combined with 30. Hall EJ. Radiobiology for the radiologist, 3rd
neuronavigation: A new concept. Neuro- edn. Philadelphia: J.B. Lippincott Co., 1988.
surgery 2001; 48: 10829. 31. Withers HR. Radiation biology and treatment
19. Matz PG, Cobbs C, Berger MS. options in radiation oncology. Cancer Res
Intraoperative cortical mapping as a guide 1999; 59: 1676s84s.
to the surgical resection of gliomas. 32. Olive PL. DNA organization affects cellular
J Neurooncol 1999; 42: 23345. radiosensitivity and detection of initial DNA
20. Danks RA, Aglio LS, Gugino LD, Black PM. strand breaks. Int J Radiat Oncol Biol Phys
Craniotomy under local anesthesia and 1992; 62: 38996.
monitored conscious sedation for the resec- 33. Ang KK. Fractionation effects in clinical
tion of tumors involving eloquent cortex. practice. In: Leibel SA, Phillips TL, eds.
J Neurooncol 2000; 49: 1319. Textbook of radiation oncology. Philadelphia:
21. Jennings CR, ODonoghue GM. Posterior W.B. Saunders Co., 1998: 2641.
fossa endoscopy. J Laryngol Otol 1998; 112: 34. Mandell LR, Kadota R, Freeman C et al.
2279. There is no role for hyperfractionated radio-
22. Savargaonkar P, Farmer PM. Utility of intra- therapy in the management of children with
operative consultations for the diagnosis of newly diagnosed diffuse intrinsic brainstem
central nervous system lesions. Ann Clin Lab tumors: results of a Pediatric Oncology
Sci 2001; 31: 1339. Group phase III trial comparing conventional
23. Bowles AP Jr, Perkins E. Long-term remission versus hyperfractionated radiotherapy. Int J
of malignant brain tumors after intracranial Radiat Oncol Biol Phys 1999; 43: 95964.

139
PRINCIPLES OF THERAPY

35. Haas-Kogan DA, Kogan SS, Yount G et al. apy of brain tumors. Int J Radiat Oncol Biol
p53 function influences the effect of fraction- Phys 1999; 43: 9216.
ated radiotherapy on glioblastoma tumors. 45. Shafman TD, Loeffler JS. Novel radiation
Int J Radiat Oncol Biol Phys 1999; 43: technologies for malignant gliomas. Curr
399403. Opin Oncol 1999; 11: 14751.
36. Fallai C, Olmi P. Hyperfractionated and 46. Adler JR Jr, Murphy MJ, Chang SD,
accelerated radiation therapy in central Hancock SL. Image-guided robotic radio-
nervous system tumors (malignant gliomas, surgery. Neurosurgery 1999; 44: 1299306.
pediatric tumors, and brain metastases). 47. Pollock BE, Gorman DA, Schomberg PJ,
Radiother Oncol 1997; 43: 23546. Kline RW. The Mayo Clinic gamma knife
37. Groves MD, Maor MH, Meyers C et al. A experience: indications and initial results.
phase II trial of high-dose bromodeoxyuridine Mayo Clin Proc 1999; 74: 513.
with accelerated fractionation radiotherapy 48. Foote KD, Friedman WA, Buatti JM, Bova
followed by procarbazine, lomustine, and FJ, Meeks SA. Linear accelerator radiosurgery
vincristine for glioblastoma multiforme. Int J in brain tumor management. Neurosurg Clin
Radiat Oncol Biol Phys 1999; 45: 12735. N Am 1999; 10: 20342.
38. Tubiana M, Eschwge F. Conformal radio- 49. Hoban PW, Jones LC, Clark BG. Modeling
therapy and intensity-modulated radio- late effects in hypofractionated stereotactic
therapy. Clinical data. Acta Oncol 2000; 39: radiotherapy. Int J Radiat Oncol Biol Phys
55567. 1999; 43: 199210.
39. Baumert BG, Lomax AJ, Miltchev V, Davis 50. Tokuuye K, Akine Y, Sumi M et al.
JB. A comparison of dose distributions of Fractionated stereotactic radiotherapy of
proton and photon beams in stereotactic small intracranial malignancies. Int J Radiat
conformal radiotherapy of brain lesions. Int J Oncol Biol Phys 1998; 42: 98994.
Radiat Oncol Biol Phys 2001; 49: 143949. 51. Loeffler JS, Kooy HM, Tarbell NJ. The
40. Barth RF, Yang WL, Rotaru JH et al. Boron emergence of conformal radiotherapy: special
neutron capture therapy of brain tumors: implications for pediatric neuro-oncology. Int
Enhanced survival and cure following J Radiat Oncol Biol Phys 1999; 44: 2378.
bloodbrain barrier disruption and intra- 52. Suh JH, Barnett GH. Brachytherapy for brain
carotid injection of sodium borocaptate and tumor. Hematol Oncol Clin N Am 1999; 13:
boronophenylalanine. Int J Radiat Oncol Biol 63550.
Phys 2000; 47: 20918. 53. Videtic GMM, Gaspar LE, Zamorano L et al.
41. Koot RW, Maarouf M, Hulshof MC et al. Use of the RTOG recursive partitioning
Brachytherapy: results of two different analysis to validate the benefit of iodine-125
therapy strategies for patients with primary implants in the primary treatment of malig-
glioblastoma multiforme. Cancer 2000; 88: nant gliomas. Int J Radiat Oncol Biol Phys
2796802. 1999; 45: 68792.
42. Lee SW, Fraass BA, Marsh LH et al. Patterns 54. Sneed PK, Stauffer PR, McDermott MW et al.
of failure following high-dose 3-D conformal Survival benefit of hyperthermia in a prospec-
radiotherapy for high-grade astrocytomas: a tive randomized trial of brachytherapy boost
quantitative dosimetric study. Int J Radiat +/ hyperthermia for glioblastoma multi-
Oncol Biol Phys 1999; 43: 7988. forme. Int J Radiat Oncol Biol Phys 1998; 40:
43. Khoo VS, Oldham M, Adams EJ et al. 28795.
Comparison of intensity-modulated tomo- 55. Glimelius B, Isacsson U, Blomquist E et al.
therapy with stereotactically guided confor- Potential gains using high-energy protons for
mal radiotherapy for brain tumors. Int J therapy of malignant tumours. Acta Oncol
Radiat Oncol Biol Phys 1999; 45: 41525. 1999; 38: 13745.
44. Kortmann RD, Becker G, Perelmouter J et al. 56. Munzenrider JE, Adams J, Liebsch NJ. Skull
Geometric accuracy of field alignment in base tumors: treatment with three-dimen-
fractionated stereotactic conformal radiother- sional planning and fractionated x-ray and

140
REFERENCES

proton radiotherapy. In: Leibel SA, Phillips 66. Hensley ML, Schuchter LM, Lindley C et al.
TL, eds. Textbook of Radiation Oncology. American Society of Clinical Oncology
Philadelphia: W.B. Saunders Co., 1998: Clinical Practice Guidelines for the Use of
34756. Chemotherapy and Radiotherapy Protectants.
57. Barth RF, Soloway AH, Goodman JH et al. J Clin Oncol 1999; 17: 333355.
Boron neutron capture therapy of brain 67. Posner JB. Neurologic complications of cancer,
tumors: an emerging therapeutic modality. 1st Edn. Philadelphia: F.A. Davis, 1995.
Neurosurgery 1999; 44: 43350. 68. Peterson K, Rosenblum MK, Powers JM,
58. Chanana AD, Capala J, Chadha M et al. Alvord EC, Walker RW, Posner JB. Effect of
Boron neutron capture therapy for glio- brain irradiation on demyelinating lesions.
blastoma multiforme: interim results from the Neurology 1993; 43: 210512.
phase I/II dose-escalation studies. Neuro- 69. Li YQ, Ballinger JR, Nordal RA, Su ZF,
surgery 1999; 44: 118292. Wong CS. Hypoxia in radiation-induced
59. Joel DD, Coderre JA, Micca PL, Nawrocky bloodspinal cord barrier breakdown. Cancer
MM. Effect of dose and infusion time on Res 2001; 61: 334854.
the delivery of p-boronophenylalanine for 70. OConnor MM, Mayberg MR. Effects of
neutron capture therapy. J Neurooncol 1999; radiation on cerebral vasculature: a review.
41: 21321. Neurosurgery 2000; 46: 13849.
60. Prados MD, Scott CB, Rotman M et al. 71. Hopewell JW, Wright EA. The nature of
Influence of bromodeoxyuridine radiosensiti- latent cerebral irradiation damage and its
zation on malignant glioma patient survival: modification by hypertension. Br J Radiol
a retrospective comparison of survival data 1970; 43: 1617.
from the Northern California Oncology 72. Lampert P, Tom MI, Rider WD.
Group (NCOG) and Radiation Therapy Disseminated demyelination of the brain
Oncology Group (RTOG) for glioblastoma following Co60 (gamma) radiation. Arch
and anaplastic astrocytoma. Int J Radiat Pathol (Chicago) 1959; 68: 32230.
Oncol Biol Phys 1998; 40: 6539. 73. Mima T, Toyonaga S, Mori K, Taniguchi T,
61. Carde P, Timmerman R, Mehta MP et al. Ogawa Y. Early decrease of P-glycoprotein in
Multicenter phase Ib/II trial of the radiation the endothelium of the rat brain capillaries
enhancer motexafin gadolinium in patients after moderate dose of irradiation. Neurol
with brain metastases. J Clin Oncol 2000; 19: Res 1999; 21: 20915.
207483. 74. Armstrong C, Ruffer J, Corn B, DeVries K,
62. Lammering G, Valerie K, Lin PS et al. Mollman J. Biphasic patterns of memory
Radiosensitization of malignant glioma cells deficits following moderate-dose partial-brain
through overexpression of dominant-negative irradiation: neuropsychologic outcome and
epidermal growth factor receptor. Clin proposed mechanisms. J Clin Oncol 1995;
Cancer Res 2001; 7: 68290. 13: 226371.
63. Valerie K, Hawkins W, Farnsworth J et al. 75. Marks JE, Wong J. The risk of cerebral
Substantially improved in vivo radiosensitiza- radionecrosis in relation to dose, time and
tion of rat glioma with mutant HSV-TK and fractionation: a follow-up study. Prog Exp
acyclovir. Cancer Gene Ther 2001; 8: 38. Tumor Res 1985; 29: 2108.
64. Gramaglia A, Loi GF, Mongioj V, Baronzio 76. Morris JG, Grattan-Smith P, Panegyres PK et
GF. Increased survival in brain metastatic al. Delayed cerebral radiation necrosis. Q J
patients treated with stereotactic radio- Med 1994; 87: 11929.
therapy, omega three fatty acids and 77. Shewmon DA, Masdeu JC. Delayed radiation
bioflavonoids. Anticancer Res 1999; 19: necrosis of the brain contralateral to original
55836. tumor. Arch Neurol 1980; 37: 5924.
65. Noel F, Tofilon PJ. Astrocytes protect against 78. Ricci PE, Karis JP, Heiserman JE et al.
X-ray-induced neuronal toxicity in vitro. Differentiating recurrent tumor from radia-
Neuroreport 1998; 9: 11337. tion necrosis: time for re-evaluation of

141
PRINCIPLES OF THERAPY

positron emission tomography? Am J 90. Randomized trial of procarbazine, lomustine,


Neuroradiol 1998; 19: 40713. and vincristine in the adjuvant treatment of
79. Vigliani MC, Duyckaerts C, Hauw JJ et al. high-grade astrocytoma: a Medical Research
Dementia following treatment of brain Council trial. J Clin Oncol 2001; 19: 50918.
tumors with radiotherapy administered alone 91. Fine HA, Dear KB, Loeffler JS, Black PM,
or in combination with nitrosourea-based Canellos GP. Meta-analysis of radiation
chemotherapy: a clinical and pathological therapy with and without adjuvant
study. J Neurooncol 1999; 41: 13749. chemotherapy for malignant gliomas in
80. Thiessen B, DeAngelis LM. Hydrocephalus in adults. Cancer 1993; 71: 258597.
radiation leukoencephalopathy results of 92. Keime-Guibert F, Napolitano M, Delattre
ventriculoperitoneal shunting. Arch Neurol J-Y. Neurological complications of radio-
1998; 55: 70510. therapy and chemotherapy. J Neurol 1998;
81. Kleinberg L, Wallner K, Malkin MG. Good 245: 695708.
performance status of long-term disease-free 93. Bartus RT, Snodgrass P, Dean RL, Kordower
survivors of intracranial gliomas. Int J Radiat JH, Emerich DF. Evidence that Cereports
Oncol Biol Phys 1993; 26: 12933. ability to increase permeability of rat gliomas
82. Johansson M, Bergenheim AT, Widmark A, is dependent upon extent of tumor growth:
Henriksson R. Effects of radiotherapy and Implications for treating newly emerging
estramustine on the microvasculature in malig- tumor colonies. Exp Neurol 2000; 161:
nant glioma. Br J Cancer 1999; 80: 1428. 23444.
83. Grill J, Couanet D, Cappelli C et al. 94. Remsen LG, Pagel MA, McCormick CI et al.
Radiation-induced cerebral vasculopathy in The influence of anesthetic choice, PaCo2,
children with neurofibromatosis and optic and other factors on osmotic bloodbrain
pathway glioma. Ann Neurol 1999; 45: barrier disruption in rats with brain tumor
3936. xenografts. Anesth Analg 1999; 88: 55967.
84. Brandes AA, Pasetto LM, Lumachi F, 95. Gobin YP, Cloughesy TF, Chow KL et al.
Monfardini S. Endocrine dysfunctions in Intraarterial chemotherapy for brain tumors
patients treated for brain tumors: incidence by using a spatial dose fractionation
and guidelines for management. J Neurooncol algorithm and pulsatile delivery. Radiology
2000; 47: 8592. 2001; 218: 72432.
85. Galanis E, Buckner JC. Chemotherapy of 96. Brem H, Piantadosi S, Burger PC et al.
brain tumors. Curr Opin Neurol 2001; 13: Placebo-controlled trial of safety and efficacy
61925. of intraoperative controlled delivery by
86. DeAngelis LM. Medical progress: Brain biodegradable polymers of chemotherapy for
tumors. N Engl J Med 2001; 344: 11423. recurrent gliomas. The Polymer-Brain Tumor
87. Huncharek M, Muscat J, Geschwind JF. Treatment Group. Lancet 1995; 345:
Multi-drug versus single agent chemotherapy 100812.
for high grade astrocytoma; results of a meta- 97. Menei P, Venier MC, Gamelin E et al. Local
analysis. Anticancer Res 1998; 18: 46937. and sustained delivery of 5-fluorouracil from
88. Shapiro WR, Shapiro JR. Biology and treat- biodegradable microspheres for the radio-
ment of malignant glioma. Oncology 1998; sensitization of glioblastoma a pilot study.
12: 23340. Cancer 1999; 86: 32530.
89. Prados MD, Scott C, Curran WJ Jr, Nelson 98. Kreuter J. Nanoparticulate systems for brain
DF, Leibel S, Kramer S. Procarbazine, lomus- delivery of drugs. Adv Drug Deliv Rev 2001;
tine, and vincristine (PCV) chemotherapy for 47: 6581.
anaplastic astrocytoma: a retrospective review 99. Zucchetti M, Boiardi A, Silvani A et al.
of Radiation Therapy Oncology Group Distribution of daunorubicin and daunorubi-
Protocols comparing survival with carmustine cinol in human glioma tumors after adminis-
or PCV adjuvant chemotherapy. J Clin Oncol tration of liposomal daunorubicin. Cancer
1999; 17: 338995. Chemother Pharmacol 1999; 44: 1736.

142
REFERENCES

100. Subach BR, Witham TF, Kondziolka D, 110. Chamberlain MC, Kormanik P. Salvage
Lunsford LD, Bozik M, Schiff D. Morbidity chemotherapy with taxol for recurrent anaplas-
and survival after 1,3-bis(2-chloroethyl)-1 tic astrocytomas. J Neurooncol 1999; 43: 718.
nitrosourea wafer implantation for recurrent 111. Freilich RJ, Balmaceda C, Seidman AD,
glioblastoma: a retrospective case matched Rubin M, DeAngelis LM. Motor neuopathy
cohort series. Neurosurgery 1999; 45: 1722. due to docetaxel and paclitaxel. Neurology
101. Esteller M, Garcia-Foncillas J, Andion E et al. 1996; 47: 1158.
Inactivation of the DNA-repair gene MGMT 112. Stewart CF, Gajjar AJ, Heideman RL,
and the clinical response of gliomas to Houghton PJ. Penetration of topotecan into
alkylating agents. N Engl J Med 2000; 343: cerebrospinal fluid after intravenous injec-
13504. tion. Onkologie 1998; 21: 224.
102. Bredel M. Anticancer drug resistance in 113. Kadota RP, Stewart CF, Horn M et al.
primary human brain tumors. Brain Res Topotecan for the treatment of recurrent or
Brain Res Rev 2001; 35: 161204. progressive central nervous system tumors
103. Ashby LS, Shapiro WR. Intra-arterial a Pediatric Oncology Group phase II study. J
cisplatin plus oral etoposide for the treatment Neurooncol 1999; 43: 437.
of recurrent malignant glioma: a phase II 114. Schtte W, Manegold C, Von Pawel JV.
study. J Neurooncology 2001; 51: 6786. Topotecan in the therapy of brain metastases
104. Shapiro WR, Green SB, Burger PC et al. A in lung cancer. Onkologie 1998; 21: 257.
randomized comparison of intra-arterial 115. Chang SM, Barker FG, II, Huhn SL et al.
versus intravenous BCNU, with or without High dose oral tamoxifen and subcutaneous
intravenous 5-fluorouracil, for newly interferon alpha-2a for recurrent glioma. J
diagnosed patients with malignant glioma. J Neurooncol 1998; 37: 16976.
Neurosurg 1992; 76: 77281. 116. Miller KD, Sweeney CJ, Sledge GW.
105. Poppenborg H, Knpfer MM, Preiss R, Wolff Redefining the target: chemotherapeutics as
JEA, Galla HJ. Cisplatin (CDDP)-induced antiangiogenics. J Clin Oncol 2001; 19:
radiation resistance is not associated with 1195206.
CDDP resistance in 86HG39 and A172 117. Los M, Voest EE. The potential role of
malignant glioma cells. Eur J Cancer 1999; antivascular therapy in the adjuvant and
35: 11504. neoadjuvant treatment of cancer. Semin
106. Friedman HS, McLendon RE, Kerby T et al. Oncol 2001; 28: 93105.
DNA mismatch repair and O6alkylguanine- 118. Lam PYP, Breakefield XO. Potential of gene
DNA alkyltransferase analysis and response therapy for brain tumors. Hum Mol Genet
to Temodal in newly diagnosed malignant 2001; 10: 77787.
glioma. J Clin Oncol 1998; 16: 38517. 119. Parney IF, Hao CH, Petruk KC. Glioma
107. Cloughesy TF, Black KL, Gobin YP et al. immunology and immunotherapy. Neuro-
Intra-arterial Cereport (RMP-7) and carbo- surgery 2000; 46: 77891.
platin: a dose escalation study for recurrent 120. Oldfield EH, Youle RJ. Immunotoxins for
malignant gliomas. Neurosurgery 1999; 44: brain tumor therapy. Curr Top Microbiol
2708. Immunol 1998; 234: 97114.
108. Dinnes J, Cave C, Huang S, Major K, Milne 121. Arab S, Murakami M, Dirks P et al.
R. The effectiveness and cost-effectiveness of Verotoxins inhibit the growth of and induce
temozolomide for the treatment of recurrent apoptosis in human astrocytoma cells. J
malignant glioma: a rapid and systematic Neurooncol 1998; 40: 13750.
review. Health Technol Assess 2001; 5: 173. 122. Fine HA, Figg WD, Jaeckle K et al. Phase II
109. Beauchesne P, Soler C, Rusch P et al. Phase trial of the antiangiogenic agent thalidomide
II study of a radiotherapy/etoposide combi- in patients with recurrent high-grade gliomas.
nation for patients with newly malignant J Clin Oncol 2000; 18: 70815.
gliomas. Cancer Chemother Pharmacol 1999; 123. Estin D, Li MW, Spray D, Wu JK. Connexins
44: 21016. are expressed in primary brain tumors and

143
PRINCIPLES OF THERAPY

enhance the bystander effect in gene therapy. throughout the course of malignant glioma
Neurosurgery 1999; 44: 3618. An evidence-based review. Cancer 2000; 89:
124. Guberman AH, Besag FMC, Brodie MJ et al. 6406.
Lamotrigine-associated rash: Risk benefit 135. Ruff RL, Posner JB. The incidence of
considerations in adults and children. systemic venous thrombosis and the risk of
Epilepsia 1999; 40: 98591. anticoagulation in patients with malignant
125. Beenen LFM, Lindeboom J, Trenit DGAK et gliomas. Trans Am Neurol Assoc 1981; 106:
al. Comparative double blind clinical trial of 2236.
phenytoin and sodium valproate as anticon- 136. Agnelli G, Piovella F, Buoncristiani P et al.
vulsant prophylaxis after craniotomy: Enoxaparin plus compression stockings
efficacy, tolerability, and cognitive effects. J compared with compression stockings alone
Neurol Neurosurg Psychiatry 1999; 67: in the prevention of venous thrombo-
47480. embolism after elective neurosurgery. N Engl
126. Martin R, Kuzniecky R, Ho S et al. Cognitive J Med 1998; 339: 805.
effects of topiramate, gabapentin, and lamo- 137. Macdonald RL, Amidei C, Lin G et al. Safety
trigine in healthy young adults. Neurology of perioperative subcutaneous heparin for
1999; 52: 3217. prophylaxis of venous thromboembolism in
127. Glantz M, Friedberg M, Cole B et al. Double- patients undergoing craniotomy. Neuro-
blind, randomized, placebo-controlled trial of surgery 1999; 45: 24551.
anticonvulsant prophylaxis in adults with 138. Constantini S, Kanner A, Friedman A et al.
newly diagnosed brain metastases. Proc Safety of perioperative minidose heparin in
ASCO 1994; 13, 176. patients undergoing brain tumor surgery: a
128. Dupond JL, Gil H, De Wazires B, Berthier prospective, randomized, double-blind study.
S, Magy N. Withdrawing corticosteroids: the J Neurosurg 2001; 94: 91821.
stress of the last milligrams. Presse Med 139. Dickinson LD, Miller LD, Patel CP, Gupta
1999; 28: 1402. SK. Enoxaparin increases the incidence of
129. Kondziolka D, Mori Y, Martinez AJ et al. postoperative intracranial hemorrhage when
Beneficial effects of the radioprotectant 21- initiated preoperatively for deep venous
aminosteroid U-74389G in a radiosurgery rat thrombosis prophylaxis in patients with brain
malignant glioma model. Int J Radiat Oncol tumors. Neurosurgery 1998; 43: 107481.
Biol Phys 1999; 44: 17984. 140. Lazio BE, Simard JM. Anticoagulation in
130. Tjuvajev J, Uehara H, Desai R et al. neurosurgical patients. Neurosurgery 1999;
Corticotropin-releasing factor decreases 45: 83847.
vasogenic brain edema. Cancer Res 1996; 56: 141. Levin JM, Schiff D, Loeffler JS et al.
135260. Complications of therapy for venous throm-
131. Glaser T, Winter S, Groscurth P et al. boembolic disease in patients with brain
Boswellic acids and malignant glioma: induc- tumors. Neurology 1993; 43: 11114.
tion of apoptosis but no modulation of drug 142. Yam D, Peled A, Shinitzky M. Suppression of
sensitivity. Br J Cancer 1999; 80: 75665. tumor growth and metastasis by dietary fish
132. De Kloet ER, Oitzl MS, Jols M. Stress and oil combined with vitamins E and C and
cognition: are corticosteroids good or bad cisplatin. Cancer Chemother Pharmacol
guys? Trends Neurosci 1999; 22: 4226. 2001; 47: 3440.
133. Gayan-Ramirez G, Vanderhoydonc F, 143. Verhoef MJ, Hagen N, Pelletier G, Forsyth P.
Verhoeven G, Decramer M. Acute treatment Alternative therapy use in neurologic diseases
with corticosteroids decreases IGF-1 and Use in brain tumor patients. Neurology
IGF-2 expression in the rat diaphragm and 1999; 52: 61722.
gastrocnemius. Am J Respir Crit Care Med 144. Kuba H, Inamura T, Ikezaki K, Kawashima
1999; 159: 2839. M, Fukui M. Thiamine-deficient lactic acido-
134. Marras LC, Geerts WH, Perry JR. The risk sis with brain tumor treatment report of
of venous thromboembolism is increased three cases. J Neurosurg 1998; 89: 10258.

144
REFERENCES

145. Giovagnoli AR. Quality of life in patients 147. Whitton AC, Rhydderch H, Furlong W,
with stable disease after surgery, radiother- Feeny D, Barr RD. Self-reported compre-
apy, and chemotherapy for malignant brain hensive health status of adult brain tumor
tumour. J Neurol Neurosurg Psychiatry 1999; patients using the health utilities index.
67: 35863. Cancer 1997; 80: 25865.
146. Weitzner MA. Psychosocial and neuropsychi- 148. Meyers CA, Weitzner MA, Valentine AD,
atric aspects of patients with primary brain Levin VA. Methylphenidate therapy improves
tumors. Cancer Invest 1999; 17: 28591. cognition, mood, and function of brain tumor
patients. J Clin Oncol 1998; 16: 25227.

145
II
Management of specific tumors
5
Glial tumors

Introduction some cranial nerves (Chapter 9). Microglia are


not true glia. Unlike the other glia, which are
Although their exact lineage is unknown, most ectodermal in origin, microglia are meso-
intrinsic brain tumors are believed to originate dermal, probably of hematopoietic origin; they
from glial cells or their precursors.1,2 Central are involved in inflammatory and immune
nervous system (CNS) glia can be divided into responses in the CNS and will not be discussed
astrocytes, oligodendrocytes, microglia and in this chapter. Ependymal cells line the
ependyma. Schwann cells, the peripheral cerebral ventricles and form the surface of the
nervous systems oligodendrocytes, are also choroid plexus, which is why choroid plexus
found within the intracranial cavity, synthesiz- tumors are discussed in this chapter. Although
ing myelin in the distal intracranial portions of the word glia (from the Greek meaning glue)

Figure 5.1
A schematic sagittal section showing
common locations for diffuse glial
tumors. (A) Most arise within the
B white matter of the hemispheres. The
tumors often infiltrate the cortex. (B)
The genu of the corpus callosum and
(C) the splenium of the corpus
A callosum are common sites of
C
gliomas. From these sites, the tumor
can infiltrate both hemispheres.
E
Gliomas can arise anywhere in the
D brainstem, usually the pons (D), but
occasionally the midbrain (E) or the
F medulla (F). Low-grade astrocytomas
commonly arise within the
cerebellum (Fig. 5.10). All are
discussed in the text.

149
GLIAL TUMORS

implies that these cells just support and hold more common in adults than in children and
neurons in place, glia have far more important generally arise in the cerebral hemispheres,
metabolic functions than just providing struc- although they may affect brainstem, optic
tural support,3 for example, they maintain the nerve, cerebellum or spinal cord. Low-grade
bloodbrain barrier (Chapter 2). tumors have a tendency to progress to a high-
Tumors of glial origin can be divided into grade phenotype. High-grade tumors, i.e.
those that infiltrate into normal brain struc- glioblastoma, may either arise by progression
tures (diffuse tumors, Fig. 5.1) and those with from a lower grade tumor or may develop de
more discrete boundaries, i.e. focal tumors novo.
(Table 5.1). The division is relative: some
diffuse glial tumors have relatively discrete
Astrocytomas
boundaries (e.g. so-called type 1 astrocytomas4)
and some focal tumors invade surrounding Introduction
normal brain (e.g. invasive ependymomas). Astrocytoma (from the Greek word astro for
star) refers to the stellate shape of some astro-
cytes. Diffuse astrocytic tumors can be divided
by histologic characteristics into astrocytoma,
Diffuse glial tumors a low-grade tumor, and anaplastic astrocytoma
Diffuse glial tumors range from very low grade and glioblastoma multiforme, both high-grade
(astrocytoma, oligodendroglioma) to extremely tumors (Fig. 5.2). Also included in this group
high grade (glioblastoma multiforme). They are are tumors that diffusely infiltrate all or much
of the brain without necessarily forming a mass
lesion (gliomatosis cerebri) and tumors
restricted to the brainstem (brainstem gliomas);
Table 5.1
both may be of either grade. Because of differ-
Glial tumors. ent age and growth characteristics, and because
their location often precludes biopsy, brainstem
gliomas are considered separately from supra-
Diffuse glial tumors
tentorial gliomas. Almost by definition, diffuse
Astrocytic tumors
Astrocytoma astrocytomas, even those that appear to be
Anaplastic astrocytoma histologically discrete, are not amenable to
Glioblastoma multiforme surgical cure.
Gliomatosis cerebri
Brainstem glioma Incidence
Oligodendroglial tumors
As indicated in Chapter 1, diffuse astrocytic
Oligodendroglioma
Anaplastic oligodendroglioma tumors represent about 25% of primary
Glioblastoma multiforme intracranial tumors encountered at autopsy,
Focal glial tumors but 35% of symptomatic primary intracranial
Pilocytic astrocytoma tumors (Table 1.3). Glial tumors affect 57
Pleomorphic xanthoastrocytoma (PXA)
new patients per 100 000 population per year,
Ependymoma
Subependymoma or about 18 000 new cases in the USA
Choroid plexus papilloma annually. In addition to being the most
common symptomatic tumor, diffuse astrocytic

150
DIFFUSE GLIAL TUMORS

(a) (b)

Figure 5.2
Photomicrographs showing the evolution of diffuse
astrocytic tumors. A glioblastoma multiforme, the
highest grade of diffuse astrocytoma, may arise by
progressive evolution of the lesion from astrocytoma
(a) showing hypercellular tumor tissue with angulated
hyperchromatic atypical astrocytic nuclei; no mitoses
are evident. The next stage is anaplastic astrocytoma
(b) showing increased nuclear atypia and the presence
of mitoses (arrows). The glioblastoma (GBM) (c)
shows highly atypical giant tumor cells and mitotic
figures (arrow). A GBM may also arise de novo.
Genetic pathways of these primary and secondary
(c) astrocytomas are outlined in Table 5.2.

Table 5.2
Comparison of World Health Organization (WHO) and St Anne/Mayo classification of astrocytomas.25

WHO grade WHO designation St Anne/Mayo Histological criteria


designation

I Pilocytic astrocytoma Astrocytoma grade 1 Rosenthal fibers, piloid cells


(low-grade diffuse) zero
criterion
II Diffuse astrocytoma Astrocytoma grade 2 One criterion, usually
nuclear atypia
III Anaplastic astrocytoma Astrocytoma grade 3 Two criteria, usually
nuclear atypia and mitosis
IV Glioblastoma multiforme Astrocytoma grade 4 Three of four criteria: the
two above plus endothelial
proliferation and/or necrosis

151
GLIAL TUMORS

tumors, unlike some other common brain Etiology


tumors such as meningiomas and pituitary
tumors, are not curable. Risk factors. With the exception of ionizing
Also as discussed in Chapter 1, diffuse radiation (Chapter 1), no risk factors have been
astrocytic tumors may be increasing in established for sporadic diffuse astrocytomas.
incidence. The most dramatic data are those Even with respect to ionizing radiation, in any
of Olney et al,5 showing the annual incidence given individual, one cannot be certain whether
of astrocytomas, anaplastic astrocytomas and a diffuse astrocytic tumor occurring in a previ-
glioblastomas between 1975 and 1992 ously irradiated patient is related or coinciden-
increasing from 45 to 53 tumors per million tal. If the tumor occurs within the radiation
population, with a dramatic increase in the portal, then the presumption of radiation as
incidence of anaplastic astrocytomas and causal is strong. In one of our patients, an
glioblastomas and a corresponding fall in anaplastic astrocytoma developed in the radia-
(low-grade) astrocytomas. The changing tion portal of a pituitary tumor 4 years after
incidence of histologic subtypes could be treatment. However, careful examination of
attributed to changes in interpretation of previous scans suggested that the tumor had
microscopic slides by neuropathologists, been there shortly following radiation therapy
although it is not known if there has been a (RT), a latency too short for it to be other than
tendency for neuropathologists to upgrade coincidence. On the other hand, we have
their interpretation of diffuse astrocytic encountered a glioblastoma in a patient irradi-
tumors (i.e. call tumors previously read as ated successfully 20 years earlier for a pineal
astrocytoma, anaplastic). If there has been region germinoma. The glioblastoma was bilat-
substantial upgrading in histologic interpreta- eral and symmetrical, effectively outlining the
tion, one would expect to see reported radiation port. The development of a radiation-
improvement in survival via the Will Rogers induced astrocytic tumor does not preclude the
effect. (Will Rogers, a humorist, stated that re-use of radiation if the patients normal brain
during the depression of the 1930s, the migra- can tolerate it.
tion of individuals from Oklahoma to As discussed in Chapter 1, two alleged
California raised the IQ of both states.) If a additional risk factors specifically linked to
tumor that was once considered low-grade is diffuse astrocytic tumors are ingestion of aspar-
now considered high-grade, and if low-grade tame and electromagnetic (non-ionizing) radia-
tumors have longer survival than high-grade tion, particularly that produced by the portable
tumors, then the change in pathologic reading cellular telephone. In portable cellular
should lead to longer survival in both patient telephones, the antenna is part of the headset
groups. To date, there has been no evidence and the users head is exposed to radiofre-
of improved survival in populations of quency energy. As discussed in Chapter 1,
patients with high-grade gliomas, so this is an studies of cellular telephones have failed to
unlikely explanation for any observed changes show a correlation with brain tumors. Also, as
in epidemiology of gliomas. In addition, discussed in Chapter 1, the aspartame data are
changes in pathologic interpretation can also unconvincing. At least one recent study failed
lead to optimistic reports of phase 2 studies to provide evidence of DNA damage after
done in an institution where the neuropathol- exposure to electromagnetic radiation within
ogist habitually upgrades. the cellular phone communication frequency

152
DIFFUSE GLIAL TUMORS

band. A study from Sweden addressing occupa- Genetic alterations


tional exposure and glioma suggests an
increased risk associated with solvents, pesti- Figure 5.3 illustrates some of the genetic changes
cides and plastic materials.6 A study from encountered in patients with diffuse astrocy-
China suggests that nutritional factors (fresh tomas.
vegetables, vitamin E and calcium protective; Genetic changes leading to the formation of
salted vegetables and salted fish deleterious) gliomas affect two major cell functions: signal
may play a role.7 transduction and the cell cycle.2 The first is
About 5% of gliomas are familial,8 most affected by overexpression of growth factors
not belonging to the recognized syndromes. A or their receptors (e.g. EGF, PDGF) that
few familial disorders that predispose to through autocrine and paracrine pathways
astrocytoma are discussed in Chapter 12. activate signaling pathways such as RAS and
Astrocytomas occasionally complicate neuro- AKT.2 Alternately, mutations of tumor
fibromatosis-1 and neurofibromatosis-2, the suppressors such as PTEN may fail to inhibit
LiFraumeni syndrome and Turcots signaling. Either change can lead to uncon-
syndrome. A specific type of astrocytoma, the trolled growth. The second general mechanism
subependymal giant cell astrocytoma, compli- is an alteration of the cell cycle. Loss of
cates tuberous sclerosis. inhibitors of the cell cycle such as INK4A, RB,

Differentiated astrocytes or neuroepithelial precursor cells

p53 Mutation (>65%) PDGF-A EGRF


PDGFR- overexpression (~60%) Overexpression, Amplification

Astrocytoma MDM2 Amplification (<10%), p53 mutation


overexpression (~50%)
LOH 19q (~65%) LOH 10/PTEN
RB alteration (~25%)
p16 deletion (3040%) ?

Anaplastic astrocytoma ?

LOH 10PTEN LOH 10q/PTEN (-80%)


DCC Loss of expression (~50%) RB alteration
PDGFR- amplification (<10%)

Secondary glioblastoma Primary glioblastoma de novo Other primary glioblastoma


including giant cell glioblastoma

Figure 5.3
Genetic change in diffuse astrocytoma. (Modified from2,25 with permission.)

153
GLIAL TUMORS

Figure 5.4
A unique patient with a symptomatic diffuse astrocytoma and a negative MR scan. The T2 (left) and T1
contrast-enhanced image (middle) were negative when the patient first presented with a generalized convulsion.
Nine months later in the face of progressive symptoms, a repeat scan showed the image on the right, which on
resection was a glioblastoma multiforme.

p53 and others can also lead to uncontrolled and/or overexpression of platelet-derived growth
growth, genetic instability and failure of factor A (PDGF-A) or platelet-derived growth
normal apoptosis (Chapter 1). factor receptor alpha (PDGFR-). Progressive
Glioblastoma (GBM) can arise de novo (Fig. genetic changes in low-grade astrocytomas,
5.4, center and right panels) or progress from including the loss of heterozygosity on 19q or
a low-grade tumor. The de novo pathway alteration in the retinoblastoma tumor suppres-
usually occurs in older patients in whom there sor gene, lead the low-grade tumor to become
is a short history of symptoms and no known anaplastic. Other genetic changes, including
pre-existing low-grade lesion. Alternatively, a mutations in the PTEN gene, loss of expression
GBM can arise through the progressive of the DCC gene, or amplification of PDGFR-
accumulation of genetic defects (Fig. 5.3, left , lead to GBM (secondary GBM). De novo or
panel) that lead first to an astrocytoma, then primary GBM arises by amplification or overex-
an anaplastic astrocytoma and finally a GBM. pression of epidermal growth factor receptor
This pathway primarily occurs in young adults (EGFR); p53 mutations are rare. In about 40%
who often have a 310 year history before a of GBMs the EGFR is truncated with an in-
GBM develops. The rate of progression to frame deletion of approximately 30 amino acids.
anaplasia correlates directly with age.10 These The truncated form is constitutively active, i.e.
alternative pathways indicate that no single active without the need for ligand binding. The
genetic change is either essential or sufficient truncated receptor gene is not found in normal
for GBM development. glial cells and thus forms a potential tumor-
Two major abnormalities that lead a differen- specific therapeutic target. PTEN inactivation
tiated astrocyte or neuroepithelial precursor cell plays an equal role in de novo and secondary
to become an astrocytoma are p53 mutations glioblastomas.11

154
DIFFUSE GLIAL TUMORS

The differentiation between primary and gliomas and may be associated with the transi-
secondary GBM may be difficult and cannot be tion to anaplasia.16 Exogenous PTEN induces
made on a molecular basis alone. If a patient G1 cell cycle arrest in glioma cells in culture.17
has a history of low-grade glioma that becomes One in vitro study suggests that tumors with
a GBM, or if histologic evidence of both low- mutated p53 are more resistant to fractionated
grade and high-grade astrocytoma is found in RT than are those with wild-type p53. Those
the same tumor specimen, the GBM is proba- tumors with mutated p53 may respond better
bly secondary. However, some apparently de to fewer, larger fractions of radiation.18
novo glioblastomas have areas of low-grade as Deletions of chromosome 22q occur in about
well as high-grade tumor. The low-grade areas 30% of astrocytic tumors and the frequency of
in these primary GBMs have more 10q these deletions increases with progressive
deletions and fewer p53 mutations than are anaplasia (17% astrocytomas, 38% GBMs).19
found in the usual low-grade astrocytoma. The nature of the suppressor gene on 22q is not
These tumors lack EGFR amplification, known.
suggesting that they represent a subset of The matrix metalloproteinases, gelatinase A
secondary GBM with unusually rapid progres- and B, are overexpressed in glioma, with
sion.12 An MR scan showing areas of non- expression of the B isoform related to higher
enhancing as well as enhancing tumor, or serial tumor grade. These substances are involved in
MR scans changing from non-enhancing to tumor invasion and angiogenesis (Chapter
enhancing, suggest a secondary GBM. The 2).20,21 The tumor suppression gene DCC
development of an intensely enhancing tumor (Deleted in Colorectal Cancer) is reduced or
on the background of a previously normal scan absent in most glioblastomas, but is usually
suggests a primary GBM. Primary and normal in low-grade tumors. Its absence
secondary GBMs may differ in biology as well predicts a poor prognosis.22 Other genetic
as genetics. Primary GBM is associated with a changes include proteosome-induced degrada-
shorter survival than is secondary GBM. tion of p27, a protein that regulates the cell
High-grade gliomas often show abnormalities cycle at G1 to S transition.23 Whether mutations
of genes coding for proteins that regulate the in p21 contribute to the development of
cell cycle (see Fig. 1.9). Control of the transi- gliomas is uncertain.24
tion from G1 to S phase is affected by mutation
or deletion of the retinoblastoma gene but also Pathology
by deletion or alterations of cyclins, including Most astrocytic tumors are gray or yellow and
deletion of CDKN2A and CDKN2B, as well as soft to the touch, but focal calcifications give
amplification or overexpression of CDK4 and a gritty feeling to some astrocytomas.
CDK6. D-type cyclin genes that regulate cyclin- Microcyst formation in low-grade tumors may
dependent kinases are overexpressed in a few cause a gelatinous appearance. Sometimes a
high-grade gliomas.13 In high-grade astrocy- single large cyst filled with clear fluid is
toma, CDKN2/p16 is decreased and associated present. However, even when the boundaries
with a poor prognosis.14 The tumor suppressor appear distinct macroscopically, microscopic
genes p53, p16, p14 and PTEN may be co- infiltration of brain is present. Grossly, it is
mutated in high-grade gliomas, each leading to often impossible to distinguish a low-grade
a different alteration in cellular pathways.15 from a high-grade diffuse astrocytoma unless
PTEN mutations are present only in high-grade necrosis, characteristic of GBM, is present.

155
GLIAL TUMORS

Owing to their infiltrative nature, diffuse astro- a diffuse astrocytoma (WHO Grade II) or
cytomas usually show blurring of gross astrocytoma grade II (St Anne/Mayo designa-
anatomic boundaries, particularly loss of the tion) shows atypical nuclei, increased cellular-
graywhite matter border. Edema surrounds ity and microcysts. No mitotic activity,
the tumor, but where tumor ends and edema endothelial proliferation or necrosis can be
begins cannot be identified grossly. GBM is identified. In the next panel, an anaplastic
more likely to appear well-demarcated from astrocytoma (WHO Grade III) or astrocytoma
surrounding normal brain. Hemorrhage and grade 3 (St Anne/Mayo) shows higher cellular-
necrosis are generally not found in lower grade ity and nuclear atypia as well as mitotic activ-
tumors but may be grossly visible in GBM. ity. GBM (WHO Grade IV) or astrocytoma
Microscopically, there may be infiltration of Grade 4 (St Anne/Mayo criteria) shows nuclear
tumor cells several centimeters from the gross atypia, mitosis, endothelial proliferation and
bulk of tumor.4 Nuclear atypia, increased cellu- necrosis. Cellular proliferation evaluated by the
larity, endothelial proliferation, mitoses and MIB-1 labeling index helps distinguish among
necrosis are used to grade diffuse astrocytic WHO Grade I (pilocytic) and Grades II and III.
tumors. Two grading systems for diffuse astro- In Grades II and III astrocytomas, the MIB-1
cytomas are widely used (Table 5.2). index is the single best prognostic factor;
The WHO uses a four-tiered system with however, for an individual patient, the labeling
Grade I reserved for pilocytic astrocytomas, index alone should not determine therapy.27
Grade II for astrocytoma (low-grade), Grade Surprisingly, the index does not give prognos-
III anaplastic astrocytoma, and Grade IV GBM tic information for recurrent tumors.28
(both high-grade), respectively.25 The St Anne/ Gemistocytes (from Latin gemma meaning
Mayo system also uses numerical grades. It is bud) are large eosinophilic cells with spheri-
often difficult to distinguish astrocytomas cal, eccentric nuclei. Gemistocytic astrocy-
from anaplastic astrocytomas, leading to some tomas, especially those with > 20%
disagreement between pathologists about gemistocytes, indicate a poor prognosis.29
interpreting individual tumors. The difficulty Occasionally, glial tumors can acquire histo-
is compounded by the recent use of stereotac- logic features suggestive of other tumor types,
tic needle biopsy, which gives the neuropathol- such as ependymal differentiation leading to
ogist a very small sample to examine. Because confusion with an ependymoma. An experi-
of the well-known heterogeneity of astrocytic enced neuropathologist can usually discern the
tumors, the smaller the sample, the less likely correct nature of the lesion; however,
it is that one can grade the tumor accurately. sometimes additional pathologic evaluation is
Intratumoral heterogeneity can be demon- necessary. For example, electron microscopy
strated not only microscopically but also at the will identify the cilia characteristic of a true
molecular level by comparative genomic ependymoma, which are absent in an astrocytic
hybridization.26 In controlled trials of the tumor with ependymal differentiation.
treatment of diffuse astrocytomas, it is impor- Two biological characteristics of diffuse
tant to have one pathologist examining all of astrocytomas are invasion of normal surround-
the tumors, ensuring consistency if not ing brain and angiogenesis (Chapter 2). Unlike
absolute accuracy. cancers elsewhere in the body, which usually
Figure 5.2 illustrates the several grades of have the capacity not only to invade surround-
diffuse astrocytomas. In the panel on the left, ing tissue but also to metastasize distantly,

156
DIFFUSE GLIAL TUMORS

astrocytomas usually invade and may spread which facilitates motility. Proteases are
widely within the CNS but rarely metastasize expressed by glioma cells and some, particu-
outside of it. Normal glial cells in vitro, and larly metalloproteinases, are probably impor-
probably in vivo as well, are motile. This tant for invasion. Growth factors, including
property is enhanced in glioma cells, and the transforming growth factor beta-1 (TGF-1),
degree of motility appears to be directly related contribute to the spread of tumor cells within
to the grade of the tumor cell. Cell migration the CNS.
appears to follow white matter pathways; the The capacity to invade not only makes it
corpus callosum and anterior commissure are impossible to completely remove diffuse astro-
among the major pathways for spread of astro- cytic tumors, but also allows them to appear
cytomas. These two routes allow tumors to in multiple places in the brain. Somewhere
spread from one hemisphere to the other. between 5% and 10% of diffuse astrocytomas
In patients with brain tumors, Kelly et al are said to be multicentric. A multicentric
have demonstrated by stereotactic needle glioma is a clinical challenge because the
biopsy of tumor and surrounding brain that imaging may resemble that of brain metasta-
tumor cells often invade normal brain to a sis and lead to an incorrect evaluation or
variable degree.4 In some tumors, there is little diagnosis. It is possible that so-called multi-
or no invasion and the tumor is grossly as well centric gliomas represent spread from a single
as microscopically reasonably well circum- focus, with areas of grossly apparent tumor
scribed. In other tumors, cells can be found being connected by bridges of microscopic
several centimeters beyond the apparent border glioma cells. In its most florid form, gliomas
of the tumor with normal brain. The series of can invade the entire brain (gliomatosis
mutations necessary to allow astrocytoma cells cerebri) without any focal regions of obvious
to invade surrounding normal brain are tumor apparent on neuroimaging. The second
discussed in Chapter 2 (see Table 2.2). Several major factor in glioma growth is angiogene-
factors apply primarily to glioma, the major sis, discussed in Chapter 2. Angiogenesis
invasive brain tumor: integrins, transmembrane supports tumor growth and facilitates clinical
glycoproteins that anchor extracellular matrix diagnosis.
components with the intracellular cytoskeleton, Both the tumor and clinician depend on
and transcriptional machinery are overex- angiogenesis. The tumor needs new blood
pressed in glioma cells. Interactions between vessels to nourish a mass beyond a few
integrins and extracellular matrix components, millimeters. The clinician relies on the fact that
including tenascin, fibronectin and osteopon- the neovasculature induced by the tumor is
tin, are necessary for invasion to occur. CD44, leaky, lacking an intact bloodbrain barrier
a glycoprotein receptor for several extracellu- and leading to contrast enhancement on the
lar matrix components, including hyaluronic MR scan.
acid, is expressed weakly in astrocytomas but
more strongly expressed in anaplastic astrocy-
Clinical findings
tomas and GBM. This receptor mediates tissue
invasion by glioma cells. Adhesion molecules Symptoms and signs
such as neural cell adhesion molecules (N- The symptoms and signs, as well as the
CAM)30 promote cellcell interaction; glioma diagnostic evaluation of diffuse astrocytomas,
cells lacking N-CAM loosen that interaction, are, for the most part, described in Chapter 3.

157
GLIAL TUMORS

Some distinctions will be highlighted here. preting the MR scan in diffuse astrocytomas
Astrocytomas are more likely than their higher- may arise in older patients who have multiple
grade counterparts to present with seizures, areas of white matter hyperintensity related to
either focal or generalized.31 Occasional vascular disease. One additional area of hyper-
seizures over many months or years, without intensity might be disregarded unless contrast
the development of other neurologic is given to reveal enhancement. However, the
symptoms, occur with astrocytomas and even hyperintensity from tumor is usually immedi-
more frequently with oligodendrogliomas (see ately subcortical and more diffuse than most
below). If seizures are the presenting symptom periventricular white matter hyperintense foci
of an anaplastic astrocytoma, they are gener- seen with vascular disease.
ally soon followed by other neurologic
dysfunction. Higher-grade lesions are more Positron emission tomography
likely to present with focal neurologic PET scans are helpful in some patients with
symptoms such as memory loss, personality putative low-grade diffuse astrocytomas prior
change, contralateral motor or sensory to biopsy. The PET scan is done with
symptoms, and visual field deficits. fluorodeoxyglucose to determine glucose
metabolic rate. If the PET scan is
Imaging hypometabolic in a patient with a non-enhanc-
In general, an infiltrative non-enhancing lesion, ing lesion who suffers only seizures control-
hyperintense on T2, connotes a low-grade lable by anticonvulsants, and has no other
glioma, whereas an enhancing lesion connotes neurologic symptoms or signs, we may elect to
a high-grade lesion. A large enhancing lesion follow that patient rather than biopsy or treat
that shifts the midline indicates a poor progno- immediately (see below). If a generally
sis.32 A normal MR scan usually excludes a hypometabolic PET scan shows an area of
diffuse astrocytoma as the cause of a patients hypermetabolism suggesting a focus of high-
symptoms, and no further workup for brain grade tumor, the neurosurgeon can direct his
tumor is required. However, we have encoun- stereotactic needle biopsy to that area, because
tered one exception to the above rule a treatment is determined by the highest grade
patient who had an entirely normal MR scan in a heterogeneous tumor. Amino acid PET
after a generalized convulsion, but several imaging, usually with methionine, can charac-
months later, after another seizure, was found terize low vs. high grade glioma with greater
to have a glioblastoma multiforme (Fig. 5.4). accuracy than fluorodeoxyglucose PET, but it
Others have encountered similar situations.33 In is currently limited to a research setting. PET
all other instances where reportedly normal scanning has also been used to distinguish
scans were found in patients with neurologic tumor recurrence from radiation necrosis,35 as
symptoms due to diffuse astrocytoma, a careful have SPECT scans. Thallium SPECT scanning
review of the scans identified either an inade- has been reported to demonstrate response to
quate study or hyperintensity on the chemotherapy better than does MRI.36
T2-weighted image that eventually became an
obvious tumor. To complicate the situation, Magnetic resonance spectroscopy
transient MRI abnormalities occasionally are Recent evidence suggests that this technique
caused by long-lasting seizures in the absence may have the capacity to distinguish diffuse
of a structural defect.34 Difficulties in inter- astrocytomas from other intrinsic tumors of the

158
DIFFUSE GLIAL TUMORS

brain and may play a role in establishing astro- Biopsy


cytoma grade non-invasively.37 The data are
still preliminary and the technique should be In most instances, if a suspected diffuse astro-
considered experimental. cytoma is encountered on MR scan,
craniotomy and removal of as much tumor as
Functional magnetic resonance imaging and is technically feasible is the best diagnostic and
cortical mapping therapeutic approach. In some instances,
As indicated in Chapter 3, fMRI prior to usually because the tumor is located in a criti-
surgery and cortical mapping during surgery cal area, surgical resection is not feasible. In
have important roles to play in the treatment that case, a stereotactic needle biopsy should
of diffuse astrocytomas. Functional magnetic be performed to establish the diagnosis before
imaging can identify not only sensory and beginning definitive treatment. It is important
motor areas, but also language areas of brain not only to establish that the abnormality on
that cannot be identified at neurosurgery the MR scan is indeed a diffuse astrocytoma,
unless the patient is operated on under local but also to grade the tumor, because grading
anesthesia. Mapping during the course of determines treatment. The limitations and
surgery can identify sensory and motor areas, complications of stereotactic biopsy were
even under general anesthesia. These tests are discussed in Chapter 3.
particularly important in patients with diffuse
astrocytomas because of their infiltrative
Differential diagnosis
nature. By delineating the exact location of
these critical areas, these tests enable the Table 5.3 lists some neoplastic and non-
surgeon to plan his approach appropriately. neoplastic lesions that are sometimes mistaken
Many patients with diffuse astrocytomas for astrocytic tumors by MRI and occasionally
suffering from focal neurologic symptoms even by biopsy. The neoplasms include glial
improve after compression of an eloquent area and non-glial tumors. For most glial tumors,
is relieved without the area itself being the exact histologic diagnosis is less important
violated. than the tumor grade; however, distinguishing

Table 5.3
Neoplastic lesions Non-neoplastic lesions Differential diagnosis
of diffuse astrocytoma.
Other glial tumors Cerebral infarction
Oligodendrogliomas Cerebral hemorrhage
Ependymoma Multiple sclerosis/Demyelinating pseudotumor
Focal astrocytic tumor Herpes simplex encephalitis
Brain abscess
Non-glial tumors
Lymphoma
Metastases
Neuronal tumors
Others

159
GLIAL TUMORS

astrocytic from oligodendroglial tumors can before hemorrhage occurred. This is apparent
affect treatment (see below). on the initial imaging procedure whereas
Other glial tumors are discussed later in this edema usually takes several days to develop
chapter. Non-glial tumors may be difficult or around a primary intracerebral hemorrhage.
impossible to distinguish from glial tumors on Thus, imaging can often suggest the presence
imaging study. Biopsy is essential to establish of an underlying tumor even when hemorrhage
the correct diagnosis and treatment. is the presentation.
A major problem is distinguishing Focal seizures can have many causes other
cerebrovascular disease, either infarction or than brain tumor. However, unlike short-lived
hemorrhage, from tumor. When an older focal seizures occurring with other epileptic
patient presents with a seizure or sudden lesions, the episodes may last many minutes to
neurologic defect, the physician usually hours and are often associated with abnormal-
suspects vascular disease, particularly if the ities of cognition and behavior. Focal seizures
patient recovers rapidly. Patients with diffuse themselves can alter brain images. One report
astrocytomas often present with transient describes enhancement and hypermetabolism
symptoms, and when they are elderly, on PET scan following multiple focal seizures
cerebrovascular disease must be a leading in a patient in clinical remission from a brain
diagnostic consideration. A CT scan without tumor. The image normalized after the seizures
contrast may show an area of hypodensity were controlled.38
that is interpreted as cerebral infarction. A non-neoplastic lesion that can mimic
Cerebral infarcts give their abnormal signal in tumor is demyelination (Fig. 5.5). Masdeu
a vascular distribution and often are triangu- and colleagues have pointed out that one
lar, with the base in the cortical gray matter radiographic distinction between demyelinat-
and the apex deep. Tumors are rounder, ing pseudotumor and GBM is that the
primarily involve white matter and do not contrast-enhancing ring is incomplete in
necessarily respect a vascular territory. When demyelinating disease, frequently where the
the cortex is involved, the gyri are expanded. lesion is adjacent to the ventricle, but
On diffusion MR scan, an infarct is hyperin- complete in GBM.39 MRS may also aid in the
tense and a tumor usually hypo- or isointense. distinction,40 but the reliability of this
A contrast-enhanced MR scan often estab- technique is unknown. Generally, the lesions
lishes the diagnosis but, too often, the patient are not easily distinguished on scan, and
is assumed to have vascular disease and no biopsy is usually necessary to establish the
further work-up is initiated beyond the initial correct diagnosis. It is the obligation of the
CT scan. neurologist and neurosurgeon to call the
Cerebral hemorrhage may announce the neuropathologists attention to the possibility
presence of a tumor. Unlike other hemorrhages, of demyelinating disease, because the
those into tumor often show enhancement of macrophages in a demyelinating lesion may be
the underlying tumor immediately after the interpreted as tumor cells, with the patient
ictus; enhancement of a hematoma can take unnecessarily subjected to major resection and
days to weeks to develop. Even without RT. RT appears to be particularly damaging
enhancement, tumors will demonstrate T2 in these patients41 and the distinction, there-
hyperintensity around the hemorrhage, often fore, is vital. Usually demyelinating pseudo-
outlining peritumoral edema that was present tumor is an isolated event and a minority of

160
DIFFUSE GLIAL TUMORS

Figure 5.5
Differential diagnosis of glioblastoma multiforme. The axial scan shows a ring-enhancing mass surrounded by
edema, suggesting a neoplasm. Note that the ring is incomplete, which might give a clue to a demyelinating
lesion (Chapter 3). A myelin stain of the lesion reveals a sharp border between the demyelinated lesion (left
half) containing many macrophages, and the adjacent brain with preserved myelin (right half).

patients go on to develop multiple sclerosis. Astrocytoma


Most patients recover and do well. Other The best treatment for astrocytomas has not
inflammatory lesions (e.g. sarcoid) may also been established.42 Surgery establishes the
mimic tumor and require biopsy. diagnosis and often relieves neurologic
Herpes simplex encephalitis usually symptoms but whether it prolongs survival is
presents acutely with headache, seizures and controversial.43 Older patients (> 40 years) with
neurologic dysfunction suggesting a temporal tumors >3 cm and with mass effect do appear
lobe lesion. Brain tumors may present in the to benefit from resection. Surgery seems to
same way. Each year we encounter at least 2 improve both quality and duration of life in
patients who recover from encephalitis but children with astrocytomas, both low- and high-
then relapse and are discovered to have a grade44 and we believe that it also does so in
diffuse astrocytoma. adults. However, surgery does not cure (pilocytic
astrocytoma is an exception see below);
surgery conventionally has been followed by RT.
Treatment
This approach remains valid for patients with
The treatment of diffuse astrocytomas depends progressive neurologic signs or intractable
on the grade, and in the paragraphs below is seizures. However, for asymptomatic patients,
divided by both grade and specific tumor type. including those with controlled seizures,

161
GLIAL TUMORS

immediate postoperative radiation therapy tumor. Patients who undergo a gross total
improves progression-free survival but not resection of a low-grade tumor are not
overall duration of survival when compared cured, but survival appears to be longer
with radiation therapy delivered after the patient and quality of life better than in those who
has developed clinical or radiographic progres- do not have resection.
sion.45 Furthermore, radiation causes dose- Patients whose tumors involve critical brain
related cognitive abnormalities and diminishes structures, e.g. language cortex, and who are
quality of life.42,46 A PET study indicated that asymptomatic save for focal seizures, partic-
RT has no effect on either methionine or glucose ularly if the PET scan is hypometabolic, can
uptake in the tumor area compared to patients be followed without treatment (an area of
not irradiated.47 However, although the normal hypermetabolism on the PET scan is an
brain areas of patients with astrocytomas who indication for biopsy and perhaps treatment
did not receive radiotherapy have lower glucose as indicated below for anaplastic tumors).
uptake than normal individuals, the glucose Some patients can be followed for many
uptake in normal brain areas was even lower in years without substantial growth of the
patients who received radiation therapy,48 lesion. At the time of clinical or radiographic
indicating radiation damage to normal brain. progression, biopsy should be performed to
When used, the best dose of radiation has ascertain whether the tumor is an astrocy-
not been established. An EORTC study toma, oligodendroglioma or something else,
comparing 45 Gy in 5 weeks with 59.4 Gy in and to determine grade. Treatment would
6.5 weeks revealed no difference in survival, then be dictated by histology.
but patients who received the higher dose Symptomatic patients should undergo
radiation tended to report lower levels of resection to the maximum extent feasible,
function and more symptom burden follow- or a stereotactic needle biopsy should be
ing completion of radiotherapy. The group performed when resection cannot be done.
differences were statistically significant for After a diagnosis is established, RT should
fatigue/malaise and insomnia immediately after be delivered to a limited field encompass-
RT, and emotional functioning at 715 months ing the visible tumor on MR scan and a
after treatment.49 Low-grade gliomas do 2 cm margin. Conventional treatment is
respond to RT with a greater than 50% delivered in fractions no larger than 1.8 Gy
decrease in tumor area on scan, but there is no to a total dose of 54 Gy. Lower doses might
statistically significant association between be equally effective (or ineffective).49 There
response as measured on scan, symptomatol- is no evidence at the present time that
ogy and progression-free survival.50 radiosurgery, brachytherapy or radiation
Each patients treatment must be individual- sensitizers have any role to play.
ized but we adhere to the following general
rules and recommendations: Anaplastic astrocytomas and glioblastoma
multiforme
If the tumor is in a relatively silent area of In most treatment series, anaplastic astrocytomas
brain (e.g. right frontal lobe), whether or (anaplasia comes from the Greek words plasso,
not it is causing neurologic symptoms and meaning to form, and ana, meaning repeat or
signs other than controllable seizures, an again; anaplasia is generally taken to mean
attempt should be made to remove the growth without appropriate form or structure)

162
DIFFUSE GLIAL TUMORS

and GBMs (glioblastoma from the Greek glia for reports describe benefit, there is at the present
glue, blastos for germ or sprout and oma for time no firm evidence that hyperfractionation,
tumor; multiforme from the Latin for polymor- radiosurgery52 or brachytherapy53 are better than
phic), also called astrocytomas grade III and IV standard RT, even when the total dose has been
by WHO and St. Anne-Mayo criteria, are usually escalated as high as 120 Gy with brachytherapy.
lumped together under the term malignant Accelerated hyperfractionation radiotherapy
glioma or malignant astrocytoma. However, (70 Gy in 44 fractions given twice daily) is
these two tumors have different prognoses (see reported to be comparable to but not better than
below), and if a clinical trial is heavily weighted standard RT.54 Re-irradiation with lower doses
with one or the other, one may observe different (34.5 Gy in 23 fractions), to treat recurrences
survival rates that are not due to treatment. that occur after initial RT and chemotherapy,
The conventional treatment of anaplastic may improve survival with little toxicity.55
astrocytoma is removal of as much of the tumor Elderly patients (> 70 years) treated with
as is surgically feasible, delivery of RT to a surgery and radiation therapy survive longer
limited field encompassing the tumor and a than those treated with less aggressive
2.53 cm margin to a dose of 59.4 Gy in 1.8-Gy regimes,56 but the prognosis is still poor. Some
fractions, and chemotherapy following the irradi- recommend reducing the dose and duration of
ation. Most studies suggest that extensive surgery radiation therapy to those over 70 (45 Gy/25
increases both duration and quality of survival. fractions).57 Some older patients in poor neuro-
RT also improves quality and duration of life. logical condition, who have extensive GBM
There is a correlation between the amount of achieve maximum palliation with a short
radiation and the duration of survival. However, course of RT, so that the decision to use a short
the maximum feasible dose appears to be the course of RT is based on the clinical situation.
equivalent of 60 Gy given in 1.82.0 Gy Two conventional chemotherapeutic regimens
fractions to a limited field. After three-dimen- are carmustine (BCNU) at a dose of 200 mg/m2
sional conformal radiotherapy to a dose of every 8 weeks to a total dose of 1500 mg/m2,
7080 Gy, recurrences still occur primarily and procarbazine, vincristine and lomustine
within the irradiated field,51 and the increased (PCV), as indicated in Table 5.4. The two are
dose does not improve survival. Although a few equally effective,58 but and because PCV is more

Table 5.4
Chemotherapy of anaplastic gliomas* and oligodendrogliomas.

Drug Dose Route Schedule

Intensivea Standardb

Carmustine (BCNU) 130 mg/m2 110 mg/m2 PO Day 1


Vincristinea 1.4 mg/m2 b 1.4 mg/m2b IV Days 8 and 28
Procarbazine 75 mg/m2/day 60 mg/m2/day PO Days 821

*Standard dose chemotherapy is used for anaplastic gliomas


aIntensive cycle is 6 weeks, standard is 8 weeks

b2 mg limit in standard dose, no limit in intensive dose

163
GLIAL TUMORS

toxic, we generally prefer BCNU as a single survival. There are some data to demonstrate
agent. Although a number of phase II trials have that pre-RT chemotherapy may be harmful,
shown that other chemotherapeutic agents are with a high rate of rapid tumor regrowth
modestly effective,59 none are superior to BCNU frequently necessitating reoperation before RT
or PCV. Increasing the dose of PCV with stem can be administered, and occasionally early
cell support increases toxicity but not efficacy.60 death. We have also had the occasional experi-
BCNU does not substantially increase the ence of profound unexpected myelosuppression
median survival, but in 2030% of patients it when BCNU was administered concurrently
prolongs survival. Good prognostic factors (e.g. with RT. Therefore, we usually start chemother-
favorable histology) do not predict benefit from apy after completion of RT.
adjuvant chemotherapy and benefit has been Inevitably, patients with anaplastic astrocy-
demonstrated for patients up to age 65.61 toma and glioblastoma multiforme relapse after
There is no evidence to indicate that treatment (Fig. 5.6). In many circumstances, a
chemotherapy given prior to RT or concomitant second surgical procedure will improve
with RT promotes either quality or duration of symptoms and increase survival for about 6

Figure 5.6
Response of a glioblastoma to radiation therapy but with recurrence outside the radiation portal. An elderly
patient presented with language difficulty and at surgery was found to have a glioblastoma multiforme. The
lesion was partially resected. The patient then received 60 Gy to the area outlined in black on the left panel.
One year later, the patient began to complain of right-sided weakness and a repeat scan revealed that the tumor
in the irradiated area had not grown but that a new lesion had appeared outside of the radiation portal (arrow).

164
DIFFUSE GLIAL TUMORS

months. As indicated above, re-irradiation at multiforme (Grade IV) is approximately one


relapse is feasible and stereotactic RT, year, with 1520% of patients surviving more
brachytherapy, other chemotherapeutic agents than 2 years but fewer than 5% more than 3
and experimental therapeutic techniques can also years.69 There are rare long-term survivors,
be considered at that point. One study indicated particularly among the young. Gliosarcomas
that BCNU wafers implanted into the tumor bed have a prognosis similar to GBM.70 Young age
at relapse may increase survival, but the study is the most important favorable prognostic
compared placebo wafers with a BCNU wafer, factor.71 Other favorable factors include perfor-
not with intravenous BCNU.62 Implantation of mance status, histology (anaplastic astrocy-
wafers may be associated with a higher risk of toma vs GBM), extent of surgery and adjuvant
postoperative complications, particularly infec- chemotherapy.69 A serum albumin level
tion and poor wound healing.63 One study < 3.4 mg/dl predicts a poor prognosis;72 the
suggests that a combination of interstitial and low levels are probably part of an acute-phase
systemic chemotherapy may promote survival.64 response regulated by cytokines that also
Temozolomide (200 mg/m2 daily for 5 days induce angiogenesis.72 In anaplastic astrocy-
followed by 23 days rest) is now the most used tomas (Grade III), the median survival is
second-line agent if the patient has originally approximately 3 years, with 1015% surviving
been treated with a nitrosourea or PCV. beyond 5 years. Diffuse astrocytomas have a
Temozolomide is given orally and has few side- wide range of behavior, but the median survival
effects, so that many physicians use it in prefer- is 58 years, with 1520% surviving 10 years.
ence to BCNU as initial chemotherapy although Good prognostic factors include young age,
data regarding its use in the adjuvant setting is good performance status and no enhancement
not yet available. A number of other chemother- on MRI.73 Prognosis for these tumors has not
apeutic agents have been reported in phase II changed substantially for the past 2030 years;
trials to have moderate activity, including topote- clearly, new therapies are required.
can, carboplatin and cisplatin (Table 4.12), but
none induce prolonged remission. Intrathecal
Gliomatosis cerebri
thiotepa may help some patients with ependymal
or leptomeningeal spread of tumor.65 Diffuse astrocytomas sometimes infiltrate the
Tamoxifen in high doses (80 mg/m2 or more) brain so widely that they involve the entire
inhibits protein kinase C (PKC). It has little brain with or without an identifiable focal
neurotoxicity, although dizziness and ataxia lesion. Such diffuse infiltration is called
may occur, but the drug has little or no gliomatosis cerebri (Fig. 2.2). Pathologically,
efficacy.66 At the present time, newer forms of the disorder is an astrocytoma that may vary
treatment, including immunotherapy, radio- from low to high grade and may differ in grade
active antibodies,67 gene therapy,68 antiangio- from area to area. Rarely, an oligodendro-
genesis therapy, and antisense therapy, have not glioma may present as gliomatosis cerebri.74
been shown to be efficacious. The symptoms may be either generalized (i.e.
headaches, papilledema, cognitive changes) or
focal, including corticospinal tract abnormali-
Prognosis
ties and seizures. Some patients present with
Table 1.11 lists the prognosis in diffuse astro- brainstem or even spinal cord signs, as the
cytoma. The median survival for glioblastoma entire neuraxis may be infiltrated occasionally.

165
GLIAL TUMORS

The MR scan is usually characterized by hyper- signs. Hydrocephalus is uncommon. The


intensity on the T2 image, either diffusely or diagnosis is made by MR scan. The tumors
multifocally. On some occasions, one or more look histologically identical to astrocytomas
areas of contrast enhancement are present in other areas of the brain. Focal brainstem
within the diffuse or multifocal non-enhancing lesions, less than 2 cm and often dorsally
areas. On rare occasions, there is no abnor- exophytic, are more likely to be pilocytic
mality of signal intensity but simply diffuse astrocytomas or gangliogliomas, and open
enlargement of the brain leading to smaller biopsy is safe. Some neurosurgeons recom-
ventricles and sulci than would be expected for mend stereotactic needle biopsy even when
the patients age. When gliomatosis cerebri is the tumor remains within the confines of the
suspected, a stereotactic needle biopsy will brainstem, but others believe that the compli-
establish the diagnosis. The needle biopsy cation rate is too high and that MR scan is
should be directed toward the most abnormal sufficiently characteristic to establish the
area of the brain. The prognosis is poor; more diagnosis. Some neurosurgeons recommend
than half the patients die within a year of partial excision if tumors are exophytic, but
symptom onset and less than a quarter survive the diffuse lesions widely infiltrate the brain-
more than 3 years. stem and cannot be approached surgically. For
Patients with gliomatosis cerebri are treated most brainstem gliomas, focal RT is the treat-
with RT.75 Unlike focal anaplastic astrocytomas ment of choice. Hyperfractionated radiother-
or glioblastoma, the radiation is delivered to apy is not more effective than conventional
the whole brain to a total dose of 60 Gy. In radiotherapy.77 Adjuvant chemotherapy, even
patients with biopsy or scan evidence of when given in high doses, has not been effec-
anaplasia, adjuvant chemotherapy with BCNU tive, although studies to find effective agents
can be given after the radiation, although its continue.78 The prognosis is better in adults
efficacy is not known. than it is for children, with 45% of adults
living longer than 5 years in our experience.79
Tumors that arise in the midbrain or medulla,
Brainstem glioma
focal or exophytic tumors of the pons and
Gliomas arising in the brainstem are hetero- tumors that are amenable to subtotal resec-
geneous76 (Fig. 5.7). They have been catego- tion have a better prognosis than the usual
rized in three ways: diffuse and focal, by diffuse pontine tumor.76 Brainstem gliomas
location, i.e. pontine, midbrain and associated with neurofibromatosis type 1
medullary, and subdivided into cystic or usually have a good prognosis and often do
exophytic lesions. Each has a different biology not require treatment80 (see Chapter 12).
and prognosis. The most common of the A peculiarly low-grade tumor sometimes
brainstem gliomas is the diffuse pontine astro- affects the midbrain. These tumors have
cytoma that usually occurs in childhood. variously been called tectal gliomas or pencil
These tumors are astrocytic and may be low gliomas of the aqueduct, depending on their
or high grade. Tumors that occur in adult- appearance on MR scan. They present in
hood differ little in signs or symptoms from adolescence or early adulthood with hydro-
those in children, usually beginning with cephalus caused by compression of the
cranial nerve palsies, especially diplopia, and aqueduct, the symptoms of which can be
subsequently evolving to cause long tract relieved by shunting of the cerebral ventricles.

166
DIFFUSE GLIAL TUMORS

Figure 5.7
A brainstem glioma in a young woman. This patient originally presented with facial myokymia and mild facial
weakness. The MRI revealed an infiltrating tumor in the brainstem (see also Fig. 3.1C). She was lost to follow-
up for approximately a year. She returned with increasing facial weakness. The scan at that time showed
diffuse hyperintensity on the T2-weighted image of the entire pons (left upper) with some contrast enhancement
(left middle). Six months after radiation, she was clinically stable and the scan showed some decrease in the
size of the T2-weighted lesion (upper right). Four years later she continued to have mild facial weakness but
the tumor on scan had largely resolved. No biopsy was performed.

Many such tumors appear to require no Subependymal giant cell


additional treatment, as they do not grow, and astrocytoma
as long as the hydrocephalus is relieved, the
patient remains asymptomatic, often for many This low-grade tumor typically arises as a
years. If there is evidence of tumor growth on single or multiple lesions lining the wall(s) of
scan or if symptoms of brainstem dysfunction the lateral ventricles.81 The tumor is believed to
develop, RT is the treatment. arise from subependymal stem cells, giving it a

167
GLIAL TUMORS

mixed neuronal and glial phenotype. The Oligodendroglial tumors


tumor usually occurs in patients suffering
from tuberous sclerosis and generally affects Introduction
children, although infants and adults have also Oligodendroglial tumors (from the Greek
been reported. Interestingly, although tuberin, words oligo meaning few, dendro meaning
the protein product of the tuberous sclerosis tree and glia meaning glue, meaning few
gene, is expressed in tubers, it is not expressed dendritic processes when compared with astro-
in these tumors.82 Their characteristic site cyts) are believed to arise from the oligoden-
within the walls of the ventricle, the calcifica- drocyte by dedifferentiation or from neoplastic
tion seen on CT scan and the stigmata of transformation of a progenitor such as the O-
tuberous sclerosis (see Chapter 12) help estab- 2A cell,83 a precursor of oligodendrocytes and
lish the diagnosis. Surgical treatment is neces- type II astrocytes. The tumors occur both in
sary only in the rare instance when the tumor pure form and mixed with neoplastic cells that
obstructs the ventricular system, causing hydro- appear astrocytic (oligoastrocytoma). Like
cephalus. Malignant degeneration is extremely diffuse astrocytomas, many tumors progress
rare. from low grade to a higher grade (Fig. 5.8).

(a) (b)

Figure 5.8
The evolution of oligodendrogliomas as indicated in
Table 5.5. Oligodendrogliomas can evolve from non-
neoplastic precursors and low-grade tumors,
anaplastic oligodendrogliomas to glioblastomas
multiforme. This figure illustrates the changes in that
sequence. (a) Uniform round nuclei with surrounding
clear halo, the so-called fried egg appearance of a
typical low-grade oligodendroglioma. (b) More
cellular tumor with brisk mitoses and atypical cells
with eosinophilic cytoplasm. (c) The oligodendrocytic
phenotype is lost and anaplastic glial forms,
(c) including giant cells, appear.

168
DIFFUSE GLIAL TUMORS

Because they are less common than astrocy- Etiology


tomas, and because of the difficulty of defini- Risk factors. There are no established risk
tive diagnosis, many aspects of the tumor are factors for the development of oligodendro-
poorly understood. For example, no unequiv- gliomas. Similar tumors can be produced in
ocal marker identifies a tumor as being oligo- experimental animals by ethylnitrosourea and
dendroglial in origin. About 50% of oligos methylnitrosourea, but chemical carcinogenesis
that are otherwise typical express glial fibril- is not established in humans. Occasional
lary acidic proteins (GFAP), usually considered families have oligodendrogliomas in more than
an astrocytic marker. In many tumors, micro- one member, but this finding could be a coinci-
gemistocytes are prominent, often leading to dence and its meaning is unclear.
an incorrect diagnosis of a gemistocytic astro-
cytoma (an astrocytoma variant, believed to Genetics
have a worse prognosis than other astrocy- Table 5.5 outlines the genetic changes thought
tomas of the same grade). In addition, the to be associated with oligodendrogliomas. The
relationship between histopathology and most important alteration is loss of hetero-
prognosis is less clear when oligoden- zygosity on the long arm of chromosome 19
drogliomas are compared with astrocytomas. and the short arm of chromosome 1. Up to
In past years, differentiating between an oligo- 80% of patients with oligodendrogliomas have
dendroglioma and an astrocytoma was the 19q deletion, representing an as yet uniden-
unimportant therapeutically, and many pathol- tified tumor suppressor gene that resides
ogists paid little attention to distinguishing between 19q 13.2 and 19q 13.4. The deletion
these tumors. Because of the recent evidence
that oligodendrogliomas are much more
chemosensitive than astrocytomas (see below), Table 5.5
more attention has been paid to the histologic Genetic alteration in oligodendroglial tumors.
differentiation; consequently, the apparent
frequency of diffuse oligodendrogliomas has Oligodendroglial cells or precursor (? 02A)
risen. In one series they represented slightly 19q LOH
under 5% of all primary brain tumors. A 1p LOH
recent study attempting to define the histologic 4q LOH
EGFR overexpression
criteria indicated that about 25% of gliomas
PDGF/PDGFR overexpression
are of oligodendroglial origin.84 Genetic Oligodendroglioma
studies (see below) may settle the issue. 9p LOH
Furthermore, the majority of oligos (approxi- 10p LOH
mately 90%) are low-grade at diagnosis, in CDCKN2A deletion
CDKN2C mutation/deletion
contrast to the astrocytic tumors that are more
VEGF overexpression
frequently malignant. TP53 mutation
Oligodendrogliomas usually occur in young P53 mutation (~10%)
individuals with a peak incidence at about age Anaplastic oligodendroglioma
30. The tumors are uncommon in childhood. ?
Unlike astrocytomas that are more common in Glioblastoma multiforme
men; oligodendrogliomas occur equally in men Modified from Kleihues and Cavenee.25
and women.

169
GLIAL TUMORS

Figure 5.9
Histologically-defined anaplastic oligodendrogliomas Response of anaplastic
oligodendrogliomas to chemotherapy
by genetic makeup. From 88 with
permission.
1p loss 1p intact

Group 1: Group 2: Group 3: Group 4:


combined, 1p loss TP53 no TP53
isolated other* mutation mutation
1p + 19q loss

# patients 23 5 8 13
age 43 51 30 52
response rate 17/17 (100%) 4/4 (100%) 2/6 (33%) 2/11 (18%)
duration response >31 months 11 months 7 months 5 months
survival from dx >123 months 71 months 71 months 16 months

on the short arm of chromosome 1 has been The swollen clear cytoplasm, surrounded by a
reported in up to 90% of oligoden- well-defined plasma membrane, has given the
drogliomas.85 Candidate genes on 1p include cells a so-called fried egg appearance, actually
the cell cycle regulator gene CDKN2C.86,87 Ino an artifact of fixation. Mitoses are rare. In the
and colleagues88 have identified 4 molecular background are delicate blood vessels, often
subtypes of anaplastic oligodendrogliomas. arranged in a chicken wire pattern. This
Those with isolated losses of 1p and 19q have delicate vasculature may account for the
a dramatic and prolonged (years) response to relatively high rate of hemorrhage even in low-
chemotherapy; those with 1p but not 19q grade oligodendrogliomas. Endothelial prolifer-
deletions respond but with shorter duration (1 ation is absent in the low-grade tumors. In
year); those without 1p deletions but with p53 anaplastic oligodendrogliomas, vascular prolif-
mutations may respond for a short time (7 eration, mitotic activity and necrosis may be
months); those with neither 1p nor p53 alter- seen. Calcifications are prominent. Table 5.6
ations do not respond (Fig. 5.9).88 details the histologic changes in oligodendro-
gliomas and their high-grade counterparts.
Pathology Although this four-tiered grading system has
Oligodendrogliomas are usually supratentorial, been developed, the only clinically useful classi-
often arising in the frontal lobe and involving fication of oligodendrogliomas is low-grade
the corpus callosum. Their gross appearance versus anaplastic. Thus, functionally, a two-
does not differ from that of astrocytomas, tiered system is used by most neuropatholo-
except that calcifications can occasionally be gists. The close relationship of cellularity,
identified in the gross specimen, a rarity in nuclear atypia and even mitotic figures to
astrocytomas. Microscopically, low-grade prognosis is less certain in oligodendroglial
oligodendrogliomas appear as a moderately tumors than in astrocytic tumors, but endo-
cellular tumor with spherical hyperchromatic thelial proliferation appears predictive of poor
nuclei surrounded by swollen clear cytoplasm. prognosis.89

170
DIFFUSE GLIAL TUMORS

Table 5.6 not relieved by psychotropic medication. When


Classification and grading of oligodendroglioma. she had a generalized convulsion, a scan
revealed a frontal tumor that proved to be an
Grade Grades based on progressive development oligodendroglioma. Because the tumor was not
resectable and the seizures were well
1 Hypercellularity, cellular anaplasia, and controlled, she was followed for several years
mitotic activity
without significant change in tumor size.
No pleomorphism
Mild hypercellularity Headaches are uncommon, because of the
Neuropil persists between cells slow-growing infiltrative nature of the tumor.
2 Moderate hypercellularity with loss of Because most of the tumors are in the frontal
neuropil between cells lobe, hemiparesis is relatively common and
Mild pleomorphism
focal parietal or occipital signs are less
3 Moderate hypercellularity and
pleomorphism common. The anaplastic oligodendroglioma
Mitoses present and usually frequent tends to manifest more lateralizing symptoms
4 Moderate hypercellularity with than the low-grade oligodendroglioma.
increasing pleomorphism and mitoses Seizures may be the presenting symptom, but
Microvascular proliferation they are often accompanied by hemiparesis,
Modified from Kleihuer and Cavenee.25 cognitive changes and hemisensory loss.

Imaging. Oligodendrogliomas cannot be distin-


guished from diffuse astrocytomas on routine
MR imaging. Characteristically, the low-grade
Clinical features tumor is hypointense on the T1-weighted
Symptoms and signs. Oligodendrogliomas image and hyperintense on the T2-weighted
grow more slowly than their astrocytic image. Most oligodendrogliomas do not
counterparts and thus are likely to cause contrast enhance, and most anaplastic oligo-
symptoms, particularly focal seizures, for a dendrogliomas do. Thus, the presence of
longer period of time before the diagnosis is contrast enhancement is a poor prognostic
made. Before the advent of MRI, many patients sign. If the tumor is heavily calcified, the
suffered focal seizures for years before a calcium can sometimes be identified on the MR
diagnosis was finally established. One of our scan as either hyper- or hypointensity; however,
patients had a 20-year history of focal seizures calcium is better seen on a CT scan. Our policy
beginning in the left foot prior to the estab- in patients with infiltrating gliomas identified
lishment of the diagnosis of what was still on MR scan who are not symptomatic, save
a low-grade oligodendroglioma. The tumor for seizures that are controlled with anticon-
subsequently became biologically more aggres- vulsants, is to perform a CT scan to look for
sive and was responsible for his death. The calcification, a fluorodeoxyglucose PET (recent
seizures are likely to be either focal motor or evidence suggests that methionine PET scans
have partial complex emotional or intellectual differentiate low-grade and high-grade tumors
components. One of our patients was treated better than glucose90) and an MRS. Low-grade
for many months at a university center for tumors are usually hypometabolic on PET scan
panic attacks that were episodes of sudden, and higher grade tumors are hypermetabolic.
overwhelming fear lasting 2030 seconds and MRS may help distinguish astrocytic tumors

171
GLIAL TUMORS

from oligodendroglial lesions as preliminary several months and, although the tumor is
data suggest different patterns. However, these still present, it is substantially smaller than
data are still early and their meaning is not yet when first treated and she is asymptomatic.
clear. Four to six cycles of chemotherapy are
given, usually with an MRI every other
Treatment. The same treatment considerations cycle to evaluate tumor size, and then the
that apply in low-grade astrocytomas also treatment is stopped. If successful, i.e. the
apply to oligodendrogliomas. There is even less tumor decreases in size and/or the patient
reason for treating asymptomatic oligoden- becomes asymptomatic; we then follow
drogliomas than for treating asymptomatic without further therapy until relapse.
astrocytomas, because the course is so much 3. If chemotherapy fails, limited-field radia-
longer and the patients usually present with tion to a dose of 54 Gy is delivered.
seizures but without other neurologic disabil- 4. Late recurrences are treated with surgery
ity. If a substantial resection can be safely done, where feasible, followed by RT if not previ-
that is the treatment of choice for low-grade ously administered and/or chemotherapy.
tumors. It ensures that the tumor is low-grade
and probably delays the onset of symptoms. For anaplastic oligodendrogliomas, the only
The following represents our therapeutic chemotherapy regimen that has been exten-
approach: sively tested is PCV94 (many physicians use
temozolomide up-front, but the only published
1. If the tumor is resectable, it should be data concern its use at recurrence95). The
resected. If the patient is asymptomatic tumors are treated with up-front chemotherapy
save for controllable seizures, no further as outlined in Table 5.4. Four to six cycles of
therapy should be administered and the chemotherapy, depending on the patients toler-
patient should be carefully followed. All ance, are delivered followed by limited-field
tumors resected or biopsied should radiation. One chemotherapy regimen is an
undergo chromosomal analysis.91 intensive version of PCV (I-PCV) with both an
2. If the tumor is not resectable and the increase in dose of all three drugs and a short-
patient is asymptomatic save for control- ened time between cycles from 8 to 6 weeks.
lable seizures, a PET scan, an MRS and a One cycle is defined as the period of therapy
CT scan should be done. If these all (i.e. 28 days) plus 2 weeks of observation for
indicate a low-grade tumor, and particu- a total of 42 days. It is unknown whether I-
larly if the tumor is calcified, no further PCV is better than standard PCV for malignant
therapy is carried out. If the tumor is oligodendrogliomas, but it is more toxic and
symptomatic, a biopsy, preferably with most patients have trouble tolerating it. We
debulking, is performed and the patient is prefer the standard-dose regimen.
treated with chemotherapy92 (see below). An experimental protocol is testing the role
Some, but not all, patients with low-grade of high-dose chemotherapy with stem cell
oligodendrogliomas respond to chemo- rescue without RT for newly diagnosed
therapy with shrinkage of the tumor and anaplastic oligodendrogliomas. This study will
resolution of symptoms.93 One of our explore the limits of chemosensitivity for
patients remains well 7 years post-treat- anaplastic oligodendrogliomas and defer RT to
ment. Her hemiparesis resolved over try and limit neurotoxicity. Recurrences after

172
FOCAL GLIAL TUMORS

Table 5.7
Low-grade High-grade Survival of
oligodendrogliomas by
Survival (%) Oligo Oligo-Astro Oligo Oligo-Astro grade and histologic type.
From Shaw et al.97
Median (year) 9.8 7.1 4.5 4.3
2-year 93 89 67 82
5-year 73 63 45 45
10-year 49 33 26 23
15-year 49 17 13 17

aggressive treatment of anaplastic oligoden- gliomas, and all of the long-term survival with
drogliomas are treated similarly to astrocy- chemotherapy was not explained by oligoden-
tomas. However, chemotherapeutic options at droglial tumors.
recurrence include melphalan, carboplatin,
cisplatin and etoposide and temozolomide.95
Oligoastrocytoma
Prognosis. The prognosis of oligoden- Tumors that contain significant elements of
drogliomas is much better than that of their both astrocytic and oligodendroglial tumor
astrocytic counterparts, with a median survival may account for 10% of diffuse gliomas.
of 16 years in our series;96 however, because of Unfortunately, no histologic markers unequiv-
the difficulty in accurate identification of these ocally distinguish tumor cells of astrocytic
tumors, their exact prognosis is not well estab- origin from those of oligodendroglial origin,
lished (Table 5.7, see also Table 1.11). making the diagnosis a subjective one about
Furthermore, genetic evidence suggests that which pathologists often disagree. However, a
only some tumors are chemosensitive and recent report suggests that a specific marker
prognosis varies accordingly (Fig. 5.9). may have been identified.98 Current evidence
Data from two randomized prospective suggests that these mixed tumors, when low
Brain Tumor Study Group (BTSG) protocols grade, respond to chemotherapy and RT but
were combined to analyze the effects of have a higher recurrence rate and shorter time
prognostic factors on survival by treatment to progression than do pure oligoden-
group. These studies assessed all malignant drogliomas. Current evidence suggests that 1p
gliomas without differentiation of astrocy- and 19q deletions do not predict response in
tomas from oligodendrogliomas. Adjuvant these mixed tumors.99
chemotherapy increased long-term survival
regardless of prognostic factors. Oligodendro-
gliomas were overrepresented among long-term
survivors, whether or not they were treated
Focal glial tumors
with chemotherapy, but the benefits of
Introduction
adjuvant chemotherapy were not restricted to
pathologically defined oligodendrogliomas.61 These tumors affect predominantly children
Thus, prognostic factors did not predict benefit and young adults and, taken together, represent
from adjuvant nitrosourea in malignant no more than a few per cent of adult brain

173
GLIAL TUMORS

Figure 5.10
A schematic sagittal section
illustrating the common location of
focal glial tumors. Focal glial tumors
include the pilocytic astrocytoma
commonly found in the cerebellum
(A), optic nerve, chiasm or
hypothalamus (B). Brainstem gliomas
E
are occasionally focal and exophytic
(C). Other focal gliomas include
ependymoma that often arise within
B A the 4th ventricle (D) and choroid
plexus papillomas (E).
C
D

tumors, although they represent a more Pilocytic astrocytoma


substantial percentage of childhood brain
tumors. They differ from diffuse astrocytomas Pilocytic astrocytomas are low-grade tumors of
in that they tend to be well-circumscribed and, astrocytic origin, corresponding to a WHO
if accessible, may be cured surgically (Fig. grade I astrocytoma (see Table 5.2). The tumor
5.10). Even though they are low-grade, is named for its bipolar fusiform cell processes
contrast enhancement tends to be the rule (piloid is from the Greek for hair). Pilocytic
rather than the exception. These tumors may astrocytoma is the most common glioma of
have a pleomorphic appearance histologically, children but also occurs in young adults; over
with mitoses and vascular proliferation which, 75% occur in those under age 20. These
when combined with contrast enhancement, tumors are found in the cerebellum (Fig.
often leads the physician to believe that one is 5.11),100 optic nerve or optic chiasm101 and
dealing with a high-grade neoplasm. A careful hypothalamus of children. They are sometimes
review by an expert neuropathologist is essen- found in the basal ganglia and in the brainstem
tial to establish the correct diagnosis. Most as a dorsally exophytic brainstem glioma. In
patients with focal gliomas should not be young adults, they are usually in the cerebral
treated with RT or chemotherapy immediately hemispheres and cerebellum (Fig. 5.11). They
after surgery, but should be followed clinically. constitute more than half of spinal astrocy-
Patients are often cured or have long remis- tomas, where they displace rather than invade
sions after surgery alone. Uncommonly, these tissue. About 15% of optic nerve pilocytic
generally low-grade tumors may recur aggres- astrocytomas are associated with neurofibro-
sively and even seed the leptomeninges. matosis-1 (NF-1).

174
FOCAL GLIAL TUMORS

Figure 5.11
A pilocytic astrocytoma of the brainstem (left) in an adult that has seeded the leptomeninges (right arrow) and
the ventricular ependymomas (right, arrowhead). The histology of leptomeningeal infiltration often remains
low-grade, as does the histology of the tumor. The photomicrograph shows hyperchromatic, pleomorphic glial
nuclei. Many Rosenthal fibers (arrows) are present.

Etiology pilocytic astrocytomas.103 A few sporadic


Pilocytic astrocytomas do not have a distinct pilocytic astrocytomas show loss of chromo-
cytogenetic pattern. Unlike diffuse astrocy- some 17q, including the region encoding the
tomas, p53 mutations are not common in NF-1 gene, but this is not the case in most
pilocytic astrocytomas. One study of 18 instances. In a cytogenetic study of 24 pilocytic
pilocytic astrocytomas (including one anaplastic astrocytomas, 12 had normal karyotypes. In the
tumor), 3 of which were NF-1 related, showed other 12, there were structural and/or numeri-
no p53 gene mutations,102 but a more recent cal abnormalities most frequently in chromo-
study demonstrated p53 mutations in 7 of 20 somes 7, 8 and 11; these genetic abnormalities

175
GLIAL TUMORS

may indicate tumor progression.104 Telomerase Clinical findings


activity, believed to occur early in glial tumori- The symptoms and signs of pilocytic astrocy-
genesis, is absent in pilocytic astrocytomas, toma depend on both the tumors location and
dysembryoplastic neuroepithelial tumors (see rate of growth. The typical cerebellar pilocytic
below) and pleomorphic xanthoastrocytomas astrocytoma is in the cerebellar hemisphere (in
(see below). With the exception of NF-1, there contradistinction to medulloblastomas, which
are no known risk factors for pilocytic astrocy- are usually in the vermis) and usually presents
tomas. with gait and ipsilateral limb ataxia. Later,
patients develop headache, nausea and vomit-
Pathology ing, diplopia from VI nerve palsy and often
Grossly, the tumors are usually well demar- papilledema. Hypothalamic pilocytic astro-
cated from surrounding normal tissue and are cytomas may present with cognitive and
often cystic. The typical tumor cell has long behavioral abnormalities or with endocrine
bipolar hair-like extensions that, taken dysfunction, especially diabetes insipidus, and
together, form a dense fibrillar background. occasionally precocious puberty. Optic nerve
The cells often contain Rosenthal fibers, a tumors present with visual field defects or
brightly eosinophilic intracytoplasmic mass, occasionally blindness. As with most brain
that helps establish the diagnosis. Microcysts tumors, symptoms are progressive, with one
are common. The tumor may show hyper- exception: a number of patients with optic
chromatism, pleomorphism and microvascular gliomas develop some visual loss and then
proliferation. In the absence of significant stabilize or improve without treatment for
mitotic activity these findings do not portend a many years.101
poor prognosis. The MIB-1 labeling index
ranges from 0% to 19% in pilocytic astro- Imaging. MR scans often show a combination
cytomas.105 Pilocytic astrocytomas with low of solid and cystic mass. Sometimes the solid
labeling indices do not appear to show progres- tumor, which usually contrast enhances, is
sion after partial resection, whereas those with small and presents as a mural nodule in a large
higher indices are more likely to progress.105 cyst, a finding characteristic of the cerebellar
Some tumors appear to be mixed with non- pilocytic astrocytoma. The tumor appears well
pilocytic components. Tumors with a demarcated and often lacks the surrounding
monomorphous pilomyxoid pattern are more edema of other neoplasms. When pilocytic
likely to recur than are more typical tumors.106 astrocytomas occur in the brainstem they tend
Although the vast majority of pilocytic astro- to be exophytic and also contrast enhancing,
cytomas are benign, some may seed the distinguishing them from the more common
leptomeninges107 or even spread distantly diffuse astrocytomas that affect the brainstem
within the CNS. Biopsy of a metastasis usually of children. The presence, absence, or degree of
reveals the same histology as the primary enhancement has no prognostic significance.
tumor, indicating that the biological behavior PET scanning with glucose or methionine
cannot always be predicted by the histologic tends to show more uptake in pilocytic astro-
appearance; the metastases may also behave in cytomas than the low-grade nature of the
a relatively indolent fashion, like the primary. tumor would suggest.109 Thus, PET scans must
Smear preparations may help in establishing be interpreted carefully in children and young
the diagnosis during surgery.108 adults with cystic tumors, recognizing that

176
FOCAL GLIAL TUMORS

hypermetabolic lesions may be biologically progressive symptoms. RT and fractionated


benign. MRS has shown a choline/N-acetyl stereotactic radiotherapy111 have been reported
aspartate ratio of 3.4, a lipid/choline ratio to have some effect. However, since many of
below 1, and a lactate peak.110 the patients are young, a number of investiga-
tors have attempted chemotherapy with a
Differential diagnosis. Although imaging variety of agents112 to delay RT. The usual
characteristics, especially a cystic tumor with chemotherapeutic regimen is carboplatin and
an enhancing nodule, and location can suggest vincristine,113 which appears to have some
the diagnosis, only biopsy will unequivocally efficacy, particularly in optic pathway and
establish it. Pilocytic astrocytomas must be hypothalamic tumors.
distinguished from other neoplasms that arise
in the same area, e.g. medulloblastoma in the Prognosis. Overall, the prognosis is usually
cerebellum, astrocytoma in the brainstem, excellent, with a 15-year survival of about
craniopharyngioma in the suprasellar area 80%.114 For cerebellar pilocytic astrocytomas,
and other glial tumors in the hemisphere. the overall 5-year survival rate is 93%, but is
Pilocytic astrocytomas must also be distin- 100% after complete resection. Younger age
guished from non-neoplastic lesions such as (< 14 years) and classic histology are good
eosinophilic granuloma in the suprasellar area prognostic factors.115 Those with complete
(Chapter 10). surgical resection do better than those with
partial resection, who in turn do better than
Treatment. Whenever possible, the treatment is those with biopsy. Brainstem involvement is a
surgical.44 Most pilocytic astrocytomas involv- negative prognostic factor.116 However, recur-
ing the cerebellum, and a few involving the rences do affect some patients, even after long
brainstem, can be successfully removed without quiescent intervals. One of our patients treated
the necessity for further treatment. Reports in childhood for a cerebellar astrocytoma
suggest that even partial removal of cerebellar relapsed after 33 years. The tumor at second
astrocytomas may yield prolonged remissions. surgery appeared identical to the initial tumor
Optic gliomas and hypothalamic pilocytic but kept recurring over several years and was
astrocytomas can sometimes be successfully ultimately responsible for the patients death,
debulked using microsurgical techniques, but although the histology remained low-grade. A
total removal is rarely possible, even though low MIB-1 labeling index is associated with a
the tumors are well circumscribed. Neverthe- reduced risk of recurrence, but a high labeling
less, after partial removal, symptoms improve index, e.g. > 5%, does not necessarily predict
in many patients and some have a prolonged recurrence.105 Leptomeningeal dissemination
remission. Accordingly, if one can remove has been reported and usually indicates aggres-
much or all of the tumor surgically, no further sive biological behavior with poor outcome
therapy should be instituted until there is even if the histology remains low-grade.107
evidence of progression. In some patients, only
a biopsy can be performed to establish the
Pleomorphic xanthoastrocytomas
diagnosis. If symptoms can be relieved by an
alternative procedure, such as ventriculoperi- This tumor is so named because it consists of a
toneal (VP) shunt, then definitive anti-tumor mixture of pleomorphic tumor cells, ranging
therapy is withheld until the patient develops from normal astrocytes to giant multinucleated

177
GLIAL TUMORS

Figure 5.12
A pleomorphic xanthoastrocytoma in
an 18-year-old man. This man
presented with increasing headache
and was found to have a large
contrast-enhancing cystic tumor. At
surgery, the tumor was originally
interpreted as glioblastoma
multiforme, but further review
suggested a pleomorphic
xanthoastrocytoma. He received no
further treatment and is free from
recurrence 5 years later. The tumor
cells exhibit pleomorphism with
scattered lipidized forms (arrow), and
a granular body (arrowhead).

cells.117 The latter contain lipid and they also Expert neurologic and neuropathologic exami-
express GFAP. The tumor occurs in children nation usually establishes the diagnosis, and no
and young adults and usually arises superficially further treatment after surgery is indicated,
in the temporal lobes, often extensively involv- other than careful follow-up. The tumors
ing the meninges.118 No specific cytogenetic or occasionally recur and sometimes seed the
molecular genetic abnormalities characterize leptomeninges, but these are the exceptions. A
this tumor. p53 missense mutations have been high mitotic index and atypical mitoses may
described in some patients, and EGFR amplifi- suggest recurrence.117 Recurrence-free survival
cation has also been described. rates are 72% at 5 years and 61% at 10 years.
Because of cortical involvement, patients Overall survival is 70% at 10 years.117
usually present with seizures that may be
present for many years before the diagnosis is
Ependymomas
made. On MR scan, the tumor appears as an
inhomogeneous mixed signal intensity mass Ependymomas are tumors that arise from the
that is well-circumscribed, often contrast ependymal cells that line the ventricular system
enhances and may be cystic (Fig. 5.12). (Fig. 5.13) and the central canal of the spinal
Surrounding edema is usually modest, as would cord.119,120 They account for about 10% of
befit a very slow-growing tumor. childhood intracranial tumors (30% of those
The treatment is surgical. Patients with arising in children younger than 3 years old)
complete resections recur less often than those and 5% of adult intracranial tumors. They are
with partial resections. The pleomorphic the most frequent neuroepithelial tumor of the
appearance of the tumor and its invasion of the spinal cord, accounting for over 50% of spinal
leptomeninges often suggest to the physician gliomas in children and adults. A slight prepon-
that he is dealing with a malignant tumor. derance of males is affected. The incidence is

178
FOCAL GLIAL TUMORS

Figure 5.13
An ependymoma of the fourth ventricle. This man presented with hydrocephalus, headache and ataxia in 1981. A
4th ventricular mass (black arrow) was discovered and resected. Because resection was incomplete, he received
radiation therapy. In 1992, a surveillance scan indicated recurrence of the tumor in the 5th ventricle. He received 2
years of carboplatin chemotherapy without a change in size of the tumor. It has remained unchanged for the
ensuing 3 years. Uniform tumor cells with round/oval nuclei and delicate processes form a perivascular
pseudorosette (white arrow).

bimodal, with the major peak at 5 years and a parenchyma of the hemisphere and may have
smaller peak at about 35 years. no obvious intraventricular component.
Ependymomas can arise anywhere ependy- Myxopapillary ependymomas are tumors of
mal cells are present. A particular site of the filum terminale of the spinal canal and are
predilection is the IVth ventricle, where they not discussed in this book.
grow within the ventricular system (Fig. 5.13). A number of cytogenetic abnormalities have
Supratentorial ependymomas also arise from been described in ependymomas, none charac-
ependymal structures but grow into the teristic.121 Loss of chromosome 22 is the most

179
GLIAL TUMORS

common. One report describes a 50% cephalus, probably by obstruction of CSF


incidence of allelic loss on the short arm of absorptive pathways due to a high CSF protein
chromosome 17 in pediatric ependymomas. concentration.125 Supratentorial tumors cause
p53 mutations are rare. p73 mRNA, which progressive focal neurologic symptoms,
shares functional and structural homology with depending on their location.
p53, is overexpressed in ependymomas.122 MR scan demonstrates a mass lesion that is
Histologically,123 ependymomas are charac- heterogeneously hypo- and isointense on T1
terized by perivascular pseudorosettes, i.e. and hyperintense on T2 and may or may not
tumor cells arranged radially around blood contrast enhance; edema may be prominent.
vessels, and ependymal or HomerWright Hemorrhage may be present. Calcification,
rosettes, i.e. columnar cells, which arrange seen best on CT scan, is present in about 15%
themselves around a central lumen. The of ependymomas. The differential diagnosis
tumors are GFAP positive. Histologic includes other tumors that can occur in the
subtypes of intracranial ependymoma include same area, e.g. medulloblastoma and brainstem
clear cell, cellular and papillary. The latter glioma.
may be mistaken for a choroid plexus papil- The initial treatment of ependymomas is
loma (see below). The diagnosis can be estab- surgical.44 A gross total resection prolongs
lished unequivocally in problematic cases by survival, but the location of the tumor, partic-
electron microscopy, where tumor cells show ularly when it involves structures at the floor
the elaborate cilia and microvilli of normal of the 4th ventricle, frequently makes that
ependyma. The tumors are generally low- impossible. Surgical resection should be
grade but may be aggressive and spread via followed by RT.126 Despite the fact that about
the cerebrospinal fluid (CSF) to seed the 10% of posterior fossa tumors eventually seed
meninges and even rarely metastasize to the leptomeninges, they usually do so in the
extraneural structures, primarily the lungs. setting of local recurrence, so that RT is
Many, but not all, studies have found histol- delivered to the site of the tumor and not to
ogy to be an unreliable prognostic factor, the whole neuraxis.127 If symptomatic lepto-
independent of tumor location, extent of meningeal seeding develops later, one can
resection and age at diagnosis. However, as prescribe additional RT. There is no role for
with many other glial tumors, mitotic figures, adjuvant chemotherapy at this time. Isolated
vascular proliferation and a high (> 4%) reports describe recurrent ependymomas
MIB-1 labeling index are encountered more responding to chemotherapy, particularly
often in those tumors destined to recur or carboplatin, and also several other chemother-
kill.124 apeutic agents. These reports make chemo-
Clinical findings depend on the site of the therapy worth trying in patients who have
tumor. Unlike brainstem gliomas, IVth ventric- failed surgery and radiation therapy, but
ular ependymomas cause hydrocephalus early response is usually poor.128 Young children
along with headache, dizziness or vertigo and, have been treated with chemotherapy initially
especially in children, nausea and vomiting as in order to delay RT129 and minimize neuro-
presenting complaints; ataxia and diplopia are toxicity.
also common. Spinal ependymomas may Unlike most glial tumors, the prognosis is
present in a similar fashion because otherwise worse in children than in adults, probably
asymptomatic tumors can cause hydro- because posterior fossa ependymomas are more

180
FOCAL GLIAL TUMORS

common in children and their IVth ventricular Choroid plexus tumors


location makes them difficult to resect; these
tumors often cause severe neurologic disability Choroid plexus papillomas and choroid plexus
leading to early death. However, the overall carcinomas are rare, accounting for less than
prognosis of intracranial ependymomas is 1% of all intracranial tumors.132,133 The tumors
better than that for most other glial tumors. originate from the epithelium of the choroid
Five-year survival rates are about 80%, and plexus of the cerebral ventricles (Fig. 5.14) and
10-year survival rates about 50%.119 Anaplastic typically occur in children. Choroid plexus
ependymomas may be invasive and more likely papilloma is the most common intracranial
to spread by CSF pathways than their lower- tumor in the first year of life. However, these
grade counterpart; however, it is not clear tumors also occur in adults. The most common
whether anaplastic features reliably confer a site in children is the lateral ventricle. In adults,
worse prognosis. Treatment does not differ for IVth ventricular tumors are more common.
an anaplastic ependymoma and for all patients, Some evidence suggests a possible role for
most eventually succumb to local recurrence simian virus-40 (SV-40) or a related DNA virus
rather than metastatic disease. in pathogenesis.134 p53 mutations have been
reported in a few tumors. The tumors are well-
demarcated intraventricular masses that may
Subependymoma be cystic and/or hemorrhagic. Microscopically,
Subependymomas are low-grade intraventricu- they resemble normal choroid plexus, but with
lar tumors, usually found in the IVth ventricle more crowded and elongated cells. Most
and, less frequently, in the lateral ventricle. choroid plexus tumors cause symptoms by
They may be multiple. Their cell of origin is hydrocephalus.
unknown. They were named because of their The hydrocephalus usually results from
histologic resemblance to areas of tumor obstruction of CSF pathways by the tumor
sometimes found in ependymomas. However, mass, but in children there may be an exces-
cytogenetic studies of subependymoma give a sive production of CSF that overwhelms spinal
normal karyotype.130 When they cause fluid absorptive pathways, leading to enlarge-
symptoms, they usually do so by ventricular ment of the ventricular system. Occasionally,
obstruction, but many are asymptomatic, choroid plexus tumors present with an intra-
encountered incidentally at autopsy, or when ventricular hemorrhage.
a scan is done. The tumors are iso- to The MR or CT scan shows an intensely
hypointense on T1 and may be heterogeneous contrast-enhancing lobulated intraventricular
due to calcification and/or hemorrhage. Some mass which may be calcified. The majority of
contrast enhance. Histologically, clusters of these tumors are histologically benign, although
GFAP- and S100-positive monomorphic cells even benign tumors may occasionally seed the
are found in a fibrillary background; sclerotic subarachnoid space. About 20% of tumors are
vessels and nuclear pleomorphism may be malignant (choroid plexus carcinoma); they
found, but mitotic figures, vascular prolifera- seed the leptomeninges along CSF pathways.
tion and necrosis are absent and the MIB-1 Choroid plexus tumors must be distinguished
labeling index is lower than that of other from other intraventricular tumors and cysts;
ependymal tumors.131 If symptomatic, surgery they must also be distinguished from metastatic
is curative. papillary carcinomas.

181
GLIAL TUMORS

Figure 5.14
A choroid plexus papilloma has seeded the leptomeninges. The scan on the left shows the contrast-enhancing
lesion involving the choroid plexus (arrow). The (middle) and (right) scans show infiltration of the
leptomeninges in the brainstem, cerebellum and cervical cord (arrows). Ribbons of regimented tumor cells with
uniform round nuclei and a cuboidal cytoplasm surround fibrovascular cores forming papillary structures.
Stromal concretions can be encountered (arrow).

The treatment of choroid plexus tumors is References


surgical. The 5-year survival rate after gross total 1. Linskey ME. Glial ontogeny and glial neopla-
removal is about 80%. Choroid plexus carcino- sia: the search for closure. J Neurooncol
mas should receive RT in the postoperative 1997; 34: 522.
period even if there has been a gross total resec- 2. Holland EC. Gliomagenesis: genetic alter-
tion.135 If there is cytologic or clinical evidence of ations and mouse models. Nat Rev Genet
2001; 2: 1209.
subarachnoid seeding, whole neuraxial radiation
3. Kettenmann H, Ransom BR. Neuroglia. New
is required.133 Chemotherapy, either postopera- York: OUP, 1999.
tively133 or preoperatively to reduce tumor 4. Kelly PJ, Daumas-Duport C, Scheithauer BW.
volume and vascularity136,137 may be helpful. Stereotactic histologic correlations of computed

182
REFERENCES

tomography and magnetic resonance imaging- 17. Adachi J, Ohbayashi K, Suzuki T, Sasaki T.
defined abnormalities in patients with glial Cell cycle arrest and astrocytic differentiation
neoplasms. Mayo Clin Proc 1987; 62: 4509. resulting from PTEN expression in glioma
5. Olney JW, Farber NB, Spitznagel E, Robins cells. J Neurosurg 1999; 91: 82230.
LN. Increasing brain tumor rates: is there a 18. Haas-Kogan DA, Kogan SS, Yount G et al.
link to aspartame? J Neuropathol Exp Neurol p53 function influences the effect of fraction-
1996; 55: 111523. ated radiotherapy on glioblastoma tumors.
6. Rodvall Y, Ahlbom A, Spannare B, Nise G. Int J Radiat Oncol Biol Phys 1999; 43:
Glioma and occupational exposure in 399403.
Sweden, a casecontrol study. Occup Environ 19. Ino Y, Silver JS, Blazejewski L et al. Common
Med 1996; 53: 52637. regions of deletion on chromosome
7. Hu J, La Vecchia C, Negri E et al. Diet and 22q12.3q13.1 and 22q13.2 in human astro-
brain cancer in adults: a case-control study in cytomas appear related to malignancy grade.
northeast China. Int J Cancer 1999; 81: 203. J Neuropathol Exp Neurol 1999; 58: 8815.
8. Malmer B, Iselius L, Holmberg E et al. 20. Forsyth PA, Wong H, Laing TD et al.
Genetic epidemiology of glioma. Br J Cancer Gelatinase-A (MMP-2), gelatinase-B (MMP-
2001; 84: 42934. 9) and membrane type matrix metallo-
9. Kleihues P, Ohgaki H. Genetics of glioma proteinase-1 (MT1-MMP) are involved in
progression and the definition of primary and different aspects of the pathophysiology of
secondary glioblastoma. Brain Pathol 1997; malignant gliomas. Br J Cancer 1999; 79:
7: 11316. 182835.
10. Shafqat S, Hedley-Whyte ET, Henson JW. 21. Nakada M, Kita D, Futami K et al. Roles of
Age-dependent rate of anaplastic transforma- membrane type 1 matrix metalloproteinase
tion in low-grade astrocytoma. Neurology and tissue inhibitor of metalloproteinases 2 in
1999; 52: 8679. invasion and dissemination of human malig-
11. Zhou XP, Li YJ, Hoang-Xuan K et al. nant glioma. J Neurosurg 2001; 94: 46473.
Mutational analysis of the PTEN gene in 22. Nakatani K, Yoshimi N, Mori H et al. The
gliomas: molecular and pathological correla- significance of the expression of tumor
tions. Int J Cancer 1999; 84: 1504. suppressor gene DCC in human gliomas. J
12. Cheng Y, Ng HK, Ding M et al. Molecular Neurooncol 1998; 40: 23742.
analysis of microdissected de novo glio- 23. Piva R, Cancelli I, Cavalla P et al.
blastomas and paired astrocytic tumors. J Proteasome-dependent degradation of
Neuropathol Exp Neurol 1999; 58: 1208. p27/kip1 in gliomas. J Neuropathol Exp
13. Bschges R, Weber RG, Actor B et al. Neurol 1999; 58: 6916.
Amplification and expression of cyclin D 24. Gran S, Tali T. p53 and p16INK4A
genes (CCND1, CCND2 and CCND3) in mutations during the progression of glomus
human malignant gliomas. Brain Pathol tumor. Pathol Oncol Res 1999; 5: 415.
1999; 9: 43542. 25. Kleihues P, Cavenee WK. World Health
14. Miettinen H, Kononen J, Sallinen P et al. Organization Classification of Tumours:
CDKN2/p16 predicts survival in oligoden- Tumours of the Nervous System Pathology
drogliomas: comparison with astrocytomas. J and Genetics. Lyon: IRAC Press, 2000.
Neurooncol 1999; 41: 20511. 26. Jung V, Romeike BFM, Henn W, Feiden W
15. Ishii N, Maier D, Merlo A et al. Frequent co- et al. Evidence of focal genetic microhetero-
alterations of TP53, p16/CDKN2A, p14ARF, geneity in glioblastoma multiforme by area-
PTEN tumor suppressor genes in human specific CGH on microdissected tumor cells.
glioma cell lines. Brain Pathol 1999; 9: J Neuropathol Exp Neurol 1999; 58: 9939.
46979. 27. Giannini C, Scheithauer BW, Burger et al.
16. Duerr EM, Rollbrocker B, Hayashi Y et al. Cellular proliferation in pilocytic and diffuse
PTEN mutations in gliomas and glioneuronal astrocytomas. J Neuropathol Exp Neurol
tumors. Oncogene 1998; 16: 225964. 1999; 58: 4653.

183
GLIAL TUMORS

28. Litofsky NS, Mix TC, Baker SP et al. Ki-67 40. De Stefano N, Caramanos Z, Preul MC et al.
(clone MIB-1) proliferation index in recurrent In vivo differentiation of astrocytic brain
glial neoplasms: no prognostic significance. tumors and isolated demyelinating lesions of
Surg Neurol 1998; 50: 57985. the type seen in multiple sclerosis using 1H
29. Kosel S, Scheithauer BW, Graeber MB. magnetic resonance spectroscopic imaging.
Genotype-phenotype correlation in gemistocytic Ann Neurol 1998; 44: 2738.
astrocytomas. Neurosurgery 2001; 48: 18793. 41. Peterson K, Rosenblum MK, Powers JM et al.
30. Maidment SL, Rucklidge GJ, Rooprai HK, Effect of brain irradiation on demyelinating
Pilkington GJ. An inverse correlation between lesions. Neurology 1993; 43: 210512.
expression of NCAM-A and the matrix- 42. Peterson K, DeAngelis LM. Weighing the
metalloproteinases gelatinase-A and gelati- benefits and risks of radiation therapy for low-
nase-B in human glioma cells in vitro. Cancer grade glioma. Neurology 2001; 56: 12556.
Lett 1997; 116: 717. 43. Bampoe J, Bernstein M. The role of surgery
31. Liigant A, Haldre S, Oun A et al. Seizure in low grade gliomas. J Neurooncol 1999; 42:
disorders in patients with brain tumors. Eur 25969.
Neurol 2001; 45: 4651. 44. Pollack IF. The role of surgery in pediatric
32. Gamburg ES, Regine WF, Patchell RA et al. gliomas. J Neurooncol 1999; 42: 27188.
The prognostic significance of midline shift at 45. Karim AB, Cornu P, Bleehen N. Immediate
presentation on survival in patients with postoperative radiotherapy in low grade
glioblastoma multiforme. Int J Radiat Oncol glioma improves progression free survival but
Biol Phys 2000; 48: 135962. not overall survival: preliminary results of an
33. Landy HJ, Lee TT, Potter P et al. Early MRI EROTC/MRC randomized phase III study.
findings in high grade glioma. J Neurooncol Proc Am Soc Clin Oncol. 17, 400a. 1998.
2000; 47: 6572. 46. Surma-Aho O, Niemela M, Vilkki J et al.
34. Amato C, Elia M, Musumeci SA et al. Adverse long-term effects of brain radio-
Transient MRI abnormalities associated with therapy in adult low-grade glioma patients.
partial status epilepticus: a case report. Eur J Neurology 2001; 56: 128590.
Radiol 2001; 38: 504. 47. Roelcke U, von Ammon K, Hausmann O et
35. Langleben DD, Segall GM. PET in differenti- al. Operated low grade astrocytomas: a long
ation of recurrent brain tumor from radiation term PET study on the effect of radiotherapy.
injury. J Nucl Med 2000; 41: 18617. J Neurol Neurosurg Psychiatry 1999; 66:
36. Roesdi MF, Postma TJ, Hoekstra OS, Van 6447.
Groeningen CJ, Wolbers JG, Heimans JJ. 48. Bruehlmeier M, Roelcke U, Amsler B et al.
Thallium-201 SPECT as response parameter Effect of radiotherapy on brain glucose
for PCV chemotherapy in recurrent glioma. J metabolism in patients operated on for low
Neurooncol 1998; 40: 2515. grade astrocytoma. J Neurol Neurosurg
37. Vigneron D, Bollen A, McDermott M et al. Psychiatry 1999; 66: 64853.
Three-dimensional magnetic resonance 49. Kiebert GM, Curran D, Aaronson NK et al.
spectroscopic imaging of histologically Quality of life after radiation therapy of
confirmed brain tumors. Magn Reson cerebral low-grade gliomas of the adult:
Imaging 2001; 19: 89101. results of a randomised phase III trial on dose
38. Quan D, Hackney DB, Pruitt AA, Lenkinski response (EORTC trial 22844). EORTC
RE, Cecil KM. Transient MRI enhancement Radiotherapy Co-operative Group. Eur J
in a patient with seizures and previously Cancer 1998; 34: 19029.
resected glioma: use of MRS. Neurology 50. Bauman G, Pahapill P, Macdonald D et al.
1999; 53: 2113. Low grade glioma: measuring radiographic
39. Masdeu JC, Quinto C, Olivera C et al. Open- response to radiotherapy. Can J Neurol Sci
ring imaging sign highly specific for atypi- 1999; 26: 1822.
cal brain demyelination. Neurology 2000; 54: 51. Lee SW, Fraass BA, Marsh LH et al. Patterns
142733. of failure following high-dose 3-D conformal

184
REFERENCES

radiotherapy for high-grade astrocytomas: a concurrent radiation in patients with newly


quantitative dosimetric study. Int J Radiat diagnosed high-grade gliomas. J Neurooncol
Oncol Biol Phys 1999; 43: 7988. 1999; 44: 7783.
52. Shrieve DC, Alexander E III, Black PM et al. 61. DeAngelis LM, Burger PC, Green SB,
Treatment of patients with primary glioblas- Cairncross JG. Malignant glioma: who
toma multiforme with standard postoperative benefits from adjuvant chemotherapy? Ann
radiotherapy and radiosurgical boost: Neurol 1998; 44: 6915.
prognostic factors and long-term outcome. J 62. Brem H, Piantadosi S, Burger et al. Placebo-
Neurosurg 1999; 90: 727. controlled trial of safety and efficacy of intra-
53. Gaspar LE, Zamorano LJ, Shamsa F et al. operative controlled delivery by biodegradable
Permanent 125iodine implants for recurrent polymers of chemotherapy for recurrent
malignant gliomas. Int J Radiat Oncol Biol gliomas. The Polymer-Brain Tumor Treatment
Phys 1999; 43: 97782. Group. Lancet 1995; 345: 100812.
54. Prados MD, Wara WM, Sneed PK et al. 63. Subach BR, Witham TF, Kondziolka D et al.
Phase III trial of accelerated hyperfractiona- Morbidity and survival after 1,3-bis(2-
tion with or without difluromethylornithine chloroethyl)-1 nitrosourea wafer implantation
(DFMO) versus standard fractionated radio- for recurrent glioblastoma: a retrospective
therapy with or without DFMO for newly case matched cohort series. Neurosurgery
diagnosed patients with glioblastoma multi- 1999; 45: 1722.
forme. Int J Radiat Oncol Biol Phys 2001; 49: 64. Boiardi A, Silvani A, Pozzi A et al. Interstitial
717. chemotherapy plus systemic chemotherapy
55. Arcicasa M, Roncadin M, Bidoli E et al. for glioblastoma patients: improved survival
Reirradiation and lomustine in patients with in sequential studies. J Neurooncol 1999; 41:
relapsed high-grade gliomas. Int J Radiat 1517.
Oncol Biol Phys 1999; 43: 78993. 65. Witham TF, Fukui MB, Meltzer CC et al.
56. Mohan DS, Suh JH, Phan JL et al. Outcome Survival of patients with high grade glioma
in elderly patients undergoing definitive treated with intrathecal thiotriethylenephos-
surgery and radiation therapy for supratento- phoramide for ependymal or leptomeningeal
rial glioblastoma multiforme at a tertiary care gliomatosis. Cancer 1999; 86: 134753.
institution. Int J Radiat Oncol Biol Phys 66. Chamberlain MC, Kormanik PA. Salvage
1998; 42: 9817. chemotherapy with tamoxifen for recurrent
57. Pierga JY, Hoang-Xuan K, Feuvret L et al. anaplastic astrocytomas. Arch Neurol 1999;
Treatment of malignant gliomas in the 56: 7038.
elderly. J Neurooncol 1999; 43: 18793. 67. Akabani G, Reist CJ, Cokgor I et al.
58. Prados MD, Scott C, Curran WJ Jr et al. Dosimetry of 131I-labeled 81C6 monoclonal
Procarbazine, lomustine, and vincristine antibody administered into surgically created
(PCV) chemotherapy for anaplastic astro- resection cavities in patients with malignant
cytoma: a retrospective review of Radiation brain tumors. J Nucl Med 1999; 40: 6318.
Therapy Oncology Group protocols compar- 68. Spear MA. Gene therapy of gliomas: receptor
ing survival with carmustine or PCV adjuvant and transcriptional targeting. Anticancer Res
chemotherapy. J Clin Oncol 1999; 17: 1998; 18: 322331.
338995. 69. Scott JN, Rewcastle NB, Brasher PMA et al.
59. Nieder C, Grosu AL, Molls M. A compari- Which glioblastoma multiforme patient will
son of treatment results for recurrent malig- become a long-term survivor? A population-
nant gliomas. Cancer Treat Rev 2000; 26: based study. Ann Neurol 1999; 46: 1838.
397409. 70. Galanis E, Buckner JC, DiNapoli RP et al.
60. Jakacki RI, Siffert J, Jamison C, Velasquez L, Clinical outcome of gliosarcoma compared
Allen JC. Dose-intensive, time-compressed with glioblastoma multiforme: North Central
procarbazine, CCNU, vincristine (PCV) with Cancer Treatment Group results. J Neurosurg
peripheral blood stem cell support and 1998; 89: 42530.

185
GLIAL TUMORS

71. Perry A, Jenkins RB, OFallon JR et al. 82. Arai Y, Ackerley CA, Becker LE. Loss of the
Clinicopathologic study of 85 similarly TSC2 product tuberin in subependymal giant-
treated patients with anaplastic astrocytic cell tumors. Acta Neuropathol (Berl) 1999;
tumors an analysis of DNA content 98: 2339.
(ploidy), cellular proliferation, and p53 83. Mao X, Jones TA, Tomlinson I et al. Genetic
expression. Cancer 1999; 86: 67283. aberrations in glioblastoma multiforme:
72. Schwartzbaum JA, Lal P, Evanoff W et al. translocation of chromosome 10 in an O-2A-
Presurgical serum albumin levels predict like cell line. Br J Cancer 1999; 79: 72431.
survival time from glioblastoma multiforme. 84. Coons SW, Johnson PC, Scheithauer BW,
J Neurooncol 1999; 43: 3541. Yates AJ, Pearl DK. Improving diagnostic
73. Bauman G, Lote K, Larson D et al. accuracy and interobserver concordance in
Pretreatment factors predict overall survival the classification and grading of primary
for patients with low-grade glioma: a recur- gliomas. Cancer 1997; 79: 138193.
sive partitioning analysis. Int J Radiat Oncol 85. Bigner SH, Rasheed K, Wiltshire RN,
Biol Phys 1999; 45: 9239. McLendon R. Morphologic and molecular
74. Tancredi A, Mangiola A, Guiducci A, genetic aspects of oligodendroglial neoplasms.
Peciarolo A, Ottaviano P. Oligodendrocytic Neurooncology 1999; 1: 5260.
gliomatosis cerebri. Acta Neurochir (Wien) 86. Pohl U, Cairncross JG, Louis DN. Homo-
2000; 142: 46972. zygous deletions of the CDKN2C/p18INK4C
75. Horst E, Micke O, Romppainen ML et al. gene on the short arm of chromosome 1 in
Radiation therapy approach in gliomatosis anaplastic oligodendrogliomas. Brain Pathol
cerebri case reports and literature review. 1999; 9: 63943.
Acta Oncologica 2000; 39: 74751. 87. Husemann K, Wolter M, Bschges R et al.
76. Farmer JP, Montes JL, Freeman CR et al. Identification of two distinct deleted regions
Brainstem gliomas: a 10-year institutional on the short arm of chromosome 1 and rare
review. Pediatr Neurosurg 2001; 34: 20614. mutation of the CDKN2C gene from 1p32 in
77. Mandell LR, Kadota R, Freeman C et al. oligodendroglial tumors. J Neuropathol Exp
There is no role for hyperfractionated radio- Neurol 1999; 58: 104150.
therapy in the management of children with 88. Ino Y, Betensky RA, Zlatescu MC et al.
newly diagnosed diffuse intrinsic brainstem Molecular subtypes of anaplastic oligoden-
tumors: results of a Pediatric Oncology droglioma: implications for patient management
Group phase III trial comparing conventional at diagnosis. Clin Cancer Res 2001; 7: 83945.
versus hyperfractionated radiotherapy. Int J 89. Giannini C, Scheithauer BW, Weaver AL et al.
Radiat Oncol Biol Phys 1999; 43: 95964. Oligodendrogliomas: Reproducibility and prog-
78. Allen J, Siffert J, Donahue B et al. A phase nostic value of histologic diagnosis and grading.
I/II study of carboplatin combined with J Neuropathol Exp Neurol 2001; 60: 24862.
hyperfractionated radiotherapy for brainstem 90. Derlon JM, Chapon F, Nol MH et al. Non-
gliomas. Cancer 1999; 86: 10649. invasive grading of oligodendrogliomas:
79. Landolfi JC, Thaler HT, DeAngelis LM. correlations between in vivo metabolic
Adult brainstem gliomas. Neurology 1998; pattern and histopathology. Eur J Nucl Med
51: 11369. 2000; 27: 77887.
80. Pollack IF, Shultz B, Mulvihill JJ. The 91. Cairncross JG, Ueki K, Zlatescu MC et al.
management of brainstem gliomas in patients Specific genetic predictors of chemotherapeu-
with neurofibromatosis 1. Neurology 1996; tic response and survival in patients with
46: 165260. anaplastic oligodendrogliomas. J Natl Cancer
81. Shepherd CW, Scheithauer BW, Gomez MR, Inst 1998; 90: 14739.
Altermatt HJ, Katzmann JA. Subependymal 92. Soffietti R, Rud R, Bradac GB, Schiffer D.
giant cell astrocytoma: a clinical, pathologi- PCV chemotherapy for recurrent oligoden-
cal, and flow cytometric study. Neurosurgery drogliomas and oligoastrocytomas. Neuro-
1991; 28: 8648. surgery 1998; 43: 106673.

186
REFERENCES

93. Mason WP, Krol GS, DeAngelis LM. Low- tumors? A cytogenetic study in 24 cases.
grade oligodendroglioma responds to Cancer Genet Cytogenet 1998; 104: 15760.
chemotherapy. Neurology 1996; 46: 2037. 105. Dirven CM, Koudstaal J, Mooij JJ, Molenaar
94. Paleologos N, Cairncross JG. Treatment of WM. The proliferative potential of the
oligodendroglioma: an update. J Neurooncol pilocytic astrocytoma: the relation between
1999; 1: 618. MIB-1 labeling and clinical and neuro-radio-
95. Chinot OL, Honore S, Dufour H et al. Safety logical follow-up. J Neurooncol 1998; 37:
and efficacy of temozolomide in patients with 916.
recurrent anaplastic oligodendrogliomas after 106. Tihan T, Fisher PG, Kepner JL et al. Pediatric
standard radiotherapy and chemotherapy. J astrocytomas with monomorphous pilomyx-
Clin Oncol 2001; 19: 244955. oid features and a less favorable outcome. J
96. Olson JD, Riedel E, DeAngelis LM. Long- Neuropathol Exp Neurol 1999; 58: 10618.
term outcome of low-grade oligoden- 107. Tamura M, Zama A, Kurihara H et al.
droglioma and mixed glioma. Neurology Management of recurrent pilocytic astro-
2000; 54: 14428. cytoma with leptomeningeal dissemination
97. Shaw EG, Scheithauer BW, OFallon JR. in childhood. Childs Nerv Syst 1998; 14:
Supratentorial gliomas: A comparative study 61722.
by grade and histologic type. J Neurooncol 108. Teo JG, Ng HK. Cytodiagnosis of pilocytic
1997; 31: 2738. astrocytoma in smear preparations. Acta
98. Marie Y, Sanson M, Mokhtari K et al. Cytol 1998; 42: 6738.
OLIG2 as a specific marker of oligoden- 109. Kaschten B, Stevenaert A, Sadzot B et al.
droglial tumour cells. Lancet 2001; 358: Preoperative evaluation of 54 gliomas by PET
298300. with fluorine-18-fluorodeoxyglucose and/or
99. Smith JS, Perry A, Borell TJ et al. Alterations carbon-11-methionine. J Nucl Med 1998; 39:
of chromosome arms 1p and 19q as predic- 77885.
tors of survival in oligodendrogliomas, astro- 110. Hwang JH, Egnaczyk GF, Ballard E et al.
cytomas, and mixed oligoastrocytomas. J Clin Proton MR spectroscopic characteristics of
Oncol 2000; 18: 63645. pediatric pilocytic astrocytomas. Am J
100. Morreale VM, Ebersold MJ, Quast LM, Neuroradiol 1998; 19: 53540.
Parisi JE. Cerebellar astrocytoma: experience 111. Debus J, Kocagnc KO, Hss A, Wenz F,
with 54 cases surgically treated at the Mayo Wannenmacher M. Fractionated stereotactic
Clinic, Rochester, Minnesota, from 1978 to radiotherapy (FSRT) for optic glioma. Int J
1990. J Neurosurg 1997; 87: 25761. Radiat Oncol Biol Phys 1999; 44: 2438.
101. Parsa CF, Hoyt CS, Lesser RL et al. 112. Kato T, Sawamura Y, Tada M et al. Cisplatin/
Spontaneous regression of optic gliomas: vincristine chemotherapy for hypothalamic/
thirteen cases documented by serial visual pathway astrocytomas in young children.
neuroimaging. Arch Ophthalmol 2001; 119: J Neurooncol 1998; 37: 26370.
51629. 113. Gropman AL, Packer RJ, Nicholson HS et al.
102. Ishii N, Sawamura Y, Tada M et al. Absence Treatment of diencephalic syndrome with
of p53 gene mutations in a tumor panel chemotherapy: growth, tumor response, and
representative of pilocytic astrocytoma diver- long term control. Cancer 1998; 83: 16672.
sity using a p53 functional assay. Int J Cancer 114. Shaw EG, Scheithauer BW, OFallon JR.
1998; 76: 797800. Supratentorial gliomas: A comparative study
103. Hayes VM, Dirven CMF, Dam A et al. High by grade and histologic type. J Neurooncol
frequency of TP53 mutations in juvenile 1997; 31: 2738.
pilocytic astrocytomas indicates role of TP53 115. Haapasalo H, Sallinen SL, Sallinen P et al.
in the development of these tumors. Brain Clinicopathological correlation of cell prolif-
Pathol 1999; 9: 4637. eration, apoptosis and p53 in cerebellar
104. Zattara-Cannoni H, Gambarelli D, Lena G et pilocytic astrocytomas. Neuropathol Appl
al. Are juvenile pilocytic astrocytomas benign Neurobiol 1999; 25: 13442.

187
GLIAL TUMORS

116. Pencalet P, Maixner W, Sainte-Rose C et al. need to be treated? Int J Radiat Oncol Biol
Benign cerebellar astrocytomas in children. J Phys 2001; 49: 75761.
Neurosurg 1999; 90: 26573. 128. Mason WP, Goldman S, Yates AJ et al.
117. Giannini C, Scheithauer BW, Burger PC et al. Survival following intensive chemotherapy
Pleomorphic xanthoastrocytoma: what do we with bone marrow reconstitution for children
really know about it? Cancer 1999; 85: with recurrent intracranial ependymoma a
203345. report of the Childrens Cancer Group. J
118. Tonn JC, Paulus W, Warmuth-Metz M et al. Neurooncol 1998; 37: 13543.
Pleomorphic xanthoastrocytoma: report of 129. Horn B, Heideman R, Geyer R et al. A multi-
six cases with special consideration of institutional retrospective study of intracra-
diagnostic and therapeutic pitfalls. Surg nial ependymoma in children: identification
Neurol 1997; 47: 1629. of risk factors. J Pediatr Hematol Oncol
119. Schwartz TH, Kim S, Glick RS et al. 1999; 21: 20311.
Supratentorial ependymomas in adult 130. Dal Cin P, Van den Berghe H, Buonamici L et
patients. Neurosurgery 1999; 44: 72131. al. Cytogenetic investigation in subependy-
120. Applegate GL, Marymont MH. Intracranial moma. Cancer Genet Cytogenet 1999; 108: 84.
ependymomas: a review. Cancer Invest 1998; 131. Prayson RA, Suh JH. Subependymomas: clini-
16: 58893. copathologic study of 14 tumors, including
121. Hamilton RL, Pollack IF. The molecular comparative MIB-1 immunohistochemical
biology of ependymomas. Brain Pathol 1997; analysis with other ependymal neoplasms.
7: 80722. Arch Pathol Lab Med 1999; 123: 3069.
122. Loiseau H, Arsaut J, Demotes-Mainard J. p73 132. Schiff D, Wen PY. Uncommon brain tumors.
gene transcripts in human brain tumors: Neurol Clin 1995; 13: 95374.
overexpression and altered splicing in ependy- 133. Chow E, Reardon DA, Shah AB et al.
momas. Neurosci Lett 1999; 263: 1736. Pediatric choroid plexus neoplasms. Int J
123. Rosenblum MK. Ependymal tumors: A Radiat Oncol Biol Phys 1999; 44: 24954.
review of their diagnostic surgical pathology. 134. Gallia GL, Gordon J, Khalili K. Tumor
Pediatr Neurosurg 1998; 28: 1605. pathogenesis of human neurotropic JC virus
124. Prayson RA. Clinicopathologic study of 61 in the CNS. J Neurovirol 1998; 4: 17581.
patients with ependymoma including MIB-1 135. Wolff JE, Sajedi M, Coppes MJ, Anderson
immunohistochemistry. Ann Diagn Pathol RA, Egeler RM. Radiation therapy and
1999; 3: 1118. survival in choroid plexus carcinoma. Lancet
125. Kordas M, Czirjak S, Doczi T. The spinal 1999; 353: 2126.
tumour related hydrocephalus. Acta Neurochir 136. St Clair SK, Humphreys RP, Pillay PK et al.
(Wien) 1997; 139: 104954. Current management of choroid plexus carci-
126. Schild SE, Nisi K, Scheithauer BW et al. The noma in children. Pediatr Neurosurg 1991;
results of radiotherapy for ependymomas: 17: 22533.
The Mayo Clinic Experience. Int J Radiat 137. Souweidane MM, Johnson JH Jr, Lis E.
Oncol Biol Phys 1998; 42: 9538. Volumetric reduction of a choroid plexus
127. Paulino AC. The local field in infratentorial carcinoma using preoperative chemotherapy.
ependymoma: Does the entire posterior fossa J Neurooncol 1999; 43: 16771.

188
6
Meningeal tumors

Introduction the intracranial cavity may spread to and


invade the leptomeninges either focally or
The meninges (from the Greek for diffusely.
membrane) consist of three layers. The dura Most primary meningeal tumors are focal.
mater (from the Latin for hard mother) is A few, such as meningeal melanoma, may
a tough, fibrous membrane with a consis- infiltrate the meninges diffusely. Meningeal
tency of canvas that forms the outer cover- tumors are classified by the World Health
ing of the brain and spinal cord. The Organization3 as listed in Table 6.1, with
arachnoid (from the Greek for spider web1) modifications. Those meningiomas generally
is a delicate fibrous membrane that is considered to be more aggressive in behavior
apposed to the dura mater and forms the are indicated by italics.
outer layer of the subarachnoid space. The Meningioma is a common tumor; all of the
pia mater (from the Latin for tender others are rare. Hemangiopericytomas were
mother) is adherent to the glial capsule of once considered a subtype of meningioma
the brain and spinal cord (pialglial (angioblastic meningioma) but are now
membrane) and surrounds arterial blood recognized as a separate and malignant
vessels as they enter the substance of the tumor. Hemangioblastoma is a tumor of
brain, leaving a potential space between the uncertain cell origin usually found in the
blood vessel and the brain. The spaces are cerebellum and often involving the
sometimes true spaces, large enough to be leptomeninges. The WHO classifies it with
identified on MRI and occasionally mistaken meningeal tumors because of its meningeal
for a cyst, tumor or infarct.2 These location, recognizing that its cellular origin
VirchowRobin spaces are potential sites of is unknown. Sarcomas of mesenchymal
brain invasion from meningeal tumors. The but non-meningothelial origin, especially
arachnoid membrane forms granulations that chondrosarcomas, may rarely arise in the
protrude into dural veins, particularly the leptomeninges. The meninges also contain
sagittal sinus, and are a site of cerebrospinal melanocytes that may give rise to either
fluid (CSF) absorption. relatively indolent melanocytomas or frankly
Tumors may arise from any cells found in malignant melanomas. Primary melanocytic
the meninges. These include not only intracranial and intraspinal tumors are less
meningeal cells, but also melanocytes and common than malignant melanomas that
connective tissue cells. Furthermore, non- originate in the skin and metastasize to the
meningeal tumors originating in or outside of leptomeninges.

189
MENINGEAL TUMORS

Table 6.1
Tumors of meninges.
Meningioma
Introduction
Tumors of meningothelial origin
Meningioma The term meningioma was coined by Cushing
Variants in 1922 to give a non-specific name to a tumor
Meningothelial WHO grade 1
that was almost always found in proximity to
Fibrous (fibroblastic) WHO grade 1
Transitional (mixed) WHO grade 1
the meninges. Fig. 6.1 illustrates the common
Psammomatous WHO grade 1 location of intracranial meningiomas. Meningeal
Angiomatous WHO grade 1 cells, actually meningothelial arachnoid cap
Microcystic WHO grade 1 cells, are also found in choroid plexus, tela
Secretory WHO grade 1 choroidea, and the arachnoid villi at the spinal
Clear cell WHO grade 2 nerve exit, explaining why tumors are common
Choroid WHO grade 2
in the spinal canal and can also occur intraven-
Lymphoplasmacyte-rich WHO grade 1
Rhabdoid WHO grade 3 tricularly and in the pineal region (Table 6.2).
Papillary WHO grade 3 Meningiomas are benign, slowly growing
Atypical WHO grade 2 tumors that compress the brain but rarely
Anaplastic (malignant) WHO grade 3 invade it (Fig. 6.2). When they compress brain
Mesenchymal, non-meningeal tumors substance, they usually cause seizures initially,
Benign neoplasms followed by focal neurologic signs. When they
Osteocartilaginous tumors
Lipoma
compress cranial nerves in the cavernous sinus
Fibrous histiocytoma or the optic nerve, they cause diplopia or visual
Others loss respectively. They also frequently evoke an
Malignant neoplasms
Hemangiopericytoma
Chondrosarcoma Table 6.2
Variant mesenchymal chondrosarcoma Sites of intracranial meningiomas.
Malignant fibrous histiocytoma
Rhabdomyosarcoma
Site Relative
Meningeal sarcomatosis
incidence (%)
Others
Primary melanocytic lesions
Parasagittal/falcine 25
Diffuse melanosis
Convexity 19
Melanocytoma
Sphenoid ridge 17
Malignant melanoma
Suprasellar (tuberculum) 9
Meningeal melanomatosis Posterior fossa 8
Tumors of uncertain histogenesis Olfactory groove 8
Hemangioblastoma Middle fossa/Meckels cave 4
(Capillary hemangioblastoma) Tentorial 3
Peritorcular (sagittal sinus) 3
Lateral ventricle 12
Foramen magnum 12
Orbit/optic nerve sheath spinal 12

Modfied from DeMonte et al.4

190
MENINGIOMA

C
B D

Figure 6.1
Typical location of meningiomas. (A) Parasagittal meningiomas may compress the sagittal sinus, causing a
pseudotumor-like syndrome without focal findings. (B) Convexity meningiomas not close to the sensorimotor
strip may grow to substantial size without causing symptoms, as may olfactory groove meningiomas (C).
(D) Meningiomas involving the sphenoid ridge may compress or enter the cavernous sinus to surround the
carotid artery and involve nerves to the ocular muscles. Meningiomas also occur in the posterior fossa,
particularly in the cerebello-pontine angle and around the foramen magnum (E).

Figure 6.2
A large olfactory groove
meningioma in a patient
who presented with a
slowly developing history
of memory loss and some
headache. (A) Note the
modest amount of edema
within the brain substance
(arrow). (B) Meningioma
showing the benign
morphology and a typical
psammoma (Greek for
sand) body (arrow) with
its concentric layers of
calcification.

191
MENINGEAL TUMORS

osteoblastic response in the surrounding bone incidentally at autopsy or on a brain CT/MR


called hyperostosis that can be identified on scan performed for an unrelated problem.10 In
plain skull films. Hyperostosis may cause a Japanese incidence study, 32% of primary
symptoms by compressing cranial nerves at the intracranial tumours were meningiomas: 39%
skull base. Meningiomas may spread along were asymptomatic, dicovered incidentally on
the dura (en plaque meningioma), impairing scan, and only 1/3 of these grew on follow-up.
multiple cranial nerves. Although the tumors can occur at any age,
As indicated in Table 6.1, the usually benign they are more common in late middle age.
meningioma has a large number of variants. Symptomatic tumors are less common in the
These are primarily of neuropathologic interest very elderly, but asymptomatic tumors found
and do not, for the most part, affect the clini- by scan or at autopsy indicate an increasing
cal course, treatment or prognosis. There are incidence with increasing age.
exceptions. The secretory meningioma,5 a Meningiomas are substantially more
tumor which often secretes vascular endothelial common in women than in men, with
growth factor (VEGF),6 may, unlike most other female/male ratios varying from 3:2 to 2:1.
meningiomas, cause edema in the underlying Africans and African-Americans show equal
brain, leading the clinician to believe that he is male/female ratios and may actually have
dealing with a malignant tumor such as a higher rates of meningiomas than their
metastasis. The course of secretory menin- Caucasian counterparts. Multiple meningiomas
giomas is, however, quite benign. The angioma- are characteristic of NF-2 and other familial
tous meningioma has prominent vascular meningioma syndromes. Multiple tumors also
channels between rather inconspicuous nests of occur sporadically but at an incidence of less
meningothelial cells. The tumor, particularly than 10%. Atypical meningiomas represent
when capillary-size vessels are numerous, may about 5% of meningiomas, and frankly malig-
mimic hemangioblastoma. The clear cell nant meningiomas about 3%.11 Meningiomas
meningioma may be mistaken for other clear are also the most common intradural but
cell tumors, e.g. hemangioblastoma, metastatic extramedullary tumors of the spinal canal.
renal cell carcinoma or neurocytoma.7 Immuno- Spinal meningiomas of the thoracic spinal cord
histochemistry and electron microscopy can virtually always occur in women.
establish the diagnosis. Clear cell meningiomas
often appear at an earlier age than other
Etiology
meningiomas and may behave more aggres-
sively than other benign variants.8 Genetics
The genetic progression of meningiomas is
depicted in Table 6.3.
Incidence
As the table indicates, multiple cytogenetic
In population-based studies, intracranial changes have been noted in meningiomas.12,13 The
meningiomas represent more than 20% of most consistent is deletion of chromosome 22,
primary intracranial neoplasms.9 When which occurs in approximately 50% of menin-
autopsy data are considered, meningiomas giomas and probably involves the NF-2 tumor
represent over 40% of intracranial neoplasms. suppressor gene and its protein product, merlin
In the Mayo Clinic series, 75% of menin- (see Chapter 12). The NF-2 gene does not appear
giomas were asymptomatic, found either to be involved in meningothelial meningiomas,14

192
MENINGIOMA

Table 6.3 mutations of PTEN, as they are in glioblas-


Genetic changes in meningiomas. toma.18 Loss of heterozygosity on 17p and 22q
is associated with aggressive tumors.19 Studies
of X-chromosome inactivation have suggested
Arachnoidal cells
that meningiomas are monoclonal tumors
when single, and when they are multiple, about
NF2 gene mutations/chromosome
half appear to be monoclonal.20,21 However,
22 q loss
? other loci on 22 other studies conclude that meningiomas are
heterogeneous in clonal composition.22
Recurrent meningiomas, whether near or
Meningiomas, WHO grade I
distant from the original lesions, are clonal,
suggesting that the meninges were seeded at the
Losses of 1p, 6q, 10q, 14q, 18q
time of surgery.23
Gains of 1q, 91 12q, 15q, 17q, 20q
Of some interest is the role of sex
hormones.13 Meningiomas rarely express estro-
Atypical meningioma, WHO grade II
gen receptors or, if they do, they do so at very
low levels,24 but more than half express estro-
Losses of 6q, 9p, 10, 14q
gen receptor mRNA.25 Approximately two-
17q amplification
rare mutations: TP53, PTEN thirds of meningiomas express progesterone
rare deletions: CDKN2A receptors, more often in female than in male
patients. The role of these receptors in the
Anaplastic (malignant) meningioma, pathogenesis of the tumor remains unknown,
WHO grade III but their presence has been an impetus for
hormonal therapy, particularly progesterone
From Kleihues and Cavenee3 with permission. receptor blockers, so far without success.26
Some have suggested that sex hormone recep-
tors account for the reportedly more rapid
growth of meningiomas during pregnancy, but
there is little scientific evidence that sex
or in radiation-induced meningiomas.15 In a few hormones function as growth factors for
studies, abnormalities of chromosome 22 have meningiomas.27 One population-based study
been detected outside the NF-2 region, perhaps showed no relationship between number of
involving the INI1 tumor suppressor gene.16 pregnancies or age at first pregnancy and the
Chromosomal changes are more extensive in development of meningiomas.28
atypical and anaplastic tumors than in the Absence of progesterone receptors is associ-
more benign variety. Allelic losses are found at ated with anaplastic tumors13 and a shorter time
1p, 3p, 6q, 9q, 10q, 14q and 17p,12 as well as to recurrence after surgery. Estramustine-binding
other sites. These genes may be responsible for protein, believed to correlate with sensitivity
progression of the tumor from a benign to a to the chemotherapeutic agent estramustine
malignant form. Complex karyotypes and (a combination of estradiol and a nitrogen
abnormalities of chromosomes 1, 3 and 6 are mustard), has been found in some menin-
present in aggressive tumors.17 Losses on giomas.29 Other abnormalities that have been
chromosome 10 are not due to deletions or described in meningiomas include co-expression

193
MENINGEAL TUMORS

of platelet-derived growth factor (PDGF) and of all grades are over-represented in Africans,
PDGF receptor genes and their protein products Asians and particularly Asians of Chinese
and perhaps EGF and IGF as well. Some menin- origin.34
giomas, especially secretory and microcystic Ionizing radiation is the only established risk
types, express VEGF.30 Neurotensin receptors factor for meningioma (Fig. 6.3). Both low-
have also been described in meningiomas. dose radiation for the treatment of tinea capitis
Growth hormone receptor mRNA is expressed and high-dose radiation for the treatment of
in most meningiomas; blockade of the receptor other brain tumors35 cause meningiomas,36
retards meningioma growth in culture.31 many of which are atypical or frankly malig-
Proteins of the JAK and STAT superfamilies, nant. Ron et al37 identified a 10-fold increase
which mediate signals from prolactin and PDGF, in meningiomas in patients who had been
are expressed in meningiomas; these proteins treated as children with low-dose radiation to
may be the mechanism by which interferon- the scalp for tinea capitis. Meningiomas also
alpha (IFN-) inhibits meningioma cell occur after high-dose radiation for brain
growth.32 Some meningiomas also express tumors such as germinomas or medulloblas-
somatostatin receptors and telomerase.13 tomas or after prophylactic cranial RT for
childhood leukemia.
Risk factors One of our patients was operated on at age
Both familial and environmental risk factors 14 in 1945 for what was originally believed to
have been implicated in the pathogenesis of be an anaplastic astrocytoma (probably a more
meningiomas.33 As mentioned above, NF-2 benign focal astrocytoma) and received post-
is a common cause of meningiomas, but there operative RT. Over 30 years later, she devel-
is also an increased incidence of meningiomas oped a meningioma within the radiation portal.
in families without NF-2 abnormalities. The tumor was at first slow growing, but
Meningiomas may be increased in Gorlin and eventually became more aggressive, leading to
Cowden syndromes (Chapter 12). Meningiomas her death.

Figure 6.3
Radiation-induced atypical
meningioma. A 30-year-old man
who had received prophylactic
cranial irradiation for leukemia
26 years previously had a CT
scan following a fall. The scan
revealed a tentorial lesion. An
MRI, left, showed the lesion
(arrow) to be uniformly contrast
enhancing. He was asymptomatic
but craniotomy revealed an
atypical meningioma with mitosis
and areas of necrosis (arrow).
Atypical meningiomas require
resection and postoperative
radiation therapy (see text).

194
MENINGIOMA

Figure 6.4
Multiple meningiomas in
a patient with breast
cancer. This
neurologically
asymptomatic patient
was scanned in 1995
(top) as part of a
screening workup for
metastatic breast cancer
(not conventional
screening for breast
cancer). Two small
contrast-enhancing
meningiomas were
identified (arrows). A
diagnosis of meningioma
was made tentatively
because of the absence
of other metastatic
lesions. Four years later
(bottom), the tumors
had not changed in size.

Trauma was suggested by Cushing as a Meningiomas are probably more common in


significant etiologic factor; he had successfully women with a history of breast cancer than in
removed a meningioma from the army chief-of- women without such a history40 (Fig. 6.4).
staff, who had suffered an injury at the same Meningiomas may be difficult to distinguish from
site years previously.38 A number of epidemio- dural metastases from breast cancer, particularly
logic studies have investigated this risk factor. when there is prominent edema surrounding the
Most have been negative, but a recent study lesion. Furthermore, breast cancer may metasta-
reported a head injury odds ratio of 1.5 for size to a meningioma.41 However, in the majority
men with meningiomas; the ratio was lower for of patients, the diagnosis can be established on
serious injury, raising a question as to MRI and the meningioma treated accordingly.
relevance.39 Head injury causing meningioma
still arises as a source of lawsuits. Others have
Pathology
implicated viral infections, particularly SV-40
virus, as a potential pathogenic cause but the Grossly, meningiomas are smooth lobulated
evidence is unconvincing. tumors with a fine vascular pattern on their

195
MENINGEAL TUMORS

surface. When benign, they separate easily colon cancer, was found to have a persistently
from underlying brain tissue, which they elevated CEA. A search of the body revealed
compress but do not invade. They may, only an enhancing dural-based lesion which
however, invade venous structures, particularly was interpreted as metastatic colon cancer.
the sagittal sinus, making surgery difficult Surgical excision revealed a secretory menin-
unless the sinus has been occluded by tumor, in gioma, and the serum CEA level fell after resec-
which case the occluded sinus can sometimes tion. The MIB-1 labeling index correlates well
be removed with the tumor. Acute occlusion of with the likelihood of recurrence after resec-
the posterior portion of the superior sagittal tion,43 although mitoses are uncommon in the
sinus, as might occur during surgery, causes usual benign variants. DNA ploidy and p53
infarction of the brain,42 and, therefore, a immunohistochemistry do not provide useful
surgeon may need to leave a small amount of prognostic information.
disease behind to avoid this complication. Although most histologic subtypes do not
Meningiomas may encase cerebral arteries, have clinical significance, some subtypes
particularly the carotid artery at the base of the behave in a more aggressive fashion than
brain. They may infiltrate the arterial wall, others. The aggressive subtypes include
making arterial reconstruction necessary if the rhabdoid44 and clear cell meningiomas.
tumor is to be resected, but this is rarely advis- Lymphoplasmacytic-rich meningiomas may be
able. Occasionally, meningiomas may penetrate associated with polyclonal gammopathy or
bone and present as a scalp mass. Some menin- anemia.
giomas grow as a flat en plaque mass infiltrat- Atypical meningiomas are characterized by
ing substantial portions of the meninges, often increased mitotic activity (4 or more/10 high
making resection impossible. power fields) and at least 3 of the following
The histologic appearance varies with the histologic features: increased cellularity, high
type of meningioma.11 Certain features are nucleus/cytoplasm ratio, prominent nucleoli,
common to all subtypes. They include a lobular necrosis, and patternless or sheet-like growth.
structure, the presence of psammoma bodies Invasion of the brain warrants a diagnosis
and calcification. Microcystic and lymphoplas- of atypical meningioma, but it alone does
macytic-rich variants contain the structures not indicate an anaplastic (malignant) menin-
that their names imply. Most meningiomas, gioma, whereas 20 mitoses per 10 high power
particularly benign meningiomas, react with fields, or histology resembling carcinoma,
epithelial membrane antigen (EMA) and sarcoma or melanoma does3 (Fig. 6.5).
vimentin on immunohistochemical studies. Occasional pleomorphic nuclei and mitoses are
EMA immunoreactivity is less apparent in noted in benign meningiomas and do not
atypical and malignant tumors. S-100 protein connote aggressive behavior. Although less
is usually absent. Estrogen receptors are rarely than 1% of meningiomas metastasize, about
found in meningiomas but there is an inverse half of malignant meningiomas metastasize,
correlation between tumor grading and the usually to liver or bone.45 One patient
intensity of reaction with progesterone recep- presented with severe hypoglycemia from liver
tors.13 Secretory meningiomas are carcino- metastases 2 years after removal of a frontal
embryonic antigen (CEA) and cytokeratin anaplastic meningioma. It is particularly
positive, and may elevate serum CEA. One of important to identify atypical and frankly
our patients, after successful treatment for malignant meningiomas, as their treatment

196
MENINGIOMA

Figure 6.5
Left frontal malignant
meningioma showing an
irregular margin
invading the brain
(arrows), typical of a
malignant meningioma.
Histologically, it was a
highly cellular tumor
with complete loss of
meningothelial
differentiation and many
mitoses.

differs from that of the benign meningioma (see a meningioma. As the tumor grows, it may
below). further compress areas of the brain, leading to
sensory and motor changes, visual field defects
or other focal symptoms (see Table 3.5).
Clinical findings Meningiomas involving the orbit, optic nerve

Symptoms and signs


Meningiomas usually grow slowly and many Table 6.4
appear not to grow even when followed for Meningioma pathophysiology of signs and symptoms.
several years.46,47 The vast majority of menin-
giomas do not invade the brain but cause Compression of CNS tissue
symptoms by: (1) compression of central Seizures
nervous system (CNS) structures, (2) shift of Cranial nerve palsies
CNS structures with or without increased Focal sensory and motor signs
Shift of CNS structures
intracranial pressure, (3) hydrocephalus, and
Cognitive and behavioral changes
(4) brain edema (Table 6.4). False localizing signs (see Table 3.6)
The symptoms of compression of CNS tissue Hydrocephalus
depend on the site of the tumor. Tumors over Cognitive failure
the convexity, particularly when they are near Gait ataxia
Urinary incontinence
the sensorimotor cortex, usually present with
Brain edema (Fig. 6.6)
focal seizures. Simple partial or complex partial Focal sensory and motor signs
seizures, including many with behavioral and Seizures
intellectual features, may mark the presence of

197
MENINGEAL TUMORS

Figure 6.6
A meningioma with extensive surrounding edema. The CT and MR scan show a lesion that is hyperdense
before contrast on the CT scan, suggesting calcification, and which uniformly contrast enhances. The MR scan
does not show the lesion well without contrast (arrow), but the tumor intensely contrast enhances. The tumor
proved to be a secretory meningioma with hyalin inclusion bodies pseudopsammoma bodies in an otherwise
typical meningioma.

or cavernous sinus present with visual loss, are recognized, particularly if they involve
ophthalmoplegia or proptosis (Fig. 6.7). relatively silent areas of brain such as the non-
Tumors at the foramen magnum may mimic dominant frontal lobe. This is because the
amyotrophic lateral sclerosis. In one unselected compressed but usually non-edematous brain
series of 59 patients examined in the modern adapts to the lesion and no significant shift of
era, headache was the most common symptom, intracranial structures occurs.
followed by visual loss and cognitive changes Headache is a common symptom in patients
(Table 6.5). with all brain tumors, including meningiomas.
Because meningiomas are so slow-growing, In many patients with meningiomas, the
they may reach a gigantic size before symptoms headaches are not related to the tumor, the

198
MENINGIOMA

Figure 6.7
A large cavernous sinus
meningioma causing a
partial non-progressive
third nerve palsy. Except
for diplopia, the patient
was well for many years
until she developed signs
of hydrocephalus requiring
a shunt. This is the same
patient illustrated in Fig.
3.5. Note that the tumor
surrounds the carotid
artery (arrow) and
partially surrounds the
basilar artery (arrowhead).

tumor being found incidentally when an patients met the clinical criteria for migraine
imaging study is performed for evaluation of headache. In all seven patients, the headache
the headache. One study49 suggests that typical ceased after surgery. As indicated in Chapter 3
migraine or cluster headaches may be related these patients all had a recent change in the
to meningiomas. The authors reviewed 514 pattern of their long-standing headache that
meningiomas, selecting patients with headache prompted further evaluation; either the
but without papilledema, vomiting or mass frequency of the attacks increased or the pain
effect on the scan. Four patients met the clini- became persistent and more intense. Patients
cal criteria for cluster headaches, and three with brain tumors and a pre-existing headache
problem are more likely to have headache
Table 6.5 associated with their brain tumor than patients
Presenting symptoms of 59 meningiomas by side of without an underlying headache problem. This
lesion. emphasizes the importance of evaluating
changing headaches even when they seem
Left Right Bilateral % of typically vascular or benign.
total As indicated above, many meningiomas are
discovered incidentally, usually when the
Headache 9 12 1 37.3 patient presents with headache or another
Visual 3 7 5 25.4 neurologic problem that leads to a scan. Thus,
Cognitive 6 3 5 23.7
one must be extremely careful in attributing
LOC 8 2 3 22.0
Weakness 6 5 18.6 coincidental symptoms to the tumor. One of
Seizure 6 3 1 16.9 our patients presented with sudden onset of
Dizziness 4 3 1 11.9 painless ptosis and diplopia. Examination
Nausea 1 1 3.3 revealed paralysis of the oculomotor nerve
LOC, loss of consciousness.
without pupillary involvement. An MR scan
From Bornstein and Witt.48 revealed a meningioma on the appropriate side
not involving the oculomotor nerve. After a

199
MENINGEAL TUMORS

glucose tolerance test, a diagnosis of diabetic of cortical veins, leading to venous hyperten-
ophthalmoplegia was made. The patient made sion and edema. The severity of peritumoral
a full recovery, and the meningioma was just edema is closely related to the labeling index,
followed. suggesting a relationship between tumor
Patients, particularly those with large frontal aggressiveness and edema.54 Tumor recurrence
lobe tumors, may present with cognitive and also correlates with the degree of edema.55
personality changes, usually apathy and depres- Meningiomas may also cause symptoms by
sion. The patient may undergo psychiatric hydrocephalus. Some meningiomas at the base
evaluation and prolonged psychiatric treatment of the brain or in the posterior fossa cause
before a tumor is suspected. A sudden person- hydrocephalus by compressing and thus
ality change should lead one to suspect a struc- obstructing the ventricular system. Others
tural lesion and evaluation by brain imaging. exude proteinaceous material(s) that interferes
Slow-growing meningiomas may also be with normal CSF absorption, causing commu-
responsible for false localizing signs. Some of nicating hydrocephalus (i.e. hydrocephalus
these signs are listed in Table 3.6. A particu- without ventricular obstruction). The hydro-
larly vexing problem before the age of cephalus is characterized by ataxia, urgency
neuroimaging was gait ataxia from a frontal incontinence, and short-term memory loss. It
meningioma. Neurosurgeons would operate on usually responds to shunting of the cerebral
the posterior fossa, expecting to find the tumor ventricles, without necessarily treating the
there rather than in the frontal lobe. underlying meningioma. We have encountered
Although most meningiomas do not cause a number of patients with large, quiescent or
brain edema, some, particularly secretory slowly-growing meningiomas at the base of the
meningiomas, can cause substantial edema brain that cause symptoms of hydrocephalus.
leading to clinical symptoms. The cause of the Characteristically, the tumors were in the
edema is not entirely established and may be cerebellopontine angle or the cavernous sinus
multifactorial. Substantial evidence implicates and caused cranial nerve palsies that were
VEGF, and perhaps other chemical substances, relatively static. After many years, the patient
as important factors.50 VEGF is secreted by would complain of gait unsteadiness and
some meningiomas, particularly those that urgency incontinence sometimes associated
receive vascular supply from the brain (most with poor short-term memory. MR scan might
meningiomas are supplied by vessels such as reveal no change in the meningioma (Fig. 6.5)
the external carotid artery that do not supply but a progressive increase in the size of the
the brain itself). The role of VEGF in angio- ventricles. The symptoms are characteristic of
genesis is described in Chapter 2, but, in hydrocephalus, and even though the ventricles
addition, if VEGF diffuses from the tumor into are dilated, the pressure is usually normal
surrounding normal brain, it causes increased (normal-pressure hydrocephalus). Symptoms
vascular permeability and edema as well as are relieved by ventricular shunting. One of our
promoting angiogenesis.51 Other chemical patients went from being unable to walk and
factors that may play a role include platelet- incontinent to full normality after the shunt.
activating factor produced by leukocyte Communicating hydrocephalus can also
infiltration into meningiomas52 and develop or be the presenting symptom of a
prostaglandins. A non-chemical factor that
53 spinal meningioma. In this situation, cranial
may play a role is compression by the tumor imaging only reveals dilated ventricles. A

200
MENINGIOMA

careful neurologic history and examination enhancing dural tail that spreads out from the
often reveals a myelopathy in addition to the body of the tumor along the dura, a less
symptoms and signs of hydrocephalus. Spinal common finding in other dural tumors, includ-
imaging will reveal the meningioma or other ing metastasis. The dural tail is often not
spinal tumor. tumor, but a hypervascular response of the
Another unusual symptom of meningioma is dura to the tumor. Thus, it is not specific for
the mimicking of pseudotumor cerebri (benign meningioma and may occur with an extra- or
intracranial hypertension); the meningioma intra-axial tumor61 and even with an aneurysm,
typically compresses or invades the posterior confusing the diagnosis. A large tumor
portion of the superior sagittal sinus,56,57 compressing brain but not causing edema is
increasing venous, and thus CSF, pressure. The probably a meningioma, although one report
patient develops headache and papilledema, describes edema in 66% of meningiomas seen
usually without change in ventricular size. on MR scan.62 Sometimes meningiomas have
The syndrome is indistinguishable from that unusual imaging features caused by hemor-
of pseudotumor cerebri, except that MR rhage, cystic degeneration or necrosis.58
venogram reveals obstruction of the sagittal Magnetic resonance spectroscopy (MRS)
sinus. The same syndrome can result from shows well-defined peaks for choline (CHO), a
compression of the dominant transverse sinus low phosphocreatine (PCr)/creatine (Cr) ratio
by a posterior fossa meningioma. and a decrease of n-acetyl aspartate (NAA).
The reduction in PCr/Cr is greater than that
Imaging seen in astrocytomas. An alanine peak is
The diagnosis of meningioma is often estab- characteristic of meningiomas, and is usually
lished by neuroimaging,58 particularly MRI. A much larger than the creatine peak.58 Nuclear
typical meningioma is hypointense or isoin- scanning is sometimes useful in the diagnosis
tense with brain on T1, usually hypointense on of meningioma. The somatostatin analog,
T2, and intensely and uniformly contrast octreotide is taken up by meningiomas but not
enhances (98%). Hyperintense T2 images most other skull-based tumors.63 PET findings
correlate with tumor invasion of cerebral of skull-based tumors have shown a much
cortex.59 In heavily calcified meningiomas higher accumulation of the amino acid methio-
(67%), contrast enhancement may sometimes nine in meningiomas compared with surround-
be minimal and CT scan may be more useful ing cerebellar tissue, making the tumor easy to
for identifying the tumor. Many meningiomas identify and demarcate. Methionine uptake
either erode bone or cause hyperostosis (27%). correlates better with proliferative potential
When hyperostosis of the base of skull is than does F18 flourodeoxyglucose (FDG) PET.64
associated with a meningioma, there is usually Neuromas show a lower uptake compared to
invasion of that bone by tumor.60 However, cerebellum,65 making a clear distinction
some meningiomas cause hyperostosis without between these two common tumors.
invasion, probably by secretion of products
such as alkaline phosphatase; that enzyme may
Differential diagnosis
also play a role in the calcifications found in
most meningiomas. Both hyperostosis and The differential diagnosis of meningioma is
bone erosion are better identified on CT than indicated in Table 6.6. It includes dural metas-
on MR scan. Meningiomas typically have an tasis, the other primary meningeal tumors

201
MENINGEAL TUMORS

Table 6.6 Sometimes only a biopsy will settle the diagno-


Differential diagnosis of meningioma. sis. However, most meningiomas are so charac-
teristic on MRI that the diagnosis is
Dural metastasis established on the basis of neuroimaging
Other primary meningeal tumors (Table 6.1) alone. This is particularly true when resection
Metastasis to leptomeninges or biopsy would be hazardous.
Meningeal lymphoma/plasmacytoma
Inflammatory pseudotumor
Granulomas (e.g. sarcoid, foreign body)
Aneurysm
Treatment
The diagnosis of meningioma is usually made
by the combination of symptoms and imaging.
listed in Table 6.1, metastasis to leptomeninges The first step in management is to determine
and primary meningeal lymphomas, plasmacy- whether the patient requires treatment. Many
tomas or melanomas as well as inflammatory meningiomas are either discovered incidentally
lesions that can mimic tumors.66 Because or cause focal seizures or cranial nerve palsies
systemic cancer can metastasize to a menin- and appear not to grow when followed over
gioma, both may be present in the same several years47 (Fig. 6.8). Calcified tumors are
patient. The diagnosis can be suspected by the less likely to grow.46 If they do not grow, surgi-
more rapid development of symptoms than cal intervention may not be necessary if the
one would expect from a meningioma and patient is asymptomatic, if symptoms are due
by neuroimaging where edema and central to another cause, or if seizures are the only
necrosis suggest a more aggressive process. symptom and are easily controlled with

Figure 6.8
No growth in a meningioma over ten years. This woman presented with focal visual seizures. A scan done in
1989 (left panel) revealed a contrast-enhancing lesion arising from the tentorium without surrounding edema. It
was presumptively diagnosed as a meningioma. She elected for no therapy other than anticonvulsants, which
have controlled her seizures for the past 10 years. A repeat scan in 1999 (right panel) shows that the tumor
has not changed over the 10-year period.

202
TREATMENT

anticonvulsants. We have followed one such cord, the surgery to remove them involves
patient for over 10 years after she presented many different approaches, ranging from the
with a seizure and was found to have a menin- relatively simple (convexity tumors) to the very
gioma involving the occipital area. Surgery was complex (skull base tumors68,69). The guiding
recommended when she was first seen, but she principle in all cases is removal of as much
refused. The seizures have been completely tumor as possible without causing or adding to
controlled with anticonvulsants and no new neurologic injury. Because of the relatively slow
symptoms have developed. The tumor has not growth rate of these tumors, a subtotal resec-
altered in size over the last decade. tion is often enough, particularly in elderly
Other meningiomas are not treated because patients. Obviously, a complete resection for
removal would be difficult and dangerous, out cure should be contemplated in every case.
of proportion to the symptoms from which Postoperative cerebral edema is a particular
the patient suffers. Typically, these are large problem in meningioma patients. This should
meningiomas at the base of the brain, often be anticipated, particularly in patients with
invading the cavernous sinus, that present with larger tumors. Patients should receive steroids
cranial nerve palsies without evidence of and be fluid-restricted in the immediate post-
increased intracranial pressure. At least two of operative period. The peak edema period can
our patients with oculomotor palsies have been be delayed, often 2448 hours after surgery.
followed for more than two decades with Edema may compress cerebral vessels, leading
stable neurologic symptoms and no change in to delayed ischemia and even infarction in the
the meningioma. An additional patient devel- days following surgery.
oped, after several years, a visual field abnor- If the tumor recurs, a second surgical proce-
mality which improved after microsurgery with dure should be carried out, if feasible, again
subtotal resection. with the intent of removing all or as much of
the tumor as possible. This should be
Surgery followed by focal RT (see below). A small
The treatment of most symptomatic menin- percentage of tumors are atypical or frankly
giomas is by surgery. In many instances, the anaplastic, either initially or at recurrence. All
entire tumor can be removed surgically and the of these should be treated with radiation
patient cured. Thus, if complete surgical removal therapy after surgery. Patients with anaplastic
can be achieved, no further therapy is indicated. tumors should receive periodic bone and body
However, in about 1520% of patients, even scans to detect asymptomatic metastases that
when the tumor has been completely removed, can be treated either surgically or by irradia-
there is recurrence. Tumors that are tion.
mushroom-shaped or lobulated are more likely Modern surgical techniques have made
to recur than those that are round. Wide dural meningioma surgery much more successful and
resection decreases the likelihood of recur- less dangerous. Preoperative embolization can
rence.67 Thus, after successful surgery, patients reduce the size and vascularity58 of the tumor,
should be followed closely with MR scans making resection easier; viable tumor in the
beginning at about 6-month intervals and gradu- perinecrotic areas may show an increased label-
ally increasing to 24-month intervals. ing index, but this is transient and does not
Because meningiomas can occur virtually indicate an increased proliferative potential of
anywhere in or around the brain or spinal the tumor.70 Techniques to preserve the carotid

203
MENINGEAL TUMORS

artery and to remove tumor from around Atypical and malignant meningiomas are
cranial nerves in the cavernous sinus have treated with 59.4 Gy.72 Conformal radiation
considerably increased the safety of operations giving a tumor dose of 54 Gy has been the
in that area. Reconstruction of the carotid standard treatment of benign meningiomas.
artery can often be carried out now as part of Evidence from uncontrolled retrospective
a procedure to remove tumor from the studies indicates that recurrence is less
cavernous sinus. frequent and survival is longer in those
Despite these advances, there is still a small patients who receive postoperative therapy.73
mortality and significant morbidity associated Thus, postoperative irradiation should be
with surgical treatment of meningiomas. In considered after subtotal resection. The size of
part, this is because many of the patients are the residual tumor predicts response to RT.
elderly and the tumors have grown to a large Progression-free survival was significantly
size prior to diagnosis, and in part because the poorer for those tumors > 5 cm than for those
tumor often directly involves important nerve < 5 cm (40% versus 93%).74 However, even
and vascular structures. Thus, in the absence of after partial resection, we may elect to follow
feasible surgery and if the tumor is growing, the patient by MRI, and if the tumor grows,
RT should be considered. re-resect and then radiate (see prognosis
In those patients in whom only partial resec- below). Skull-based meningiomas often can
tion or no resection is possible, RT may either only be subtotally resected. Radiation therapy
prevent further growth or cause some shrink- can produce long-term control of unresected
age but rarely, if ever, eradicates the tumor. or partially resected meningiomas with few
Unlike the case with every other type of side-effects.75
primary brain tumor, biopsy is not necessary Radiosurgery is another option which has
for tissue diagnosis in unresectable meningioma received increasing attention.76,77 The role of
prior to the administration of RT. The clinical radiosurgery in the treatment of meningiomas
and radiographic appearance are usually so is still not established, but many radiation
characteristic, and the differential diagnosis so oncologists are enthusiastic about the ability to
clear, that surgery is performed only if it can treat meningiomas with high doses of radiation
be helpful therapeutically, and a diagnostic while sparing surrounding brain. Radiosurgery
biopsy is limited to the unusual patient who is limited to tumors 3 cm in size or less and
requires tissue confirmation before treatment may be particularly useful in elderly or infirm
can start. patients who cannot easily tolerate surgery. In
one series 93% of patients followed for 510
Radiation therapy years required no other therapy.76 Radiosurgery
RT is applied as primary treatment to inoper- is being increasingly advocated for small
able tumors and as secondary treatment after tumors in surgically difficult areas such as near
partial surgery in some patients, after recur- a patent sagittal sinus78 or at the base of the
rence, after surgery and in all instances of brain.79 The benefit of radiosurgery may be
atypical or malignant meningioma even if delayed, and improvement may take months to
completely excised.71 In patients whose tumor years. Thus, radiosurgery is not a good option
is benign and totally resected, postoperative for patients with rapidly progressive symptoms.
RT is unnecessary. If the tumor recurs, it is Side effects of radiosurgery both acute80 (brain
resected again and postoperative RT given. edema) and delayed81 (cranial nerve palsy,

204
TREATMENT

Figure 6.9
Side effects of radiosurgery. This patient received radiosurgery for treatment of a meningioma at the base of the
brain. The meningioma decreased somewhat in size but she developed severe headache. The MR scan revealed
a contrast-enhancing area in the frontal lobe (the meningioma had not directly involved the frontal lobe)
surrounded by edema (left). The T2-weighted image (middle) revealed marked edema with mass effect. She was
treated with steroids with amelioration of symptoms. When a routine scan was done 2 years later (right), the
radiation toxicity had resolved.

visual loss, radiation necrosis), are uncommon Prognosis


but do occur (Fig. 6.9).
The prognosis after treatment of meningiomas
Chemotherapy depends on the histology of the tumor and on
The role of chemotherapy for meningiomas the adequacy of resection. Age at diagnosis,
is not established.82 Isolated reports have tumor size and postoperative RT may also play
suggested that octreotide, hydroxyurea,83 a role.86 A low-grade meningioma that is totally
tamoxifen, doxorubicin-based regimens, IFN- resected has recurrence rates of about 20% in 5
84 and mifepristone (RU486) have controlled years86 and 25% in 10 years.87 Tumors with less
the growth of some meningiomas. Hydroxy- than gross total resection have about a 60%
urea has not produced responses in our experi- chance of progressing without postoperative RT.
ence. A number of investigative protocols for Overall, younger patients do better than the
the treatment of meningioma are underway, elderly. The 5-year survival rate of patients aged
including growth factor and angiogenesis 2164 is 81%, whereas it is 56% for patients 65
inhibitors and gene therapy, but none has and older. The 5-year survival rate for benign
proved efficacious as yet. Retinoic acid is tumors overall is 70%, whereas it is 55% with
reported to stimulate meningioma cell adhesion malignant tumors, even with aggressive surgery
and inhibit invasion in vitro.85 and adjuvant radiation.88 The higher the MIB-1

205
MENINGEAL TUMORS

labeling index at initial surgery, the shorter the chromosome 9p, suggesting that the p16-
time to recurrence.89 In general quality of life mediated cell cycle regulatory pathway may be
remains good after treatment of either young or involved in transformation or progression of
elderly patients.90,91 these tumors. Unlike meningiomas, there is no
alteration in the NF2 gene. Somatostatin recep-
tors are expressed by some tumors.95 There are
no known risk factors for the development of
Hemangiopericytoma meningeal hemangiopericytomas.
Introduction
Originally called hemangioblastic meningioma
Pathology
(as were hemangioblastomas see below), or Hemangiopericytomas are highly vascular but
angioblastic meningioma, this tumor is now look no different grossly from meningiomas.96
recognized as a distinct tumor of uncertain Microscopically, the tumor consists of
cellular origin that behaves much more aggres- uniformly plump or polygonal cells with oval
sively than meningiomas. The dural-based nuclei and scant ill-defined cytoplasm. There
highly vascular tumor is indistinguishable is often a dense intercellular pattern of retic-
histologically from hemangiopericytomas ular staining surrounding vascular spaces that
arising in soft tissues elsewhere in the body. are lined by normal endothelium. The wide
Hemangiopericytomas represent less than 1% and branching vascular spaces that distinguish
of primary CNS tumors and are about 2% as this tumor from meningiomas have been
common as meningiomas.92 About 8% of all called stag horn sinusoids. They separate the
hemangiopericytomas are meningeal, and tumor into small nodules. Unlike menin-
about two-thirds of intracranial hemangio- giomas, there is no calcification, and no
pericytomas are supratentorial. Their peak psammoma bodies. Occasionally, the tumor
incidence is between 30 and 50 years of age invades the brain. Hemangiopericytomas do
and they are slightly more common in men not react with epithelial membrane antigen,
than in women. Like meningiomas, they are but are immunoreactive for vimentin and
often found in the parasagittal area, particu- CD34. VEGF is produced by some tumors,
larly around and attached to the torcular probably causing brain edema by a paracrine
herophili (from Latin for wine press the mechanism. Mitotic activity is usually promi-
confluence of dural venous sinuses), and, very nent, with MIB-1 labeling indices varying
rarely, in the ventricle.93 from 0.6% to 36%.

Etiology Clinical findings


Several cytogenetic abnormalities have been The signs and symptoms of meningeal hem-
found in hemangiopericytomas. The most angiopericytomas are similar to those of the
common is the rearrangement of chromosome more common meningiomas (Table 6.7).
12q13. Cytogenetic alterations have also been The common occurrence of these tumors
found in 19q, 6p and 7p. Approximately compressing the torcula may lead to intra-
25% of meningeal hemangiopericytomas have cranial hypertension with headache and
homologous deletions of CDKN2/p1694 on papilledema but no other neurologic signs.

206
HEMANGIOPERICYTOMA

Table 6.7 Except for the fact that meningeal hem-


Presenting signs and symptoms in 44 patients with angiopericytomas are rarely calcified, their
intracranial hemangiopericytoma.
CT and MR appearance is identical to that
of meningiomas. However, MRS yields a
Location % myoinositol peak that distinguishes them from
meningiomas. Hemangiopericytomas are dural-
Supratentorial
based, isodense before contrast, and uniformly
Headache 68
Papilledema 50 and sharply contrast enhance. The tumors may
Hemiparesis 40 also differ slightly from meningiomas in that
Visual field defect 20 they tend to cause lytic destruction of adjacent
Seizure 16 bone rather than hyperostosis. The presence of
Altered sensorium 15 hyperostosis probably excludes hemangioperi-
Infratentorial
cytoma. PET scans are characterized by low
Gait disturbance 56
Ataxia 44 glucose utilization but increased methionine
Headache, papilledema 44 uptake, similar to meningiomas.99 Octreotide
Hypacusis 33 scans (for somatostatin receptors) may be
Vertigo, dizziness 33 positive.
Neck pain 33

From Guthrie et al.96


Treatment
Whenever possible, the treatment is surgical.100
However, despite apparent complete removal
of tumor, local recurrence occurs in the major-
Hemangiopericytomas are occasionally associ- ity of patients.101 Approximately a quarter of
ated with two different paraneoplastic patients suffer recurrence after 5 years and
syndromes. One is hypoglycemia, probably a two-thirds after 10 years. Metastases are also
result of tumor production of insulin-like common, usually to bones, lungs and liver.
growth factor.97 The second is osteomalacia.95 Because of the likelihood of local recurrence,
Patients may present with fractures, and are and later distant metastases, all patients,
found to have hypophosphatemia and low even those with apparently complete surgical
1,25-dihydroxy vitamin D3 with normocal- removal, should be irradiated in the post-
cemia and normal levels of alkaline operative period.102 RT to a dose of 59.4 Gy
phosphatase. The tumor is believed to produce appears to reduce the local recurrence rate,
some humoral factor inhibiting 25-hydroxy prolonging disease-free and overall survival
vitamin D-1 -hydroxylase activity and from 65 months with surgery alone to 96
phosphorus reabsorption in the kidney. The months with surgery plus RT.103 Adjuvant
factor may act via parathyroid hormone- chemotherapy has no role.
related protein receptors,98 but is unrelated to When tumor recurs, it should be reoperated
cyclic AMP production in the proximal renal if feasible.92 This should be followed by exter-
tubules. One of our patients had symptoms of nal beam radiation, if not previously given. For
osteomalacia for over a decade before patients with small tumors at recurrence, radio-
headache led to cranial imaging and the discov- surgery may give a complete response in
ery of the indolent meningeal tumor. 30%.92,104 In previously irradiated patients,

207
MENINGEAL TUMORS

radiosurgery produced partial responses in renal cell carcinoma, pheochromocytoma,


70%, with 15% remaining stable. endolymphatic sac tumors109 and cysts in
Doxorubicin-based chemotherapy regimens various visceral organs (Chapter 12).
were not particularly effective.92 In general, Hemangioblastomas are the most common
chemotherapy is believed be ineffective in the primary tumor of the cerebellum in adults;
treatment of recurrent or metastatic hemangio- however, they are much less common than
pericytomas.105 metastases to the cerebellum. Sporadic
hemangioblastomas usually appear in adult-
hood, primarily between the ages of 30 and
Prognosis
60. Hemangioblastomas associated with VHL
Unlike meningioma, where the prognosis is disease occur at an earlier age, the mean being
excellent, the prognosis of hemangioperi- 29 years. The mean age for sporadic retinal
cytoma is poor, albeit prolonged. In several hemangioblastomas is 48, whereas that
series, the recurrence rate after 15 years is associated with VHL is 24. The age difference
about 90% for local recurrence and about fits a formula that requires only a single
70% for metastases, usually to bones, lung or somatic mutation for the tumor to develop in
liver. Once metastases occur, survival is about VHL disease (the first mutation having been
2 years. RT probably delays recurrence about inherited in the germline), but two indepen-
two-fold. Recurrent hemangiopericytomas can dent mutations for the sporadic form, thus
sometimes be treated by reoperation. The supporting Knudsons two-hit hypothesis110
median survival after first recurrence is about (Chapter 1). Sporadic hemangioblastomas,
5 years.92 There is some, but not total, corre- which can occur anywhere in the nervous
lation between markers of cell proliferation system, are usually single, whereas patients
and likelihood of recurrence.106 with VHL disease usually have multiple
hemangioblastomas along the neuraxis.

Hemangioblastoma Etiology
The VHL gene is located on chromosome 3q
Introduction 25.111 It is a tumor suppressor gene with three
Hemangioblastoma,107 a tumor that typically exons. The gene is widely expressed in normal
involves the leptomeninges, was once classi- adult tissues. The protein product consists of 213
fied as a hemangioblastic meningioma. amino acids and binds to the catalytic subunit of
Hemangioblastomas are benign tumors of elongin, thus interfering with the activity of RNA
uncertain origin that usually occur in the polymerase. The protein also regulates the
cerebellum but may appear anywhere in the expression of VEGF, platelet-derived growth
nervous system, including the cerebral factor beta (PDGF-) and Glut-1. The tumor
hemispheres, spinal cord, optic nerve108 and cells synthesize VEGF, leading
peripheral nerves. In about 25% of patients, to the exuberant growth of endothelial cells
hemangioblastomas occur as part of the Von associated with the tumor. VEGF expression is
HippelLindau (VHL) syndrome, an auto- low in solid tumors, moderate in microcystic
somal dominant disorder characterized by tumors and high in macrocystic tumors.112
hemangioblastomas in the CNS and retina, Because mutations may occur in any one of the

208
HEMANGIOBLASTOMA

exons, the clinical manifestations vary from in the wall of a cyst. The tumor consists of two
affected family to affected family; they may also cell types, the stromal cells and vascular cells.
vary within affected members of a given family. The stromal cells are believed to be the neoplas-
Allelic loss, or mutations of the VHL gene, is tic cells. The vascular cells are believed to repre-
found in stromal cells of most sporadic cerebel- sent a non-neoplastic proliferation of capillary
lar hemangioblastomas.113 One series suggests endothelial cells as the result of VEGF produc-
that more than 10% of patients with sporadic tion by the stromal cells. Stromal cell nuclei
hemangioblastoma show germline mutations of vary in size and have inconspicuous nucleoli.
VHL,114 suggesting that the range of VHL disease The nuclei may occasionally be atypical and
may be wider than previously recognized. There hyperchromatic. The cytoplasm is eosinophilic
are no known environmental risk factors for the and lipid-rich, with lipid-containing vacuoles
development of sporadic hemangioblastomas. giving the tumor a clear cell appearance,7
similar to metastatic renal cell carcinoma (Table
6.8), an important distinction in VHL patients.
Pathology
The stromal cells are vimentin and neuron-
Grossly, the tumor is seen as a well-circum- specific enolase positive; they may show glial
scribed, highly vascularized red nodule, usually fibrillary acidic protein (GFAP) expression but

Table 6.8
Clear cell lesions of the CNS: differential diagnosis.

General list of lesions Differential diagnosis

Hemangioblastoma Metastatic renal cell carcinoma


Microcystic meningioma
Meningioma variants Meningeal hemangioblastoma
Metastatic carcinoma
Chordoma
Clear cell pituitary adenoma
Germinoma Metastatic carcinoma
Clear cell pituitary adenoma
Large cell lymphoma
Oligodendroglioma Mixed glial neoplasms
Dysembryoplastic neuroepithelial tumor infarct
Demyelinating disorder
Neoplasms with clear cell foci (central neurocytoma, medulloblastoma,
ependymoma, choroid plexus carcinoma, pilocytic astrocytoma)
Clear cell carcinoma
Xanthomatous lesions Pleomorphic xanthoastrocytoma
Lipid-rich glioblastoma multiforme
Malakoplakia
Xanthomatous reactions to Langerhans cell histiocytosis and colloid cyst
Xanthomatous changes in Schwannoma, choroid plexus, meningioma

From Gokden et al7 with permission.

209
MENINGEAL TUMORS

are negative for EMA. The stromal cells are in the spinal cord or optic nerve, they cause
distributed within an intricate network of slowly progressive symptoms appropriate to
capillaries. The tumor is low-grade with their location.
mitoses absent or infrequent; labeling indices The MR scan of hemangioblastoma is
are usually less than 1%. In keeping with their characteristic (Fig. 6.10). The tumor is an
low-grade nature, the tumors may show intensely contrast-enhancing nodule, usually
Rosenthal fibers. surrounded by a large cyst. The cyst fluid is
hyperintense on the T2 image. The tumor
often contains hypointense flow voids as a
Clinical findings
result of its hypervascularity. When more than
Tumors in the cerebellar hemisphere usually one tumor is seen on cranial imaging, the
cause ipsilateral cerebellar signs, including gait patient probably has VHL disease, and a
and unilateral appendicular ataxia.107 By complete workup should include an enhanced
compression of the 4th ventricle, they can spine MRI, complete ophthalmologic exami-
cause hydrocephalus with attendant symptoms nation and body CT scan. Renal cell carci-
of headache, nausea and vomiting, cognitive noma, pheochromocytoma and multiple cysts
change, ataxia and urgency incontinence. of other organs are found in patients with
Because the tumors synthesize erythropoietin in VHL disease; renal cell cancer is the primary
about 20% of patients, erythrocytosis, an cause of death in these patients. Furthermore,
elevated hemoglobin without a change in the renal cell carcinoma metastatic to the cerebel-
white cell or platelet count, may be the present- lum can have an identical MR appearance to
ing manifestation, often identified on a blood a hemangioblastoma and has a predilection to
count done for other reasons. When tumors are metastasize to the posterior fossa as opposed

Figure 6.10
A hemangioblastoma
of the cerebellum.
The lesion is cystic
(arrow) and
intensely contrast
enhances. Headache
and cerebellar
symptoms resolved
after surgery.
Histologically, one
can identify foamy
stromal cells (arrow)
embedded in a rich
capillary network.

210
MELANOCYTIC TUMORS

to supratentorial structures. We encountered may adversely effect prognosis. The overall 5-


one patient with hemangioblastoma of one year survival is over 80%.
cerebellar hemisphere and a renal cell metas-
tasis in the other cerebellar hemisphere. The
two looked alike on neuroimaging.
Melanocytic tumors
Treatment Introduction
The treatment is surgical.107 Complete excision Melanocytes normally found in the
results in cure in most instances; recurrence leptomeninges can give rise to either benign or
after total excision is less than 10%. Tumors malignant tumors.117 The tumors may either
in the spinal cord can often be completely form mass lesions or diffusely involve the
excised. Some brainstem tumors can also be leptomeninges. The lesions are divided into
excised, but the morbidity is high. For tumors three groups: diffuse melanosis, a cytologically
that cannot be completely excised, RT should benign, diffuse proliferation of melanocytes
follow subtotal resection. Radiosurgery may involving the leptomeninges;117 melanocytoma,
give better results than standard RT.115 Those a low-grade but ultimately fatal tumor of
patients with VHL disease should have imaging the leptomeninges (Table 6.9); and primary
of the entire nervous system, because tumors melanoma, a malignant meningeal tumor.
are often multiple. Small, asymptomatic tumors Combined, these lesions account for less
might warrant treatment before symptoms than 0.1% of primary CNS tumors. When they
develop, but tumors are frequently so numer- occur as mass lesions, they involve the poste-
ous that many are followed and not treated rior fossa and the spinal canal more frequently
until symptoms develop. Early detection of than supratentorial areas. Primary melanoma
tumors is crucial to ensure a good prognosis, of the leptomeninges is far less common than
and periodic screening of patients with VHL metastatic melanoma and is usually easily
disease is indicated to look for recurrence and distinguished clinically.
to detect new tumors.107 Once-yearly should Melanocytomas and primary meningeal
suffice. Screening should include first-degree melanomas are believed to arise from
relatives.

Prognosis Table 6.9


Clinical findings in 16 patients with intracranial
The prognosis of hemangioblastomas of the meningeal melanocytomas.119
cerebellum depends in part on the effectiveness
of surgery and in part on whether the lesion is Median age at diagnosis 38
sporadic or associated with VHL disease. Female/Male ratio 2.2:1
Complete surgical removal ensures cure. Partial Age at presentation (years) 971
surgical removals are often well controlled with Duration of symptoms (years) 0.114
Location of tumor Posterior fossa
either RT or radiosurgery. Sporadic tumors
Postoperative survival (years) 0.0435
do not become malignant or metastasize.
However, in VHL disease, other tumors may Modified from Clarke et al.118
appear either within or outside the CNS and

211
MENINGEAL TUMORS

melanocytes that are derived from the neural mass lesion. Alternatively, diffuse invasion of
crest. In the normal CNS, melanocytes are the leptomeninges may interfere with normal
localized around the base of the brain, the CSF absorption, leading to hydrocephalus and
ventral medulla oblongata, and along the upper signs of generalized brain dysfunction, includ-
cervical spinal cord, which may explain the ing headache, mental status changes and
predilection for tumors to occur in these areas. ataxia. Symptoms of hydrocephalus may also
There are no known risk factors, and genetic develop later in the course of these tumors.
abnormalities have not been established. MR scan aids in the diagnosis when the
leptomeninges are diffusely involved.117 The
meninges appear thickened and contrast
Pathology
enhance. Mass lesions are either isointense or
Grossly, the lesions may appear as black hyperintense on the T1-weighted image with
tumors involving the subarachnoid space or intense enhancement, and hypointense on the
may give the entire subarachnoid space a dark T2-weighted image. Melanin is paramagnetic
appearance. The tumors may also appear as and may yield hyperintensity on short echo
non-pigmented, fleshy masses. Histologically, time MR sequences. Because the tumors are
melanocytomas consist of monomorphic cells highly vascular, they may hemorrhage, leading
with low mitotic activity. Small amounts of to hyperintensity on the T1-weighted image.
necrosis or hemorrhage may be present. They Lumbar puncture may establish the diagnosis
occur mostly in the spinal cord but occasion- by revealing abnormal melanin-containing cells
ally in the posterior fossa.119 In melanomas, on cytologic examination.121 The cells may show
the cells are pleomorphic with multinucleated immunoreactivity for S100 protein, HMB 45 and
giant cells. Many mitoses, necroses and vimentin. HMB 45 positivity establishes the
hemorrhages are found. The tumors do not diagnosis but does not distinguish melanocytomas
express GFAP, neurofilament, cytokeratins or from malignant melanomas, either primary or
EMA, but most express HMB 45. About 25% metastatic. If there is sufficient melanin in the
of patients with diffuse meningeal melanocy- CSF, it may appear black. More commonly, the
tosis have concomitant cutaneous lesions CSF is xanthochromic, a result of multiple small
(neurocutaneous melanosis),120,121 and approx- hemorrhages. Red cells may or may not be
imately 10% of patients with large melano- present at the time of the examination.
cytic nevi of the skin have CNS melanocytosis. When the patient presents with mass lesions,
Occasionally, melanosis may be associated the diagnosis is usually not made until surgery,
with a melanoma.122 when the surgeon, expecting to encounter an
intrinsic brain tumor, instead finds a black
tumor that involves the leptomeninges123 with
Clinical findings
secondary brain invasion. Biopsy then estab-
The signs and symptoms depend on the lishes the diagnosis. Microscopically, melano-
location of greatest growth. The tumors cytomas and melanomas must be distinguished
begin as leptomeningeal involvement but may from other pigmented lesions of the meninges,
involve the brain either by invasion down including melanotic meningiomas and melan-
VirchowRobin spaces or by compression. otic schwannomas.
Thus, patients may present with seizures, Two important problems in differential
ataxia or spinal cord signs suggesting a focal diagnosis are to distinguish the more benign

212
MISCELLANEOUS TUMORS

melanocytomas from melanomas, and to distin- The prognosis in all of these tumors is poor.
guish primary melanomas of the leptomeninges Although long survival in patients with
from metastatic melanomas. The former can be melanocytoma is possible, including one
done by careful pathologic evaluation, and the patient alive 35 years after total excision and
latter by a search for a primary tumor of the RT of an intracranial melanocytoma, most
skin, either by history or examination at the patients with melanocytomas succumb within
time of diagnosis. Accordingly, all patients with several years and most patients with melanoma
melanotic tumors of the leptomeninges should within several months. Even diffuse melanosis
be examined both for congenital melanotic has a poor prognosis.
lesions of the skin (neurocutaneous melanosis)
or for malignant melanomas that have not been
detected previously.
Miscellaneous tumors
Several other tumors can occasionally arise from
Treatment
the meninges. These include both benign and
If a mass lesion is present, it should be removed malignant tumors. The benign tumors include
to the maximum extent possible. Patients with chondromas, osteomas, lipomas, fibrous histio-
melanomas should receive RT to the involved cytomas, solitary plasmacytomas126 (Chapter 11)
area in the postoperative period. It is not clear and myxomas.127 Malignant tumors include
whether melanocytomas are best treated with chondrosarcomas,128 Ewings sarcoma,
radiation or simply followed until recurrence. rhabdomyosarcoma, 129 leiomyosarcoma130 and
However, the tumors are highly radioresistant, fibrosarcoma. 131 Both high- and low-grade
and it is questionable whether RT has any gliomas can either arise in or metastasize to the
substantial effect. Radiosurgery has been leptomeninges.132 The low-grade gliomas include
reported to be highly effective in the treatment pilocytic astrocytomas133 (Chapter 5). All of these
of brain metastases from melanoma and might tumors can present with meningeal mass lesions
be considered if only a few small areas of the that, when malignant, grow more rapidly than
nervous system are involved in primary most meningiomas. Primary CNS lymphomas
melanocytoma or malignant melanoma.124 characteristically involve the leptomeninges but
Chemotherapy has largely been restricted to rarely as mass lesions (Chapter 11).
the use of chemotherapeutic agents for Sometimes, the diagnosis may be suspected
metastatic melanoma and most are ineffective. by MR or CT scan. For example, lipomas have
Individual reports of dacarbazine, given a characteristic hyperintensity on both T1- and
intrathecally for the treatment of meningeal T2-weighted images and do not contrast
melanoma, and immunotherapeutic agents enhance. Lipomas characteristically involve the
such as Interleukin-2 (IL-2), are limited to a corpus callosum or the cerebellopontine angle.
few case reports.125 Some case reports are In the latter case, they present with hearing
suspect because of the failure to distinguish loss, dizziness and tinnitus. Surgical resection
between melanocytoma and malignant may not remove the entire tumor and may lead
melanoma. Symptomatic hydrocephalus can be to postoperative deficits. Observation may be
relieved by ventriculoperitoneal (VP) shunt the best approach134 until either the patient
even if the underlying tumor cannot be treated becomes symptomatic, or the tumor is seen to
effectively. be growing on serial scans.

213
MENINGEAL TUMORS

Intracranial chondromas are usually found at of the central nervous system. Semin Diagn
the base of the skull, most commonly in the sella Pathol 1997; 14: 25369.
8. Pimentel J, Fernandes A, Pinto AE et al. Clear
and parasellar regions with varying degrees of
cell meningioma variant and clinical aggres-
involvement of the cavernous sinus.135 Rarely, siveness. Clin Neuropathol 1998; 17: 1416.
they are confined to the cavernous sinus. The 9. CBTRUS (2000). Statistical Report: Primary
tumors are slowly growing and are best treated Brain Tumors in the United States,
surgically. Fibrous histiocytomas may be either 19921997. 2000. Central Brain Tumor
benign or malignant.136 Malignant rhabdoid Registry of the United States.
10. Radhakrishnan K, Mokri B, Parisi JE et al.
tumors are a rare, but extremely aggressive, The trends in incidence of primary brain
tumor of childhood of unclear histogenesis but tumors in the population of Rochester,
characteristic histology.137 These lesions are Minnesota. Ann Neurol 1995; 37: 6773.
usually approached surgically and, if malignant, 11. Perry A, Scheithauer BW, Stafford SL et al.
followed by RT. Despite treatment, outcome is Malignancy in meningiomas a clinicopatho-
logic study of 116 patients, with grading
poor due to rapid relapse.
implications. Cancer 1999; 85: 204656.
12. Cerd-Nicols M, Lpez-Gines C, Prez-
Bacete M et al. Histopathological and
References cytogenetic findings in benign, atypical and
anaplastic human meningiomas: a study of 60
1. Sanan A, van Loveren HR. The arachnoid tumors. J Neurooncol 2000; 47: 99108.
and the myth of Arachne. Neurosurgery 13. Sanson M, Cornu P. Biology of meningiomas.
1999; 45: 1525. Acta Neurochir (Wien) 2000; 142: 493505.
2. Bastos AC, Andermann F, Melancon D et al. 14. Evans JJ, Jeun SS, Lee JH et al. Molecular
Late-onset temporal lobe epilepsy and dilata- alterations in the neurofibromatosis Type 2
tion of the hippocampal sulcus by an enlarged gene and its protein rarely occurring in
VirchowRobin space. Neurology 1998; 50: meningothelial meningiomas. J Neurosurg
7847. 2001; 94: 1117.
3. Kleihues P, Cavenee WK. World Health 15. Shoshan Y, Chernova O, Jeun SS et al.
Organization Classification of Tumours: Radiation-induced meningioma: a distinct
Tumours of the Nervous System Pathology molecular genetic pattern? J Neuropathol Exp
and Genetics. Lyon: IRAC Press, 2000. Neurol 2000; 59: 61420.
4. DeMonte F, Marmon E, Al-Mefty O. 16. Schmitz U, Mueller W, Weber M et al. INI1
Meningiomas. In: Kaye AH, Laws ERJ, eds. mutations in meningiomas at a potential
Brain tumors. 2nd edn New York: Churchill hotspot in exon 9. Br J Cancer 2001; 84:
Livingstone, 2001: 71954. 199201.
5. Buhl R, Hugo HH, Mihajlovic Z, Mehdorn 17. Perry A, Jenkins RB, Dahl RJ et al.
HM. Secretory meningiomas: clinical and Cytogenetic analysis of aggressive menin-
immunohistochemical observations. Neuro- giomas: possible diagnostic and prognostic
surgery 2001; 48: 297301. implications. Cancer 1996; 77: 256773.
6. Lamszus K, Lengler U, Schmidt NO et al. 18. Bostrom J, Cobbers JM, Wolter M et al.
Vascular endothelial growth factor, hepato- Mutation of the PTEN (MMAC1) tumor
cyte growth factor/scatter factor, basic fibro- suppressor gene in a subset of glioblastomas
blast growth factor, and placenta growth but not in meningiomas with loss of chromo-
factor in human meningiomas and their some arm 10q. Cancer Res 1998; 58: 2933.
relation to angiogenesis and malignancy. 19. Kim JH, Lee SH, Rhee CH et al. Loss of
Neurosurgery 2000; 46: 93847. heterozygosity on chromosome 22q and 17p
7. Gokden M, Roth KA, Carroll SL et al. Clear correlates with aggressiveness of menin-
cell neoplasms and pseudoneoplastic lesions giomas. J Neurooncol 1998; 40: 1016.

214
REFERENCES

20. Zhu J, Frosch MP, Busque L et al. Analysis receptor antagonist. J Neurosurg 1999; 91:
of meningiomas by methylation- and 939.
transcription-based clonality assays. Cancer 32. Magrassi L, De Fraja C, Conti L et al.
Res 1995; 55: 386572. Expression of the JAK and STAT superfami-
21. Zhu JJG, Maruyama T, Jacoby LB et al. lies in human meningiomas. J Neurosurg
Clonal analysis of a case of multiple menin- 1999; 91: 4406.
giomas using multiple molecular genetic 33. Bondy M, Ligon BL. Epidemiology and etiol-
approaches: pathology case report. ogy of intracranial meningiomas: a review.
Neurosurgery 1999; 45: 40916. J Neurooncol 1996; 29: 197205.
22. Wu JK, MacGillavry M, Kessaris C et al. 34. Das A, Tang WY, Smith DR. Meningiomas
Clonal analysis of meningiomas. Neuro- in Singapore: demographic and biological
surgery 1996; 38: 1196200. characteristics. J Neurooncol 2000; 47:
23. von Deimling A, Larson J, Wellenreuther R 15360.
et al. Clonal origin of recurrent meningiomas. 35. Nishio S, Morioka T, Inamura T et al.
Brain Pathology 1999; 9: 64550. Radiation-induced brain tumours: potential
24. Moresco RM, Scheithauer BW, Lucignani G late complications of radiation therapy for
et al. Oestrogen receptors in meningiomas: a brain tumours. Acta Neurochir (Wien) 1998;
correlative PET and immunohistochemical 140: 76370.
study. Nucl Med Commun 1997; 18: 36. Pollak L, Walach N, Gur R, Schiffer J.
60615. Meningiomas after radiotherapy for tinea
25. Carroll RS, Zhang J, Black PM. Expression capitis still no history. Tumori 1998; 84:
of estrogen receptors alpha and beta in 658.
human meningiomas. J Neurooncol 1999; 42: 37. Ron E, Modan B, Boice JD Jr et al. Tumors
10916. of the brain and nervous system after radio-
26. Koide SS. Mifepristone. Auxiliary therapeutic therapy in childhood. N Engl J Med 1988;
use in cancer and related disorders. J Reprod 319: 10339.
Med 1998; 43: 55160. 38. Kotzen RM, Swanson RM, Milhorat TH,
27. Roelvink NC, Kamphorst W, August H, van Boockvar JA. Post-traumatic meningioma:
Alphen M, Rao BR. Literature statistics do case report and historical perspective. J
not support a growth stimulating role for Neurol Neurosurg Psychiatry 1999; 66: 796.
female sex steroid hormones in haeman- 39. Preston-Martin S, Pogoda JM, Schlehofer B
giomas and meningiomas. J Neurooncol et al. An international casecontrol study of
1991; 11: 24353. adult glioma and meningioma: the role of head
28. Lambe M, Coogan P, Baron J. Reproductive trauma. Int J Epidemiol 1998; 27: 579-86.
factors and the risk of brain tumors: a 40. Markopoulos C, Sampalis F, Givalos N,
population-based study in Sweden. Int J Gogas H. Association of breast cancer with
Cancer 1997; 72: 38993. meningioma. Eur J Surg Oncol 1998; 24:
29. Karlsson AE, Bergenheim AT, Bjrk P, 3324.
Henriksson R. Estramustine-binding protein 41. Lee A, Wallace C, Rewcastle B, Sutherland G.
in meningioma. Acta Neuropathol (Berl) Metastases to meningioma. Am J Neuroradiol
1999; 98: 13540. 1998; 19: 11202.
30. Christov C, Lechapt-Zalcman E, Adle- 42. Kondziolka D, Flickinger JC, Perez B.
Biassette H, Nachev S, Gherardi RK. Judicious resection and/or radiosurgery for
Vascular permeability factor/vascular endo- parasagittal meningiomas: outcomes from
thelial growth factor (VPF/VEGF) and its a multicenter review. Gamma Knife
receptor flt-1 in microcystic meningiomas. Meningioma Study Group. Neurosurgery
Acta Neuropathol (Berl) 1999; 98: 41420. 1998; 43: 40513.
31. Friend KE, Radinsky R, McCutcheon IE. 43. Perry A, Stafford SL, Scheithauer BW, Suman
Growth hormone receptor expression and VJ, Lohse CM. The prognostic significance of
function in meningiomas: effect of a specific MIB-1, p53, and DNA flow cytometry in

215
MENINGEAL TUMORS

completely resected primary meningiomas. 56. Oka K, Go Y, Kimura H, Tomonaga M.


Cancer 1998; 82: 22629. Obstruction of the superior sagittal sinus
44. Perry A, Scheithauer BW, Stafford SL, Abell- caused by parasagittal meningiomas: the role
Aleff PC, Meyer FB. Rhabdoid meningioma: of collateral venous pathways. J Neurosurg
an aggressive variant. Am J Surg Pathol 1998; 1994; 81: 5204.
22: 148290. 57. Anegawa S, Hayashi T, Torigoe R, Furukawa
45. Enam SA, Abdulrauf S, Mehta B, Malik GM, Y. Diffuse calvarial meningioma case report
Mahmood A. Metastasis in meningioma. and review of the literature. J Neurosurg
Acta Neurochir (Wien) 1996; 138: 11727. 1999; 90: 9703.
46. Go RS, Taylor BV, Kimmel DW. The natural 58. Engelhard HH. Progress in the diagnosis and
history of asymptomatic meningiomas in treatment of patients with meningiomas. Part
Olmsted County, Minnesota. Neurology I: diagnostic imaging, preoperative emboliza-
1998; 51: 171820. tion. Surg Neurol 2001; 55: 89101.
47. Braunstein JB, Vick NA. Meningiomas: the 59. Ildan F, Tuna A, Ger AI et al. Correlation
decision not to operate. Neurology 1997; 48: of the relationships of braintumor interfaces,
145962. magnetic resonance imaging, and angio-
48. Bornstein RA, Witt NJ. Are right-hemisphere graphic findings to predict cleavage of menin-
lesions really larger? Lesion size and lateral- giomas. J Neurosurg 1999; 91: 38490.
ity in meningioma patients. Acta Neurol 60. Pieper DR, Al-Mefty O, Hanada Y, Buechner
Scand 1984; 69: 17681. D. Hyperostosis associated with meningioma
49. Talacchi A, Lombardo C, Bricolo A. Vascular of the cranial base: secondary changes or
headache due to intracranial meningioma: a tumor invasion. Neurosurgery 1999; 44:
curable form of headache. Lancet 1997; 350: 7426.
10045. 61. Pierallini A, Bonamini M, Di Stefano D,
50. Bitzer M, Opitz H, Popp J et al. Angiogenesis Siciliano P, Bozzao L. Pleomorphic xantho-
and brain oedema in intracranial meningiomas: astrocytoma with CT and MRI appearance of
influence of vascular endothelial growth factor. meningioma. Neuroradiology 1999; 41: 304.
Acta Neurochir (Wien) 1998; 140: 33340. 62. Kizana E, Lee R, Young N, Dorsch NW, Soo
51. Bitzer M, Opitz H, Popp J, Morgalla M, YS. A review of the radiological features of
Gruber A, Heiss E, Voigt K. Angiogenesis and intracranial meningiomas. Australas Radiol
brain oedema in intracranial meningiomas: 1996; 40: 45462.
influence of vascular endothelial growth factor. 63. Hildebrandt G, Scheidhauer K, Luyken C et
Acta Neurochir (Wein) 1998; 140: 33340. al. High sensitivity of the in vivo detection of
52. Hirashima Y, Hayashi N, Fukuda O et al. somatostatin receptors by 111indium (DTPA-
Platelet-activating factor and edema octreotide)-scintigraphy in meningioma
surrounding meningiomas. J Neurosurg 1998; patients. Acta Neurochir (Wien) 1994; 126:
88: 3047. 6371.
53. Constantini S, Tamir J, Gomori MJ, Shohami 64. Iuchi T, Iwadate Y, Namba H et al. Glucose
E. Tumor prostaglandin levels correlate with and methionine uptake and proliferative
edema around supratentorial meningiomas. activity in meningiomas. Neurol Res 1999;
Neurosurgery 1993; 33: 20410. 21: 6404.
54. Ide M, Jimbo M, Yamamoto M et al. MIB-1 65. Nyberg G, Bergstrom M, Enblad P, Lilja A,
staining index and peritumoral brain edema Muhr C, Langstrom B. PET-methionine of
of meningiomas. Cancer 1996; 78: 13343. skull base neuromas and meningiomas. Acta
55. Mantle RE, Lach B, Delgado MR, Baeesa S, Otolaryngol (Stockh) 1997; 117: 4829.
Blanger G. Predicting the probability of 66. Yamaki T, Ikeda T, Sakamoto Y, Ohtaki M,
meningioma recurrence based on the quantity Hashi K. Lymphoplasmacyte-rich menin-
of peritumoral brain edema on computerized gioma with clinical resemblance to inflamma-
tomography scanning. J Neurosurg 1999; 91: tory pseudotumor. Report of two cases. J
37583. Neurosurg 1997; 86: 898904.

216
REFERENCES

67. Nakasu S, Nakasu Y, Nakajima M, Matsuda cavernous sinus meningiomas. J Neurosurg


M, Handa J. Preoperative identification of 2000; 93 Suppl 3: 6873.
meningiomas that are highly likely to recur. J 78. Kondziolka D, Flickinger JC, Perez B,
Neurosurg 1999; 90: 45562. Gamma Knife Meningioma Study Group.
68. Lee JH, Jeun SS, Evans J, Kosmorsky G. Judicious resection and/or radiosurgery for
Surgical management of clinoidal menin- parasagittal meningiomas: outcomes from a
giomas. Neurosurgery 2001; 48: 10129. multicenter review. Neurosurgery 1998; 43:
69. Selesnick SH, Nguyen TD, Gutin PH, Lavyne 40513.
MH. Posterior petrous face meningiomas. 79. Roche PH, Regis J, Dufour H et al. Gamma
Otolaryngol Head Neck Surg 2001; 124: knife radiosurgery in the management of
40813. cavernous sinus meningiomas. J Neurosurg
70. Patsouris E, Laas R, Hagel C, Stavrou D. 2000; 93 Suppl 3: 6873.
Increased proliferative activity due to 80. Singh VP, Kansal S, Vaishya S, Julka PK,
necroses induced by pre-operative emboliza- Mehta VS. Early complications following
tion in benign meningiomas. J Neurooncol gamma knife radiosurgery for intracranial
1998; 40: 25764. meningiomas. J Neurosurg 2000; 93: 5761.
71. Hug EB, DeVries A, Thornton AF et al. 81. Stelzer KJ. Acute and long-term complica-
Management of atypical and malignant tions of therapeutic radiation for skull base
meningiomas: role of high-dose, 3D-confor- tumors. Neurosurg Clin N Am 2000; 11:
mal radiation therapy. J Neurooncol 2000; 597604.
48: 15160. 82. Kyritsis AP. Chemotherapy for meningiomas.
72. Milosevic MF, Frost PJ, Laperriere NJ, J Neurooncol 1996; 29: 26972.
Wong CS, Simpson WJ. Radiotherapy for 83. Schrell UMH, Rittig MG, Anders M et al.
atypical or malignant intracranial menin- Hydroxyurea for treatment of unresectable
gioma. Int J Radiat Oncol Biol Phys 1996; and recurrent meningiomas. II. Decrease in
34: 81722. size of meningiomas in patients treated with
73. Maire JP, Caudry M, Guerin J et al. hydroxyurea. J Neuro Surg 1997; 86: 8404.
Fractionated radiation therapy in the treat- 84. Kaba SE, DeMonte F, Bruner JM et al. The
ment of intracranial meningiomas: local treatment of recurrent unresectable and
control, functional efficacy, and tolerance in malignant meningiomas with interferon
91 patients. Int J Radiat Oncol Biol Phys alpha-2B. Neurosurgery 1997; 40: 2715.
1995; 33: 31521. 85. Pereda MP, Hopfner U, Pagotto U et al.
74. Connell PP, Macdonald RL, Mansur DB, Retinoic acid stimulates meningioma cell
Nicholas MK, Mundt AJ. Tumor size predicts adhesion to the extracellular matrix and
control of benign meningiomas treated with inhibits invasion. Br J Cancer 1999; 81: 3816.
radiotherapy. Neurosurgery 1999; 44: 11949. 86. McCarthy BJ, Davis FG, Freels S et al.
75. Dufour H, Muracciole X, Mtellus P et al. Factors associated with survival in patients
Long-term tumor control and functional with meningioma. J Neurosurg 1998; 88:
outcome in patients with cavernous sinus 8319.
meningiomas treated by radiotherapy with 87. Stafford SL, Perry A, Suman VJ et al.
or without previous surgery: is there an Primarily resected meningiomas: outcome and
alternative to aggressive tumor removal? prognostic factors in 581 Mayo Clinic
Neurosurgery 2001; 48: 28594. patients, 1978 through 1988. Mayo Clin Proc
76. Kondziolka D, Levy EI, Niranjan A, 1998; 73: 93642.
Flickinger JC, Lunsford LD. Long-term 88. Dziuk TW, Woo S, Butler EB et al. Malignant
outcomes after meningioma radiosurgery: meningioma: an indication for initial aggres-
physician and patient perspectives. J sive surgery and adjuvant radiotherapy. J
Neurosurg 1999; 91: 4450. Neurooncol 1998; 37: 17788.
77. Roche PH, Regis J, Dufour H et al. Gamma 89. Nakaguchi H, Fujimaki T, Matsuno A et al.
knife radiosurgery in the management of Postoperative residual tumor growth of

217
MENINGEAL TUMORS

meningioma can be predicted by MIB-1 100. Brunori A, Delitala A, Oddi G, Chiappetta F.


immunohistochemistry. Cancer 1999; 85: Recent experience in the management of
224954. meningeal hemangiopericytomas. Tumori
90. Kalkanis SN, Quiones-Hinojosa A, Buzney E, 1997; 83: 85661.
Ribaudo HJ, Black PM. Quality of life follow- 101. Spitz FR, Bouvet M, Pisters PW, Pollock RE,
ing surgery for intracranial meningiomas at Feig BW. Hemangiopericytoma: a 20-year
Brigham and Womens Hospital: a study of single-institution experience. Ann Surg Oncol
164 patients using a modification of the 1998; 5: 3505.
functional assessment of cancer therapybrain 102. Dufour H, Mtellus P, Fuentes S et al.
questionnaire. J Neurooncol 2000; 48: 23341. Meningeal hemangiopericytoma: A retrospec-
91. Tucha O, Smely C, Lange KW. Effects of tive study of 21 patients with special review
surgery on cognitive functioning of elderly of postoperative external radiotherapy.
patients with intracranial meningioma. Br J Neurosurgery 2001; 48: 75663.
Neurosurg 2001; 15: 1848. 103. Bastin KT, Mehta MP. Meningeal hemangio-
92. Galanis E, Buckner JC, Scheithauer BW, pericytoma: defining the role for radiation
Kimmel DW, Schomberg PJ, Piepgras DG. therapy. J Neuro oncology 1992; 14: 27787.
Management of recurrent meningeal hem- 104. Kocher M, Voges J, Staar S et al. Linear
angiopericytoma. Cancer 1998; 82: 191520. accelerator radiosurgery for recurrent malig-
93. Abrahams JM, Forman MS, Lavi E, Goldberg nant tumors of the skull base. Am J Clin
H, Flamm ES. Hemangiopericytoma of the Oncol 1998; 21: 1822.
third ventricle. Case report. J Neurosurg 105. Younis GA, Sawaya R, DeMonte F, Hess KR,
1999; 90: 35962. Albrecht S, Bruner JM. Aggressive meningeal
94. Ono Y, Ueki K, Joseph JT, Louis DN. tumors: review of a series. J Neurosurg 1995;
Homozygous deletions of the CDKN2/p16 82: 1727.
gene in dural hemangiopericytomas. Acta 106. Nakaguchi H, Fujimaki T, Matsuno A,
Neuropathol (Berl) 1996; 91: 2215. Matsuura R, Asai A, Suzuki I, Sasaki T,
95. Sandhu FA, Martuza RL. Craniofacial hem- Kirino T. Postoperative residual tumor
angiopericytoma associated with oncogenic growth of meningioma can be predicted by
osteomalacia: case report. J Neurooncol 2000; MIB-1 immunohistochemistry. Cancer 1999;
46: 2417. 85: 224954.
96. Guthrie BL, Ebersold MJ, Scheithauer BW, 107. Conway JE, Chou D, Clatterbuck RE et al.
Shaw EG. Meningeal hemangiopericytoma: Hemangioblastomas of the central nervous
histopathological features, treatment, and system in von HippelLindau syndrome and
long-term follow-up of 44 cases. sporadic disease. Neurosurgery 2001; 48: 5562.
Neurosurgery 1989; 25: 51422. 108. Richard S, Campello C, Taillandier L, Parker
97. Hoekman K, van Doorn J, Gloudemans T, F, Resche F. Haemangioblastoma of the
Maassen JA, Schuller AG, Pinedo HM. central nervous system in von HippelLindau
Hypoglycaemia associated with the produc- disease. French VHL Study Group. J Intern
tion of insulin-like growth factor II and Med 1998; 243: 54753.
insulin-like growth factor binding protein 6 109. Kawahara N, Kume H, Ueki K et al. VHL
by a haemangiopericytoma. Clin Endocrinol gene inactivation in an endolymphatic sac
(Oxf) 1999; 51: 24753. tumor associated with von HippelLindau
98. Nelson AE, Namkung HJ, Patava J et al. disease. Neurology 1999; 53: 20810.
Characteristics of tumor cell bioactivity in 110. Chang JH, Spraul CW, Lynn ML, Drack A,
oncogenic osteomalacia. Mol Cell Endocrinol Grossniklaus HE. The two-stage mutation
1996; 124: 1723. model in retinal hemangioblastoma.
99. Kracht LW, Bauer A, Herholz K et al. Ophthalmic Genet 1998; 19: 12330.
Positron emission tomography in a case of 111. Kondo K, Kaelin WG, Jr. The von Hippel
intracranial hemangiopericytoma. J Comput Lindau tumor suppressor gene. Exp Cell Res
Assist Tomogr 1999; 23: 3658. 2001; 264: 11725.

218
REFERENCES

112. Vaquero J, Zurita M, Oya S, Coca S, Salas 123. Barut S. Primary leptomeningeal melanoma
C. Vascular permeability factor expression simulating a meningioma. Neurosurg Rev
in cerebellar hemangioblastomas: correlation 1995; 18: 1437.
with tumor-associated cysts. J Neurooncol 124. Lavine SD, Petrovich Z, Cohen-Gadol AA et
1999; 41: 37. al. Gamma knife radiosurgery for metastatic
113. Lee JY, Dong SM, Park WS et al. Loss of melanoma: an analysis of survival, outcome,
heterozygosity and somatic mutations of the and complications. Neurosurgery 1999; 44:
VHL tumor suppressor gene in sporadic 5964.
cerebellar hemangioblastomas. Cancer Res 125. Fathallah-Shaykh HM, Zimmerman C,
1998; 58: 5048. Morgan H et al. Response of primary
114. Olschwang S, Richard S, Boisson C et al. leptomeningeal melanoma to intrathecal
Germline mutation profile of the VHL gene recombinant interleukin-2 a case report.
in von HippelLindau disease and in sporadic Cancer 1996; 77: 154450.
hemangioblastoma. Hum Mutat 1998; 12: 126. Vaicys C, Schulder M, Wolansky LJ,
42430. Fromowitz FB. Falcotentorial plasmacytoma
115. Jawahar A, Kondziolka D, Garces YI et al. case report. J Neurosurg 1999; 91: 1325.
Stereotactic radiosurgery for hemangioblas- 127. Graham JF, Loo SYT, Matoba A. Primary
tomas of the brain. Acta Neurochir (Wien) brain myxoma, an unusual tumor of
2000; 142: 6414. meningeal origin: case report. Neurosurgery
116. Brat DJ, Giannini C, Scheithauer BW, Burger 1999; 45: 1669.
PC. Primary melanocytic neoplasms of the 128. Forbes RB, Eljamel MS. Meningeal
central nervous system. Am J Surg Pathol chondrosarcomas, a review of 31 patients. Br
1999; 23: 74554. J Neurosurg 1998; 12: 4614.
117. Byrd SE, Darling CF, Tomita T et al. MR 129. Kobayashi S, Hirakawa E, Sasaki M,
imaging of symptomatic neurocutaneous Ishikawa M, Haba R. Meningeal
melanosis in children. Pediatr Radiol 1997; rhabdomyosarcoma. Report of a case with
27: 3944. cytologic, immunohistologic and ultra-
118. Clarke DB, Leblanc R, Bertrand G, Quartey structural studies. Acta Cytol 1995; 39:
GR, Snipes GJ. Meningeal melanocytoma. 42834.
Report of a case and a historical comparison. 130. Bejjani GK, Stopak B, Schwartz A, Santi R.
J Neurosurg 1998; 88: 11621. Primary dural leiomyosarcoma in a patient
119. Prabhu SS, Lynch PG, Keogh AJ, Parekh HC. infected with human immunodeficiency virus:
Intracranial meningeal melanocytoma: a case report. Neurosurgery 1999; 44:
report of two cases and a review of the liter- 199202.
ature. Surg Neurol 1993; 40: 51621. 131. Donnet A, Figarella-Branger D, Grisoli F.
120. DeDavid M, Orlow SJ, Provost N et al. Primary meningeal fibrosarcoma: a particular
Neurocutaneous melanosis: clinical features neuroradiological presentation. J Neurooncol
of large congenital melanocytic nevi in 1999; 42: 7983.
patients with manifest central nervous system 132. Ng HK, Poon WS. Primary leptomeningeal
melanosis. J Am Acad Dermatol 1996; 35: astrocytoma. Case report. J Neurosurg 1998;
52938. 88: 5869.
121. Alameda F, Lloreta J, Galito E, Roquer J, 133. Tamura M, Zama A, Kurihara H et al.
Serrano S. Meningeal melanocytoma: a case Management of recurrent pilocytic astro-
report and literature review. Ultrastruct cytoma with leptomeningeal dissemination in
Pathol 1998; 22: 34956. childhood. Childs Nerv Syst 1998; 14:
122. Murray C, DIntino Y, MacCormick R, 61722.
Nassar B, Walsh N. Melanosis in association 134. Bigelow DC, Eisen MD, Smith PG et al.
with metastatic malignant melanoma: report of Lipomas of the internal auditory canal and
a case and a unifying concept of pathogenesis. cerebellopontine angle. Laryngoscope 1998;
Am J Dermatopathol 1999; 21: 2830. 108: 145969.

219
MENINGEAL TUMORS

135. Brownlee RD, Sevick RJ, Rewcastle NB, 137. Yachnis AT, Neubauer D, Muir D.
Tranmer BI. Intracranial chondroma. Am J Characterization of a primary central nervous
Neuroradiol 1997; 18: 88993. system atypical teratoid/rhabdoid tumor and
136. Martinez-Salazar A, Supler M, Rojiani AM. derivative cell line: immunophenotype and
Primary intracerebral malignant fibrous histi- neoplastic properties. J Neuropathol Exp
ocytoma: immunohistochemical findings and Neurol 1998; 57: 96171.
etiopathogenetic considerations. Mod Pathol
1997; 10: 14954.

220
7
Neuronal, mixed neuronalglial and embryonal tumors

Introduction
Since Lhermitte and Duclos first described a
neuronal tumor of the cerebellum in 1920, the
field of neuronal and mixed neuronalglial
C
neoplasms has been fraught with controversy. D
One reason is that until recently neurologists A
and neuropathologists have had a strong bias
against the diagnosis of neuronal neoplasm
because of the view that all neurons were
postmitotic and neuronal precursors did not
exist in the adult brain. Current evidence B F
indicates that this is untrue and may lead
neuropathologists to reconsider this preconcep-
tion. A second reason has been that glial
E
tumors may infiltrate cerebral cortex widely,
and it often becomes difficult to distinguish
neurons that are trapped within a glial tumor
from neurons that are themselves neoplastic.
Nevertheless, a number of advances in
Figure 7.1
neurology, neuroimaging and neuropathology Location of neuronal, mixed neuronalglial and
have led neuropathologists to diagnose these embryonal tumors. Central neurocytomas are found
rare neuronal tumors more frequently (Fig. in or close to the lateral ventricle (A). DNTs are
7.1). Those advances include non-invasive found in the cortex of the temporal lobe (B).
imaging techniques that allow diagnosis of Gangliogliomas and ganglioneurocytomas are found
in the frontal lobe, involving both cortex and
neoplasms in patients who are asymptomatic
subcortex (C). Subependymal giant cell astrocytomas
save for seizures. A second advance has been arise from the wall of the lateral ventricles (D).
the surgical treatment of epilepsy. Early on it Medulloblastomas are found in the inferior vermis
was discovered that resections for temporal of the cerebellum (E). Other primitive
lobe epilepsy often identified cortically based, neuroectodermal tumors can arise anywhere in the
otherwise asymptomatic tumors, most of which central nervous system, particularly in the cerebral
hemispheres (F).
were probably dysembryoplastic neuroepithe-
lial tumors (DNT) (see below). The recent

221
NEURONAL, MIXED NEURONALGLIAL AND EMBRYONAL TUMORS

Table 7.1
Gangliocytoma Neuronal, mixed neuronalglial
Dysplastic gangliocytoma of cerebellum (LhermitteDuclos) (Chapter 12) and neuroblastic tumors.
Desmoplastic infantile astrocytoma/ganglioglioma
Dysembryoplastic neuroepithelial tumor
Ganglioglioma
Anaplastic (malignant) ganglioglioma
Central neurocytoma
Paraganglioma of the filum terminale
Olfactory neuroblastoma (esthesioneuroblastoma) (Chapter 9)
Variant: olfactory neuroepithelioma

more aggressive surgical approach to epilepsy This chapter also considers tumors classified
is identifying more such tumors. A third by the WHO as embryonal in origin because
advance has been in neuropathology and some evidence suggests these tumors may be
immunohistochemistry. The identification of neuronal or neuronal precursor in origin.
synaptic vesicles in neoplastic cells by electron Unlike the other neuronal tumors, they are not
microscopy allows the neuropathologist to rare, but they also primarily affect the young.
distinguish the rare central neurocytoma from The neuronal and embryonal tumors
the more common similarly appearing oligo- discussed in this chapter generally affect
dendroglioma. The immunohistochemical stain children and young adults, with no particular
for synaptophysin also reveals synaptic activity sex or ethnic predilection. Their exact
in neuronal tumors. Absence of glial fibrillary incidence is unknown, but they may constitute
acidic proteins (GFAP) supports the diagnosis. as many as 5% of primary brain tumors and,
Perhaps more important has been the discov- in selected populations such as children and
ery of two neuronal nuclear markers, Neu-N1 adults with pharmaco-resistant epilepsy, the
and anti-Hu,2 which appear, when positive, to incidence may be much higher.
unequivocally identify neoplastic cells as Unlike gliomas, neuronal and neuronalglial
neuronal in origin. The lack of a reaction with tumors are usually low-grade and have a
these neuronal markers, however, does not relatively benign prognosis. They often present
exclude the possibility that the tumor arose with seizures and, before modern imaging
from a neuronal precursor. techniques, patients could suffer seizures for
Table 7.1 shows the World Health many years (as long as 39 years in one report)
Organization (WHO) classification of tumors before a diagnosis was made. Embryonal
believed to be of neuronal or mixed neuronal tumors, although probably neuronal in origin
and glial origin. These tumors usually affect (at least, often expressing neuronal markers),2
children or young adults and, if in the cerebral differ from the relatively benign neuronal and
hemisphere, usually present as epilepsy. The neuronalglial tumors in that they are much
neuropathologic classification has undergone a more aggressive and do not respond well to
dramatic change between the first WHO current therapeutic endeavors. The characteris-
edition in 1979 and the current edition in tics of neuronal and neuronalglial tumors are
20003 for some of the reasons indicated above. shown in Table 7.2.

222
NEURONAL AND NEURONALGLIAL TUMORS

Table 7.2
Characteristics of neuronal and neuronalglial tumors.

Age Preferential/ Imaging characteristics


(years) at pathognomonic pathognomonic
presentation site

Ganglioglioma/gangliocytoma < 30 Temporal lobe Cystic, calcified, sharply


demarcated, frequently
enhancing
Dysplastic Ganz gliocytoma 2040 Cerebellum Striated pattern, not enhancing
of the cerebellum
(LhermitteDuclos)
Desmoplastic infantile <2 Hemispheric Massive, multicystic,
ganglioglioma multidensity/intensity, solid
portion enhancing meningeal
attachment
Central neurocytoma 2040 Intraventricular Multicystic, attached to
septum pellucidum/lateral
ventricle wall
Prominent enhancement
Dysembryoplastic < 30 Temporal lobe Cystic with 12 of tumors
neuroepithelial tumor enhancing
Cortical
Multinodular

From Krouwer18 with permission.

Neuronal and neuronalglial and, with the exception of one tumor associ-
ated with von HippelLindau (VHL) disease,
tumors no familial incidence. The genetics of central
Central neurocytoma neurocytomas are not established but chromo-
somal imbalances have been found on 2p, 10q,
This tumor was identified as a separate 18q.7 Gain of chromosome 7 has been
histopathological entity in 1982. Central observed in a few patients but epidermal
neurocytomas arise from subependymal matrix growth factor receptor located on chromosome
cells close to a lateral ventricle and usually 7, is not amplified.8 p53 mutations have not
present as an intraventricular mass within the been detected.9
frontal horn of one or both lateral ventricles4
(Fig. 7.2) or, rarely, the 4th ventricle5 or Pathology
elsewhere.6 They represent less than 1% of Central neurocytomas look like oligodendro-
intracranial tumors and have a peak incidence gliomas, including the presence of gross calcifi-
between 20 and 40 years of age. They affect cation and occasionally hemorrhage. The tumors
both sexes equally. There are no known consist of round cells with round or oval nuclei
environmental risk factors for neurocytoma and finely speckled chromatin. There is a promi-

223
NEURONAL, MIXED NEURONALGLIAL AND EMBRYONAL TUMORS

Figure 7.2
A central neurocytoma. An unenhanced (left) and enhanced (right) MRI showing the typical honeycombed
appearance of a central neurocytoma obstructing the ventricular system and causing hydrocephalus. The upper
photomicrograph shows the typical H&E histology of a central neurocytoma. The cellular elements are bland
round cells with clear halos mimicking oligodendroglioma cells, neuropil and a hypocellular zone (arrow). The
intense neuronal nuclear staining with the anti-Hu antibody (lower photomicrograph) indicates that this is a
neuronal tumor, not an oligodendroglioma.

nent nucleus and scanty cytoplasm. The correct filaments are not seen, but vascular endothelial
diagnosis is made by either immunohisto- growth factor (VEGF) and the neurotrophin
chemistry or electron microscopy.10 Synapto- receptor protein, Trk11 immunoreactivity may be
physin stains reveal the tumor to be of neuronal present, as may photoreceptors.12 Electron
origin, as do the more recently used markers, microscopy identifies features typical of neurons,
Neu-N1 and anti-Hu.2 GFAP is usually absent, including synapses in some instances.10
but occasional tumor cells and reactive astrocytes Mitoses and necrosis are uncommon. The
may be GFAP and vimentin positive.10 Neuro- MIB-1 labeling index ranges from 0.1 to 6.0,

224
NEURONAL AND NEURONALGLIAL TUMORS

with little correlation between histologic MR scan. T2-weighted images are usually
atypia and the labeling index. Whether histo- hyperintense but may have areas of
logic atypia (i.e. cellular pleomorphism, hypointensity representing hemosiderin. The
endothelial proliferation and necrosis) or the tumors intensely contrast enhance, revealing a
labeling index predicts outcome is controver- sharply demarcated mass within the ventricle.
sial.1315 SPECT scan has been reported to demonstrate
significant uptake of technetium-99-labeled
hexamethylpropyleneamine oxine (HMPAO)
Clinical findings and thallium-201 chloride.17
Because the tumor arises in the ventricular Because of their pathological appearance,
system, it usually presents with hydrocephalus central neurocytomas were once thought to be
due to blockage of the foramen of Monro. intraventricular oligodendrogliomas or ependy-
Headache, nausea, vomiting, diplopia and momas. Also included in the differential
visual field defects secondary to compression of diagnosis are subependymomas, giant cell
the optic chiasm are common. Some patients astrocytomas, choroid plexus papillomas,
develop pituitary insufficiency as a result of meningiomas and other clear cell lesions.
compression of the pituitary stalk. Other
hormonal dysfunctions, including gigantism in
children and inappropriate secretion of anti- Treatment
diuretic hormone, have been associated with The treatment of neurocytoma and other
neurocytomas involving the septum, 3rd ventri- neuronal or mixed neuronalglial tumors is
cle and hypothalamus. The tumor does have outlined in Fig. 7.3. Surgery is the treatment
the capacity to invade the parenchyma and of choice. The surgeon should aim for a
cause focal neurologic deficits, including complete resection, after which no further
seizures and hemiparesis. Although benign in therapy is indicated. Even partial resections
their behavior and histologic characteristics, may be associated with prolonged survival.
central neurocytomas occasionally seed the Accordingly, we recommend follow-up with
leptomeninges.16 Because of the generalized careful monitoring for patients with both
symptoms of hydrocephalus, the history is complete and partial resections who are
usually of short duration before the diagnosis otherwise asymptomatic. Follow-up should
is established. be especially scrupulous in patients whose
The MRI of neurocytomas is characteristic. tumor shows an elevated labeling index.13
T1-weighted images demonstrate an intra- Recurrence can be treated by further surgery
ventricular mass which is usually isointense or followed by RT or by RT without re-resec-
hyperintense compared to surrounding brain. tion.19 Radiosurgery has also been reported to
Speckled areas of hypointensity represent be effective.20 If the patient is symptomatic
calcifications that occur in 5060% of after surgery from residual tumor, external
tumors. These are better seen on CT scan. beam radiation therapy to a dose of 54 Gy is
Microcystic regions may also appear as probably the best approach. Responses to
hypointense areas, giving the tumor a honey- chemotherapy have been reported, but
combed appearance. The tumors sometimes chemotherapy should probably be reserved
hemorrhage, although usually not symptomat- for recurrence after surgery and RT or
ically, and evidence of blood may be found on leptomeningeal dissemination.21

225
NEURONAL, MIXED NEURONALGLIAL AND EMBRYONAL TUMORS

Figure 7.3
Algorithm for treatment
Surgical resection, as
radical as possible
of neuronal and mixed
neuronalglial tumors.
Patients with a recurrent
central neurocytoma
should have an MR scan
Neurologic and neuroradiologic monitoring of the spine without and
with contrast. (From
Krouwer18 with
permission.)

Recurrence

Radical resection (MRI


Subtotal resection/biopsy
documented)

Neurologic and neuroradiologic Radiation therapy (or


monitoring chemotherapy if patient <12
years)

Prognosis epilepsy surgery for the treatment of long-


The tumor is relatively benign, with a 5-year standing partial, usually temporal lobe,
actuarial survival rate of 81% and a 5-year seizures (Fig. 7.4). These tumors represent
local control rate of 100% after gross total only a fraction of 1% of intracranial tumors.
resection,22 but recurrences and meningeal DNTs are of mixed glial and neuronal origin
dissemination may occur.23 The tumor may but, unlike most other neuroepithelial tumors,
become more aggressive in pathologic appear- predominantly involve the temporal lobe
ance with each recurrence. cerebral cortex rather than the white matter.
Occasionally, the tumors are found elsewhere,
including the septum pellucidum26 and poste-
rior fossa. DNTs usually occur in infants and
Dysembryoplastic neuroepithelial children but have been reported in young and
tumor (DNT) occasionally elderly adults. Among patients
undergoing epilepsy surgery in whom a
This low-grade cortical-based tumor24,25 was tumor is encountered, DNTs are relatively
originally identified in patients undergoing common.25

226
NEURONAL AND NEURONALGLIAL TUMORS

Figure 7.4
A DNT in a young
adult with seizures.
The panel on the left
is a T1-weighted MRI
demonstrating the
typical cortical
involvement and
temporal lobe location
of this lesion (arrow).
Contrast enhancement
may be present. The
panel on the right is a
T2-weighted image.
The photomicrograph
shows sparse
collections of
oligodendroglia-like
cells and neurons
(arrow) floating in a
myxoid matrix. The
neural elements stain
with neuronal markers
such as anti-Hu.

Etiology and non-specific have been described. In the


The only established risk factor for DNT is its simple form, glioneuronal elements oriented
occasional occurrence in patients with neuro- perpendicular to the cortical surface are
fibromatosis type-1. Genetic abnormalities present, and columns are formed by bundles of
have not been identified. Telomerase expres- axons lined by small oligodendrocyte-like cells.
sion27 and PTEN mutations28 are not present. Between the neoplastic columns, neurons
The apoptosis associated proteins, bcl and bax, appear to float within an interstitial fluid.31
are expressed in tumor cells, suggesting that The complex form, in addition to these
they may play a role in pathogenesis.29 Brain changes, has a micronodular architecture
neurotrophic factor (BNDF) and its receptor consisting of neoplastic oligodendrocytes and
(TrkB) are overexpressed in DNTs.30 These astrocytes with foci of cortical dysplasia. The
agents promote neuronal survival and may third form, said to be without specific morpho-
increase neuronal excitability, perhaps playing logic features, cannot be distinguished on histo-
a role in both tumorigenesis and in the seizures logic criteria alone from astrocytomas or other
that so commonly characterize these tumors. gliomas.25 Here the diagnosis is made based on
clinical history, location and MR findings. It is
Pathology not clear that the non-specific form is indeed a
Grossly, the tumor expands the cortex and DNT. In one such patient, a mixed DNT and
appears as multiple firm nodules within the ganglioglioma was identified.32 The MIB-1
cortex. Three histologic types simple, complex labeling index is usually quite low, although

227
NEURONAL, MIXED NEURONALGLIAL AND EMBRYONAL TUMORS

figures up to 8% have been reported. The additional therapy is required. The tumor
tumors react with the neuronal markers, anti- usually does not recur unless the initial resec-
Hu and Neu-N, differentiating even the non- tion is incomplete.37 At 5 years about 60% of
specific type from pure glioma. Hemorrhage is DNT patients are seizure-free38 Rarely, patients
occasionally present histologically.33 develop psychotic symptoms with paranoid
and depressive features,39 postoperatively after
Clinical findings resection of DNT or temporal lobe ganglio-
As indicated above, most patients present with glioma.
long-standing, usually drug-resistant, partial
seizures with or without secondary generaliza-
tion.34 The seizures usually begin in childhood, Gangliogliomas and
although they have been reported to begin in
gangliocytomas
adulthood. Seizures are usually unassociated
with other neurologic symptoms or signs, These low-grade tumors are composed either
although, rarely, hemorrhage may cause acute of neoplastic neurons (gangliocytoma) or a
neurologic dysfunction.33 combination of neoplastic neurons and glial
On MR scan, the lesions are located corti- cells (ganglioglioma)40 (Fig. 7.5). A rare
cally and usually subcortically as well.35 variant, called ganglioglioneurocytoma, has
About half the tumors are poorly demarcated, also been described.41 Although almost
while the others are sharply demarcated. always low-grade, the glial component has
Tumors are cystic, with most having multiple the capacity to become anaplastic and behave
cysts. There may be a mass effect but aggressively, even disseminating throughout
surrounding edema is usually absent. the leptomeninges. Together, the tumors are
Calcification and occasionally hemorrhage believed to represent about 1% of primary
may be present.33 On T1 images, the lesion intracranial tumors, but up to 4% of
usually appears as a well-defined, sharply pediatric tumors.42 However, in one institu-
marginated, hypointense mass; on T2 it is tion43 the incidence of these tumors was 5%
hyperintense. About half the tumors contrast of intrinsic brain tumors. In selected popula-
enhance, either in a nodular or ring-like tions of resistant focal epilepsy, the incidence
pattern. In long-standing tumors of child- may be even higher. The tumor occurs mostly
hood, deformity of the skull overlying the in children and young adults,44 but the age
mass may be identified on CT scan or even range is from 3 days to 80 years and there is
plain skull films. The deformity results from an equal sex distribution. The tumors may be
chronic pressure of the tumor molding the found anywhere in the central nervous system
childs pliable skull. The tumors are metabol- (CNS) but the majority are found super-
ically inactive on PET scans, either with ficially in the temporal lobe. In one series,
glucose or methionine,36 differentiating them 84% of tumors were in the temporal lobe,
from other childhood low-grade tumors. 10% in the frontal lobe and 4% in the brain-
stem.43 Tumors located in the hemisphere are
Treatment generally diagnosed at an older age than
The treatment is surgical.35 A total resection is those located in midline structures such as the
indicated if possible, but even partial resection optic chiasm, hypothalamus, brainstem and
may control seizures and tumor growth. No spinal cord.

228
NEURONAL AND NEURONALGLIAL TUMORS

Figure 7.5
A ganglioglioma in a young woman with complex partial seizures. At the original resection this tumor was
believed to be an anaplastic astrocytoma, and radiation therapy and chemotherapy were recommended. Careful
histologic review (right) revealed a mixture of neoplastic ganglion cells (arrow) and glial cells embedded in a
collagen-rich matrix with foci of lymphocytic infiltrates (arrowhead). The patient has been followed for several
years without recurrence.

Etiology Pathology
There are no known environmental risk factors. Gangliocytomas are composed of large pleo-
Gangliocytomas and gangliogliomas are usually morphic neurons within a network of collagen
sporadic, but have been reported in association fibers. Perivascular lymphocytic infiltration is
with NF-1.45 Gangliogliomas are usually common, and a few mitotic figures are
karyotypically (from Greek for nucleus, thus sometimes present. The lymphocytic infiltrates
the chromosomal pattern) normal,46 but abnor- may be so prominent as to suggest the diagno-
mal karyotypes, including ring chromosome 1, sis of encephalitis. Eosinophilic granular bodies,
trisomy of chromosomes 5, 6 and 7, and which are liposomes filled with cellular debris,
deletion of chromosome 6, have been reported. are a characteristic feature. Their presence
One report describes aberrant p53 protein suggests the diagnosis of a low-grade ganglion
expression in 20 of 21 specimens.47 Deletions of cell tumor, even when other elements look
portions of chromosome 17p, where p53 aggressive and suggest a diagnosis such as
resides, have been observed in an anaplastic fibrosarcoma. In gangliogliomas, the neural
ganglioglioma with leptomeningeal spread.48 elements may be abundant or sparse. The glial
No PTEN mutations were detected in 16 elements may contain eosinophilic granular or
gangliogliomas.49 As in DNTs (see above) hyaline bodies, and the tumor often contains
BNDF and TrkB are overexpressed.30 microcysts, calcification and an occasional
Immunoreactivity of the stem cell marker CD34 desmoplastic focus. The glial component is often
is present in most gangliogliomas.50 GFAP positive. The neuronal component is anti-

229
NEURONAL, MIXED NEURONALGLIAL AND EMBRYONAL TUMORS

Hu positive. No cells with intermediate histo- seizure type. In the days before MR scanning,
logic features were identified in one series,47 but the seizures were often present for many years,
we have seen cells that are both GFAP and anti- in one case up to 45 years prior to diagnosis.
Hu reactive.2 Electron microscopy identifies Electroencephalograms are often performed to
dense core granules and sometimes synaptic evaluate the seizure disorder; most are abnor-
junctions, indicating the neuronal component. mal but may misidentify tumor location.51
Labeling indices are usually low, with MIB-1 Those tumors that present in the midline often
varying from 1% to 3%. Malignant ganglion present with hydrocephalus and signs of
cells are rare. Extreme care should be taken in increased intracranial pressure, including
evaluating these tumors neuropathologically. headache, nausea and vomiting. Brainstem
Inexperienced neuropathologists may interpret tumors present with cranial nerve dysfunction
the mildly pleomorphic cells as representing an and ataxia. When the lesion is in the midline,
anaplastic astrocytoma rather than a ganglio- symptoms are only present for a short time
glioma. Careful review, considering the clinical before diagnosis is made.
symptomatology, the pre- and postoperative MR On MR scan, most tumors are hypointense
scans, and the immunohistochemistry, will on the T1-weighted image and hyperintense on
usually reveal the correct diagnosis.47 the T2-weighted image. About half of the
tumors have a cystic component. The tumors
Clinical findings usually contrast enhance but not all do. The
The symptoms and signs depend on the tumors most characteristic finding is a cystic tumor
location. The vast majority of patients present without much mass effect or surrounding
with seizures, either focal or generalized (Table edema, with an area of contrast enhancement
7.3). near the margin of the cyst. Calcification may
In various series, seizures occurred in be present either on scan or microscopically.
45100% of patients. Because of the tumors Hemorrhage and necrosis are rare.
usual location in the temporal lobe, Interestingly, both thallium SPECT and PET
partialcomplex seizures are the most common scans (high uptake) and MRS (high choline
peak relative to creatine and N-acetyl aspar-
tate) may suggest high-grade tumor despite a
Table 7.3 very low labeling index.52
Signs and symptoms in 51 patients with
gangliogliomas.
Treatment
The treatment is surgical. A total resection can
Signs/symptoms Patients be achieved in most cases and is usually
curative.40 Even if total resection is not
No. %
possible, intractable seizures may improve,
Epilepsy 47 92 especially if the epileptogenic focus is identified
Cranial nerve deficit 1 2 by corticography and resected. The best
Hemiparesis 1 2 outcome with respect to seizure control is
Quadriparesis 2 4 associated with younger age, shorter duration
Increased intracranial pressure 2 4
of epilepsy, absence of generalized seizures and
From Zentner et al,43 with permission. no epileptiform discharge on postoperative
EEG.51 Often, particularly when the tumor is

230
EMBRYONAL TUMORS

located in a midline structure, substantial resec- 80% of patients become seizure-free after
tion is not possible. If the patient is asymp- surgery.38,51 Four-year progression-free survival
tomatic apart from seizures, careful follow-up rates after gross total resection of low- and
without further treatment is recommended high-grade tumors were 78% and 75%, respec-
even after partial resection. If the tumor recurs tively. After subtotal resection, the figures were
after complete or partial resection, a second 63% and 25%.40
operation should be considered, followed by
conformal RT to 54 Gy.
Gangliogliomas with an anaplastic-appearing Embryonal tumors
glial element should be treated in the same way,
without adjuvant RT. Survival of 34 years Introduction
has been reported with one such tumor.53 Some
The WHO lists several tumors as being of
authors recommend a more aggressive
embryonal origin. Table 7.4 represents a slight
approach to these tumors when they occur in
modification. Many of these tumors, including
midline structures such as hypothalamus or
medulloblastoma, neuroblastoma and primitive
brainstem, when the glial component has an
neuroectodermal tumors, probably originate from
anaplastic component.54 Patients have a higher
neurons or neuronal precursors and thus are
recurrence rate and a shorter median survival;
considered in this chapter. Ependymoblastomas
thus, some investigators recommend adjuvant
are rare primitive neuroectodermal tumors of
RT in this high-risk group. However, a recent
young children that are considered to be embry-
report suggests that RT of low-grade ganglio-
onal tumors by the WHO. They differ from
gliomas may promote malignant degeneration.55
anaplastic ependymomas (Chapter 5). Because
The concept is interesting but the issue is unset-
they are so rare and so little is known about
tled. Recurrence of anaplastic gangliogliomas
them, they are not discussed further. The classifi-
may also be associated with histologic degener-
cation of medulloblastomas is controversial.
ation to a glioblastoma multiforme. This is
Because the tumors appear histologically identical
another reason to consider re-resection at recur-
to both peripheral and central nervous system
rence; subsequent postoperative treatment is
determined by histology.
The role of chemotherapy in anaplastic or Table 7.4
recurrent low-grade tumor is unknown. One Embryonal tumors.
might also consider it in rapidly progressive
recurrent tumors in very young children to
Medulloepithelioma
avoid RT, or in older patients in whom RT has Ependymoblastoma
failed. Nitrosoureas, retinoic acid56 and cisplat- Medulloblastoma
inum have all been reported to have some Variants: desmoplastic medulloblastoma
effect. Occasionally, gangliogliomas seed the Medullomyoblastoma
leptomeninges, in which case chemotherapy Melanotic medulloblastoma
Supratentorial primitive neuroectodermal
should be considered.
tumors (PNET)
Neuroblastoma
Prognosis Ganglioneuroblastoma
The prognosis of low-grade gangliocytomas Atypical teratoid/rhabdoid tumor
and gangliogliomas is excellent. Sixty to

231
NEURONAL, MIXED NEURONALGLIAL AND EMBRYONAL TUMORS

tumors called primitive neuroectodermal tumors, the 4th ventricle, causing hydrocephalus. In
as well as pineal region tumors, and central and adults, the tumor can arise in the cerebellar
peripheral neuroblastomas, some pathologists hemisphere (Fig. 7.6), resulting in a somewhat
consider all of these tumors together as primitive different clinical picture than in children.
neuroectodermal tumors. The WHO has chosen
not to do so. One reason for keeping these Etiology
tumors separate is that the prognosis for medul- The disorder is usually sporadic but a number of
loblastoma is relatively good, whereas similar cases of medulloblastoma in monozygotic and
appearing tumors occurring elsewhere in the dizygotic twins and siblings have been reported.61
brain usually have a dismal prognosis (see below). Medulloblastomas have also been reported in
Another reason is that medulloblastomas differ several familial cancer syndromes, including
genetically from other primitive neuroectodermal LiFraumeni syndrome, the nevoid basal cell
tumors (see below). carcinoma syndrome, and Turcots syndrome
Taken together, embryonal tumors constitute (Chapter 12). Some patients with extraneural
an important fraction of pediatric brain malignancies, including Wilms tumor and malig-
tumors. Most appear similar, with a nant renal rhabdoid tumors, have also been
background of undifferentiated round cells observed to develop medulloblastomas. Con-
showing a variety of divergent patterns of versely, some studies suggest that relatives of
differentiation, including glial and neuronal. patients with medulloblastoma are at increased
Genetic markers may eventually reveal their risk of developing childhood leukemia or
origin and lead to reclassification. lymphoma. No environmental risks have been
identified. One study suggested that exposure to
SV40, which contaminated the polio vaccine in
Medulloblastoma
the 1950s, might be a risk factor, but a more
The medulloblastoma is a malignant childhood recent study refutes that finding.62
tumor located in the vermis of the cerebellum A number of chromosomal abnormalities have
but with a tendency to seed the leptomeninges been identified;63 the most common is isochro-
and cause widespread metastases within the mosome 17q.64 (An isochromosome is a chromo-
CNS and occasionally extraneural metastases, some that has split transversely and fused so that
especially to bone.57 Medulloblastoma is the one arm, in this case the long arm of chromo-
most common intracranial tumor of children, some 17, is doubled.) This abnormality can be
accounting for about 25% of childhood brain found in as many as 60% of medulloblastomas,
tumors.58 The incidence of medulloblastoma is but not in most other pediatric tumors.65,66
0.50.6/100 000 children59 per year, peaking at Abnormalities in several other chromosomes
the age of 7. Seventy percent of medulloblas- have also been reported.67 Loss of 9q and loss of
tomas occur before age 16. In one epidemio- 22 occur in a minority of tumors,64 and PTEN
logic series, 34% of medulloblastomas occurred mutations are rare in this disorder. A recent study
in patients over age 15, but the highest implicates the DMBT1 tumor suppressor gene in
incidence in adults 1519 years old was only almost 50% of medulloblastomas.67 Mutations
0.23/100 000/year.60 Males outnumber females of the PTCH gene, the causal mutation in medul-
by about 2:1. These childhood tumors almost loblastomas associated with the nevoid basal cell
always arise in the cerebellar vermis (often carcinoma syndrome (Chapter 12), are found in
inferior vermis) and may compress or invade only about 10% of sporadic medulloblastomas.68

232
EMBRYONAL TUMORS

Figure 7.6
A medulloblastoma. This young woman presented with headache and ataxia and was found to have two
lesions, one in the left and one in the right cerebellar hemisphere. The larger lesion enhanced with contrast (left
panel). The smaller lesion enhanced poorly (arrow). Both are apparent on the T2-weighted image. She was
treated with the protocol outlined in Figure 7.2. Three years later, a small enhancing lesion appeared in the
right cerebellum away from the original tumor (right panel, arrow). Although initially a recurrence was feared,
follow-up has shown no change in the lesion over 2 years, suggesting that it represents a vascular anomaly
caused by the radiation. The photomicrograph shows dense collections of undifferentiated neuroepithelial cells
forming nodules. Almost 90% of the tumors show neuronal differentiation as indicated by anti-Hu staining.
Some others show glial differentiation. These differentiations have no prognostic significance.

Pathology tumor (desmoplastic tumors are infiltrated by


Medulloblastomas vary in their gross appear- hyperplastic fibroblasts and collagen; the term
ance. Some, particularly in adults, are hemor- is from the Greek desmo meaning band and
rhagic and necrotic; others appear discrete and plastikos related to molding), medullo-
relatively firm. The histologic variants of myoblastoma, melanotic medulloblastoma and
medulloblastoma include classic desmoplastic cerebellar neuroblastoma. These variants may

233
NEURONAL, MIXED NEURONALGLIAL AND EMBRYONAL TUMORS

lead to some histologic confusion but they have Because the patient is too ill to go to school,
little bearing on the clinical signs, treatment or but seems fine later in the day, a diagnosis of
prognosis. school phobia may delay the appropriate
The typical medulloblastoma is composed of evaluation. When the tumor pushes the cere-
densely packed cells with round, oval or carrot- bellar tonsils through the foramen magnum
shaped hyperchromatic nuclei surrounded by (tonsillar herniation), patients may complain of
scanty cytoplasm (Fig. 7.6). Homer Wright neck pain. Nausea may or may not precede
rosettes are found in about 40% of patients; the vomiting. Some patients receive extensive
mitoses are frequent and areas of necrosis with gastrointestinal evaluation before a correct
pseudopalisading can sometimes be found. The diagnosis is made. However, the delay in
desmoplastic variety is more common in adults diagnosis does not seem to affect the progno-
than in children. The term cerebellar neuro- sis.69 In infants, medulloblastoma may present
blastoma is used to describe the desmoplastic simply as failure to thrive associated with
variety with extreme lobularity. In large cell vomiting and irritability.
medulloblastoma, the tumor is composed of Gait ataxia is usually the first sign. Because
cells with large round nuclei and prominent the posterior vermis controls balance, children
nucleoli; it contains large areas of necrosis and either complain of trouble walking or seem to
resembles atypical rhabdoid teratoid tumors of be unsteady on their feet, staggering as if drunk
the cerebellar region. when they walk. Early in the evolution of
Medulloblastomas can show differentiation symptoms, point-to-point testing in the extrem-
along several lines, including neural, glial and ities, including the finger-to-nose and the heel-
ependymal. The labeling indices are usually to-knee-to-shin tests, may be normal and lead
high, MIB-1 often being higher than 20%. The the physician to believe that there is no cerebel-
anti-Hu antibody is positive in almost 90% of lar dysfunction and that the reeling gait has a
cases,2 indicating that some degree of neuronal psychogenic basis. Additional signs include
differentiation is found in most medulloblas- diplopia from unilateral or bilateral abducens
tomas, prompting us to classify them under paralysis, transient loss of vision (visual obscu-
neuronal or mixed neuronal glial tumors. rations) related to papilledema and head tilt.
Cerebellar fits, episodic extensor spasms of
Clinical findings trunk and extremities, also called decerebrate
rigidity, are an ominous sign, suggesting tonsil-
Symptoms and signs. In children, headache and lar herniation.
vomiting are the most common presenting Because leptomeningeal dissemination may
symptoms. The headache may be frontal or occur early in the course of the disease,
occipital, and unilateral or bilateral. When patients may present with more widespread
unilateral and occipital, it defines the site of the symptoms, including seizures in 510% of
lesion. When unilateral but not occipital, it children. Cranial nerve palsies, with the
defines the side of the lesion. Early in the exception of the abducens nerve, back pain,
evolution of the tumor, headache is often absent deep tendon reflexes in the legs, and
present in the morning upon waking, only radicular sensory or motor changes suggest
to disappear for the rest of the day. leptomeningeal dissemination, the symptoms
Disappearance of the headache may be associ- resulting from direct infiltration of cranial
ated with vomiting, particularly in children. nerves or spinal roots.

234
EMBRYONAL TUMORS

Hydrocephalus from compression or invasion All patients in whom a diagnosis of medullo-


by tumor of the 4th ventricle is usually present blastoma is suspected on imaging should be
at diagnosis. Hydrocephalus may evolve quite evaluated for metastatic disease. This can be
rapidly, in some patients requiring emergency done by gadolinium-enhanced MR scan of the
decompression of the ventricular system before entire CNS.72 Areas of contrast enhancement,
definitive therapy. In some patients the ataxia is either nodular or linear in the cerebral
relieved by treating hydrocephalus with a shunt, hemispheres or spinal cord, suggest metastatic
suggesting that frontal lobe dysfunction may disease. In the absence of symptoms, systemic
play a role in the gait disorder. Rarely, medul- staging with a bone scan or bone marrow
loblastoma may invade the leptomeninges biopsy is probably unnecessary.73
without causing a mass lesion. We have encoun- The CSF should be examined for malignant
tered a few patients with hydrocephalus associ- cells, but because of the dangers of herniation
ated with cerebrospinal fluid (CSF) pleocytosis from a posterior fossa mass, this procedure is
but without abnormalities on MR scan. Because not indicated until the patients tumor has been
the cells in the spinal fluid were interpreted resected surgically. Preoperative imaging of the
cytologically as lymphocytes and the patients spine is important for the surgeon, because
had hydrocephalus, they were shunted. In one radical resection of the focal tumor, with its
instance, the diagnosis was not made until the attendant risk, does not confer survival advan-
patient, whose symptoms had been relieved by tage in a patient who already has disseminated
a ventriculoperitoneal (VP) shunt, developed a CNS disease. CSF examination should proba-
pelvic mass that on biopsy proved to be a bly be carried out 23 weeks after resection,
metastatic medulloblastoma that had spread via allowing time for a few cells that may have
the shunt. floated into the CSF at surgery to either seed,
giving a positive cytology, or disappear, indicat-
Imaging ing that the neuraxis is free of metastasis.
The diagnosis is strongly suspected on Sampling of ventricular fluid via a shunt is
neuroimaging. In children, the tumor is usually inadequate. Ventricular CSF may not contain
in the vermis of the cerebellum and appears malignant cells even when they are abundant
hyperdense on CT scan, hypointense on T1 and in the lumbar CSF.74 Both imaging and CSF
hyperintense on T2-weighted MR sequences. examination are required; some leptomeningeal
The tumor enhances intensely and homoge- metastases will be missed with either alone.75
neously, with no evidence of cyst formation or
necrosis;70 rarely, the tumor is non-enhancing. Treatment
Hydrocephalus is quite common. Treatment is determined in part by preopera-
In adults, the tumor is usually in the cerebellar tive staging (Table 7.5) and in part by the
hemisphere rather than the vermis, and hydro- completeness of surgical resection of the
cephalus occurs in less than half of the patients.71 tumor.76,77 If the patient presents with marked
The CT scan shows a hyperdense tumor, often hydrocephalus and plateau waves suggesting
with low-density areas suggesting cysts or necro- imminent herniation, a ventriculostomy or
sis. The same cysts or necrosis can be seen on MR ventriculoperitoneal (VP) shunt should be
scan, where the tumor is hypointense on T1 and placed prior to definitive surgery. If the patient
hyperintense on T2. Enhancement is variable but has leptomeningeal disease, shunting for hydro-
usually less than that seen in children. cephalus may lead to systemic dissemination of

235
NEURONAL, MIXED NEURONALGLIAL AND EMBRYONAL TUMORS

the tumor. A number of devices have been of masses on total neuraxial MR scan and
designed to reduce the incidence of ventricular negative cytologic examination of lumbar CSF
shunt metastases; how well these work is performed 23 weeks after surgery. Patients
unclear.57 with brainstem involvement are still considered
In all patients, a maximally feasible resection to be at average risk. High risk includes
of the tumor should be carried out. If the tumor patients with residual tumor > 1.5 cm or staged
presents as a single focus without MR evidence as M1M4 (Table 7.5). Most investigators
of dissemination, a radical resection is indicated. believe that staging after surgery, as indicated
If there is evidence of widespread dissemination, above, is more important than staging before
a debulking operation attempting to decrease surgery for determining further therapy and for
tumor volume to < 1.5 cm, without attempt at predicting prognosis.
radical resection, is the preferred choice. Figure 7.7 outlines a standard treatment
After the surgical procedure, the patient is scheme.79 All patients require whole-neuraxial
defined as being either average or high risk. An radiation.80 The procedure should be
average-risk patient is a patient: (1) who is > 3 performed by a skilled and experienced radia-
and )21 years old at diagnosis; (2) whose tion oncologist; the quality of the radiation
postoperative MR scan is either free of tumor targeting correlates with outcome.81 A standard
or has residual tumor < 1.5 cm in largest dose is 36 Gy to the entire nervous system,
dimension (the MR should be a contrast- with a boost to the tumor site of approximately
enhanced image performed within 72 h of 18 Gy (total to tumor 54 Gy). Some physi-
surgery); (3) with no evidence of metastatic cians do not reduce the craniospinal axis dose
disease in the neuraxis, as defined by absence of RT for average-risk patients (Fig. 7.7), but

Table 7.5
Staging of medulloblastoma.78

T1 Tumor < 3 cm in diameter


T2 Tumor * 3 cm in diameter
T3a Tumor > 3 cm in diameter with extension into the aqueduct of Sylvius and/or into foramen of
Luschka
T3b Tumor > 3 cm in diameter with unequivocal extension into the brainstem.
T3b can be defined by intraoperative demonstration of tumor extension into the brainstem in the
absence of radiographic evidence.
T4a Tumor > 3 cm in diameter with extension up past the aqueduct of Sylvius and/or down past the
foramen magnum
No consideration is given to the number of structures invaded or the presence of hydrocephalus.
M0 No evidence of subarachnoid cells found in CSF
M1a Microscopic tumor cells found in CSF
M2a Gross nodular seeding in the cerebellar, cerebral subarachnoid space, or the 3rd or lateral ventricles
M3a Gross nodular seeding in spinal subarachnoid space
M4a Metastasis outside the cerebrospinal axis
Residual tumor > 1.5 cm postoperatively*

*High risk.

236
EMBRYONAL TUMORS

Figure 7.7
Treatment of patients
Maximal resection
with medulloblastoma.a
Cisplatin begins 6 weeks
post-RT.b Vincristine
Stage (MRI entire neuraxis, LP) begins during RTb,
maximum dose 2 mg/m2.

High risk Average risk

36 Gy Craniospinal RT 54 Gy 23.4 Gy Craniospinal 55.8 Gy


to posterior fossa RT to posterior fossa

Lomustine 75 mg/m2 every 6 weeksa


Cisplatin 68 mg/m2 every 6 weeks
Vincristine 1.5 mg/m2 every week for 8 weeksb

most do when RT is combined with chemo- cyclophosphamide is underway. To date, the


therapy, as there appears to be no difference in addition of adjuvant chemotherapy for
progression-free survival at 56 years and average-risk patients has not improved
cognitive deficits are less pronounced.82,83 One survival. However, many pediatric neuro-
study suggests 55.8 Gy to the tumor and oncologists are treating all patients with
23.4 Gy to the remainder of the nervous system adjuvant therapy, given the sensitivity of
in children with localized tumors.83 Hyper- medulloblastomas to these agents, the 40%
fractionationed radiation does not appear to incidence of relapse over 5 years with RT
lengthen survival, at least in good-risk alone, and the opportunity to reduce the dose
patients.84 Patients at high risk (see Table 7.5) of neuraxis RT.
should be treated with adjuvant chemotherapy. In infants, some investigators have suggested
Several chemotherapeutic protocols have been that RT be deferred if chemotherapy can
suggested,73 but none has proved superior to control the growth of the tumor after surgery.
the protocol outlined in Fig. 7.7. This Several such protocols exist. Busulphan is effec-
chemotherapy regimen is usually well tolerated tive for treating medulloblastoma and/or primi-
by children, but the cumulative amount of tive neuroectodermal tumor implanted into
vincristine (VCR) is not tolerated by most athymic mice,85 but its utility in patients has
adults. Consequently, we usually administer the not been tested.
VCR only once every other week during RT to Surveillance MRI scans after therapy detect
adults, and even that dose may have to be asymptomatic recurrences, and may make
modified because of peripheral nerve toxicity. subsequent therapy easier, but whether they
A controlled trial comparing carmustine with increase survival is controversial.86 We have

237
NEURONAL, MIXED NEURONALGLIAL AND EMBRYONAL TUMORS

encountered patients whose surveillance scans develops fever, headache and sometimes hydro-
revealed contrast enhancement not caused by cephalus. The CSF shows a pleocytosis with
tumor.87 an elevated protein. Cultures are negative. An
increased dose of corticosteroids usually
Treatment complications suppresses the inflammation and patients then
The successful treatment of medulloblastoma recover spontaneously. Antibiotics are not
comes at a price.88 (See Fig. 7.8.) Several signif- required. Bacterial meningitis is an extremely
icant complications of treatment, both acute rare complication of clean (i.e. surgery
and delayed, are relatively common. Among through an uncontaminated field) posterior
the acute complications are postoperative fossa surgery. A CSF lactate level, which is high
aseptic meningitis (hemogenic meningitis) and in bacterial but not elevated hemogenic menin-
the cerebellar mutism syndrome. Hemogenic gitis, may assist in the diagnosis.91 On rare
meningitis follows some posterior fossa opera- occasions, particularly in patients on high
tions, when blood is spilled into the cisterna doses of steroids, the symptoms of hemogenic
magna.89,90 Elements in the blood (probably red meningitis do not develop until the patient has
cell membranes) irritate the leptomeninges and been discharged and the steroids tapered to low
initiate an inflammatory response. The patient doses. We have seen a few patients in whom

Figure 7.8
Failure of radiation therapy in a patient with a medulloblastoma. The patients cerebellum was successfully
treated, but a routine surveillance scan 18 months after treatment showed contrast-enhancing masses at the
inferior tips of the temporal lobes (arrows). On biopsy, this proved to be metastatic medulloblastoma.
Evidently, the radiation portals had been poorly drawn and did not include the inferior temporal lobes.

238
EMBRYONAL TUMORS

the symptoms of hemogenic meningitis Thyroid dysfunction, resulting from radiation


persisted for many weeks, requiring a very to the thyroid gland within the spinal cord
gradual tapering of the steroids. The same radiation portal, can occur at any time, even
syndrome sometimes follows spinal surgery many years after radiation has been completed.
and supratentorial surgery when the lateral Accordingly, patients should be followed with
ventricle has been entered. Rarely, chronic thyroid stimulating hormone (TSH) and T4
subclinical bleeding after surgery for cerebellar levels for the rest of their lives. In some
tumors may lead to siderosis of the meninges, children, T4 remains normal and patients are
with hearing loss, ataxia and cognitive dysfunc- clinically euthyroid but TSH is high. Some have
tion appearing years after surgery. The diagno- suggested that such patients should be treated
sis is established by the hypointense coating of with thyroid hormone to diminish the possibil-
the brainstem and cerebellum on the MRI.92 ity that high TSH levels may drive their irradi-
The cerebellar mutism syndrome is virtually ated thyroid to become neoplastic.
always restricted to children who undergo Many but not all children treated for medul-
cerebellar surgery.93 In the postoperative loblastoma demonstrate a decreased IQ.82
period, after a latent period of a few days, Cognitive dysfunction is probably related to
during which time the child speaks normally, both the posterior fossa RT97,98 and the total
the child becomes mute, often refuses to dose to the brain.82,83 Some have difficulty at
swallow and may have other behavioral abnor- school. Several risk factors increase the likeli-
malities resembling autism (e.g. irritability and hood of such patients developing cognitive
lack of social interaction). Visual impairment, dysfunction:
probably cortical blindness, may also be
present. Most patients recover speech sponta- 1. The presence of hydrocephalus in either the
neously after days to weeks. The pathogenesis pre- or postoperative period. Accordingly,
of this syndrome is unknown, and postopera- patients should be followed and hydro-
tive MR scans fail to demonstrate brainstem cephalus corrected as soon as detected.
or hemispheric abnormalities. However, the 2. Postoperative infectious meningitis, proba-
cerebellum contributes to cognitive functions, bly because it increases intracranial
and, therefore, intellectual, language and pressure and the likelihood of developing
behavorial disturbances affect many children hydrocephalus. It is not known if
after posterior fossa surgery.94,95 hemogenic meningitis is a risk factor.
Delayed manifestations of treatment toxicity 3. RT. One study found a full-scale IQ drop
after medulloblastoma include growth failure, of 14 points 2 years after treatment for
endocrine disturbances, cognitive dysfunction, medulloblastoma. Patients with posterior
vasculopathy and secondary tumors. Growth fossa tumors who were not irradiated (e.g.
failure results in part from radiation to the cerebellar astrocytoma) had no significant
spinal column, which inhibits vertebral growth, decline in IQ, but most did not suffer
and in part from radiation to the hypothala- from hydrocephalus in the preoperative
mus and pituitary gland, which causes growth period, as do children with medullo-
hormone deficiency.96 As a result, most children blastoma. Both the dose of radiation and
treated for medulloblastoma remain small. the age of the patient (young age) are
Growth hormone will restore long bone risk factors for chronic radiation
growth, but spinal growth remains impaired. encephalopathy.

239
NEURONAL, MIXED NEURONALGLIAL AND EMBRYONAL TUMORS

4. The posterior fossa surgery itself. As patients. For average-risk patients, progression-
indicated above, some patients who free survival in one series was 86% at 3 years
undergo operations on the cerebellum for and 79% at 7 years. Sex is an important predic-
tumors develop mutism, pseudobulbar tor for survival, with girls having a much better
symptoms and behavioral changes after outcome than boys.103 Very young children have
surgery. Although the mutism and swallow- a worse prognosis, perhaps due to the lower
ing difficulty always improve, behavioral radiation doses often given.104 Interestingly, the
symptoms may persist and interfere with duration of symptoms is inversely related to
cognitive function later in life.99 disease stage and, thus, survival.105 Among
adults late relapse is common and systemic
Late post-irradiation occlusive vasculopathy failure occurs even in low-risk patients: both
is a rare complication of childhood medullo- problems require prolonged and careful surveil-
blastoma.100 Vessels in the posterior fossa lance.106
become stenotic and may occlude, leading to Children should be followed with a uniform
occipital lobe and posterior fossa infarction. surveillance program which includes repeat
Hyperfractionated radiotherapy is associated lumbar punctures and MR scans. In 2530%
with MR evidence of necrosis, telangiectasia of patients, surveillance scans will reveal
and white matter changes.101 Finally, RT to the asymptomatic relapse, but current evidence
CNS may lead to secondary tumors. Children suggests that these patients do not benefit from
treated for brain tumors have a nine-fold higher salvage therapy more than those with
risk for developing a second cancer. In one symptomatic relapses.86 The outlook following
series of 187 children treated for medulloblas- recurrence of medulloblastoma is poor. Salvage
toma, the actuarial incidence of death due to a chemotherapy produces some response in
second malignant tumor was 13% at 30 years. about half of the patients, and a complete
The tumors may occur either in the radiation response in about a fourth. In one series, the
field, e.g. sarcoma of the occipital bone, be part median survival was only several months and
of a familial tumor syndrome such as neurofi- only a few patients lived longer than 2 years.
bromatosis, occur as a result of alkylating High-dose chemotherapy with autologous stem
chemotherapeutic agents, e.g. leukemia, or cell rescue for relapse yields a 30% progres-
occur from radiation of the neck, e.g. thyroid sion-free survival at a median of 54 months.107
cancer, as indicated above. Children cured of Early detection of relapse and aggressive treat-
medulloblastoma must be followed life-long for ment seem to be warranted to prevent the
these potential complications. development of neurologic symptoms.
How long it is necessary to follow patients
Prognosis who are free from recurrence is unclear.
Medulloblastoma is one of the intracranial Collins law states that for embryonal tumors
tumors in which there has been a substantial of childhood the risk of recurrence ends when
improvement in survival as a direct result of the child is free of disease for years equal to
therapy. The 5-year survival rate in the 1960s his age when the tumor appeared plus 9
was about 30%, and was as low as 8% in one months. The law, with some exceptions, seems
series.102 It is now closer to about 70% using to apply to childhood medulloblastomas108 and
modern protocols combining surgery and radia- other brain tumors of young children, although
tion along with chemotherapy for high-risk rare late relapses have been observed.109

240
EMBRYONAL TUMORS

Other tumors of embryonal prognosis is much worse. Children with non-


origin pineal supratentorial PNETs have an overall 5-
year survival rate of 34%.
Not all cerebellar lesions are medulloblast- Atypical teratoid/rhabdoid tumors are
omas, or even tumors (Fig. 7.9). Other tumors uncommon tumors of childhood found in the
in this category, including CNS neuroblastoma cerebral hemispheres, cerebellum or cerebello-
and ganglioneuroblastoma, primitive neuroec- pontine angle.110 They all contain rhabdoid
todermal tumors (PNET) and rhabdoid tumors, cells (a cell with an apparent autoplasmic
are all quite rare. Collectively, these tumors inclusion consisting of bundles of intermediate
represent less than 1% of childhood brain filaments) and represent about 2% of child-
tumors and usually occur before 2 years of age. hood neuroepithelial tumors. They usually
The tumors are similar in neuroimaging and occur in young children; the median age is 20
histologic appearance to cerebellar medullo- months but a few adults have been reported.
blastomas. The treatment is the same but the They differ from other embryonal tumors in

Figure 7.9
Pitfalls in the diagnosis of neuronal tumors. This young woman presented to medical attention
with a two week history of ataxia and mild vertigo. Her examination was normal save for a
slightly unsteady gait and a vertiginous sensation when she moved suddenly. An MR scan
showed a non-contrast enhancing mass in the vermis of the cerebellum. The mass was
hypointense on the T1 (left) and hypointense on the T2 (right) image. Medulloblastoma was
considered but because she was so mildly symptomatic it was decided to follow her. Two months
later the lesion completely disappeared. Its nature remains a mystery.

241
NEURONAL, MIXED NEURONALGLIAL AND EMBRYONAL TUMORS

their histopathology. Some consist only of particularly RT, is likely to produce significant
rhabdoid cells, while others consist of a combi- neurologic sequelae in these young survivors.
nation of rhabdoid and PNET-appearing cells.
The tumor may be vimentin, GFAP, epithelial Neuroblastoma, a childhood tumor of the
membrane agonist (EMA) and smooth muscle adrenal gland and sympathetic nervous
actin positive. They are sometimes mistaken for system, is the third most common childhood
medulloblastoma when they occur in the poste- tumor after leukemia and brain tumors. This
rior fossa. Unlike other embryonal tumors, tumor may occasionally arise within the
most show abnormalities of chromosome 22, intracranial cavity, usually deep in the cerebral
either monosomy or deletion of 22q.111 The hemisphere. The tumors have a histologic
tumors are highly malignant, and with treat- appearance identical to their peripheral
ment similar to that of medulloblastoma, counterparts, with Homer Wright rosettes and
median survival is about 6 months. About 30% expression of neurofilament protein and
show leptomeningeal spread at diagnosis. synaptophysin. The tumor must be differenti-
High-dose chemotherapy with autologous ated from metastatic neuroblastoma, which
bone marrow or stem cell support, and RT usually spreads to the dura rather than the
have yielded improved survival in some parenchyma of the brain, and must be differ-
children;112,113 however, vigorous treatment, entiated histologically from other PNETs and

Figure 7.10
The MRI of a young child presenting with headache. A dural-based tumor with a large cyst
was identified on the MR scan. At surgery, the tumor appeared to be a primitive
neuroectodermal tumor with sheets of uniform round cells with small amounts of
cytoplasm. Genetic evaluation revealed it to be a Ewings sarcoma. Extraskeletal Ewings
sarcomas are uncommon. Those arising in the meninges are extremely rare.

242
REFERENCES

medulloblastoma, which the neuroblastoma 4. Maiuri F, Spaziante R, De Caro ML et al.


resembles histologically. Central neurocytoma: clinico-pathological
study of 5 cases and review of the literature.
Clin Neurol Neurosurg 1995; 97: 21928.
Primitive neuroectodermal tumor (Fig. 7.10) is 5. Warmuth-Metz M, Klein R, Srensen N,
a term that some apply to a number of histo- Solymosi L. Central neurocytoma of the
logically identical tumors that in the brain fourth ventricle case report. J Neurosurg
include cerebral neuroblastoma, medulloblas- 1999; 91: 5069.
toma and pinealoblastoma (see Chapter 8). 6. Ashkan K, Casey ATH, DArrigo C,
Harkness WF, Thomas DGT. Benign central
PNETs above the tentorium cerebelli are differ- neurocytoma a double misnomer? Cancer
ent from medulloblastoma of the cerebellum 2000; 89: 111120.
both in their genetic composition and in their 7. Yin XL, Pang JCS, Hui ABY, Ng H. Detection
clinical course. Isochromosome 17q observed of chromosomal imbalances in central neuro-
in medulloblastoma is not found in supra- cytomas by using comparative genomic
hybridization. J Neurosurg 2000; 93: 7781.
tentorial PNETs. Loss of chromosome 14q,
8. Tong CY, Ng HK, Pang JC et al. Central
detected in about 40% of supratentorial neurocytomas are genetically distinct from
PNETs, is not detected in medulloblastoma.114 oligodendrogliomas and neuroblastomas.
Loss of chromosome 3p is common in supra- Histopathology 2000; 37: 1605.
tentorial PNETs but not in medulloblastoma.64 9. Nozaki M, Tada M, Matsumoto R,
The clinical symptoms obviously differ, because Sawamura Y, Abe H, Iggo RD. Rare occur-
rence of inactivating p53 gene mutations in
PNETs arise in the cerebral hemisphere, primary nonastrocytic tumors of the central
whereas medulloblastomas arise in the cere- nervous system: reappraisal by yeast
bellum. PNETs disseminate throughout the functional assay. Acta Neuropathol (Berl)
neuraxis, and whole-neuraxial radiation is 1998; 95: 2916.
recommended for all. However, the 5-year 10. Ishiuchi S, Tamura M. Central neurocytoma:
an immunohistochemical, ultrastructural and
recurrence-free survival is much lower in
cell culture study. Acta Neuropathol (Berl)
patients with supratentorial PNETs115,116 than 1997; 94: 42535.
in those with medulloblastoma. 11. Nishio S, Morioka T, Hamada Y, Fukui
M, Nakagawara A. Immunohistochemical
expression of tyrosine kinase (Trk) receptor
References proteins in mature neuronal cell tumors of the
central nervous system. Clin Neuropathol
1. Wolf HK, Buslei R, Schmidt-Kastner R et al. 1998; 17: 12330.
NeuN: a useful neuronal marker for diagnos- 12. Mena H, Morrison AL, Jones RV, Gyure KA.
tic histopathology. J Histochem Cytochem Central neurocytomas express photoreceptor
1996; 44: 116771. differentiation. Cancer 2001; 91: 13643.
2. Gultekin SH, Dalmau J, Graus Y, Posner JB, 13. Favereaux A, Vital A, Loiseau H et al.
Rosenblum MK. Anti-Hu immunolabeling as Histopathological variants of central neuro-
an index of neuronal differentiation in human cytoma: Report of 10 cases. Ann Pathol
brain tumors a study of 112 central 2000; 20: 55863.
neuroepithelial neoplasms. Am J Surg Pathol 14. Mackay IR. Central neurocytoma: histologic
1998; 22: 195200. atypia, proliferation potential, and clinical
3. Kleihues P, Cavenee WK. World Health outcome. Cancer 1999; 85: 160610.
Organization Classification of Tumours: 15. Mackenzie IRA. Central neurocytoma
Tumours of the nervous system pathology histologic atypia, proliferation potential, and
and genetics. Lyon: IRAC Press, 2000. clinical outcome. Cancer 1999; 85: 160610.

243
NEURONAL, MIXED NEURONALGLIAL AND EMBRYONAL TUMORS

16. Eng DY, DeMonte F, Ginsberg L, Fuller GN, 29. Prayson RA. Bcl-2, bcl-x, and bax expression
Jaeckle K. Craniospinal dissemination of in dysembryoplastic neuroepithelial tumors.
central neurocytoma. Report of two cases. Clin Neuropathol 2000; 19: 5762.
J Neurosurg 1997; 86: 54752. 30. Aronica E, Leenstra S, Jansen GH et al.
17. Oguchi M, Higashi K, Taniguchi M et al. Expression of brain-derived neurotrophic
Single photon emission CT images in a case factor and tyrosine kinase B receptor proteins
of intraventricular neurocytoma. Ann Nucl in glioneuronal tumors from patients with
Med 1998; 12: 1614. intractable epilepsy: colocalization with N-
18. Krouwer HGJ. Neuronal and mixed methyl-D-aspartic acid receptor. Acta
neuronalglial tumors. In: Vecht C, ed. Neuropathol (Berl) 2001; 101: 38392.
Neurooncology, part 2. Amsterdam: Elsevier, 31. Kleihues P, Cavenee WK. Pathology and
1997: 13757. genetics of tumours of the nervous system.
19. Wharton SB, Antoun NM, Macfarlane R, Lyons: International Agency for Research on
Anderson JR. The natural history of a Cancer, 1997.
recurrent central neurocytoma-like tumor 32. Hirose T, Scheithauer BW. Mixed dysembryo-
Clin Neuropathol 1998; 17: 13640. plastic neuroepithelial tumor and ganglio-
20. Anderson RC, Elder JB, Parsa AT, Issacson glioma. Acta Neuropathol (Berl) 1998; 95:
SR, Sisti MB. Radiosurgery for the treatment 64954.
of recurrent central neurocytomas. Neuro- 33. Thom M, Gomez-Anson B, Revesz T et al.
surgery 2001; 48: 12317. Spontaneous intralesional haemorrhage in
21. Brandes AA, Amist P, Gardiman M et al. dysembryoplastic neuroepithelial tumours: a
Chemotherapy in patients with recurrent and series of five cases. J Neurol Neurosurg
progressive central neurocytoma. Cancer Psychiatry 1999; 67: 97101.
2000; 88: 16974. 34. Schiff D, Wen PY. Uncommon brain tumors.
22. Schild SE, Scheithauer BW, Haddock MG et Neurol Clin 1995; 13: 95374.
al. Central neurocytomas. Cancer 1997; 79: 35. Lee DY, Chung CK, Hwang YS et al.
7905. Dysembryoplastic neuroepithelial tumor:
23. Sgouros S, Carey M, Aluwihare N, Barber P, radiological findings (including PET, SPECT,
Jackowski A. Central neurocytoma: a correla- and MRS) and surgical strategy. J Neuro-
tive clinicopathologic and radiologic analysis. oncol 2000; 47: 16774.
Surg Neurol 1998; 49: 197204. 36. Kaplan AM, Lawson MA, Spataro J et al.
24. Cabiol J, Acebes JJ, Isamat F. Dysembryo- Positron emission tomography using [18F]
plastic neuroepithelial tumor. Crit Rev fluorodeoxyglucose and [11C] L-methionine to
Neurosurg 1999; 9: 11625. metabolically characterize dysembryoplastic
25. Daumas-Duport C, Varlet P, Bacha S et al. neuroepithelial tumors. J Child Neurol 1999;
Dysembryoplastic neuroepithelial tumors: 14: 6737.
nonspecific histological forms a study of 40 37. Prayson RA, Morris HH, Estes ML, Comair
cases. J Neurooncol 1999; 41: 26780. YG. Dysembryoplastic neuroepithelial tumor:
26. Baisden BL, Brat DJ, Melhem ER et al. Dys- a clinicopathologic and immunohistochemical
embryoplastic neuroepithelial tumor-like study of 11 tumors including MIB1 immuno-
neoplasm of the septum pellucidum: a lesion reactivity. Clin Neuropathol 1996; 15: 4753.
often misdiagnosed as glioma: report of 10 38. Aronica E, Leenstra S, Van Veelen CWM
cases. Am J Surg Pathol 2001; 25: 4949. et al. Glioneuronal tumors and medically
27. Chong EY, Lam PY, Poon WS, Ng HK. intractable epilepsy: a clinical study with
Telomerase expression in gliomas including long-term follow-up of seizure outcome after
the nonastrocytic tumors. Hum Pathol 1998; surgery. Epilepsy Res 2001; 43: 17991.
29: 599603. 39. Andermann LF, Savard G, Meencke HJ et al.
28. Duerr EM, Rollbrocker B, Hayashi Y et al. Psychosis after resection of ganglioglioma or
PTEN mutations in gliomas and glioneuronal DNET: evidence for an association. Epilepsia
tumors. Oncogene 1998; 16: 225964. 1999; 40: 837.

244
REFERENCES

40. Selch MT, Goy BW, Lee SP et al. 52. Kumabe T, Shimizu H, Sonoda Y, Shirane
Gangliogliomas: experience with 34 patients R. Thallium-201 single-photon emission
and review of the literature. Am J Clin Oncol computed tomographic and proton magnetic
1998; 21: 55764. resonance spectroscopic characteristics of
41. Alkhani AM, Bilbao JM, Medline P, intracranial ganglioglioma: three technical
Ogundimu FA. Ganglioglioneurocytoma of case reports. Neurosurgery 1999; 45: 1837.
the posterior fossa. Can J Neurol Sci 1999; 53. Celli P, Scarpinati M, Nardacci B, Cervoni L,
26: 20710. Cantore GP. Gangliogliomas of the cerebral
42. Johnson JH Jr, Hariharan S, Berman J et al. hemispheres. Report of 14 cases with long-
Clinical outcome of pediatric gangliogliomas: term follow-up and review of the literature.
ninety-nine cases over 20 years. Pediatr Acta Neurochir (Wien) 1993; 125: 527.
Neurosurg 1997; 27: 2037. 54. Haddad SF, Moore SA, Menezes AH,
43. Zentner J, Wolf HK, Ostertun B et al. VanGilder JC. Ganglioglioma: 13 years of
Gangliogliomas: clinical, radiological, and experience. Neurosurgery 1992; 31: 1718.
histopathological findings in 51 patients. J 55. Rumana CS, Valadka AB. Radiation therapy
Neurol Neurosurg Psychiatry 1994; 57: and malignant degeneration of benign supra-
1497502. tentorial gangliogliomas. Neurosurgery 1998;
44. Hakim R, Loeffler JS, Anthony DC, Black 42: 103843.
PM. Gangliogliomas in adults. Cancer 1997; 56. Kaba SE, Langford LA, Yung WK, Kyritsis
79: 12731. AP. Resolution of recurrent malignant gangli-
45. Geraci AP, de Csepel J, Shlasko E, Wallace oglioma after treatment with cis-retinoic acid.
SA. Ganglioneuroblastoma and ganglioneu- J Neurooncol 1996; 30: 5560.
roma in association with neurofibromatosis 57. Halperin EC, Samulski T, Oakes WJ,
type I: report of three cases. J Child Neurol Friedman HS. Fabrication and testing of a
1998; 13: 3568. device capable of reducing the incidence of
46. Bhattacharjee MB, Armstrong DD, Vogel H, ventricular shunt promoted metastasis. J
Cooley LD. Cytogenetic analysis of 120 Neurooncol 1996; 27: 3946.
primary pediatric brain tumors and literature 58. Packer RJ. Childhood medulloblastoma:
review. Cancer Genet Cytogenet 1997; 97: progress and future challenges. Brain Dev
3953. 1999; 21: 7581.
47. Hirose T, Scheithauer BW, Lopes MBS et al. 59. Agerlin N, Gjerris F, Brincker H et al.
Ganglioglioma. An ultrastructural and Childhood medulloblastoma in Denmark
immunohistochemical study. Cancer 1997; 19601984. A population-based retrospective
79: 9891003. study. Childs Nerv Syst 1999; 15: 2936.
48. Wacker MR, Cogen PH, Etzell JE et al. 60. Giordana MT, Schiffer P, Lanotte M,
Diffuse leptomeningeal involvement by a Girardi P, Chio A. Epidemiology of adult
ganglioglioma in a child. Case report. J medulloblastoma. Int J Cancer 1999; 80:
Neurosurg 1992; 77: 3026. 68992.
49. Duerr EM, Rollbrocker B, Hayashi Y, et al. 61. Bayani J, Zielenska M, Marrano P et al.
PTEN mutations in gliomas and glioneuronal Molecular cytogenetic analysis of medul-
tumors. Oncogene 1998; 16: 25964. loblastomas and supratentorial primitive
50. Blumcke I, Lobach M, Wolf HK, Wiestler neuroectodermal tumors by using conven-
OD. Evidence for developmental precursor tional banding, comparative genomic
lesions in epilepsy-associated glioneuronal hybridization, and spectral karyotyping. J
tumors. Microsc Res Tech 1999; 46: 538. Neurosurg 2000; 93: 43748.
51. Morris HH, Matkovic Z, Estes ML et al. 62. Strickler HD, Rosenberg PS, Devesa SS,
Ganglioglioma and intractable epilepsy: clini- Fraumeni JFJ, Goedert JJ. Contamination of
cal and neurophysiologic features and predic- poliovirus vaccine with SV40 and the
tors of outcome after surgery. Epilepsia 1998; incidence of medulloblastoma. Med Pediatr
39: 30713. Oncol 1999; 32: 778.

245
NEURONAL, MIXED NEURONALGLIAL AND EMBRYONAL TUMORS

63. Reardon DA, Michalkiewicz E, Boyett JM randomized phase III study. J Clin Oncol
et al. Extensive genomic abnormalities in 1999; 17: 83245.
childhood medulloblastoma by comparative 74. Gajjar A, Fouladi M, Walter AW et al.
genomic hybridization. Cancer Res 1997; 57: Comparison of lumbar and shunt
40427. cerebrospinal fluid specimens for cytologic
64. Nicholson JC, Ross FM, Kohler JA, Ellison detection of leptomeningeal disease in
DW. Comparative genomic hybridization and pediatric patients with brain tumors. J Clin
histological variation in primitive neuroecto- Oncol 1999; 17: 18258.
dermal tumours. Br J Cancer 1999; 80: 75. Fouladi M, Gajjar A, Boyett JM et al.
132231. Comparison of CSF cytology and spinal
65. Giordana MT, Migheli A, Pavanelli E. magnetic resonance imaging in the detection
Isochromosome 17q is a constant finding in of leptomeningeal disease in pediatric medul-
medulloblastoma. An interphase cytogenetic loblastoma or primitive neuroectodermal
study on tissue sections. Neuropathol Appl tumor. J Clin Oncol 1999; 17: 32347.
Neurobiol 1998; 24: 2338. 76. Chintagumpala M, Berg S, Blaney SM.
66. Avet-Loiseau H, Vnuat AM, Terrier- Treatment controversies in medulloblastoma.
Lacombe MJ et al. Comparative genome Curr Opin Oncol 2001; 13: 1549.
hybridization detects many recurrent imbal- 77. Greenberg HS, Chamberlain MC, Glantz MJ,
ances in central nervous system primitive Wang S. Adult medulloblastoma: multiagent
neuroectodermal tumours in children. Br J chemotherapy. Neurooncol 2001; 3: 2934.
Cancer 1999; 79: 18437. 78. Packer RJ, Sutton LN, Elterman R et al.
67. Yin XL, Pang JC, Liu YH et al. Analysis of Outcome for children with medulloblastoma
loss of heterozygosity on chromosomes 10q, treated with radiation and cisplatin, CCNU,
11, and 16 in medulloblastomas. J Neurosurg and vincristine chemotherapy. J Neurosurg
2001; 94: 799805. 1994; 81: 6908.
68. Dong J, Gailani MR, Pomeroy SL, Reardon 79. Packer RJ, Sutton LN, Goldwein JW et al.
D, Bale AE. Identification of PATCHED Improved survival with the use of adjuvant
mutations in medulloblastomas by direct chemotherapy in the treatment of medul-
sequencing. Hum Mutat 2000; 16: 8990. loblastoma. J Neurosurg 1991; 74: 43340.
69. Halperin EC, Friedman HS. Is there a 80. Paulino AC. Radiotherapeutic management
correlation between duration of presenting of medulloblastoma. Oncology (Huntingt)
symptoms and stage of medulloblastoma at 1997; 11: 81323.
the time of diagnosis? Cancer 1996; 78: 81. Carrie C, Hoffstetter S, Gomez F et al. Impact
87480. of targeting deviations on outcome in medul-
70. Tortori-Donati P, Fondelli MP, Rossi A et al. loblastoma: Study of the French Society of
Medulloblastoma in children: CT and MRI Pediatric Oncology (SFOP). Int J Radiat
findings. Neuroradiology 1996; 38: 3529. Oncol Biol Phys 1999; 45: 4359.
71. Becker RL, Becker AD, Sobel DF. Adult 82. Mulhern RK, Kepner JL, Thomas PR et al.
medulloblastoma: review of 13 cases with Neuropsychologic functioning of survivors
emphasis on MRI. Neuroradiology 1995; 37: of childhood medulloblastoma randomized
1048. to receive conventional or reduced-dose
72. Harrison SK, Ditchfield MR, Waters K. craniospinal irradiation: a pediatric oncol-
Correlation of MRI and CSF cytology in the ogy group study. J Clin Oncol 1998; 16:
diagnosis of medulloblastoma spinal metas- 17238.
tases. Pediatr Radiol 1998; 28: 5714. 83. Packer RJ, Goldwein J, Nicholson HS et al.
73. Zeltzer PM, Boyett JM, Finlay JL et al. Treatment of children with medulloblastomas
Metastasis stage, adjuvant treatment, and with reduced-dose craniospinal radiation
residual tumor are prognostic factors for therapy and adjuvant chemotherapy: A
medulloblastoma in children: conclusions childrens cancer group study. J Clin Oncol
from the Childrens Cancer Group 921 1999; 17: 212736.

246
REFERENCES

84. Prados MD, Edwards MSB, Chang SM et al. cerebellar tumour resection in children:
Hyperfractionated craniospinal radiation cerebellar cognitive affective syndrome in a
therapy for primitive neuroectodermal paediatric population. Brain 2000; 123 (Pt 5):
tumors: results of a phase II study. Int J 104150.
Radiat Oncol Biol Phys 1999; 43: 27985. 96. Clarson CL, Del Maestro RF. Growth failure
85. Boland I, Vassal G, Morizet J et al. after treatment of pediatric brain tumors.
Busulphan is active against neuroblastoma Pediatrics 1999; 103: E3716.
and medulloblastoma xenografts in athymic 97. Grill J, Renaux VK, Bulteau C et al. Long-
mice at clinically achievable plasma drug term intellectual outcome in children with
concentrations. Br J Cancer 1999; 79: posterior fossa tumors according to radiation
78792. doses and volumes. Int J Radiat Oncol Biol
86. Bouffet E, Doz F, Demaille MC et al. Phys 1999; 45: 13745.
Improving survival in recurrent medullo- 98. Copeland DR, DeMoor C, Moore BD III,
blastoma: earlier detection, better treatment or Ater JL. Neurocognitive development of
still an impasse? Br J Cancer 1998; 77: 13216. children after a cerebellar tumor in infancy: a
87. Fouladi M, Heideman R, Langston JW et al. longitudinal study. J Clin Oncol 1999; 17:
Infectious meningitis mimicking recurrent 347686.
medulloblastoma on magnetic resonance 99. Ersahin Y, Mutluer S, agli S, Duman Y.
imaging case report. J Neurosurg 1999; 91: Cerebellar mutism: report of seven cases and
499502. review of the literature. Neurosurgery 1996;
88. Sutton LN, Phillips PC, Molloy PT. Surgical 38: 606.
management of medulloblastoma. J Neuro- 100. Grenier Y, Tomita T, Marymont MH, Byrd
oncol 1996; 29: 921. S, Burrowes DM. Late postirradiation occlu-
89. Forgacs P, Geyer CA, Freidberg SR. sive vasculopathy in childhood medullo-
Characterization of chemical meningitis after blastoma. Report of two cases. J Neurosurg
neurological surgery. Clin Infect Dis 2001: 1998; 89: 4604.
32: 17985. 101. Russo C, Fischbein N, Grant E, Prados MD.
90. Carmel PW, Greif LK. The aseptic meningitis Late radiation injury following hyperfraction-
syndrome: A complication of posterior fossa ated craniospinal radiotherapy for primitive
surgery. Pediatr Neurosurg 1993; 19: 27680. neuroectodermal tumor. Int J Radiat Oncol
91. Leib SL, Boscacci R, Gratzl O, Zimmerli W. Biol Phys 1999; 44: 8590.
Predictive value of cerebrospinal fluid (CSF) 102. Agerlin N, Gjerris F, Brincker H et al.
lactate level versus CSF/blood glucose ratio for Childhood medulloblastoma in Denmark
the diagnosis of bacterial meningitis following 19601984. A population-based retrospective
neurosurgery. Clin Infect Dis 1999; 29: 6974. study. Child Nerv Syst 1999; 15: 2936.
92. Anderson NE, Sheffield S, Hope JKA. 103. Weil MD, Lamborn K, Edwards MS, Wara
Superficial siderosis of the central nervous WM. Influence of a childs sex on medul-
system a late complication of cerebellar loblastoma outcome. JAMA 1998; 279:
tumors. Neurology 1999; 52: 1639. 14746.
93. Liu GT, Phillips PC, Molloy PT et al. Visual 104. Saran FH, Driever PH, Thilmann C et al.
impairment associated with mutism after Survival of very young children with medul-
posterior fossa surgery in children. Neuro- loblastoma (primitive neuroectodermal tumor
surgery 1998; 42: 2536. of the posterior fossa) treated with
94. Riva D, Giorgi C. The cerebellum contributes craniospinal irradiation. In J Radiat Oncol
to higher functions during development: Biol Phys 1998; 42: 95967.
evidence from a series of children surgically 105. Halperin EC, Watson DM, George SL.
treated for posterior fossa tumours. Brain Duration of symptoms prior to diagnosis is
2000; 123: 105161. related inversely to presenting disease stage in
95. Levisohn L, Cronin-Golomb A, Schmahmann children with medulloblastoma. Cancer 2001;
JD. Neuropsychological consequences of 91: 144450.

247
NEURONAL, MIXED NEURONALGLIAL AND EMBRYONAL TUMORS

106. Chan AW, Tarbell NJ, Black PM et al. Adult chromosome 22q dosage in distinguishing
medulloblastoma: Prognostic factors and atypical teratoid/rhabdoid tumors from medul-
patterns of relapse. Neurosurgery 2000; 47: loblastoma/central primitive neuroectodermal
62331. tumors. Hum Pathol 2001; 32: 15662.
107. Dunkel IJ, Boyett JM, Yates A et al. High- 112. Hilden JM, Watterson J, Longee DC et al.
dose carboplatin, thiotepa, and etoposide with Central nervous system atypical teratoid
autologous stem-cell rescue for patients with tumor rhabdoid tumor: response to intensive
recurrent medulloblastoma. J Clin Oncol therapy and review of the literature. J
1998; 16: 2228. Neurooncol 1998; 40: 26575.
108. Brown WD, Tavare CJ, Sobel EL, Gilles FH. 113. Olson TA, Bayar E, Kosnik E et al. Successful
The applicability of Collins Law to child- treatment of disseminated central nervous
hood brain tumors and its usefulness as a system malignant rhabdoid tumor. J Pediatr
predictor of survival. Neurosurgery 1995; 36: Hematol Oncol 1995; 17: 715.
10936. 114. Russo C, Pellarin M, Tingby O et al.
109. Sala F, Colarusso E, Mazza C, Talacchi A, Comparative genomic hybridization in
Bricolo A. Brain tumors in children under 3 patients with supratentorial and infratentorial
years of age recent experience (19871997) primitive neuroectodermal tumors. Cancer
in 39 patients. Pediatr Neurosurg 1999; 31: 1999; 86: 3319.
1626. 115. Paulino AC, Melian E. Medulloblastoma
110. Ho DMT, Hsu CY, Wong TT, Ting LT, and supratentorial primitive neuroectodermal
Chiang H. Atypical teratoid/rhabdoid tumor tumors an institutional experience. Cancer
of the central nervous system: a comparative 1999; 86: 1428.
study with primitive neuroectodermal 116. Reddy AT, Janss AJ, Phillips PC, Weiss HL,
tumor/medulloblastoma. Acta Neuropathol Packer RJ. Outcome for children with supra-
(Berl) 2000; 99: 4828. tentorial primitive neuroectodermal tumors
111. Bruch LA, Hill DA, Cai DX et al. A role for treated with surgery, radiation, and chemo-
fluorescence in situ hybridization detection of therapy. Cancer 2000; 88: 218993.

248
8
Pineal region tumors

Introduction Tumors of the pineal gland and pineal


region are often detected when they are very
The pineal gland (from the Latin for pine) is small, because of their tendency to compress
a pine-cone-shaped structure measuring about the underlying brainstem tectum and
8 4 mm in the adult and weighing about obstruct the aqueduct of Sylvius, causing
200 mg. It is located in the midline of the hydrocephalus (Fig. 8.1). Many different
brain, just above the tectum of the midbrain. tumors, some benign and some malignant,
It is tethered to the posterior portion of the arise in or near the pineal gland (Fig. 8.1),4
3rd ventricle by a neuroepithelial stalk that is including germ cell tumors (GCTs), pineal
partially ependymal-lined. The pineal gland is parenchymal tumors,5 glial tumors, and
a neuroendocrine gland that is solid in child- meningiomas. In addition, several non-
hood and has a tendency to calcify later in neoplastic mass lesions, including cysts6 and
life. Sometimes cysts form; these are usually vascular malformations, may mimic pineal
but not always asymptomatic.1 The pineal region tumors (Table 8.1). The most common
gland, often referred to as a third eye, recog- pineal region tumor, the GCT, can also occur
nizes light and darkness via the sympathetic in the suprasellar area. All together, tumors
nervous system. Retinal fibers project of the pineal region represent about 1% of
indirectly to the pineal gland; retinal photore- intracranial tumors in adults and about 8%
ceptors project to the suprachiasmatic nucleus in children (Table 8.2).
of the hypothalamus, and then via the median Several non-neoplastic lesions found in the
forebrain bundle to the brainstem, the spinal pineal region include epidermoid8 and
cord and the superior cervical ganglion. dermoid cysts (see Chapter 12) and vascular
Sympathetic fibers from the superior cervical anomalies, in particular aneurysms of the vein
ganglion innervate the pineal gland. In of Galen. About one-third of all pineal region
amphibians, there are photoreceptors within masses are benign. The other two-thirds are
the pineal gland, making it truly a third eye. malignant tumors, but the single most
The pineal gland produces the hormone common tumor type, the germinoma, is
melatonin, which appears to play an impor- curable. Pineal parenchymal tumors, GCTs
tant role in the sleepwake cycle.2 The pineal and cysts are discussed in this chapter.
gland is enlarged in some patients with Gliomas arising in the pineal region, usually
systemic cancer but the cause of that enlarge- from the tectum of the midbrain, were
ment is not known.3 discussed in Chapter 5.

249
PINEAL REGION TUMORS

Figure 8.1
Distribution of tumors that commonly arise
in the pineal region. The tumors discussed
in this chapter mostly arise in or around the
pineal gland (A), compressing the aqueduct
and causing hydrocephalus. A few, especially
germinomas in females, arise in the
suprasellar area (B) and cause visual and
endocrinologic disturbances. Occasional
A
B
tumors invade the hypothalamus.

Table 8.2
Relative frequency of pineal region tumors in US series.

Tumor % of total
US European
Series* Series**
Table 8.1
Germ-cell 37 35
Pineal region tumors.
Germinomas 17
Teratomas 6
a
NGGCT 14
Pineal parenchymal tumors
Pinealoma 22 28
Pineocytoma
Pineocytoma 12 7
Pineoblastoma
Pineoblastoma or mixed 10 21
Intermediate differentiation (see text)
Glial 28 27
Germ cell tumors
Astrocytoma 19
Germinoma
Oligodendroglioma and 3
Non-germinomatous germ cell tumors:
mixed glioma
Embryonal carcinoma
Ependymoma 5
Yolk sac tumor (endodermal sinus tumor)
Other 13 10
Choriocarcinoma
Cyst 3
Teratoma
Meningioma 6 7
Mature or immature mixed tumors
Melanoma 1
Others
Metastasis 1
Meningioma
Other 4
Tectal astrocytoma
Vein of Galen aneurysms aNon-germinomatous germ cell tumors.
Dermoid and epidermoid cysts *Data in US series from Paker and Cohen7
Pineal cysts **Data from European patients, from Fauchon et al.5

250
PINEAL PARENCHYMAL TUMORS

Figure 8.2
A pineocytoma in an adult. The panel on the top left shows an enhancing tumor of the pineal compressing the
tectum. The lesion cannot be distinguished from other pineal region tumors by its MR characteristics alone.
Sometimes, if the tumor is surrounded by normal pineal calcification, one can infer that the tumor arose within the
pineal gland and is either a pineocytoma or a pineoblastoma. Histologically, one sees solid sheets of moderately-sized
cells with fibrillar processes occasionally forming pineocytomatous rosettes. No mitoses are identified.

Pineal parenchymal tumors diate differentiation9 with an unpredictable


growth rate and clinical behavior. In addition,
Introduction cysts of the pineal gland can occasionally be large
enough to become symptomatic.6 Pineal
There are three types of pineal parenchymal parenchymal tumors occasionally complicate
tumors (Table 8.1), the relatively low-grade inherited retinoblastoma in children,10 the so-
pineocytoma, the high-grade pineoblastoma (Fig. called trilateral retinoblastoma. The higher-grade
8.2) and pineal parenchymal tumors of interme- tumors account for approximately one-third of

251
PINEAL REGION TUMORS

parenchymal pineal tumors, have a slight male


preponderance, and are usually found in children
and young adults but occasionally occur in older
individuals.11 The lower-grade tumors tend to
occur in adults and account for about 40% of
pineal parenchymal tumors. The remaining
2530% are intermediate or mixed tumors. Both
pineocytomas and pineoblastomas can behave
aggressively, and either can seed the
leptomeninges at diagnosis or at relapse,
although pineocytomas do so rarely.12 Both
pineal parenchymal tumors are derived from
pineocytes and neuroepithelial cells that have Figure 8.3
both photosensory and neuroendocrine The histology of a pineoblastoma. The tumor is
functions. Pineocytoma cells label with synapto- typically more cellular with small hyperchromatic
nuclei, and has many mitoses and areas of necrosis.
physin and for neurofilaments, demonstrating An MRI of a pineoblastoma cannot be distinguished
neuronal differentiation. Pineal parenchymal from that of a pineocytoma.
tumors cannot be distinguished from each other
clinically or by imaging.

and for neurofilaments, evidence of neuronal


Etiology differentiation. These neoplastic cells may be
Aside from hereditary retinoblastoma, no other capable of synthesizing serotonin and
environmental risk factors for pineal parenchy- melatonin, the neurotransmitters secreted by
mal tumors have been unequivocally identified. the normal pineal gland, although this does
Experiments in mice have implicated SV-40 not cause clinical syndromes of hypersecre-
virus in the development of trilateral retino- tion. The pineocytoma is a slowly growing
blastoma.13 Multiple chromosomal abnormali- tumor that can demonstrate neuronal, glial,
ties have been described in pineal tumors with melanocytic, photoreceptor and mesenchymal
higher grade tumors, showing gains of 12q and differentiation.
loss of 22.14 Pineoblastomas are much more cellular, are
more primitive appearing, and have increased
mitotic activity and sometimes necrosis; they
Pathology
tend to resemble cerebellar medulloblastomas
Grossly, pineocytomas tend to be well-circum- and may have HomerWright rosettes. Some
scribed. They may show hemorrhage or cystic consider them a supratentorial primitive
change but usually no necrosis. Pineo- neuroectodermal tumor (Chapter 7). They are
blastomas are usually larger, and hemorrhage often included in that category in clinical trials.
and necrosis are more common. Neither Fauchon et al5 recognize four grades of
tumor calcifies. Pineocytomas are moderately pineal parenchymal tumors that correlate with
cellular with delicate connective tissue stroma prognosis: grade 1a pineocytoma, without
and HomerWright rosettes15 (Fig. 8.3). mitosis and positive immunostaining for
Pineocytoma cells label with synaptophysin neuron-specific enolase, synaptophysin,

252
PINEAL PARENCHYMAL TUMORS

chromogranin A and neurofilaments; grade 2, Table 8.3


a transitional tumor with fewer than 6 Symptoms and signs of pineal parenchymal tumors.
mitoses/10 high power fields (HPF) and
positive immunostaining; grade 3, either fewer Generalized
than 6 mitoses/10 HPF but negative or weak Headache
immunostaining or more than 6 mitoses/HPF Nausea/vomiting
Visual obscurations
with immunostaining; grade 4 pineoblastoma,
Papilledema
with many mitoses and weak or absent Tinnitus
immunostaining. Confusion
Vertigo/dizziness
Cognitive dysfunction
Clinical findings Endocrine
Insomnia
Patients with pineal tumors usually present Precocious puberty
with hydrocephalus (63/76 patients5) caused by Focal
the mass compressing the Sylvian aqueduct. Ataxia
Because the superior and inferior colliculi Parinauds syndrome
immediately underlie the pineal gland, the Hearing loss
compressing mass also causes eye movement
abnormalities, specifically Parinauds syndrome
(paralysis of upward gaze, pupils that fail to
respond to light but constrict when the patient can cause visual loss, diplopia or abrupt loss
accommodates, i.e. light/near pupillary dissoci- of consciousness, mimicking a subarachnoid
ation and convergenceretraction nystagmus). hemorrhage.
Rarely, the tumor causes hearing loss from Pineocytomas are usually relatively small
compression of auditory fibers in the inferior when they present (less than 3 cm in diameter).
colliculus (Fig. 8.1). On MR scan, obstructive hydrocephalus is
In one series of 30 patients,16 headaches were almost always present; and the lesion can be
present in 73%, impaired vision in 47%, seen compressing the Sylvian aqueduct. The
nausea and vomiting in 40%, impaired tumors are hypointense on the T1-weighted
ambulation (ataxia) in 37%, cognitive dysfunc- image and hyperintense on the T2-weighted
tion in 27%, vertigo and dizziness in 23%, image with homogeneous enhancement after
fatigue in 17%, dysarthria in 13%, tinnitus in contrast. Pineoblastomas may be larger and
13% and changes in consciousness in 13%. appear more heterogeneous after contrast
Signs included papilledema (60%), ataxia enhancement but otherwise cannot be distin-
(50%), and Parinauds syndrome (30%) (Table guished radiographically. Leptomeningeal
8.3). dissemination can often be identified by
Other signs sometimes result from brainstem enhancement of the cerebral or spinal
or cerebellar compression. Insomnia is a rare meninges. Because these tumors cannot be
symptom of a pineal region tumor2 or its treat- distinguished from GCTs of the region,
ment;17 melatonin relieves this symptom. Like if hydrocephalus or increased intracranial
pituitary tumors (Chapter 10), pineal tumors pressure do not contraindicate, a lumbar
may bleed, although they do so rarely. A puncture to measure cerebrospinal fluid (CSF)
sudden hemorrhage, called pineal apoplexy, markers for GCT should be performed in the

253
PINEAL REGION TUMORS

preoperative period. Usually, it is not safe to setting of persistent or recurrent primary


perform a lumbar puncture, and diagnosis is tumor.16 Following surgical diagnosis, the
made by biopsy. If hydrocephalus is a major entire neuraxis is irradiated with a focal boost
problem threatening herniation, and a shunting to the pineal region. Chemotherapy, either
procedure is required, ventricular CSF should preceding or following RT, using drugs such
be assessed for the presence of both tumor cells as etoposide, cisplatin or carboplatin,
and CSF markers. vincristine and cyclophosphamide, improves
prognosis.24 The 1-, 3- and 5-year survival
rates for pineoblastoma are 88%, 78% and
Treatment
58%.
Because of the potential danger of surgery in Pineocytomas and pineoblastomas are most
and around the pineal region, some investiga- likely to recur locally. Spinal dissemination
tors have recommended radiation therapy (RT) occurs, but usually when there is also residual
without biopsy. Others have suggested stereo- or recurrent local tumor. Chemotherapy, either
tactic needle biopsy,18 but sampling error can conventional or experimental, is the first
occur. Because of the dangers of incorrect treatment modality used at relapse. Recurrent
histologic diagnosis using either CSF cytology disease is treated with radiotherapy if it was
or small samples achieved by stereotactic not administered at diagnosis.
needle biopsy, and because of the relative safety Pineal cysts are usually asymptomatic and
of modern neurosurgery, we recommend that have a high incidence in young women.25 Most
all lesions be biopsied and, if possible, resected are identified inadvertently when an MR scan
by an open surgical approach. In hydrocephalic is performed for another reason or the patient
patients, ventriculoperitoneal shunts may be has non-specific headache which is unrelated
placed prior to biopsy.19 Frameless stereotactic to the cyst. However, pineal cysts can reach
surgical techniques and endoscopic approaches a large size and occasionally become
have rendered surgery much safer.20 symptomatic.6 Symptomatic cysts average over
Several different surgical approaches are avail- 15 mm in diameter, whereas asymptomatic
able.21,22 One is a combined supra-infratentorial cysts are usually less than 10 mm. The
trans-sinus approach.23 Alternatively, a supra- symptoms are similar to those of a pineal
cerebellar, infratentorial approach can facilitate region tumor, including hydrocephalus and
tumor resection. For pineocytomas, the treat- Parinauds syndrome. A sudden increase in cyst
ment following surgery is not established. These size from hemorrhage may lead to acute hydro-
are almost always benign tumors that rarely cephalus with loss of consciousness and, rarely,
metastasize and have 1-, 3- and 5-year survival sudden death. The cysts differ from pineal
rates of 100%, 100% and 67% respectively, parenchymal tumors on MR scan in that they
after complete resection alone.16 RT should be are hypointense on T1 and hyperintense on T2,
delivered to tumors that are incompletely have a fluid-filled appearance and usually a
resected. Radiosurgery has been reported to be thin rim of contrast enhancement defining the
useful for the treatment of pineocytomas as well cyst wall. They are restricted to the pineal
as some other tumors within the pineal region.18 gland and, if symptomatic, can be treated by
Pineoblastomas are highly malignant. They ventricular shunt, 3rd ventriculostomy alone,
infiltrate surrounding structures and often surgical removal or drainage;26 however, simple
seed the leptomeninges, but usually in the cyst drainage risks recurrence.1

254
GERM CELL TUMORS

Germ cell tumors GCTs arise in midline structures, mostly in


the pineal region and the suprasellar cistern,
Introduction and sometimes in both areas simultaneously.
Less common sites of GCTs include the
GCTs mainly occur in children and adoles- cerebral ventricle, basal ganglia, thalamus and
cents27 and are identical in appearance to GCTs cerebellar hemisphere (Fig. 8.4). Germinomas
elsewhere, e.g. the ovary and testes. Their represent about 50% of intracranial GCTs
biological behavior is also similar but treat- (Table 8.4). Teratomas are the next most
ment differs.28 Most GCTs, except for mature common tumor (20%), with mature (relatively
teratomas, are highly malignant but do respond benign) teratoma being slightly more common
to therapy. Cure is usual in germinomas but than the more aggressive immature teratoma29
rare in the other malignant tumors. Therefore, or teratoma with malignant transformation.
GCTs are divided into germinomas and non- Mixed GCTs, yolk sac tumor (endodermal
germinomatous germ cell tumors (NGGCTs). sinus tumor),30 embryonal carcinoma and

Figure 8.4
A germinoma of the basal ganglia. This patient was 35 in 1990 when he noted that his vision was not right.
About a year later he first noticed diplopia on upward gaze (while playing tennis). His pupils were noted to be
poorly reactive to light. When he attempted to gaze upward, there was less than 5 of movement and there
was retractory nystagmus. Convergence was poor. An MR of the brain showed a lesion in the tectum and right
thalamus. A lumbar puncture showed nine white cells with normal biochemical markers and a negative
cytologic examination. A stereotactic needle biopsy revealed the lesion to be a granulomatous germinoma (see
Fig. 8.8). He was treated with chemotherapy. The tumor and symptoms disappeared and have not recurred.

255
PINEAL REGION TUMORS

Table 8.4
Histologic subtypes of central nervous system germ
Etiology
cell tumors. Specific environmental risk factors have not
been identified. There is an increased risk of
Type Approximate intracranial GCTs in patients with Klinefelters
percentage syndrome, a disorder characterized by 2 X
chromosomes, i.e. a 47 XXY genotype. Such
Germinoma 43 patients are also predisposed to mediastinal
Teratoma 20
Mixed tumors 20
GCTs. Less well established are GCTs associ-
Yolk sac tumors 2 ated with Downs syndrome, neurofibromato-
Embryonal carcinoma 4 sis type-1 (NF-1) and prior gonadal GCTs.
Choriocarcinoma 2 Interestingly, 6/14 brain tumors reported in
patients with Downs syndrome were GCTs.
From Campos et al,29 with permission.
Chromosomal imbalances include losses of
13q, 18q, 9q, 11q and gains on 12p, 8q and
1q;32 p53 mutations are absent,33 but telo-
choriocarcinomas together represent about merase activity is present.34
25% of tumors. Intracranial GCTs are believed by many to
GCTs, rare in Caucasians, are more common arise from embryonic cell rests that lie in the
in Asians. Taken together, GCTs represent only midline of the brain. However, unlike the
about 0.51% of all primary intracranial epithelial cell rests of Rathkes pouch that are
neoplasms encountered in the USA, and only found in some normals and are believed to give
3% of such neoplasms in children and adoles- rise to craniopharyngiomas (Chapter 10), no
cents. In Japan and Taiwan, however, GCTs such embryonic germ cells have been found in
account for at least 2% of all primary intra- the nervous system. Sano35 has proposed that
cranial neoplasms and as many as 15% of when the primitive streak begins to form,
pediatric intracranial neoplasms. In Japan and embryonic germ cells enter the primitive groove
Korea, GCTs represent 7080% of pineal and migrate with the moving cells of the
region tumors.31 About 90% of these tumors mesoderm to the neural plate. They may then
occur in those 20 years of age and younger, be enfolded into the neuraxis.
with a peak incidence in the second decade of The origin of the various kinds of intra-
life. Less than 10% of these tumors occur in cranial GCTs is also in dispute. The germ cell
patients over age 20, and probably less than theory of tumors posits that primordial germ
2% in those over age 30. Overall, the tumors cells give rise to either germinomas or totipo-
are 22.5 times more common in males than in tential cells, the latter giving rise to the
females, with an even higher male-to-female NGGCTs. Sano has pointed out that such a
ratio in the NGGCTs. There is also a sex scheme would make germinoma the most
predilection for location of GCTs: pineal primitive, and therefore the most malignant, of
tumors primarily occur in boys, and suprasellar GCTs, although its biological behavior is
tumors are more common in girls. In one actually among the most benign. He has
series, 76/78 (97%) pineal region GCTs were proposed the scheme outlined in Fig. 8.5, in
found in males, whereas 25/46 (54%) supra- which each of the various GCTs arises from a
sellar tumors were found in females.29 different stage of embryonic development.35

256
GERM CELL TUMORS

Figure 8.5
Hypothesis concerning
Ovum Sperm the origin of germ cell
tumors. Sano suggests
that each of the
Zygote individual tumors arises
at a different site in the
course of embryologic
development; the most
Morula primitive arising from
the blastocyst, forming
Trophoblast choriocarcinoma; the
Blastocyst (Cytotrophoblast Choriocarcinoma most well-formed arising
Syncytiotrophoblast) during the embryonic
+ period, forming a
Extraembryonic teratoma. From Sano35
Embryoblast mesoderm
with permission.
Primary
yolk sac

Embryonic Chorion
disc Endodermal
Secondary
yolk sac sinus tumour

Triploblast Embryonal
embryo carcinoma

Primordial Germinoma
germ cells

Embryonic Teratoma
period

Pathology frequent. Lymphocytes often infiltrate the


tumor, giving it a granulomatous appearance.
GCTs are solid, although many, particularly The inflammatory infiltrate may be so
the more malignant ones, show necrosis and pronounced that it leads to a misdiagnosis of
hemorrhage. Microscopically, the germinoma granuloma rather than neoplasm.36 Most
is identical to the testicular seminoma and germinomas label with placental alkaline
consists of large germ cells with a vesicular phosphatase but this may be hard to demon-
nucleus and prominent nucleoli. The cytoplasm strate in highly inflammatory tumors. Some
is abundant and may appear pale or clear germinomas contain syncytiotrophoblastic
because of glycogen accumulation. The cells giant cells that produce human chorionic
may appear as sheaths or lobules. Mitoses are gonadotropin (HCG). The presence of these

257
PINEAL REGION TUMORS

cells and the concomitant elevation of CSF for HCG, which distinguishes them from other
HCG is believed to diminish the usually excel- GCTs; they may also be positive for placental
lent prognosis of germinoma,37,38 although one alkaline phosphatase and cytokeratins.
study reports no difference in prognosis.39 Mixed tumors are relatively common and
Teratomas are composed of cells of different consist of cell types from any of the above-
tissue types. Small uniform cells with incon- listed tumors. These tumors carry the progno-
spicuous nucleoli and moderate eosinophilic sis of the most aggressive histology contained
cytoplasm may differentiate along neuronal or, within the specimen. An accurate histopatho-
rarely, astrocytic lines, expressing appropriate logic diagnosis is essential to determine treat-
immunohistochemical markers. Mitoses are ment. Only germinomas and teratomas usually
rare or absent, but occasional multinucleated occur as pure tumors, the others being mixed.
giant cells are identified. Mature teratomas The diagnosis is made both by routine histol-
contain fully differentiated tissue elements, and ogy and immunohistochemistry
may contain cartilage, bone, hair or even teeth.
Mitotic figures are sparse or absent. Immature
Clinical findings
teratomas show the same cell types but incom-
pletely differentiated, resembling fetal tissues. The symptoms and signs of GCTs depend on
Mitoses are common. Some teratomas undergo the location of the tumor (i.e. pineal or
malignant degeneration, with evidence of a suprasellar).40 Those tumors that arise in the
sarcoma or carcinoma within what would pineal region present in a fashion similar to
otherwise be a mature teratoma. Teratomas primary parenchymal pineal tumors (Table 8.5).
may be positive for -fetoprotein or cytoker- They usually present with a combination of
atins, the cytokeratin reactivity being a feature the generalized symptoms of hydrocephalus
of the epithelial components and the -fetopro- and the focal symptoms of tectal compression,
tein of enteric-type glandular components. e.g. Parinauds syndrome. Those tumors that
The yolk sac or endodermal sinus tumor is arise in the suprasellar area have symptoms
composed of primitive epithelial-like cells, indistinguishable from those of pituitary
sometimes growing in a background of myxoid region tumors (Chapter 10), e.g. bitemporal
tissue. The tumor has eosinophilic inclusions hemianopia, pituitary failure and often diabetes
within the cytoplasm that contain -fetopro- insipidus. Cognitive abnormalities (e.g. low IQ)
tein, a characteristic that distinguishes it from are common in patients with suprasellar GCTs,
a germinoma or embryonal carcinoma. The but not in those with pineal region tumors.41
tumors may also contain placental alkaline Those tumors that arise within the parenchyma
phosphatase and cytokeratins. of the brain, particularly the basal ganglia,
The embryonal carcinoma contains primitive cause focal symptoms appropriate to their
epithelial cells, usually with a clear cytoplasm. location (Fig. 8.5). One of our patients devel-
Mitoses are frequent and necrosis is common. oped a spastic left hemiparesis associated with
The cells stain for placental alkaline an enhancing lesion in the thalamus and inter-
phosphatase but they also stain for cytokeratin, nal capsule. The tumor was thought by clinical
which distinguishes them from the sometimes and MR criteria to be a glioma, but biopsy
similarly appearing germinomas. The chorio- revealed a germinoma.
carcinomas consist of cells resembling GCTs are occasionally associated with preco-
embryonic trophoblast. The cells are positive cious puberty, in either girls or boys. This is

258
GERM CELL TUMORS

Table 8.5
Frequency (%) Presenting symptoms and
signs clustered by
Pineal Suprasellar primary tumor location.
From Kretschmar27 with
Headache 4778 21 permission.
Diplopia; visual deficits 33 33
Parinauds syndrome 3442 14
Papilledema; hydrocephalus 4761 21
Lethargy; obtundation 2226 15
Ataxia 20 9
Diabetes insipidus 1018 41
Hypothalamicpituitary dysfunction 19 33
Precocious puberty 6 3
Growth delay 4 9
Menstrual abnormalities 34 16

probably a result of tumor involvement of the in boys, but not girls, harboring HCG-produc-
hypothalamus, releasing the immature gonads ing intracranial germ cell neoplasms.
from tonic inhibitory control. Alternatively, GCTs cannot be distinguished from other
HCG, elaborated by the tumor, can act as a pineal region tumors on MR scan (Fig. 8.6).42
stimulant of testosterone production. This They usually are iso- or hypointense on the
mechanism would explain precocious puberty T1-weighted image and isointense with brain

Figure 8.6
Meningioma of the pineal region. A large isointense mass on the T1-weighted image appears to be within the
pineal region, compressing the tectum (left panel, arrow). The lesion contrast enhances and one sees a dural tail
(right panel, arrow) which suggests a meningioma rather than a germ cell tumor.

259
PINEAL REGION TUMORS

Table 8.6
Biological tumor markers in germ cell tumors.

Tumor -Fetoprotein -Human Human Lactate Placental Cytokeratins


histology chorionic placental dehydrogenase alkaline
gonadotropin lactogen isoenzymes phosphatase

Teratoma +/ +
Germinoma + +
Embryonal carcinoma +/ +/ + +
Choriocarcinoma ++ + +/ +
Yolk sac tumor ++ +
(endodermal sinus)

Modified from Kleihues et al.45

on the T2-weighted image with fairly intense HCG. Low levels of HCG in the spinal fluid
homogeneous contrast enhancement. One (< 2000 mIU/ml) may be seen in germinomas
radiographic feature that differentiates GCTs which contain syncytiotrophoblastic giant cells.
from primary pineal tumors is that the However, the presence of detectable HCG in
GCT appears to surround the pineal gland, the CSF may predict a higher rate of recurrence
compressing pineal calcifications, whereas than if no HCG is present.37 Absence of all
parenchymal pineal tumors explode the markers in the spinal fluid suggests that the
pineal gland, dispersing pineal calcifications. lesion is a germinoma or a teratoma. A grossly
Teratomas are often cystic and may have elevated level of HCG establishes the diagnosis
calcified regions as well as areas of lipid that of choriocarcinoma; a grossly elevated
are hyperintense on both T1- and T2- -fetoprotein suggests a yolk sac tumor, and
weighted images. When germinomas occur in an elevated placental alkaline phosphatase, a
the basal ganglia or thalamus, they are gener- germinoma. These markers are quite reliable,
ally cystic and some are hemorrhagic. The but because of mixed tumors an accurate
signal intensity is heterogeneous on T1- and histopathologic diagnosis is essential to deter-
T2-weighted images and the tumor usually mine treatment.
enhances.43
Examination of the CSF for biochemical
Treatment
markers often helps establish the diagnosis
(Table 8.6). Cytologic examination demonstrates Treatment depends on the histologic type and
tumor cells in only 1015% of patients.44 stage of the disease (Fig. 8.7). Prior to therapy
Important markers include -fetoprotein, - a spinal MR scan with contrast to search for
HCG and placental alkaline phosphatase. subarachnoid metastases (mature teratomas do
Lactate dehydrogenase isozymes are less impor- not require further imaging), lumbar and, if a
tant. As Table 8.6 indicates, germinomas shunt or ventriculostomy is present, ventricular
normally do not excrete either -fetoprotein or CSF should be examined for malignant cells.

260
GERM CELL TUMORS

MR head Pineal tumor

Hydrocephalus No hydrocephalus

Consider shunt or Lumbar CSF for


ventriculostomy CSF markers and cytology
markers and cytology

Surgery

Meningioma Mature teratoma Pineocytoma Pineoblastoma Germinoma NGGCT

Total Subtotal Neuraxis Neuraxis Neuraxis


resection resection MR MR MR

CSF if CSF if CSF if


not done not done not done

Follow Follow Follow RT focal RT + chemo RT focal Chemo + focal


neuraxis RT (neuraxis
RT if the
leptomeninges
are involved)

Figure 8.7
Approach to pineal region tumor. RT, radiation therapy.

Although some physicians will treat on the common and a small sample may prevent
basis of imaging studies and spinal fluid exami- accurate histologic identification of all compo-
nation, most require tumor histology to deter- nents in a mixed tumor; the most aggressive
mine treatment. The larger the tissue sample histology identified in such a lesion determines
the better, because mixed tumors are relatively the therapeutic approach.

261
PINEAL REGION TUMORS

We recommend a surgical approach to all and is probably related to tumor vascularity


pineal region tumors with the intent of achiev- and malignancy. Risk factors for neurologic
ing a maximally feasible resection, except for deficit include highly malignant and vascular
pure germinomas (see below). Pineal region tumors, preoperative neurologic deficit and
tumors can be approached surgically in several preoperative RT.
ways.20,22 They can be approached supratento- Diagnostic stereotactic needle biopsy is the
rially via a parietal interhemispheric approach, preferred approach for some patients, such as
or an occipital transtentorial approach. Tumors those with basal ganglia or thalamic tumors, or
can also be approached via an infratentorial, with disseminated tumor, in whom CSF does
supracerebellar approach; the technique is not yield a definitive diagnosis. Stereotactic
performed with the patient in the prone biopsies in the pineal region carry a greater risk
position, with the head lower than the heart. of hemorrhage than in other areas of the brain,
This avoids the complication of air embolism particularly when the tumor is highly vascular.
that can occur with patients in the sitting However, both morbidity and mortality are
position. Some tumors can be approached low and there are times when stereotactic
endoscopically.20 Most surgeons believe that biopsy should be performed in preference to
radical debulking of the more malignant open surgery. Endoscopic biopsy via the 3rd
tumors leads to a better prognosis.46 In one ventricle may be safer in some tumors.
series of operations for pineal region tumors, After the histologic diagnosis is established,
gross total removal was possible in 90% of patients can be divided into those with pure
benign tumors and 25% of malignant tumors. germinoma (Fig. 8.8), those with mature
Furthermore, for NGGCTs, survival depends teratomas, and those with other NGGCTs. All
on the extent of resection. In one series, the patients, except those with pure mature
3-year survival was 73% after complete re- teratomas, should have a complete staging
section, 32% after partial resection and 0% evaluation if not performed preoperatively.
after biopsy.46 Because germinomas are curable This should include a complete spine MRI with
with radiation, radical resection is not gadolinium and a CSF examination. In those
required. If the diagnosis of germinoma is few patients with preoperative studies which
strongly suspected clinically and on the basis of show tumor dissemination, the diagnosis may
pre-operative CSF data, and the diagnosis can be established by cytologic examination of
be established by biopsy, many believe that no ventricular or lumbar CSF. Furthermore, serum
additional risks should be taken by continuing or CSF tumor markers may delineate the
the resection.47 pathologic diagnosis, particularly when only a
With modern techniques, operative mortality biopsy was obtained. For example, a biopsy
varies from 0%8%, usually 12%, and demonstrating teratoma associated with a very
permanent morbidity from 0%12%, usually high CSF -fetoprotein level suggests that the
56%. The common complications include tumor contains endodermal sinus components
ocular movement disorders, postoperative not sampled by biopsy, and the patient should
confusion that may precede permanent cogni- be treated accordingly. Spinal MRI should be
tive dysfunction, ataxia and aseptic meningitis performed before CSF is obtained to avoid
(Chapter 7). Many of the neurologic deficits possible meningeal enhancement induced by
are transient. Hemorrhage into the tumor bed the lumbar puncture, which can be confused
may occur up to several days postoperatively with subarachnoid tumor dissemination. The

262
GERM CELL TUMORS

Figure 8.8
A pineal region germinoma. This 30-year-old man presented to medical attention with headache but no other
symptoms. An MR scan revealed hydrocephalus with marked periventricular hyperintensity, indicating
transependymal spinal fluid absorption (right panel). A uniformly contrast-enhancing tumor was found in the
pineal region, obstructing the ventricular system (left panel). Biopsy revealed a pure germinoma. The tumor was
treated with radiation therapy with complete resolution. This patient had an unusual but occasionally described
symptom during radiation. Each time the beam was turned on, he was able to see blue light through his closed
eyes in a darkened room. He was told by the technician and a junior radiation oncologist that it must be a
psychological reaction, but the symptom is well-described in the literature.65 Even rarer is the ability to smell
the radiation beam, which this patient did not experience. The photomicrograph shows neoplastic germ cells
with large vesicular nuclei and clear cytoplasm, interspersed with typical collections of mature T-cells (arrow).

CSF should be examined about 2 weeks additional 30.6 Gy to the tumor bed in 1.8-Gy
postoperatively to allow time for biochemical fractions (Fig. 8.9). Some advocate lower doses
markers and cells shed by the resected tumor of radiation, either independently or combined
but not representing dissemination to be with chemotherapy.48,49 If either CSF cytology
cleared from CSF. or a contrast-enhanced MRI of the spine
In patients with germinoma, surgery should suggests leptomeningeal dissemination, cranio-
be followed by RT to a portal that includes the spinal irradiation is required.50 Radiosurgery
site of the tumor, either pineal or suprasellar, may have a role by providing a local boost to
and the lateral and 3rd ventricles. A total tumor the pineal region tumor to reduce overall radia-
dose of 50.4 Gy is usually sufficient. The usual tion exposure of the remainder of the brain.
dose of RT is 19.8 Gy delivered to Rarely, transient spontaneous or steroid-
the lateral 3rd and 4th ventricles, with an induced regression of intracranial germinomas

263
PINEAL REGION TUMORS

Figure 8.9
Radiation portal for
treatment of a germinoma.
Focal treatment to the pineal
region alone is not sufficient
to control tumor recurrence.
A radiation portal that
encompasses both the pineal
and the supratentorial
ventricular system, as seen in
this illustration, cures more
than 90% of patients.
Whole-neuraxial radiation is
unnecessary.

has been described;51,52 steroid regression may radiation dosage, particularly in patients with
be related to lymphocytic elements within the leptomeningeal dissemination at diagnosis.56
tumor53 (Chapter 11). NGGCTs require maximal surgical resection,57
Some investigators have recommended RT and chemotherapy in all cases.57 RT is deliv-
chemotherapy as the primary treatment for ered to a total dose of 54 Gy to a focal field
germinomas54 in an effort to eliminate cranial involving the 3rd and 4th ventricles. Any
RT and its potential for neurotoxicity. Most evidence of leptomeningeal dissemination
tumors respond, but the regimens are toxic and mandates neuraxis RT. In these instances, pre-
there is a 50% relapse rate within 2 years, irradiation chemotherapy is probably indicated,
requiring radiation.55 RT is efficacious even using multiagent chemotherapy, usually cisplatin,
after failed chemotherapy. Accordingly, in etoposide, vincristine and cyclophosphamide.46
adolescents and adults, we recommend RT, If there is a response after two cycles of
which has a 10-year progression-free survival chemotherapy, it should be continued for six
of about 90%. Furthermore, RT appears to be cycles. Some suggest high-dose chemotherapy
well tolerated in this population, with few with stem cell or bone marrow rescue.58 The
significant long-term neurologic sequelae.38 efficacy of this approach is not known,
Chemotherapy may allow for a reduction in because the number of patients so treated is

264
GERM CELL TUMORS

Table 8.7
Histology 5-year survival (%) 10-year survival (%) Prognosis of intracranial
germ cell tumors.
Good prognosis
Pure germinoma 95 92
Mature teratoma 93 93
Intermediate prognosis
Germinoma with STGCa 83 83
Immature teratoma 70 70
Mixed tumorb 53 35
Poor prognosis
Choriocarcinoma
Yolk sac tumor 9
Embryonal tumor
Other mixed tumor

Modified from Tagliabue et al,3 with permission.


aSTGC, syncytiotrophoblastic giant cell.

bMainly germinoma or teratoma.

small but some reports are enthusiastic.58 Prognosis


Whether or not successful, chemotherapy
should be followed by RT. The dose of 54 Gy The major factor that determines prognosis is
is the same as that for NGGCTs. Adjuvant the tumor type (Table 8.7); a lesser factor is the
chemotherapy is usually not well tolerated extent of the surgical resection (Fig. 8.10).38,46
because of limited bone marrow reserve For mixed tumors, the amount of malignant
following neuraxis RT. tissue is also prognostic. For example, a mature
Mature teratomas require no treatment teratoma with a small amount of choriocarci-
beyond surgery if they can be successfully noma has a better prognosis than a mature
resected. Some mature teratomas involve the teratoma containing a large amount of chorio-
hypothalamus or hypophyseal region, and only carcinoma.46
partial removal or biopsy is possible. In those Yolk sac tumors, embryonal carcinomas and
cases, some have advocated adjuvant choriocarcinomas have a poor prognosis; these
chemotherapy without radiation.59 The recom- tumors usually relapse within 2 years, and
mended regimen is etoposide and cisplatin. dissemination through the leptomeninges is
Immature teratomas require postoperative RT common. Some children with pineal region
combined with chemotherapy. One regimen tumors are at risk for cognitive deficits leading
includes ifosfamide, cisplatin, etoposide and to school failure. The complications of irradi-
local radiation to a dose of 50 Gy. The progno- ation are discussed in Chapter 4. However,
sis of immature teratomas is relatively good, recent evidence suggests that such children
with one series yielding 5- and 10-year survival have an excellent quality of life as adults.61
rates of 70%.60 Because the cure rate for germinomas is also so

265
PINEAL REGION TUMORS

Figure 8.10
A pineal region primary central nervous system melanoma. This patient presented with headache and
hydrocephalus. The tumor was a melanocytoma with elements of melanoma (Chapter 6). The CSF and spinal
MRI were normal. Two years after focal radiation therapy, the patient is free of disease.

high, both these groups are at risk for very late of local recurrence as well. Systemic metastases
complications (often greater than 20 years), occur but are rare. Recurrent disease is treated
including secondary tumors within the radia- with chemotherapy if the patient has not
tion portal. These include malignant menin- received chemotherapy before, or with differ-
giomas and malignant astrocytomas. ent agents if the patient has received prior
The prognosis for survival in patients with chemotherapy. Focal spinal RT may be an
suprasellar germinomas is also excellent, but option for symptomatic spinal leptomeningeal
many suffer cognitive and endocrine dysfunction metastases, depending upon prior neuraxis RT.
despite successful RT.62 However, quality of life
seems good; many patients go on to complete
college and even graduate school.63 The role that
radiotherapy plays in causing cognitive dysfunc- References
tion is unclear because many patients with
suprasellar GCTs have cognitive abnormalities 1. Mena H, Armonda RA, Ribas JL, Ondra SL,
preceding treatment.41 Other complications are Rushing EJ. Nonneoplastic pineal cysts: a clini-
seen rarely. One patient with a suprasellar copathologic study of twenty-one cases. Ann
Diagn Pathol 1997; 1: 118.
germinoma developed a vascular malformation
2. Etzioni A, Luboshitzky R, Tiosano D et al.
of the spinal cord 5 years after neuraxis RT.64 Melatonin replacement corrects sleep distur-
GCTs usually recur locally. Spinal dissemi- bances in a child with pineal tumor. Neurology
nation may occur, but usually in the presence 1996; 46: 2613.

266
REFERENCES

3. Tagliabue M, Lissoni P, Barni S et al. A radio- 16. Schild SE, Scheithauer BW, Schomberg PJ et al.
logic study by CT scan of pineal size in cancer Pineal parenchymal tumors. Clinical, patho-
patients: correlation to melatonin blood levels. logic, and therapeutic aspects. Cancer 1993;
Tumori 1989; 75: 2268. 72: 87080.
4. Schild SE, Scheithauer BW, Haddock MG et al. 17. Jan JE, Tai J, Hahn G, Rothstein RR.
Histologically confirmed pineal tumors and Melatonin replacement therapy in a child with
other germ cell tumors of the brain. Cancer a pineal tumor. J Child Neurol 2001; 16:
1996; 78: 256471. 13940.
5. Fauchon F, Jouvet A, Paquis P et al. 18. Kreth FW, Schatz CR, Pagenstecher A et al.
Parenchymal pineal tumors: A clinicopatho- Stereotactic management of lesions of the
logical study of 76 cases. Int J Radiat Oncol pineal region. Neurosurgery 1996; 39: 2809.
Biol Phys 2000; 46: 95968. 19. Brandes AA, Pasetto LM, Monfardini S. The
6. Engel U, Gottschalk S, Niehaus L et al. Cystic treatment of cranial germ cell tumours. Cancer
lesions of the pineal region MRI and pathol- Treat Rev 2000; 26: 23342.
ogy. Neuroradiology 2000; 42: 399402. 20. Oi S, Shibata M, Tominaga J et al. Efficacy
7. Packer RJ, Cohen BH. Germ-cell tumors and of neuroendoscopic procedures in minimally
pineal tumors. In: Vecht CJ, ed. Neuro- invasive preferential management of pineal
Oncology. Part II. Amsterdam: Elsevier 1997: region tumors: a prospective study. J
22956. Neurosurg 2000; 93: 24553.
8. Konovalov AN, Spallone A, Pitzkhelauri DI. 21. Saenz A, Zamorano L, Matter A, Bucius R,
Pineal epidermoid cysts: diagnosis and manage- Diaz F. Interactive image guided surgery of
ment. J Neurosurg 1999; 91: 3704. the pineal region. Minim Invasive Neurosurg
9. Kleihues P, Cavenee WK. World Health 1998; 41: 2730.
Organization Classification of Tumours: 22. Fukui M, Natori Y, Matsushima T, Nishio S,
Tumours of the nervous system pathology Ikezaki K. Operative approaches to the pineal
and genetics. Lyon: IRAC Press, 2000. region tumors. Child Nerv Syst 1998; 14: 4952.
10. Kivela T. Trilateral retinoblastoma: a meta- 23. Ziyal IM, Sekhar LN, Salas E, Olan WJ.
analysis of hereditary retinoblastoma associ- Combined supra/infratentorial-transsinus ap-
ated with primary ectopic intracranial proach to large pineal region tumors. J
retinoblastoma. J Clin Oncol 1999; 17: Neurosurg 1998; 88: 10507.
182937. 24. Kurisaka M, Arisawa M, Mori T et al.
11. Chang SM, Lillis-Hearne PK, Larson DA et al. Combination chemotherapy (cisplatin,
Pineoblastoma in adults. Neurosurgery 1995; vinblastin) and low-dose irradiation in the
37: 38390. treatment of pineal parenchymal cell tumors.
12. Ito T, Takahashi H, Ikuta F, Sato H. Childs Nerv Syst 1998; 14: 5649.
Metastatic pineocytoma of the spinal cord after 25. Sawamura Y, Ikeda J, Ozawa M et al.
long-term dormancy. Pathol Int 1994; 44: Magnetic resonance images reveal a high
8604. incidence of asymptomatic pineal cysts in
13. Marcus DM, Brooks SE, Leff G et al. Trilateral young women. Neurosurgery 1995; 37: 115.
retinoblastoma: insights into histogenesis and 26. Kang HS, Kim DG, Han DH. Large glial cyst of
management. Surv Ophthalmol 1998; 43: 5970. the pineal gland: a possible growth mechanism.
14. Rickert CH, Simon R, Bergmann M, Case report. J Neurosurg 1998; 88: 13840.
Dockhorn-Dworniczak B, Paulus W. 27. Kretschmar CS. Germ cell tumors of the brain
Comparative genomic hybridization in pineal in children: a review of current literature and
parenchymal tumors. Genes Chromosom new advances in therapy. Cancer Invest 1997;
Cancer 2001; 30: 99104. 15: 18798.
15. Fvre-Montange M, Jouvet A, Privat K et al. 28. Hooda BS, Finlay JL. Recent advances in the
Immunohistochemical, ultrastructural, bio- diagnosis and treatment of central nervous
chemical and in vitro studies of a pineocytoma. system germ-cell tumours. Curr Opin Neurol
Acta Neuropathol (Berl) 1998; 95: 5329. 1999; 12: 6936.

267
PINEAL REGION TUMORS

29. Matsutani M, Sano K, Takakura K et al. 41. Kitamura K, Shirato H, Sawamura Y et al.
Primary intracranial germ cell tumors: a clini- Preirradiation evaluation and technical assess-
cal analysis of 153 histologically verified cases. ment of involved-field radiotherapy using
J Neurosurg 1997; 86: 44655. computed tomographic (CT) simulation and
30. Chandy MJ, Damaraju SC. Benign tumours of neoadjuvant chemotherapy for intracranial
the pineal region: a prospective study from 1983 germinoma. Int J Radiat Oncol Biol Phys 1999;
to 1997. Br J Neurosurg 1998; 12: 22833. 43: 7838.
31. Oi S, Matsuzawa K, Choi JU et al. Identical 42. Satoh H, Uozumi T, Kiya K et al. MRI of
characteristics of the patient populations with pineal region tumours: relationship between
pineal region tumors in Japan and in Korea tumours and adjacent structures. Neuro-
and therapeutic modalities. Childs Nerv Syst radiology 1995; 37: 62430.
1998; 14: 3640. 43. Kim DI, Yoon PH, Ryu YH, Jeon P, Hwang
32. Rickert CH, Simon R, Bergmann M, Dockhorn- GJ. MRI of germinomas arising from the basal
Dworniczak B, Paulus W. Comparative genomic ganglia and thalamus. Neuroradiology 1998;
hybridization in pineal germ cell tumors. J 40: 50711.
Neuropathol Exp Neurol 2000; 59: 81521. 44. Paulino AC, Wen B-C, Mohideen MN.
33. Nozaki M, Tada M, Matsumoto R et al. Rare Controversies in the management of intra-
occurrence of inactivating p53 gene mutations in cranial germinomas. Oncology (Huntingt)
primary nonastrocytic tumors of the central 1999; 13: 51330.
nervous system: reappraisal by yeast functional 45. Kleihues P, Cavenee WK. Pathology and genet-
assay. Acta Neuropathol (Berl) 1998; 95: 2916. ics of tumours of the nervous system. Lyons:
34. Hiraga S, Ohnishi T, Izumoto S et al. International Agency for Research on Cancer,
Telomerase activity and alterations in telomere 1997.
length in human brain tumors. Cancer Res 46. Schild SE, Haddock MG, Scheithauer BW et al.
1998; 58: 211725. Nongerminomatous germ cell tumors of the
35. Sano K. Pathogenesis of intracranial germ cell brain. Int J Radiat Oncol Biol Phys 1996; 36:
tumors reconsidered. J Neurosurg 1999; 90: 55763.
25864. 47. Sawamura Y, de Tribolet N, Ishii N, Abe H.
36. Kraichoke S, Cosgrove M, Chandrasoma PT. Management of primary intracranial germino-
Granulomatous inflammation in pineal germi- mas: diagnostic surgery or radical resection? J
noma. A cause of diagnostic failure at stereo- Neurosurg 1997; 87: 2626.
taxic brain biopsy. Am J Surg Pathol 1988; 12: 48. Shibamoto Y, Sasai K, Oya N, Hiraoka M.
65560. Intracranial germinoma: radiation therapy with
37. Inamura T, Nishio S, Ikezaki K, Fukui M. tumor volume-based dose selection. Radiology
Human chorionic gonadotrophin in CSF, not 2001; 218: 4526.
serum, predicts outcome in germinoma. J 49. Brandes AA, Pasetto LM, Monfardini S. The
Neurol Neurosurg Psychiatry 1999; 66: 6547. treatment of cranial germ cell tumours. Cancer
38. Matsutani M, Sano K, Takakura K et al. Treat Rev 2000; 26: 23342.
Primary intracranial germ cell tumors: a clini- 50. Shibamoto Y, Oda Y, Yamashita J et al. The role
cal analysis of 153 histologically verified cases. of cerebrospinal fluid cytology in radiotherapy
J Neurosurg 1997; 86: 44655. planning for intracranial germinoma. Int J
39. Shin KH, Kim IH, Choe C. Impacts of elevated Radiat Oncol Biol Phys 1994; 29: 108994.
level of HCG in serum on clinical course and 51. Ide M, Jimbo M, Yamamoto M et al.
radiotherapy results in the histology-confirmed Spontaneous regression of primary intracranial
intracranial germinomas. Acta Oncol 2001; 40: germinoma a case report. Cancer 1997; 79:
98101. 55863.
40. Salzman KL, Rojiani AM, Buatti J et al. 52. Fujimaki T, Mishima K, Asai A, Suzuki I, Kirino
Primary intracranial germ cell tumors: clinico- T. Spontaneous regression of a residual pineal
pathologic review of 32 cases. Pediatr Pathol tumor after resection of a cerebellar vermian
Lab Med 1997; 17: 71327. germinoma. J Neurooncol 1999; 41: 6570.

268
REFERENCES

53. Mascalchi M, Roncaroli F, Salvi F, Frank G. considerations based on 34 cases. J Neurosurg


Transient regression of an intracranial germ 1998; 89: 72837.
cell tumour after intravenous steroid adminis- 60. Matsutani M, Sano K, Takakura K et al.
tration: a case report. J Neurol Neurosurg Primary intracranial germ cell tumors: A clini-
Psychiatry 1998; 64: 6702. cal analysis of 153 histologically verified cases.
54. Balmaceda C, Heller G, Rosenblum M, et al. J Neurosurg 1997; 86: 44655.
Chemotherapy without irradiation a novel 61. Bamberg M, Kortmann R, Calaminus G et al.
approach for newly diagnosed CNS germ cell Radiation therapy for intracranial germinomas:
tumors: results of an international cooperative results of the German Cooperative Prospective
trial. J Clin Oncol 1996; 14: 290815. Trials MAKEI 83/86/89. J Clin Oncol 1999;
55. Merchant TE, Davis BJ, Sheldon JM, Leibel 17: 258592.
SA. Radiation therapy for relapsed CNS germi- 62. Oka H, Kawano N, Tanaka T et al. Long-term
noma after primary chemotherapy. J Clin functional outcome of suprasellar germinomas:
Oncol 1998; 16: 2049. usefulness and limitations of radiotherapy. J
56. Buckner JC, Peethambaram PP, Smithson WA et Neurooncol 1998; 40: 18590.
al. Phase II trial of primary chemotherapy 63. Sutton LN, Radcliffe L, Goldwein JW et al.
followed by reduced-dose radiation for CNS Quality of life of adult survivors of germino-
germ cell tumors. J Clin Oncol 1999; 17: 93340. mas treated with craniospinal irradiation.
57. Robertson PL, DaRosso RC, Allen JC. Neurosurgery 1999; 45: 12927.
Improved prognosis of intracranial non-germi- 64. Maraire JN, Abdulrauf SI, Berger S, Knisely J,
noma germ cell tumors with multimodality Awad IA. De novo development of a cavernous
therapy. J Neurooncol 1997; 32: 7180. malformation of the spinal cord following
58. Tada T, Takizawa T, Nakazato F et al. spinal axis radiation case report. J Neurosurg
Treatment of intracranial nongerminomatous 1999; 90: 2348.
germ-cell tumor by high-dose chemotherapy 65. Steidley KD, Eastman RM, Stabile RJ.
and autologous stem-cell rescue. J Neurooncol Observations of visual sensations produced by
1999; 44: 716. cerenkov radiation from high-energy electrons.
59. Sawamura Y, Kato T, Ikeda J et al. Teratomas Int J Radiat Oncol Biol Phys 1989; 17:
of the central nervous system: treatment 68590.

269
9
Tumors of cranial nerves and skull base

Introduction large categories: tumors that arise from the


nerve sheath and directly invade cranial nerves,
Several intracranial tumors cause their and tumors that arise from bones or other
symptoms by invading or compressing cranial structures at the skull base and compress
nerves (Fig. 9.1). These tumors fall into two cranial nerves either within the intracranial
cavity or as they exit the skull to innervate the
various cranial organs (Table 9.1).
Although the two groups of tumors differ
with respect to histogenesis, biology, treatment
and prognosis, they are both considered in this
chapter because they often present to the physi-
cian with similar symptoms. Moreover, neither
group is restricted to cranial nerves or the base
of the skull. Nerve sheath tumors can arise
anywhere in the body and are particularly
common along spinal roots. Most of the
tumors that affect the base of the skull can also

C B
A Table 9.1
Tumors of cranial and spinal nerves and skull base.

Nerve sheath tumors


Schwannoma (neurilemmoma, neurinoma)
Neurofibroma
Malignant schwannoma
Skull base tumors
Paraganglioma (chemodectoma)
Figure 9.1 Chordoma
Location of cranial nerve tumors. Although any Chondroma or chondrosarcoma
cranial nerve can be involved, the most common are Carcinoma/lymphoma of the paranasal sinuses
the acoustic and trigeminal. (A) Vestibular Esthesioneuroblastoma
schwannoma in internal auditory canal. Meningioma (see Chapter 6)
(B) Vestibular schwannoma in cerebellopontine angle. Sarcoma (Ewings, rhabdomyosarcoma)
(C) Trigeminal schwannoma.

270
NERVE SHEATH TUMORS

affect the calvarium or other bones. account for 8% of intracranial tumors and
Nevertheless, cranial nerves are frequent represent about 30% of primary spinal tumors.
targets, and this chapter is a convenient place Incidental acoustic neuromas found at autopsy
to discuss the tumors as a group. from individuals dying of unrelated causes are
even more common. One autopsy study suggests
that acoustic neuromas may be present in over
2% of autopsies.1 The incidence of vestibular
Nerve sheath tumors schwannomas has increased from 7.8/106 per
year in 197683 to 12/106 per year in 199095,
Introduction
probably a result of easier and better diagnosis
The WHO recognizes four groups of nerve with MRI. The peak age is between 40 and 60.
sheath tumors: schwannomas, neurofibromas, Intracranial schwannomas are twice as common
malignant peripheral nerve sheath tumors, in women as in men.
also called neurogenic sarcoma or malignant Unilateral schwannomas are, for the most
schwannoma, and the rare benign perineuri- part, sporadic; bilateral vestibular schwanno-
noma, not discussed here. Nerve sheath tumors mas are pathognomonic of NF-2 (Chapter 12).
can occur as sporadic tumors or as part of the All familial and 60% of sporadic schwannomas
familial tumor syndromes neurofibromatosis-1 have a defect in the NF-2 tumor suppressor
(NF-1) and neurofibromatosis-2 (NF-2). When gene, but only patients with NF-2 have a
they occur sporadically, they are usually single germline mutation as well. The NF-2 gene is
and the vast majority are benign. When they inactivated either by a point mutation leading
occur as part of a familial syndrome, they are to a truncated protein, or by loss of the short
more likely to be multiple, and to undergo arm of chromosome 22 leading to complete
malignant degeneration (Chapter 12). Nerve loss of the protein. The NF-2 gene codes for a
sheath tumors can occur along any cranial or protein called merlin, a protein that regulates
peripheral nerve root or nerve except the olfac- cell proliferation and motility.2 Other common
tory and optic nerves, which are not truly nerves chromosomal abnormalities include a deletion
but extensions of the brain, and thus not suscep- at 1p.3 No p53 mutations have been encoun-
tible to the development of nerve sheath tumors. tered.4 Most acoustic neuromas express trans-
(Gliomas do arise in the optic nerve, particularly forming growth factor beta-1 (TGF-1) in the
as part of the NF-1 syndrome.) Nerve sheath cytoplasm of Schwann cells, tumor cells, blood
tumors are more common in the spinal canal vessel walls and the tumor capsule. This
and peripheral nerves than they are in the growth factor could promote tumor growth.5
intracranial cavity. However, when they arise in Other than NF-2, there are no established risk
the intracranial cavity, they are often responsi- factors for vestibular schwannomas or other
ble for significant symptoms. The major benign schwannomas of the peripheral or
intracranial nerve sheath tumor is the vestibular central nervous systems. In particular, head
schwannoma, also called acoustic neuroma or injury does not appear to be a risk factor.6
acoustic neurinoma (Fig. 9.2); it is a benign
tumor that arises within the vestibular portion
Vestibular schwannoma
of the VIIIth cranial nerve. Pure schwannomas
may also arise in other cranial nerves, particu- Vestibular schwannomas are thought to arise
larly the trigeminal nerve. Schwannomas near the zone of ObersteinerRedlich. This is

271
TUMORS OF CRANIAL NERVES AND SKULL BASE

the area where oligodendroglial myelin gives variants may occasionally be difficult to
way to Schwann cell myelin. The zone may distinguish from other tumors, especially the
occur near the exit of the vestibular nerve from melanotic schwannoma, and particularly when
the brain or further out laterally within the it occurs in the spinal canal.
internal auditory canal. Current evidence The tumors are slow growing and, even if
suggests that about 5060% of tumors arise untreated, some may be stable or even regress
from the superior vestibular nerve, 4050% over time.7 On average, vestibular schwanno-
from the inferior vestibular nerve, and less than mas are thought to increase at a rate of less
10% from the cochlear nerve. Acoustic neuro- than 2 mm per year, but growth can be slower
mas are classified by imaging into (1) small, or faster. Growth rate is usually slower in
less than 2-cm intracanalicular tumors, (2) older than in younger patients. Serial imaging
medium-sized, extending beyond the internal can sometimes predict growth rate. SPECT
auditory meatus but less than 3 cm, and (3) scanning using thallium chloride has been
large, greater than 3 cm. Macroscopically, the reported to be particularly useful in assessing
tumors are typically firm, well-circumscribed tumor growth. It was found to be superior to
and encapsulated. If they involve brain (i.e. MRI because it better quantifies tumor vascu-
cerebellum), they compress rather than invade larity, essential for tumor growth.8 Slow or
it. They are relatively avascular, although on absent growth over a period of 18 months to 3
occasion they are hemorrhagic. The tumors are years is said to make it unlikely that subsequent
usually solid, although small cysts sometimes enlargement will be significant. Such patients
occur. Microscopically, the tumors are can then be followed clinically and with scans
composed of what appear to be mature at infrequent intervals, i.e. every 2 years.
Schwann cells with relatively abundant, faintly Whether growth rate is controlled by
eosinophilic cytoplasm. Nuclear pleomorphism hormones is unclear. Many of these tumors
and mitotic figures are sometimes seen but do have estrogen and progesterone receptors and
not indicate malignancy. The tumors consist some are said to increase in size during
of so-called Antoni-A areas, represented by pregnancy. Human vestibular schwannomas
closely packed tumor cells, and Antoni-B areas, implanted into nude mice grew more rapidly
where cell density is much less. The Antoni-B when treated with estrogen, and this estrogen
areas dominate in vestibular schwannomas. effect was blocked by tamoxifen.9 Other
Immunohistochemically, the tumors express growth factors may increase the tumor size by
S100 protein and sometimes glial fibrillary an autocrine mechanism.
acidic protein (GFAP). MIB-1 labeling indices
range from 0.3 to 6.6, with a mean of 1.7. In Clinical findings
subtotally resected tumors there is a correlation The symptoms and signs of a schwannoma
between the labeling index and the rapidity depend on its location. The vestibular schwan-
of recurrence. Several variants have been noma (Fig. 9.2) usually presents with uni-
described, including the cellular schwannoma, lateral hearing loss, sometimes preceded or
the plexiform schwannoma and the melanotic accompanied by tinnitus, and a vague feeling
schwannoma. Although all three may occur in of dizziness and unsteadiness when walking
vestibular schwannoma, the cellular is the most (Table 9.2).
common, and the histologic variant does not In most patients, the symptoms of vestibular
affect treatment or prognosis. However, these schwannoma begin with hearing loss without

272
NERVE SHEATH TUMORS

Figure 9.2
A vestibular schwannoma. This middle-aged woman presented with progressive hearing loss and mild tinnitus.
She was an athlete and noticed that she was slightly more unsteady than she had been previously but not
sufficient to interfere with function. Her neurologic examination was normal, save for hearing loss; at surgery,
an acoustic neuroma was found (arrow). The photomicrograph shows the typical biphasic histological
appearance with Antoni B-type loosely textured tissue (upper half) and Antoni A-type dense tissue (lower half).

Table 9.2
Symptomatology Percentage of Average symptom Presenting symptoms in
patients duration (years) 491 patients with
vestibular schwannoma.10
Hearing loss
Gradual 79 4.5
Sudden 10 2.0
Tinnitus 51 4.0
Dysequilibrium 41 3.6
Facial numbness 19 4.1
Headache 15 4.0
Facial weakness 9 6.0
Trigeminal neuralgia 5 8.8

From Janetta,10 with permission.

273
TUMORS OF CRANIAL NERVES AND SKULL BASE

other symptoms. The history is one of gradu- Table 9.3


ally declining hearing over many years. The Preoperative cranial nerve and other neurologic
deficits in 190 patients with large vestibular
patient may completely fail to notice the schwannomas.
hearing loss until it is quite severe. If the
schwannoma is on the left, the patient may
recognize that he has unconsciously shifted the Cranial nerve Percentage of
abnormality patients
telephone from the accustomed left ear (to keep
the dominant hand free for writing) to the right Vth nerve
ear. Hearing loss usually begins in the high Corneal reflex 45.3
tones and often affects speech discrimination Facial numbness 31.6
disproportionately. Accordingly, a few patients VIIth nerve 4.7
IXth nerve 1.1
with even normal audiograms may complain of
Xth nerve 1.6
difficulty understanding what is said on the Cerebellar ataxia 12.1
telephone. In about 10% of patients, the Nystagmus 20.0
hearing loss begins suddenly and mimics the Raised intracranial pressure 2.1
more common condition of idiopathic sudden Neurofibromatosis type-2 14.2
hearing loss.10 Accordingly, all patients with the Other diseases 8.4
sudden onset of unilateral hearing loss require From Lanman et al,12 with permission.
evaluation for vestibular schwannoma.11
Because of the tumors slow growth, the
vestibular system is affected so slowly that
central compensation for the peripheral
vestibular loss often occurs without symptoms. Signs other than hearing loss and vestibular
In a minority of patients, the tumor may cause failure are associated with large tumors
vertigo at onset or a feeling of dysequilibrium, (> 3 cm) (Table 9.3). Ipsilateral decreased
as if being pushed to one side (away from the sensation in the face and particularly a
side of the lesion) when walking. Occasionally decreased corneal reflex are common signs of
hyperventilation can induce nystagmus. Despite a large tumor. Facial weakness, often subtle
the lack of symptoms, by the time the patient and manifested only by slowed or slightly
presents for medical care, ipsilateral vestibular decreased blinking on the involved side or
function, as tested by stimulating the semicir- failure of the ipsilateral platysma muscle to
cular canals, is usually absent. Other symptoms contract when the patient grimaces, occurs in
are extremely uncommon. About 1% of some patients. Cerebellar ataxia, nystagmus
patients present with hemifacial spasm. About and evidence of increased intracranial pressure
1020% complain of numbness in the face. occur as the tumors grow larger and compress
Facial weakness, particularly a delayed blink the cerebellum and the IVth ventricle.
response, may sometimes be apparent on The diagnosis is established by MRI. The
examination but is rarely noticeable by the tumors are characteristically isointense with
patient. Very large tumors have presented as nerve on the T1-weighted image but they
contralateral facial numbness, facial pain, or enlarge the involved nerve. They are isointense
facial spasm due to brainstem compression. with cerebrospinal fluid (CSF) on the T2-
Intratumoral hemorrhage or cyst formation weighted image and thus may not be visible.
may cause acute symptoms. They contrast enhance intensely, making even

274
NERVE SHEATH TUMORS

small tumors clearly visible on most MR scans. evoked responses (which yield a pattern of
Magnetic resonance cisternography using a retrocochlear loss with consistent abnormality
heavily T2-weighted two-dimensional fast of wave 5), stapedial reflex testing, electro-
spinecho technique allows one to see the inter- corticography and electroneuronography.
nal auditory meatus and identify the spatial Although all of these tests can be used to evalu-
relationships between the facial nerve and the ate hearing function, none is as sensitive or
superior vestibular nerve as well as the specific for the diagnosis as the MR scan. Of
relationship between blood vessels and cranial patients with vestibular schwannomas, 45%
nerves in the internal auditory canal and in the have NF-2. About 1020% of patients with
cerebellopontine cistern. This technique should NF-2 present with an apparently sporadic
be of considerable benefit to the surgeon.13,14 A unilateral vestibular schwannoma. All patients
recent report has suggested that screening by with unilateral vestibular schwannomas require
fast T2 magnetic resonance (MR) may be as evaluation for other stigmata of NF-2 (Chapter
efficacious and substantially cheaper than 12)15
gadolinium-enhanced MR scan.11 This is clini-
cally relevant, because although about 10% of Differential diagnosis
patients with vestibular schwannomas present The clinical differential diagnosis includes all
with sudden deafness, far less than 1% of causes of hearing loss, vestibular dysfunction
sudden deafness is caused by vestibular or unilateral ataxia (Fig. 9.3). All of these are
schwannomas. Thus, a large number of excluded by a high-resolution gadolinium-
patients must be screened, to identify the rare enhanced MR scan. Intracanalicular tumors
patient with a tumor. are virtually always vestibular schwannomas.
Several other laboratory tests are sometimes However, we recently encountered a patient
used in the diagnosis of vestibular schwan- with slowly progressive facial weakness and
noma, including audiometry (which is usually slight enhancement of the facial and acoustic
abnormal but non-specific), caloric testing (also nerve in the internal auditory canal. The tumor
abnormal but non-specific), brainstem auditory grew over 3 years and was believed to be an

Figure 9.3
This patient
presented with
progressive facial
weakness followed
by hearing loss 2
years later (see text).
Contrast enhancing
tumor on the left
involved the acoustic
canal. A second
tumor was found at
the jugular foramen
(arrow). Both turned
out to be metastatic
carcinomas.

275
TUMORS OF CRANIAL NERVES AND SKULL BASE

unusual vestibular schwannoma (Fig. 9.3). At Although microsurgical techniques can


surgery, it turned out to be a carcinoma. The frequently preserve hearing and facial nerve
primary tumor was not identified but was function, total extirpation of tumors, particu-
believed by histologic characteristics to arise larly large tumors, often requires sacrifice of the
from an occult parotid or skin tumor that acoustic nerve and may damage the facial and
tracked back along the facial nerve into the trigeminal nerves. Total removal of a vestibular
internal auditory canal (neurotropic cancer).16 schwannoma cures the patient. A variety of
Another patient with a similar clinical story surgical approaches have been recommended,
had a lymphoma. including a translabyrinthine approach,12 a
When the tumor is located more proximally middle cranial fossa approach,18 and a sub-
near the cerebellum, the differential diagnosis occipital retrosigmoid approach for hearing
includes other tumors that arise there, predom- preservation.19,20 The approach depends largely
inantly meningiomas and epidermoids (Chapter on the location of the tumor and the presence
12).17 The distinction cannot always be made or absence of preserved hearing. In expert
preoperatively between a cerebellopontine angle hands, the size of the tumor is irrelevant and
acoustic tumor or meningioma, but menin- even large tumors can be resected in most
giomas are usually larger and do not track along instances. The operating microscope and more
the cranial nerve toward the internal auditory recently the endoscope21 are useful tools.
meatus. They do not enlarge the internal Sometimes the capsule is adherent to cranial
auditory meatus on CT scan, as do vestibular nerves, making complete resection difficult. In
schwannomas. Symptoms of CPA meningiomas those instances, a partial resection may preserve
include ataxia and many present with hydro- facial nerve function and the tumor may cease
cephalus; only two-thirds have hearing loss.17 growing. Intraoperative monitoring of acoustic
and facial nerves helps to preserve those
Treatment functions.2225 Successful surgery does not
The treatment of vestibular schwannomas has improve hearing but, when hearing function is
two goals. The first is to prevent the tumor present, it can be preserved in approximately
from causing increased symptoms over time, 50% of patients with small tumors. Facial nerve
i.e. cure the tumor. Unchecked, a tumor in the function can be preserved in up to 100% of
internal auditory canal that presents with patients with small tumors but in less than 50%
hearing loss, with or without tinnitus, may of patients with tumors greater than 3 cm in
later cause facial paralysis and eventually, as it size. Some patients develop severe facial paral-
grows more medially, dysfunction of other ysis despite anatomic preservation of the nerve.
cranial nerves. A medially placed tumor will Most recover; if no recovery has occurred
also compress the cerebellum, causing ataxia within a year, hypoglossal-facial anastomosis
and eventually hydrocephalus. The larger the should be considered.26
tumor, the more difficult the treatment. Operative mortality is extremely low.
The second goal is to preserve remaining Operative morbidity, however, is common.
intact neurologic function. Often, hearing loss Headache, not present prior to surgery, occurs in
is not complete and the goal is to maintain about 50% of patients after vestibular schwan-
residual hearing function. In addition, preser- noma surgery and it can sometimes be quite
vation of facial nerve function is an important severe and persistent.27 Its cause is unknown.28
goal. These two goals are sometimes in conflict. The headache usually improves with time and

276
NERVE SHEATH TUMORS

responds to medical treatment that includes preferred approach for most patients. A dose of
valproic acid and verapamil29 but, if intractable, 1015 Gy is delivered to the tumor periphery
may be improved by a cranioplasty to repair the and a maximum of 1525 Gy to the center of
skull defect,30 even in the absence of a CSF leak. the tumor. The entire dose can be delivered
Headache usually does not occur following a either as a single fraction or a few fractions.
translabyrinthine approach. CSF leakage occurs Radiosurgery has been delivered both by
in approximately 12% of patients, although gamma knife and by linear accelerator,37 the
figures vary from 120%.31 Patients with CSF former giving a more restricted field. Local
leakage, which is usually rhinorrhea, should be control, i.e. no growth of tumor, has been
treated conservatively with a lumbar drain. If the reported in over 90% of patients. Useful
leak is from the wound, additional sutures are hearing has been preserved acutely in 75% of
placed. If the leakage does not repair itself by patients, a greater proportion than usual after
conservative treatment, re-exploration and surgery. Lower doses (14 Gy) improve hearing
occasionally CSF diversion may be necessary. preservation.36 Facial and trigeminal function are
Infectious meningitis is a very rare complication. also reported to be better preserved after radio-
It can occur either in the presence or absence of surgery. However, acute hearing loss, delayed
CSF leakage. Aseptic meningitis is more hearing loss, delayed facial paresis, either
common; this complication is described in detail transient or permanent, and delayed trigeminal
in Chapter 7. Decreased CSF pressure from the sensory loss occur in some patients within 23
surgery or from lumbar drainage can cause years after the procedure, particularly with
transient low tone hearing loss, believed to be higher doses of radiation.36 Fractionated stereo-
related to decreased perilymphatic pressure.32 tactic radiosurgery may be as effective as other
Patients generally recover spontaneously. treatments, with less cranial nerve dysfunction.38
Transverse sinus thrombosis leading to the This approach may be most appropriate for
syndrome of pseudotumor cerebri (increased patients with NF-2 and bilateral vestibular
venous and thus intracranial pressure resulting in schwannomas, where even partial hearing preser-
headache, papilledema and visual obscuration) vation is a critical issue.
followed translabyrinthine or suboccipital Undoubtedly, both surgery and radiosurgery
craniectomy in 5% of 107 patients.33 Two will continue to play a role in the management
serious but rare complications of surgery are of patients with vestibular schwannomas.
hematoma in the operative cavity and brainstem Many tumors are too large to be treated by
infarction. Infarction results from vasospasm of radiosurgery and demand a surgical approach.
small vessels supplying the brainstem that are Sometimes surgery fails or the tumor regrows
traumatized during the surgery. Less serious after surgery. Pathologic features including
but common operative complications include hyaline degeneration, cell density and the label-
tinnitus, not present preoperatively and rarely a ing index predict an increased likelihood of
significant problem,34 taste dysfunction (reduced regrowth after surgery. In those instances,
or distorted) and abnormal tearing (dry eye or radiosurgery may be effective.39 Conversely,
tearing when eating crocodile tears).35 radiosurgery sometimes fails, and those
In recent years, a number of physicians have patients should be considered for surgery.40
recommended radiosurgery for acoustic neuro- One note of caution is that tumors can
mas smaller than 3 cm in diameter and some temporarily expand after radiosurgery, and one
have suggested it is superior to surgery36 as the must be certain that the tumor is increasing in

277
TUMORS OF CRANIAL NERVES AND SKULL BASE

size over time before approaching it surgically. women than men. The schwannomas may
Chemotherapy has no role to play in the treat- arise from Schwann cells within the trigeminal
ment of these tumors. (gasserian) ganglion, the nerve root, or one of
Not all vestibular schwannomas grow. In the three divisions of the trigeminal nerve.
one study, only half of the tumors grew over a About half are within the middle cranial fossa,
3-year period.41 Thus, patients who refuse 30% are within the posterior fossa, and 20%
surgical or radiosurgical treatment, or who are dumbbell-shaped with extension into both
have a tumor in the sole hearing ear, or who cranial fossae.45 Rarely, the tumor arises from
are medically unable to undergo treatment, extradural portions of the trigeminal nerve,
can be followed. No radiologic test, except presenting in the maxillary sinus, orbit or
perhaps SPECT scans,8 predicts rate of growth. retropharyngeal space.46
Because the trigeminal nerve supplies somatic
sensation, pain is much more common with
Other schwannomas
schwannomas of the trigeminal than of the
Schwannomas can arise from other cranial vestibular nerve. Most patients present with
nerves. Ocular nerve schwannomas are sensory loss or paresthesias in the face, usually
rare.42,43 The most common involved nerve, involving all three divisions, although the
other than the vestibular, is the trigeminal (Fig. sensory loss is not necessarily complete in the
9.4), which represents only about 5% of all distribution of any of the divisions. In some
intracranial schwannomas and less than 1% of patients, paroxysms of facial pain resembling
primary intracranial tumors (Fig. 9.4).44 Like trigeminal neuralgia are the presenting
vestibular schwannomas, they occur during complaint.47 Usually, such patients can be found
middle life and are slightly more common in to have sensory abnormalities on examination,

Figure 9.4
A trigeminal neurinoma
(arrow). This 40-year-old man
had an MRI after a mild
concussion. He had no
neurologic symptoms or signs.
The tumor has not grown in 2
years.

278
NERVE SHEATH TUMORS

Table 9.4
Symptoms and signs of trigeminal schwannomas.

Symptom Percentage Sign Percentage

Trigeminal nerve dysfunction 55 Trigeminal nerve


Numbness 27 Decreased sensation 74
Pain 23 Diminished or absent corneal reflex 68
Paresthesias 5 Pain 38
Headache 15 Motor weakness 39
Diplopia 10 Other cranial nerve deficits
Hearing loss/tinnitus 8 II 10
Visual loss 5 III 14
Ear pain 3 IV 7
Othera 8 VI 35
VII 23
VIII 32
IX, X 8
XI 1

From Shrivastava et al,49 with permission.


Only 27 patients (21%) had abnormal findings limited to the trigeminal nerve.
aOther symptoms, subarachnoid hemorrhage, vertigo, seizure, exophthalmus, gait difficulty, hemifacial spasm. Two patients had more

than one initial symptom.

a finding not present with typical trigeminal many as three-quarters of patients have
neuralgia. Furthermore, the pain may persist findings referable to other cranial nerves (Table
between paroxysms and the pain usually lacks 9.4).
the trigger points characteristic of trigeminal The diagnosis is made by MR scan.48 The
neuralgia. Although sensory loss and pain tumor is usually hyperintense on the T2-
limited to one division of the trigeminal nerve weighted image and hypo or isointense on the
suggest that the lesion is within that division T1-weighted image with enhancement that is
rather than the ganglion or the root, there is typically uniform but may show areas of necro-
enough variability that the tumor cannot be sis, particularly in large tumors. The differen-
localized by clinical examination alone.48 As the tial diagnosis includes other mass lesions that
tumor grows, it can compress other cranial may arise along the distribution of the trigem-
nerves, causing diplopia, hearing loss, headache inal nerve. In the posterior fossa and Meckels
and ear pain (Table 9.4). cave, meningiomas, epidermoid tumors and
At the time of examination, most patients aneurysms may mimic trigeminal schwanno-
have sensory loss to a variable degree in all mas. In the cavernous sinus, meningiomas,
three sensory divisions of the trigeminal nerve, lymphomas, metastases and aneurysms must
and about 40% of patients have some be considered in the differential diagnosis.
weakness of the muscles of mastication. As Neurotropic metastatic tumors (Chapter 13)

279
TUMORS OF CRANIAL NERVES AND SKULL BASE

may mimic peripheral trigeminal schwanno- Table 9.5


mas, and other base of skull tumors may Signs and symptoms of facial nerve schwannomas.
present with trigeminal symptoms (see
below).48 Symptom Percentage
Schwannomas of the trigeminal nerve do not
differ in their pathologic findings from other Hearing loss 4191
Tinnitus 60
schwannomas. The tumors are benign, but they
Vertigo 34
may occasionally undergo malignant degenera- Facial weakness 4690
tion. Intratumoral hemorrhage can sometimes Sudden onset 20
be found microscopically or grossly.
From Shrivastava et al49 with permission.
The treatment is surgical.47 Total resection
results in cure, whereas partial resection may be
followed by tumor recurrence. The surgical
mortality is low, less than 2%, but morbidity
includes injury to other cranial nerves, CSF leak so close to the acoustic nerve through most of
with meningitis, and sometimes, as in vestibu- its course, it may be impossible to distinguish
lar schwannomas, hydrocephalus, probably a on scan whether the tumor has arisen in the
consequence of blood spilled into the sub- facial or the vestibular nerve. However, clinical
arachnoid space. Most patients are left with symptoms help the differentiation: vestibular
permanent trigeminal dysfunction. Although schwannomas almost always begin with
experience is limited, radiosurgery has been hearing loss, whereas facial schwannomas
reported to be safe and efficacious, and tumors usually begin with facial weakness. The differ-
may either cease growing or actually regress.50,51 ential diagnosis includes meningiomas in the
Facial nerve schwannomas represent less than posterior fossa, vestibular schwannomas and
2% of intracranial schwannomas. The patients other base of skull tumors.
present with facial weakness, tinnitus, hearing Facial schwannomas, like vestibular schwan-
loss and, sometimes, vertigo (Table 9.5). nomas, may not enlarge over time and thus can
Because the facial nerve acquires its Schwann be followed conservatively in some patients.52
cell sheath closer to the brainstem than does When appropriate, the treatment is surgical.
the vestibular nerve, tumors can arise closer to Complete resection leads to cure. With modern
the brainstem and cause brainstem compres- microsurgical techniques, mortality is low.
sion as their initial symptom. The facial nerve Facial paralysis is very common and hearing
also extends through the internal auditory loss, either transient or permanent, occurs in
canal along with the acoustic nerve, and these about one-third of patients. The complications
tumors can be mistaken for vestibular schwan- are similar to those of surgery for vestibular
nomas when they arise intracanalicularly. The schwannomas. Because of the rarity of these
tumor can also present as a visible mass in tumors, the role of radiosurgery is not yet
the middle ear, causing conductive rather than established. Some evidence suggests that radio-
sensorineural hearing loss. Otalgia and surgery is safe and efficacious.50
otorrhea may also be present. Schwannomas occasionally arise from the
The diagnosis is made by MR scan. The MR nerves exiting the jugular foramen.53 This
characteristics are the same as those of vestibu- includes the glossopharyngeal, vagus and
lar schwannomas. Because the facial nerve lies spinal accessory nerves. As with the facial

280
NERVE SHEATH TUMORS

nerve, these nerves acquire the Schwann cell Table 9.6


sheath close to the brainstem (spinal cord in Signs and symptoms of hypoglossal nerve
schwannomas.
the case of the spinal accessory nerve), so the
tumors may arise either within the posterior
fossa or within the jugular foramen extra- Major findings Percentage Less
cranially.53 Posterior fossa tumors often common
findings
present with hearing loss, facial numbness or
occasionally hemifacial spasm resulting from Headache 73 Tongue
compression of other cranial nerves. These hemiatrophy
tumors can grow to a large size and may cause IX, X, XI nerve 67 Vertigo
brainstem or cerebellar dysfunction as their dysfunction
initial symptom. Cranial nerve dysfunction, Limb weakness 66 Nystagmus
(spastic)
including hoarseness, dysphagia, weakness or
Ataxia Facial weakness
atrophy of the sternocleidomastoid and trapez- Dysarthria Hearing loss
ius muscles, is more likely to occur from
tumors within the jugular foramen than those From Shrivastava et al49 with permission.

nearer the brainstem. Unilateral tongue


weakness and atrophy may occur from
extracranial tumors compressing the hypo-
glossal nerve. Sometimes the jugular vein is
compressed. Pressure on the vein can result in
Neurofibromas
turbulent flow causing tinnitus,54 or, if the Neurofibromas are not found in the intracra-
compressed jugular vein is dominant, i.e. nial cavity. They involve spinal or peripheral
substantially larger than its partner, compres- nerves but not cranial nerves. They are of
sion may sufficiently interfere with blood flow interest when considering intracranial tumors
to cause increased intracranial pressure with because most are associated with NF-1
papilledema.55 The tumors are benign. (Chapter 12). NF-1, in turn, is associated with
Diagnosis by MR imaging and treatment are a variety of intracranial tumors including
similar to those for vestibular schwannomas. schwannomas.
The most serious postoperative complication is
dysphagia, which may require a feeding tube,
usually temporarily.56 Dysphagia can follow
Malignant nerve sheath tumors
either surgery or RT.56 Malignant nerve sheath tumors, also called
Hypoglossal schwannomas are fairly rare malignant schwannomas, are found primarily
(Table 9.6). Patients present with unilateral in peripheral nerves, including the brachial
tongue dysfunction which may cause dysarthria plexus and sciatic nerve. They may arise from
but may also be asymptomatic. As the mass a pre-existing neurofibroma or, much more
grows, it compresses brainstem, cerebellum or rarely, a schwannoma, or they may arise de
adjacent cranial nerves. Headache is common, novo. Rarely, they are found in the intra-
along with dysfunction of adjacent vagus, cranial cavity, involving cranial nerves.57
glossopharyngeal and spinal accessory nerves. The trigeminal nerve is most commonly
The diagnosis and treatment are similar to affected, but the acoustic nerve, the facial
those of other schwannomas. nerve, including the nervus intermedius, and

281
TUMORS OF CRANIAL NERVES AND SKULL BASE

occasionally other cranial nerves can be neuroblastoma), carcinomas or lymphomas


involved. The tumors are often sporadic, but of the paranasal sinuses, as well as para-
may be associated with NF-1, in which case gangliomas that often arise from the glomus
they may be multiple. The tumors cannot be jugulare (glomus jugulare tumor) and rhabdo-
identified as malignant prior to surgery. They myosarcomas.59 Chordomas characteristically
are treated with surgery followed by radiation arise either in the clivus, thus affecting cranial
but, unlike their more benign counterparts, nerves, or in the sacrum, causing a cauda
they usually recur and may be responsible for equina syndrome. They can, however, arise
the patients death. almost anywhere along the vertebral column,
although less commonly than the two places
cited above. Chondromas and chondrosarco-
mas, as well as tumors discussed elsewhere in
Skull base tumors this book, including meningiomas, schwanno-
mas and pituitary adenomas, can also affect the
skull base. Table 9.7 classifies these tumors by
Introduction
their relative incidence and common location.
Several tumors, both benign58 and malignant, Most skull base tumors are treated surgically
have a predilection to involve cranial nerves by if possible. All surgery of skull base tumors is
affecting the base of the skull (Fig. 9.5). These difficult. The location of these tumors makes
include esthesioneuroblastomas (olfactory them hard to reach, and involvement of cranial

Figure 9.5
Some of the tumors that
characteristically occur at the skull
base. (A) Clivus chordoma.
(B) Nasopharyngeal cancer.
(C) Paranasal sinus (frontal)
carcinoma.

282
SKULL BASE TUMORS

Table 9.7
Cranial base tumors: type and relative occurrence.

Tumors Percentage of Other data


intracranial tumors

Meningioma 20% 4050% in cranial base


Vestibular schwannoma 68% 6078% of cerebellopontine angle tumor
Trigeminal schwannoma 0.070.36% 0.88% of intracranial schwannomas
Jugular foramen schwannoma < 1% 2.9% of intracranial schwannoma
Facial schwannoma < 1% 0.8% of intrapetrous mass
1.9% of all intracranial schwannomas
Other cranial nerve schwannoma < 1% Oculomotor, trochlear, abducens,
hypoglossal: rare, case reports
Pituitary adenoma 715% 25% in autopsy of general population
Paraganglioma 0.01% 0.6% of all headneck tumors
Chordoma 0.151% 3% of malignant bone tumors
Chondrosarcoma < 1% 5% of cranial base tumors
Esthesioneuroblastoma 3% of all intranasal tumors
Nasopharyngeal carcinoma 3% of all headneck cancers
Adenoid cystic carcinoma 1% of all headneck cancers
6% of salivary neoplasms
Primary sarcoma (rhabdomyosarcoma, 4%
Ewings, fibrosarcoma, etc.)
Juvenile angiofibroma < 0.5% of all head/neck neoplasms

From Morita and Piepgras with permission.

nerves makes increased dysfunction of those vestibular schwannomas.61 The operating


nerves a major operative risk. When the tumors microscope has been a major advance in allow-
are in the cavernous sinus, they can also involve ing neurosurgeons to separate tumor tissue
the carotid artery, usually compressing rather from intertwined cranial nerves.
than invading it. CSF leak after surgery repre- RT is also difficult for tumors at the skull
sents another major risk because it is often hard base. Their intimate association with the
to repair the bone and dura at the skull base. cranial nerves and other intracranial structures
Nevertheless, there have been considerable sometimes prohibits the delivery of sterilizing
advances in skull base surgery that have doses to the tumor without side-effects. Often,
allowed neurosurgeons who specialize in that these tumors appear to respond better and
area to carry out substantial resection with more safely to radiosurgery or heavy particle
reduced but still significant morbidity and radiation. Chemotherapy plays an important
mortality. In some instances of carotid artery role in some tumors, but most of the tumors
involvement, a graft can be placed and the that arise at the base of the skull are not sensi-
tumor better resected. Endoscopy of the poste- tive to chemotherapy. However, the combina-
rior fossa sometimes allows tumor removal tion of surgery and RT has substantially
with minimal invasiveness, particularly for improved survival in recent years.

283
TUMORS OF CRANIAL NERVES AND SKULL BASE

Esthesioneuroblastoma (olfactory to fifth decades. However, small infants and


neuroblastoma) elderly patients have occasionally been
reported.
Esthesioneuroblastoma is a rare malignant Little is known of the genetic alterations
neuroectodermal tumor that is believed to because of its rarity, but one study indicated
originate from neurons of the olfactory multiple chromosomal changes with overrep-
epithelium high in the nasal cavity, usually resentation of chromosomes 4, 8, 11 and 14.
involving the cribriform plate (Fig. 9.6). The Partial DNA gains on chromosomes 1 and
tumor shows a slight male predominance and 17, deletions of the entire chromosomes 16,
has a peak age at presentation in the second 18, 19 and X, and partial losses of chromo-

Figure 9.6
An esthesioneuroblastoma involving the cribriform plate. The patient did not have neurologic symptoms, only
nasal obstruction and epistaxis. After surgery, he completely lost his sense of smell but was not particularly
bothered by this. The photomicrograph shows monomorphic nests of round cells in a fibrillary background
with occasional rosette formation (arrow).

284
SKULL BASE TUMORS

somes 5q and 17p have been reported. These the T2-weighted image. It uniformly contrast
are unusual chromosomal aberrations.62 enhances. CT scan provides the best informa-
Esthesioneuroblastomas do not have gene tion concerning bone destruction, particularly
translocations similar to those that occur in of the cribriform plate and the orbit.
Ewings sarcoma, suggesting that the tumor Radionuclide scanning with octreotide, a
is not a primitive neuroectodermal tumor.63 somatostatin analog, shows uptake similar to
Mutations in p53 have not been detected but that with other neural crest tumors, helping
there appears to be overexpression of wild- distinguish esthesioneuroblastoma from other
type p53 protein in subsets of the tumor that neoplasms that may arise in the paranasal
show aggressive behavior and a tendency for sinuses. The differential diagnosis includes all
recurrence.64 other tumors of the paranasal sinuses or nasal
Grossly, the tumor is highly vascular and cavity, e.g. carcinomas and lymphomas.
may contain intratumoral hemorrhage. Micro- Treatment includes radical surgical resection
scopically, the tumor appears as a small round and postoperative RT.65 Chemotherapy with
cell tumor, such as a poorly differentiated carci- cisplatin-based regimens improves survival, partic-
noma, melanoma or other neuroectodermal ularly in those with high-grade tumors.66 Some
tumor. Immunohistochemical stains are helpful investigators have advocated preoperative neo-
because all of the neuroendocrine markers are adjuvant therapy, including RT and chemotherapy
positive. These include neuron-specific enolase, prior to radical craniofacial resection. Such treat-
chromogranins, synaptophysin and the ment has led to 5- and 10-year overall survival
neuronal marker anti-Hu. Epithelial markers rates of 81% and 54.5% respectively.67
such as cytokeratin and epithelial membrane The prognosis is guarded. In most series,
antigen (EMA) are absent. The MIB-1 index local recurrence appears in 3070% of patients
can be as high as 50%. and regional lymph node or distant metastasis
The tumor usually presents with nasal rather is as high as 40%. Recurrence may appear after
than neurologic symptoms. Patients complain many years in remission. Unfavorable prog-
of either nasal obstruction, epistaxis or both. nostic factors include widespread or metastatic
Anosmia, either unilateral or bilateral, is disease, a proliferative index greater than
present but the patient is often unaware of the 10%, overexpression of wild-type p53, and
symptom. As the tumor grows, it infiltrates aneuploid or polyploid chromosomes.
adjacent structures, including the cribriform
plate and base of the skull, sometimes
Paraganglioma
compressing or invading the frontal lobes. It
also invades paranasal sinuses and the oronasal The paraganglioma is usually a benign tumor
pharynx. The tumor can metastasize to other that originates in autonomic ganglia anywhere
organs and can seed the leptomeninges, in in the body. The cells are of neural crest origin
which case neurologic symptoms are inevitable. and, pertinent to this book, can arise from
CT, MR and radionuclide imaging are all paraganglionic tissues at the base of the skull.68
helpful in the diagnosis. MRI gives the best The most common is the glomus jugulare
estimate of tumor spread into the surrounding tumor.69 The tumors can arise from the glomus
soft tissue areas and intracranial space. The tympanicum and are found in the middle ear
tumor is hypo- or hyperintense on the T1- and mastoid; they can also arise from chemo-
weighted image and usually hyperintense on receptor cells in the adventitia of the jugular

285
TUMORS OF CRANIAL NERVES AND SKULL BASE

vein or the glomus intravagale, a collection T2-weighted image; multiple flow voids and
of chemoreceptor cells on the auricular branch a heterogeneously contrast-enhancing mass
of the vagus nerve. are characteristic. An MR angiogram reveals
Paragangliomas are found largely in middle- the extreme vascularity of these tumors.
aged women, with a peak incidence in the 5th Scintigraphy using either iodine-123-labeled
decade. Bilateral glomus tumors occur in about metaiodobenzylguanidine or indium-111-
12% of patients. Familial occurrence is seen labeled pentetreotide suggests a neuroendocrine
in about 10% of patients. In both familial and tumor. In secreting tumors, the diagnosis can
sporadic tumors, deletions occur on 11q.70 In be suggested by a 24-h urinary assay for
one study, mutations of p53 and p16 were catechols. For such patients, catecholamine
noted at relapse but not in the initial tumor.71 secretion must be blocked prior to surgery,
Grossly, the tumors are red-brown, highly usually by administration of -adrenergic
vascular and reasonably well-circumscribed. blocking agents before and during surgery. The
Histologically, they resemble a normal differential diagnosis can include any tumor
paraganglion and consist of pale chief cells that involves the skull base, but the high vascu-
that are arranged in nests and lobules. larity and slow progression strongly suggest a
Sustentacular (supporting cells from Latin to glomus tumor.
hold upright) cells surround the lobules. Surgery and radiation are the modes of treat-
Neuroendocrine markers, such as neuron- ment. Total resection can sometimes be
specific enolase and chromogranin, identify the achieved and, if so, the patient is usually
chief cells. Sustentacular cells react with S-100 cured.72 In lesions not amenable to surgery,
protein and sometimes show immunoreactivity local control can be achieved in over 90% of
with GFAP. patients.73 For subtotal resection, RT in the
Tinnitus is the typical presenting complaint postoperative period delays recurrence.
of the glomus jugulare tumor. The tinnitus is Embolization prior to surgery will decrease the
usually pulsatile and can often be heard by an vascularity and sometimes make total excision
observer who places a stethoscope over the possible. Vocal cord paralysis, swallowing
mastoid or jugular vein. Other common findings difficulty, facial paralysis, hearing loss and CSF
include hearing loss and hoarseness due to leak are among the major complications of
involvement of the vagus nerve with vocal cord surgery.
dysfunction. Ear pain occurs in a significant The tumors are usually benign but recur-
number of patients. In some individuals, a bluish rence is common after partial resection, even
tumor can be seen behind the eardrum on with postoperative RT. A few tumors become
otoscopic inspection. The tumors are quite slow malignant and metastasize. Radiosurgery has
growing, and progression of symptoms is been reported to decrease tumor volume and
usually slow. About 1% of tumors may secrete vascularity in some patients.
catecholamines, causing palpitations, intermit-
tent hypertension and anxiety. The tumor
Chordoma
occasionally occludes the transverse sinus, but
rarely presents as pseudotumor cerebri. Chordomas are tumors of notochordal origin.
The diagnosis is suggested by MR scan. The They characteristically arise in the clivus (Fig.
lesions have variable intensity on the T1- 9.7) or sacral spinal cord. Although benign
weighted image but are hyperintense on the and slow growing, they tend to recur and

286
SKULL BASE TUMORS

Figure 9.7
A clival chordoma. This young woman presented in 1979 with progressive quadriparesis. A
clinical diagnosis of multiple sclerosis was made, but when CT scanning became available,
a mass was revealed that was believed to be intrinsic to the brainstem. At exploration,
nothing was found. Subsequently, as imaging improved, a mass was found to be
compressing the brain from the clivus and was partially resected. It turned out to be a
chordoma. She was treated with multiple operations, standard radiation therapy and finally
proton beam radiation. She lived for over 20 years but suffered multiple recurrences. The
photomicrograph shows nodules of vacuolated physaliphorous tumor cells (arrow).

sometimes late in their course may metastasize entity) demonstrated a rearrangement between
to bone or lungs. This tumor usually occurs in chromosomes 1 and 9, with variable losses of
middle age without any specific sex prepon- chromosome 1p. Other cytogenetic abnormali-
derance. ties have been identified in sporadic chordo-
Chordomas constitute less than 0.5% of mas; 1p appears to be involved frequently.
intracranial tumors. They are believed to arise The tumor has a characteristic appearance
from vestigial remnants of the interosseous consisting of lobules of pleomorphic, vacuo-
primitive notochord. Little is known about the lated, physaliphorous (from Greek for bubble
genetics of chordomas. One study demon- and bearing) cells scattered in a mucoid or
strated loss of heterozygosity in intron 17 of myxoid matrix. Some workers have divided
the retinoblastoma (RB) gene in two of seven chordomas into typical chordomas and
chordomas which may serve as a marker for chondroid chordomas, the latter with some
aggressive behavior.74 Another study of famil- evidence of cartilage formation. Chondroid
ial clivus chordomas (an exceedingly rare chordomas are believed to have a somewhat

287
TUMORS OF CRANIAL NERVES AND SKULL BASE

better prognosis. Histochemically, the tumors Table 9.8


react with cytokeratin, S-100 protein and Differential diagnosis of chordomas and
chondrosarcomas.
vimentin. The typical and chondroid tumors do
not differ in labeling index. The MIB-1 index
of recurrent tumors is considerably higher than Pituitary adenoma
that of primary tumors. p53 and cyclin D1 Craniopharyngioma
Meningioma
labeling, which indicate mutations in these
Schwannoma
genes, also correlate with recurrence and a high Nasopharyngeal carcinoma
MIB-1 labeling index.75 Salivary gland tumors
The most common presenting symptoms of Metastasis
clival chordomas are diplopia, headache and,
less commonly, visual disturbances and ataxia.
Because the tumors often arise from the base
of the clivus, bilateral VIth nerve palsies are a Women treated with radiation do less well than
highly suggestive sign. Other lower cranial men. Radiosurgery has been suggested for small
nerves can be affected, either singly or in lesions.77 Others have recommended charged
combination. Symptoms are often subtle at particle radiation.78 Ten-year control rates for
onset and progress insidiously, so that tumors skull base chordomas using mixed-beam
are usually large when the diagnosis is estab- protonphoton therapy are 65% for men and
lished. 42% for women.79 Reoperation at recurrence
Imaging usually establishes the diagnosis. sometimes helps but the tumors relentlessly
The MR image varies on the T1-weighted recur, eventually causing death.
scan but the tumor is hyperintense on the T2- As indicated above, the prognosis is poor.
weighted image and irregular enhancement Age is the most significant factor associated
is usually seen. Contrast material enhances with longer overall and disease-free survival,
chordomas more slowly than it does other with younger patients doing better than older
tumors at the skull base, often taking several patients. Diplopia is associated with longer
minutes to reach maximum enhancement.76 survival, probably because it is a symptom
Therefore, if the post-contrast images are which initiates early medical evaluation. The
obtained too quickly, the enhancement may be degree of resection and postoperative RT also
missed. On CT scan there is clear evidence of predict longer survival.
bone destruction and sometimes calcification
in the lesion. Rarely, the bone is not involved.
Chondrosarcomas
The location of the intracranial chordoma
is characteristic. Other tumors of bone, Both the more benign chondroma and the more
however, can occur in a similar position malignant chondrosarcoma can develop at the
(Table 9.8), and thus definitive diagnosis skull base. These are tumors of cartilage cells
requires biopsy. or their precursors. The chondrosarcomas are
The primary treatment is surgical. Patients divided into two categories: myxoid and
who undergo total or near-total resection mesenchymal. The classic chondrosarcoma is
survive substantially longer than patients with a composed of mature cartilage plus malignant
partial resection. Whatever the degree of resec- chondrocytes that are frequently multinucle-
tion, RT is required in the postoperative period. ated. The de-differentiated chondrosarcoma is

288
SKULL BASE TUMORS

Figure 9.8
An osteoblastoma
destroying the base of
the skull and causing
hearing loss. The tumor
presented with tinnitus
and hearing loss and
progressed very slowly.
The CT scan revealed
bone destruction
(arrow). Biopsy revealed
an osteoblastoma, an
unusual base of skull
tumor. The
photomicrograph shows
an osteoid trabecula
lined by uniform large
osteoblasts, and stromal
giant cells (arrow).

histologically indistinguishable from a malig- and hemangiomas can become malignant, and
nant fibrohistiocytoma. The mesenchymal Ollier and Maffucci syndromes have also been
chondrosarcoma is composed of undifferenti- associated with gliomas.
ated round or small cells which may be The clinical picture depends on where the
anti-Hu positive, suggesting neuronal differen- tumor arises. The middle and posterior fossae
tiation; they tend to occur in younger people are common sites. MR images show high inten-
and often originate from the sphenoid or sity on T2-weighted images, and enhancement
petrous bone. Chondrosarcomas may be is irregular. Chondrosarcomas differ from
subclassified histologically into grades IIII, chordomas in that they tend to be more later-
with the higher grades behaving in a more ally placed. The lateral placement makes them
aggressive fashion. Rarely, osteoblastomas also more accessible to surgery and probably leads
affect the skull base (Fig. 9.8). to a better prognosis than that of chordomas.
Chondrosarcomas (and even osteogenic The treatment, whatever the grade, is
sarcomas) are occasionally found within the surgery. The lowest-grade tumors are often
parenchyma of the brain or the dura.80 They cured by surgery alone. Partial resection or
are usually a result of metastasis from a distant complete resection of a high-grade tumor
tumor but, rarely, they are primary brain requires postoperative RT. Charged particle
tumors. Lower-grade chondrosarcomas are radiation has been recommended, as it has
sometimes associated with Olliers disease,81 a for chordomas, because it can deliver highly
rare syndrome characterized by multiple focused radiation which will spare surrounding
enchondromas; when Olliers disease is associ- critical structures. Protonphoton therapy
ated with multiple hemangiomas, it is called yields a 10-year control rate of 94%.79 Because
Maffucci syndrome.82 Both the enchondromas the tumor grows slowly, recurrences may be

289
TUMORS OF CRANIAL NERVES AND SKULL BASE

late. The 5-year recurrence-free survival rate Tumors of nasopharynx and


may be as high as 90% following surgery and
conventional RT. One study reports a 10-year paranasal sinuses
local control rate of 98% after 6480 Gy of A group of tumors arise in the nasopharynx or
conformal radiation therapy,83 but not without the paranasal sinuses and may involve the skull
hypothalamic/pituitary dysfunction.84 There is base, presenting with neurologic symptoms
no established chemotherapy for this tumor; and signs (Fig. 9.9). The tumors vary in their
one report describes a durable response to histology and include squamous cell cancers,
ifosfamidedoxorubicin chemotherapy.85 adenocarcinomas and nasopharyngeal cancers,

Figure 9.9
This 40-year-old woman developed left facial pain in 1993. She was treated for Lyme disease but the pain
persisted and she developed left-sided headaches and eventually some hearing loss on the left, which was
believed to be conductive. An MRI in 1995 (left) showed an enhancing tumor in the ethmoid sinus invading
the base of the brain and surrounding the carotid arteries (arrows). The tumor was resected and she received
postoperative radiation therapy. She was followed with serial scans and felt reasonably well until June 1998,
when she began feeling face pain on the right side; again, diagnosis was delayed but, by December, she noticed
that her right eye was bulging. There was no diplopia. A repeat scan at that time (right) showed recurrent
tumor invading the orbit, causing proptosis and lateral deviation of the right eye (arrow). A reoperation was
done. The tumor was a low-grade fibrosarcoma. It recurred several months later. The photomicrograph shows
interlacing fascicles of spindle cells with atypical nuclei and low mitotic index.

290
REFERENCES

including lymphoepithelial carcinoma and Paranasal sinus carcinomas are best treated
lymphoma. They also differ in their epidemiol- by surgery if possible.89 In some individuals,
ogy. Tobacco plays a role in the pathogenesis preoperative radiation and/or chemotherapy
of squamous cell cancers, as do some occupa- may shrink the tumor sufficiently to make
tional hazards such as nickel refining. Asbestos surgery feasible. If not administered prior to
inhalation and woodworking play a role in surgery, resection should be followed by RT
adenocarcinoma. Nasopharyngeal carcinoma is and chemotherapy. Nasopharyngeal carcino-
20 times more common in Asians than in mas are generally not surgically accessible. RT
North Americans, and has a particular and chemotherapy are the treatment modalities
predilection in those of southern Chinese of choice. Even when radiation is successful,
origin. Two major risk factors appear to be several months may elapse before biopsies
salted fish as a dietary staple and exposure to become negative.90 Some have suggested that
the EpsteinBarr virus (EBV). Interleukin-10, concurrent chemo-radiotherapy, followed by
whose sequence is homologous to an open adjuvant chemotherapy, may be more effec-
reading frame of EBV, is expressed in about tive.91
one-half of nasopharyngeal carcinomas and is The prognosis depends upon the extent and
negatively associated with survival.86 Genetic degree of malignancy of the tumor. In nasopha-
changes have been described and depend on the ryngeal carcinoma, disease-free survival can be
specific tumor.87,88 as high as 65% with aggressive treatment.92 It
Most patients present with local symptoms, is substantially lower, about 35%, in paranasal
but involvement of cranial nerves can be the sinus tumors. RT of nasal and paranasal tumors
first symptom. Local symptoms depend on the may lead to delayed retinopathy.93 Conformal
tumor site. Paranasal sinus and nasal tumors radiation therapy reduces the dose to optic
cause nasal obstruction and epistaxis, as can nerves and chiasm, thus diminishing the risk of
nasopharyngeal carcinomas. If the tumor late complications.94 Temporal lobe necrosis is
invades the orbit, proptosis can be an early a late complication of treatment of nasopha-
symptom, usually associated with diplopia but ryngeal carcinoma. MRS may show temporal
not visual loss. These tumors affect the anterior lobe abnormalities before the MRI does.95
base of the skull and, in addition to headache,
can cause other neurologic symptoms such as
anosmia, visual disturbances from compression
of the optic nerves or diplopia from compres-
References
sion of ocular motor nerves in or near the 1. Wiegand DA, Ojemann RG, Fickel V. Surgical
cavernous sinus. Facial pain and sensory distur- treatment of acoustic neuroma (vestibular
schwannoma) in the United States: report from
bance may also occur. Hearing loss with the Acoustic Neuroma Registry. Laryngoscope
nasopharyngeal carcinoma is usually unilateral 1996; 106: 5866.
and can be conductive, due to occlusion of the 2. Reed N, Gutmann DH. Tumorigenesis in
eustachian tubes by the tumor. neurofibromatosis: new insights and poten-
The diagnosis is suggested by MR scanning. tial therapies. Trends Mol Med 2001; 7:
15762.
In those patients who present with cranial
3. Leone PE, Bello MJ, Mendiola M et al. Allelic
nerve palsies, a CT scan will often better status of 1p, 14q, and 22q and NF2 gene
identify bone destruction of the skull base. The mutations in sporadic schwannomas. Int J Mol
diagnosis can only be established by biopsy. Med 1998; 1: 88992.

291
TUMORS OF CRANIAL NERVES AND SKULL BASE

4. Monoh K, Ishikawa K, Yasui N et al. p53 unilateral vestibular schwannoma. J Neurol


tumor suppressor gene in acoustic neuromas. Neurosurg Psychiatry 1999; 66: 7647.
Acta Otolaryngol Suppl (Stockh) 1998; 537: 16. McCord MW, Mendenhall WM, Parsons JT,
115. Flowers FP. Skin cancer of the head and
5. Cardillo MR, Filipo R, Monini S et al. neck with incidental microscopic perineural
Transforming growth factor-beta1 expression invasion. Int J Radiat Oncol Biol Phys 1999;
in human acoustic neuroma. Am J Otol 1999; 43: 5915.
20: 658. 17. Mallucci CL, Ward V, Carney AS,
6. Inskip PD, Mellemkjaer L, Gridley G, Olsen ODonoghue GM, Robertson I. Clinical
JH. Incidence of intracranial tumors following features and outcomes in patients with non-
hospitalization for head injuries (Denmark). acoustic cerebellopontine angle tumours. J
Cancer Causes Control 1998; 9: 10916. Neurol Neurosurg Psychiatry 1999; 66:
7. Charabi S, Tos M, Thomsen J, Charabi B, 76871.
Mantoni M. Vestibular schwannoma growth: 18. Gjuric M, Wigand ME, Wolf SR. Enlarged
the continuing controversy. Laryngoscope middle fossa vestibular schwannoma surgery:
2000; 110: 17205. experience with 735 cases. Otol Neurotol
8. Charabi S, Thomsen J, Tos M et al. Acoustic 2001; 22: 22330.
neuroma vestibular schwannoma growth: past, 19. Magliulo G, Sepe C, Varacalli S, Fusconi M.
present and future. Acta Otolaryngol (Stockh) Cerebrospinal fluid leak management follow-
1998; 118: 32732. ing cerebellopontine angle surgery. J
9. Stidham KR, Roberson JBJ. Effects of estrogen Otolaryngol 1998; 27: 25862.
and tamoxifen on growth of human vestibular 20. Irving RM, Jackler RK, Pitts LH. Hearing
schwannomas in the nude mouse. Otolaryngol preservation in patients undergoing vestibular
Head Neck Surg 1999; 120: 2624. schwannoma surgery: comparison of middle
10. Jannetta PJ. Vestibular neurilemmomas. Clin fossa and retrosigmoid approaches. J
Neurosurg 1997; 44: 52948. Neurosurg 1998; 88: 8405.
11. Daniels RL, Shelton C, Harnsberger HR. Ultra 21. Gksu N, Bayazit Y, Kemaloglu Y. Endoscopy
high resolution nonenhanced fast spin echo of the posterior fossa and dissection of acous-
magnetic resonance imaging: cost-effective tic neuroma. J Neurosurg 1999; 91: 77680.
screening for acoustic neuroma in patients with 22. Mullatti N, Coakham HB, Maw AR, Butler
sudden sensorineural hearing loss. Otolaryngol SR, Morgan MH. Intraoperative monitoring
Head Neck Surg 1998; 119: 3649. during surgery for acoustic neuroma: benefits
12. Lanman TH, Brackmann DE, Hitselberger WE, of an extratympanic intrameatal electrode. J
Subin B. Report of 190 consecutive cases of Neurol Neurosurg Psychiatry 1999; 66: 5919.
large acoustic tumors (vestibular schwannoma) 23. Roberson JB Jr, Jackson LE, McAuley JR.
removed via the translabyrinthine approach. Acoustic neuroma surgery: absent auditory
J Neurosurg 1999; 90: 61723. brainstem response does not contraindicate
13. Kumon Y, Sakaki S, Ohue S et al. Usefulness attempted hearing preservation. Laryngoscope
of heavily T2-weighted magnetic resonance 1999; 109: 90410.
imaging in patients with cerebellopontine angle 24. Axon PR, Ramsden RT. Intraoperative electro-
tumors. Neurosurgery 1998; 43: 133843. myography for predicting facial function in
14. Ryu H, Tanaka T, Yamamoto S et al. vestibular schwannoma surgery. Laryngoscope
Magnetic resonance cisternography used to 1999; 109: 9226.
determine precise topography of the facial 25. Colletti V, Fiorino FG. Advances in monitor-
nerve and three components of the eighth ing of seventh and eighth cranial nerve
cranial nerve in the internal auditory canal and function during posterior fossa surgery. Am J
cerebellopontine cistern. J Neurosurg 1999; 90: Otol 1998; 19: 50312.
62434. 26. Kunihiro T, Kanzaki J, Shiobara R, Inoue Y,
15. Evans DGR, Lye R, Neary W et al. Probability Kurashima K. Long-term prognosis of profound
of bilateral disease in people presenting with a facial nerve paralysis secondary to acoustic

292
REFERENCES

neuroma resection. ORL J Otorhinolaryngol 39. Pollock BE, Lunsford LD, Flickinger JC, Clyde
Relat Spec 1999; 61: 98102. BL, Kondziolka D. Vestibular schwannoma
27. Levo H, Pyykk I, Blomstedt G. Postoperative management. Part I. Failed microsurgery and
headache after surgery for vestibular schwan- the role of delayed stereotactic radiosurgery.
noma. Ann Otol Rhinol Laryngol 2000; 109: J Neurosurg 1998; 89: 9448.
8538. 40. Pollock BE, Lunsford LD, Kondziolka D et al.
28. Levo H, Blomstedt G, Pyykk I. Vestibular Vestibular schwannoma management. Part II.
schwannoma surgery and headache. Acta Failed radiosurgery and the role of delayed
Otolaryngol (Stockh) 2000; 235. microsurgery. J Neurosurg 1998; 89: 94955.
29. Hanson MB, Glasscock ME III, Brandes JL, 41. Selesnick SH, Johnson G. Radiologic surveil-
Jackson G. Medical treatment of headache lance of acoustic neuromas. Am J Otol 1998;
after suboccipital acoustic tumor removal. 19: 8469.
Laryngoscope 1998; 108: 11114. 42. Asaoka K, Sawamura Y, Murai H, Satoh M.
30. Wazen JJ, Sisti M, Lam SM. Cranioplasty in Schwannoma of the oculomotor nerve: a case
acoustic neuroma surgery. Laryngoscope 2000; report with consideration of the surgical treat-
110: 12947. ment. Neurosurgery 1999; 45: 6303.
31. Brennan JW, Rowed DW, Nedzelski JM, Chen 43. Kawasaki A. Oculomotor nerve schwannoma
JM. Cerebrospinal fluid leak after acoustic associated with ophthalmoplegic migraine. Am
neuroma surgery: influence of tumor size and J Ophthalmol 1999; 128: 65860.
surgical approach on incidence and response to 44. Friedman RA, Pensak ML, Osterhaus D, Tew
treatment. J Neurosurg 2001; 94: 21723. JMJ, van Loveren HR. Trigeminal schwanno-
32. Stoeckli SJ, Bohmer A. Persistent bilateral mas: the role of the neurotologist in multi-
hearing loss after shunt placement for hydro- disciplinary management. Otolaryngol Head
cephalus. Case report. J Neurosurg 1999; 90: Neck Surg 1999; 120: 35560.
7735. 45. Yoshida K, Kawase T. Trigeminal neurinomas
33. Keiper GL Jr, Sherman JD, Tomsick TA, Tew extending into multiple fossae: surgical
JM, Jr. Dural sinus thrombosis and pseudo- methods and review of the literature. J
tumor cerebri: unexpected complications of Neurosurg 1999; 91: 20211.
suboccipital craniotomy and translabyrinthine 46. Krishnamurthy S, Holmes B, Powers SK.
craniectomy. J Neurosurg 1999; 91: 1927. Schwannomas limited to the infratemporal
34. Levo H, Blomstedt G, Pyykko I. Tinnitus fossa: report of two cases. J Neurooncol 1998;
and vestibular schwannoma surgery. Acta 36: 26977.
Otolaryngol Suppl 2000; 543: 289. 47. Samii M, Migliori MM, Tatagiba M, Babu R.
35. Irving RM, Viani L, Hardy DG, Baguley DM, Surgical treatment of trigeminal schwannomas.
Moffat DA. Nervus intermedius function after J Neurosurg 1995; 82: 7118.
vestibular schwannoma removal: clinical 48. Nemzek WR. The trigeminal nerve. Top Magn
features and pathophysiological mechanisms. Reson Imaging 1996; 8: 13254.
Laryngoscope 1995; 105: 80913. 49. Shrivastava RK, Strauss R, Post KD. Other
36. Flickinger JC, Kondziolka D, Niranjan A, schwannomas of cranial nerves. In: Kaye AH,
Lunsford LD. Results of acoustic neuroma Laws Jr ER eds. Brain tumors: An encyclopedic
radiosurgery: an analysis of 5 years experience approach. Edinburgh: Churchill Livingstone,
using current methods. J Neurosurg 2001; 94: 2001: 687709.
16. 50. Mabanta SR, Buatti JM, Friedman WA, Meeks
37. Spiegelmann R, Lidar Z, Gofman J et al. SL, Mendenhall WM, Bova FJ. Linear accelera-
Linear accelerator radiosurgery for vestibular tor radiosurgery for nonacoustic schwannomas.
schwannoma. J Neurosurg 2001; 94: 713. Int J Radiat Oncol Biol Phys 1999; 43: 5458.
38. Fuss M, Debus J, Lohr F et al. Conventionally 51. Huang CF, Kondziolka D, Flickinger JC,
fractionated stereotactic radiotherapy (FSRT) Lunsford LD. Stereotactic radiosurgery for
for acoustic neuromas. Int J Radiat Oncol Biol trigeminal schwannomas. Neurosurgery 1999;
Phys 2000; 48: 13817. 45: 1116.

293
REFERENCES

52. Van Den Abbeele T, Viala P, Francois M, 65. Eich HT, Staar S, Micke O et al. Radiotherapy
Narcy P. Facial neuromas in children: delayed of esthesioneuroblastoma. Int J Radiat Oncol
or immediate surgery? Am J Otol 1999; 20: Biol Phys 2001; 49: 15560.
2536. 66. McElroy EA Jr, Buckner JC, Lewis JE. Chemo-
53. Kanemoto Y, Ochiai C, Yoshimoto Y, Nagai therapy for advanced esthesioneuroblastoma:
M. Primary extracranial jugular foramen neuri- The Mayo Clinic experience. Neurosurgery
noma manifesting with marked hemiatrophy of 1998; 42: 10237.
the tongue: case report. Surg Neurol 1998; 49: 67. Polin RS, Sheehan JP, Chenelle AG et al. The
5347. role of preoperative adjuvant treatment in
54. Meador KJ, Swift TR. Tinnitus from intra- the management of esthesioneuroblastoma:
cranial hypertension. Neurology 1984; 34: The University of Virginia experience.
125861. Neurosurgery 1998; 42: 102937.
55. Graus F, Slatkin NE. Papilledema in the 68. Reithmeier T, Gumprecht H, Stolzle A,
metastatic jugular foramen syndrome. Arch Lumenta CB. Intracerebral paraganglioma.
Neurol 1983; 40: 81618. Acta Neurochir (Wien) 2000; 142: 10636.
56. Pri S, Coiffier L, Laccourreye L et al. 69. Robertson JH, Gardner G, Cocke EWJ.
Swallowing disorders in paralysis of the lower Glomus jugulare tumors. Clin Neurosurg
cranial nerves: a functional analysis. Ann Otol 1994; 41: 3961.
Rhinol Laryngol 1999; 108: 60611. 70. Bikhazi PH, Messina L, Mhatre AN, Goldstein
57. Akimoto J, Ito H, Kudo M. Primary intra- JA, Lalwani AK. Molecular pathogenesis in
cranial malignant schwannoma of trigeminal sporadic head and neck paraganglioma.
nerve. A case report with review of the litera- Laryngoscope 2000; 110: 13468.
ture. Acta Neurochir (Wien) 2000; 142: 5915. 71. Gran S, Tali T. p53 and p16INK4A mutations
58. Eisenberg MB, Al-Mefty O, DeMonte F, during the progression of glomus tumor.
Burson GT. Benign nonmeningeal tumors of Pathol Oncol Res 1999; 5: 415.
the cavernous sinus. Neurosurgery 1999; 44: 72. Sen C, Hague K, Kacchara R et al. Jugular
94954. foramen: Microscopic anatomic features and
59. Paulino AC. Role of radiation therapy in implications for neural preservation with refer-
parameningeal rhabdomyosarcoma. Cancer ence to glomus tumors involving the temporal
Invest 1999; 17: 22330. bone. Neurosurgery 2001; 48: 83847.
60. Morita A, Piepgras DG. Tumors of the skull 73. Springate SC, Weichselbaum RR. Radiation or
base. In: Vecht CJ, ed. Handbook of clinical surgery for chemodectoma of the temporal
neurology, Vol. 68: Neuro-oncology Part 2. bone: a review of local control and complica-
Amsterdam: Elsevier Science, 1997: 46596. tions. Head Neck 1990; 12: 3037.
61. Jennings CR, ODonoghue GM. Posterior fossa 74. Eisenberg MB, Woloschak M, Sen C, Wolfe D.
endoscopy. J Laryngol Otol 1998; 112: 2279. Loss of heterozygosity in the retinoblastoma
62. Szymas J, Wolf G, Kowalczyk D, Nowak S, tumor suppressor gene in skull base chordomas
Petersen I. Olfactory neuroblastoma: detection and chondrosarcomas. Surg Neurol 1997; 47:
of genomic imbalances by comparative 15660.
genomic hybridization. Acta Neurochir (Wien) 75. Matsuno A, Sasaki T, Nagasima T et al.
1997; 139: 83944. Immunohistochemical examination of prolifer-
63. Mezzelani A, Tornielli S, Minoletti F et al. ative potentials and the expression of cell cycle-
Esthesioneuroblastoma is not a member of the related proteins of intracranial chordomas.
primitive peripheral neuroectodermal tumour Hum Pathol 1997; 28: 7149.
Ewings group. Br J Cancer 1999; 81: 58691. 76. Ikushima I, Korogi Y, Hirai T et al.
64. Papadaki H, Kounelis S, Kapadia SB et al. Chordomas of the skull base: dynamic MRI. J
Relationship of p53 gene alterations with Comput Assist Tomogr 1996; 20: 54750.
tumor progression and recurrence in olfactory 77. Muthukumar H, Kondziolka D, Lunsford LD,
neuroblastoma. Am J Surg Pathol 1996; 20: Flickinger JC. Stereotactic radiosurgery for
71521. chordoma and chondrosarcoma: further

294
LAST A HEAD

experiences. Int J Radiat Oncol Biol Phys patients with nasopharyngeal carcinoma.
1998; 41: 38792. Cancer 1999; 85: 143945.
78. Hug EB, Loredo LN, Slater JD et al. Proton 87. Hui AB, Lo KW, Leung SF et al. Detection of
radiation therapy for chordomas and recurrent chromosomal gains and losses in
chondrosarcomas of the skull base. J primary nasopharyngeal carcinoma by compar-
Neurosurg 1999; 91: 4329. ative genomic hybridisation. Int J Cancer 1999;
79. Munzenrider JE, Liebsch NJ. Proton therapy 82: 498503.
for tumors of the skull base. Strahlenther 88. Caruana SM, Zwiebel N, Cocker R et al. p53
Onkol 1999; 175 Suppl 2: 5763. alteration and human papilloma virus infection
80. Oruckaptan HH, Berker M, Soylemezoglu F, in paranasal sinus cancer. Cancer 1997; 79:
Ozcan OE. Parafalcine chondrosarcoma: an 13208.
unusual localization for a classical variant. 89. Le QT, Fu KK, Kaplan M et al. Treatment of
Case report and review of the literature. Surg maxillary sinus carcinoma a comparison of
Neurol 2001; 55: 1749. the 1997 and 1977 American Joint Committee
81. Hofman S, Heeg M, Klein JP, Krikke AP. on Cancer staging systems. Cancer 1999; 86:
Simultaneous occurrence of a supra- and an 170011.
infratentorial glioma in a patient with Olliers 90. Kwong DLW, Nicholls J, Wei WI et al. The
disease: more evidence for non-mesodermal time course of histologic remission after treat-
tumor predisposition in multiple enchondro- ment of patients with nasopharyngeal carci-
matosis. Skeletal Radiol 1998; 27: 68891. noma. Cancer 1999; 85: 144653.
82. Ramina R, Coelho NM, Meneses MS, Pedrozo 91. Tan EH, Chua ET, Wee J et al. Concurrent
AA. Maffuccis syndrome associated with a chemoradiotherapy followed by adjuvant
cranial base chondrosarcoma: case report and chemotherapy in Asian patients with nasopha-
literature review. Neurosurgery 1997; 41: ryngeal carcinoma: toxicities and preliminary
26972. results. Int J Radiat Oncol Biol Phys 1999; 45:
83. Rosenberg AE, Nielsen GP, Keel SB et al. 597601.
Chondrosarcoma of the base of the skull: a 92. Lin JC, Jan JS. Locally advanced nasopharyn-
clinicopathologic study of 200 cases with geal cancer: long-term outcomes of radiation
emphasis on its distinction from chrodoma. therapy. Radiology 1999; 211: 5138.
Am J Surg Pathol 1999; 23: 13708. 93. Takeda A, Shigematsu N, Suzuki S et al. Late
84. Pai HH, Thornton A, Katznelson L et al. retinal complication of radiation therapy for
Hypothalamic/pituitary function following nasal and paranasal malignancies: relationship
high-dose conformal radiotherapy to the base between irradiated-dose area and severity. Int
of skull: demonstration of a dose-effect J Radiat Oncol Biol Phys 1999; 44: 599605.
relationship using dose-volume histogram 94. Brizel DM, Light K, Zhou SM, Marks LB.
analysis. Int J Radiat Oncol Biol Phys 2001; Conformal radiation therapy treatment
49: 107992. planning reduces the dose to the optic struc-
85. La Rocca RV, Morgan KW, Paris K, Baeker tures for patients with tumors of the paranasal
TR. Recurrent chondrosarcoma of the cranial sinuses. Radiother Oncol 1999; 51: 2158.
base: a durable response to ifosfamide-doxoru- 95. Chong VFH, Rumpel H, Aw YS et al.
bicin chemotherapy. J Neurooncol 1000; 41: Temporal lobe necrosis following radiation
2813. therapy for nasopharyngeal carcinoma: 1H MR
86. Fijieda S, Lee K, Sunaga H et al. Staining of spectroscopic findings. Int J Radiat Oncol Biol
interleukin-10 predicts clinical outcome in Phys 1999; 45: 699705.

295
10
Pituitary and sellar region tumors

Introduction trigeminal nerve. In about 20% of individuals,


the diaphragma sella is incompetent, potentially
The pituitary (Latin for phlegm) or hypophysis allowing cerebrospinal fluid (CSF) from the
(Greek for undergrowth) sits in a bony inden- suprasellar cistern to enter the sella turcica.
tation of the skull base called the sella turcica. Lateral to the pituitary gland are the paired
Its superior surface is covered by dura called the cavernous sinuses, which contain the carotid
diaphragma sella, through which passes the artery, the nerves to the extraocular muscles
pituitary stalk connecting the pituitary gland (oculomotorcranial nerve III; trochlearcranial
with the hypothalamus (Fig. 10.1). The nerve IV; and abducenscranial nerve VI) and
diaphragm sella is pain-sensitive; its nerve the first division of the trigeminal nervecranial
supply is from the ophthalmic division of the nerve V. Below the floor of the sella is the

C
A
B C
C

D A

C B

Figure 10.1
The pituitary gland and its relationships to
surrounding structures. Left horizontal plane: (A)
adenohypophysis, (B) neurohypophysis, (C) cavernous
sinus with carotid artery. Right coronal plane: (A)
optic chiasm, (B) cavernous sinus, (C) sphenoid sinus,
(D) hypothalamus.

296
ANTERIOR PITUITARY TUMORS

sphenoid sinus. Above the diaphragma sella and region, but non-pituitary, tumors are also
within the suprasellar cistern is the optic chiasm, discussed in this chapter (Table 10.1).
which contains crossing fibers of the optic nerve
from each of the retinas. Above the optic chiasm
is the hypothalamus, that secretes the releasing
factors that regulate pituitary function and are Anterior pituitary tumors
transmitted to the pituitary gland via the stalk. Introduction
The pituitary gland is actually two struc-
tures: an anterior portion (adenohypophysis), a Pituitary tumors are the third most common
true endocrine gland, and a posterior portion primary intracranial neoplasms, exceeded only
(neurohypophysis), an extension of the by gliomas and meningiomas; they account for
hypothalamus, i.e. neural tissue; both secrete 1015% of all primary intracranial tumors
hormones. The anterior pituitary secretes encountered in clinical practice. Their incidence
growth hormone (GH), prolactin (PRL), ranges from 114/100 000 annually in various
corticotropin (ACTH), thyroid-stimulating clinical series, and their prevalence at autopsy
hormone (TSH) and the gonadotropins, varies from 5% to 25%. In one autopsy series,
luteinizing hormone (LH), and follicle-stimu- incidental pituitary adenomas larger than
lating hormone (FSH). The posterior pituitary 2 mm, and thus identifiable on MR scan, were
secretes arginine vasopressin, an antidiuretic found in 1.7% of the population.4 An
hormone (ADH), and oxytocin, a regulator of additional 4.4% of pituitary glands had other
smooth muscle contraction important for lacta- lesions greater than 2 mm (i.e. focal hyperpla-
tion and uterine contraction. Tumors of the sia, Rathke cysts). Another series reported a
anterior pituitary are common and almost 9% incidence of lesions of unspecified size.5
uniformly benign.1,2 Tumors of the posterior The most recent study reported adenomas in
pituitary are rare3 and, with few exceptions 24% of 100 glands, 10 of which were
(e.g. granular cell tumors), do not differ from lactotrophs and 12 were nonfunctioning.
those that arise elsewhere in the brain. Tumors Granular cell tumors were found in 9%.6 Most
not of pituitary origin can affect either the of the tumors were < 3 mm. Thus, all pituitary
gland itself (e.g. metastases) or areas in lesions identifiable on MR scan are not neces-
proximity to the sella (e.g. in the suprasellar sarily symptomatic. Clinically, pituitary tumors
cistern or the optic chiasm). These sellar are more common in women than in men, but
in autopsy series they appear to be equally
common in both sexes.
Table 10.1 Pituitary tumors can be classified in several
Classification of pituitary and sellar region tumors
ways:

I Pituitary adenomas and carcinomas 1. By size. Microadenomas are tumors less


II Posterior pituitary tumors than 1 cm in diameter and macroadenomas
III Metastases to pituitary gland and stalk
are those larger than 1 cm. Microadenomas
IV Germ cell tumors (Chapter 8)
V Craniopharyngiomas cause symptoms by excessive secretion of
VI Optic nerve and chiasm tumors (Chapter 5) hormones not under normal feedback
VII Rathke cleft cysts control from the hypothalamus.
Macroadenomas cause their symptoms

297
PITUITARY AND SELLAR REGION TUMORS

either by excessive hormone secretion or by microscopy are very useful because they
compressing normal glandular or neural can define the hormones secreted by the
structures. tumor and thus allow for a functional
2. By endocrine function. Pituitary adenomas classification.7
are divided into those that secrete
hormones and those that are chemically
Pituitary tumors arise clonally from a single
inactive. Chemically inactive adenomas can
cell-type within the pituitary gland (Table
cause symptoms only when they grow large
10.2). Some pituitary adenomas secrete the
enough to compress other structures, i.e.
hormone(s) from their cell of origin, whereas
become macroadenomas.
others lose the capacity to secrete hormones.
3. By clinical findings.1
Virtually all pituitary adenomas escape
4. By histology.7 Almost all pituitary adeno-
feedback control and thus, if they secrete
mas are benign. They are identified micro-
hormones, they do so excessively, leading to
scopically by uniformity of cells with
endocrine abnormalities.
disruption of the normal acinar patterns.
The tumors are usually well demarcated by
a pseudocapsule consisting of reticulin and
Etiology
compressed normal gland. Mitoses and
proliferative indices are usually low, but Pituitary adenomas are monoclonal tumors
invasiveness is common. Dural invasion, whose only established genetic defect occurs in
cellular pleomorphism, increased cellular- patients with multiple endocrine neoplasia
ity, necrosis or aneuploidy do not neces- syndrome type I (MEN I).2,10 This autosomal
sarily imply aggressive biological activity.9 dominant condition is characterized by
Mitotic activity and p53 immunoreactivity spontaneous development of tumors of the
may imply decreased likelihood of cure. anterior pituitary, pancreatic islets and
Immunohistochemistry and electron parathyroid glands. Pituitary adenomas,

Table 10.2
Classification of pituitary adenomas.

Cell type Clinical Hormone secreted Endocrine syndrome


prevalencea
(%)

Lactotroph 27 Prolactin Galactorrhea, amenorrhea (women)


Impotence (men)
Somatotroph 21 Growth hormone prolactin Acromegaly
Corticotroph 8 ACTH Cushings syndrome, Nelsons syndrome
Thyrotroph 1 Thyroid stimulating hormone Hyperthyroidism
Gonadotroph 6 FSH, LH None or hypogonadism
Null cell 35 None Hypopituitarism

a Data from Thapar and Laws based on 3000 surgically treated pituitary adenomas.9

298
ANTERIOR PITUITARY TUMORS

usually associated with GH, ACTH or PRL Endocrine abnormalities


hypersecretion, occur in about 25% of such
patients. MEN I is associated with both micro- Table 10.2 lists the various syndromes caused
and macroadenomas, mostly PRL secreting. by hypersecretion of particular hormones.
The genetic abnormality is allelic loss of a The clinical symptoms are detailed in the
tumor suppressor gene called menin at paragraphs on specific tumor types (see below).
11q13.10 The function of the menin protein is Macroadenomas, by compressing surrounding
unknown, but it reacts with the metastasis- normal glandular elements, can lead to hypo-
suppressor nm23.10 MEN I patients account secretion of one or more pituitary hormones
for no more than a few percent of pituitary (Table 10.3).
tumors, but deletions of the 11q13 locus, Gonadotrophs are most vulnerable to
where menin resides, have been found in compression. Loss of libido, amenorrhea, and
sporadic pituitary tumors.9 The G-protein impotence are often the presenting symptoms
oncogene, GSPT1 is mutated in 3040% of in patients with large pituitary adenomas. TSH
patients with acromegaly. This mutation in the and GH are the next most vulnerable cell types.
-chain of the G-protein results in constitutive
activity of adenyl cyclase. Adenomas with G-
Table 10.3
protein mutations are significantly smaller
Some symptoms and signs of pituitary failure.
than those without mutations and may be
more sensitive to inhibitory factors such as
somatostatin or dopamine. Amplification of C- Gonadotroph failure
Loss of libido
fos and point mutations in H-ras oncogenes
Impotence
have been shown in isolated prolactinoma Osteoporosis
cases. A number of growth factors have been Thyrotroph failure
identified in the pituitary and it is believed Fatigue, malaise
that growth factor mediated autocrine and Apathy, slow cognition
paracrine stimulation may be important in Myalgias
Carpal tunnel syndrome
pituitary tumor development. Constipation
Delayed reflexes
Weight gain
Deep voice
Clinical findings diagnosis Somatotroph failure
Reduced strength (muscle mass)
Pituitary adenomas cause their symptoms by
Central obesity
abnormal hormone secretion (found in about Premature atherosclerosis
70% of pituitary adenomas) or by compression Depression, anxiety, emotional lability
of normal glandular and neural structures.11,12 Corticotroph failure
As macroadenomas outgrow the sella turcica, Fatigue
they compress neural structures including Weight gain
Hypoglycemia
ocular motor nerves laterally and the optic Vasopressin (posterior pituitary)
chiasm and hypothalamus superiorly. An Polydipsia
occasional pituitary adenoma erodes the floor Frequent urination (especially nocturia)
of the sella turcica and presents as a mass in Thirst
the sphenoid sinus or nasopharynx.

299
PITUITARY AND SELLAR REGION TUMORS

GH deficiency has subtle but important effects Table 10.4


in adults,13 and GH deficiency can mimic other Non-endocrine signs and symptoms.
diseases because of its psychiatric and cardiac
manifestations. Hypothyroidism with its vague Symptoms Signs
and protean symptoms of fatigue and malaise
may be a confusing presenting complaint. Headache Extraocular muscle
paresis
Hypoadrenalism is rare. Prolactin levels may be
Peripheral visual loss Bitemporal hemianopsia
elevated in patients with non-secreting pituitary or unilateral optic
adenomas, because the hypothalamus controls neuropathy
PRL by releasing inhibitors, of which Sudden blindness Optic neuropathy
dopamine is the most important. Thus, Loss of consciousness
compression of the pituitary stalk by a
macroadenoma decreases the output of most
anterior pituitary hormones, but may increase
output of PRL. PRL levels caused by stalk
compression rarely rise above 200 ng/ml,
which is easily distinguished from the usually
high PRL levels (2500 ng/ml) seen with
PRL-secreting adenomas. Diabetes insipidus,
although a common finding in pituitary metas-
tases and seen transiently after pituitary
surgery, is almost never a presenting complaint
of pituitary adenoma. Sudden pituitary failure
may be caused by pituitary apoplexy (see
below).

Non-endocrine findings (Table 10.4)


Headache is frequently an early symptom,
probably due to stretching of the diaphragma
sella. The headache is non-specific, usually felt
as a dull bifrontal or vertex pain and, like
headache from other brain tumors, is often
more intense in the morning. In some patients,
as the tumor grows and the diaphragma sella
is ruptured, the headache disappears. As the Figure 10.2
tumor grows superiorly, above the diaphragma Large prolactin-secreting pituitary adenoma. The
sella, it encounters the optic chiasm (Fig. 10.2). patient is a middle-aged physician who developed
impotence but had no other symptoms. The tumor
Compression of that structure may lead to
responded completely to bromocriptine. The
bitemporal hemianopia of which the patient is photomicrograph shows loss of acinar architecture
often unaware. Thus, the first symptom of a with sheets of neoplastic cells with round nuclei,
pituitary tumor may be an automobile accident basophilic cytoplasm and distinct cell borders.

300
ANTERIOR PITUITARY TUMORS

because of the patients lack of peripheral Laboratory evaluation


vision. Depending on the exact location of the
optic structures and the site of suprasellar If a pituitary abnormality is suspected, high-
tumor growth, one optic nerve anterior to the resolution MRI with gadolinium enhance-
chiasm may be compressed, causing unilateral ment and 1 mm sections through the pituitary
visual loss with optic atrophy and rarely a fossa will establish the diagnosis in most
temporal visual defect in the opposite eye, a instances, even when microadenomas are as
junctional scotoma.14 If the tumor grows later- small as 23 mm (Fig. 10.3). The physician
ally rather than anteriorly, it compresses the who suspects a pituitary adenoma should tell
cavernous sinus, causing ptosis (oculomotor the radiologist that he requires fine sections
nerve), diplopia (oculomotor, trochlear or and rapid imaging during bolus contrast
abducens nerve), or facial numbness (trigemi- administration, particularly for a suspected
nal nerve). Very large pituitary adenomas can microadenoma. With macroadenomas, the
compress the hypothalamus, causing dementia, MRI also establishes the spatial relationship
drowsiness, hypo- or hyperphagia, emotional between the tumor and important surround-
disturbances and obstructive hydrocephalus ing neural structures. Characteristically, the
from compression of the 3rd ventricle. tumor is isointense on T1 and variably iso-
For reasons not entirely understood, both to hypointense on T2. Immediately after
large and small pituitary adenomas have a injection of contrast, the tumor is hypo-
tendency to either bleed or infarct sponta- intense on T1 compared to normal gland,
neously, causing pituitary apoplexy.15 The which has no bloodbrain barrier and there-
vascular insult usually occurs spontaneously fore enhances diffusely. Later it becomes
but may result from cerebral angiography or isointense with the contrast-enhanced normal
during provocative endocrine testing; it also gland. About 10% of prolactinomas calcify,
occurs with greater frequency during preg- which is best seen on CT scan. A CT scan of
nancy. The result is sudden enlargement of the the pituitary fossa is particularly valuable in
pituitary tumor and spillage of hemorrhagic or identifying acute hemorrhages in patients
necrotic material into the suprasellar cistern with pituitary apoplexy and may help with
and the subarachnoid space. The clinical the differential diagnosis of pituitary tumor
symptoms include sudden onset of visual loss, versus meningioma by identifying the hyper-
ocular palsies, acute hypopituitarism, and ostosis of bone characteristic of a menin-
alteration in consciousness varying from confu- gioma.
sion to coma. Fever and nuchal rigidity may A second approach to diagnosis is measure-
suggest the diagnosis of bacterial or viral ment of hormones in the blood. Both hyper-
meningitis. The CSF may contain blood or and hyposecretion of pituitary hormones can
white cells, the latter in response to spilled be identified by blood measurement. Hormone
necrotic material. Most patients respond to levels not only help establish the diagnosis
conservative treatment, replacing absent of the tumor cell type, but are also useful
hormones and using corticosteroids to suppress to follow tumor response to treatment and
inflammation. In some patients, emergency to identify post-treatment hypopituitarism.
decompression of the enlarged pituitary is Careful neuro-ophthalmologic examination
necessary to relieve symptoms. Untreated with special attention to visual fields and
pituitary apoplexy can be fatal. ocular movements will determine whether

301
PITUITARY AND SELLAR REGION TUMORS

Figure 10.3
A prolactin-secreting microadenoma in an adult male with sexual difficulties. Microadenomas are generally
found on MR scan by their failure to enhance as rapidly as normal pituitary during dynamic injection of
contrast material. This particular prolactinoma, which responded to bromocriptine therapy with normalization
of the prolactin level, was actually seen before contrast as a hypointense 5 mm lesion (arrow lower right). It
was also hyperintense on the T2-weighted image (arrow upper right), possibly because of cystic degeneration.
The photomicrograph shows a chromophobe adenoma. A psammomatous concretion is also present (arrow).

macroadenomas have substantially interfered nasal fluid glucose is helpful and easily accom-
with neurologic function. An occasional plished using urine dipsticks. If measurable
patient whose tumor has eroded the floor of glucose is present, the fluid is CSF; if absent,
the sella may develop a CSF leak. All patients it may or may not be CSF. Transferrin, a
with macroadenomas should be questioned protein present only in CSF, is more specific,16
concerning the presence of rhinorrhea. If but requires collection of an adequate fluid
rhinorrhea is present, measurement of the specimen.

302
ANTERIOR PITUITARY TUMORS

Differential diagnosis
In most instances, MR scans will distinguish
pituitary neoplasms from other masses of the
sella turcica (Table 10.5).
MRI cannot distinguish pituitary adenomas
from the much rarer pituitary carcinomas;9 that
distinction requires biopsy. Tumors of neuro-
hypophyseal origin, which are quite rare, may
be hard to distinguish from those of adeno-
hypophyseal origin. Tumors of non-pituitary
origin, such as craniopharyngiomas, germ
cell tumors, meningiomas and hypothalamic
gliomas, are generally easily distinguishable
because they either spare the pituitary fossa or
invade it only after filling the suprasellar area.
Dermoids and epidermoids, Rathkes cleft cysts
and the empty sella syndrome (from CSF in the
sella) have different signal intensity on MRI
from pituitary tumors. Carotid aneurysms
rarely mimic pituitary adenoma, and MR scan
characteristics easily differentiate these. Figure 10.4
One diagnostic problem involves metastasis A pituitary metastasis in a man with advanced colon
to the pituitary gland (Fig. 10.4).20 Carcinomas cancer. He presented with rapidly developing visual
loss. A diagnosis of pituitary apoplexy was made. At
of the breast have a predilection to metastasize
surgery, both the pituitary tumor and the dural
to the pituitary, but other cancers may also do tumor (arrow) were metastases.
this. Unlike pituitary adenomas, the first
symptom is usually diabetes insipidus from
invasion of either the pituitary stalk or the
Table 10.5 neurohypophysis. Sometimes the tumor also
Sellar and suprasellar masses: differential diagnosis. invades the subarachnoid space, allowing one
to make a diagnosis of metastatic tumor by
examining the CSF. The clinical situation also
Pituitary adenoma
Craniopharyngioma helps establish the diagnosis of pituitary metas-
Meningioma tasis, which usually occurs in the setting of
Rathke cleft cyst disseminated tumor. Occasionally a diagnosis is
Carotid aneurysm not possible on the basis of imaging or CSF
Langerhans cell histiocytosis (see Chapter 11) examination, and biopsy is the only option.
Lymphocyte hypophysitis
The most difficult differential diagnosis is
Pituitary granuloma
Teratoma17 (Chapter 9) between certain inflammatory conditions and
Pituitary abscess18 pituitary adenoma. Lymphocytic hypophysitis
Pituitary lymphoma19 and giant cell granulomas of the pituitary
often present with hypopituitarism, particularly

303
PITUITARY AND SELLAR REGION TUMORS

Figure 10.5
Lymphocytic hypophysitis mimicking a pituitary tumor. The intense enhancement of the infundibulum (arrows)
was suggestive of inflammation rather than tumor, as was the fact that 2 months previously (upper left) the
patient had a scan for non-specific headaches which was negative.

hypothyroidism; diabetes insipidus is common enlarging sella turcica to be suffering from a


(Fig. 10.5).21,22 A CSF pleocytosis may be possible inflammatory condition and consider
present.23 The inflammatory lesion enlarges the a trial of steroids.
sella and mimics pituitary adenomas on MR
scan; however, the frequent enhancement of the
Treatment
stalk and inferior hypothalamus is often a clue
that the process is inflammatory and not an Surgery
adenoma. Because their treatment is non-surgi- For most pituitary adenomas, the treatment
cal, i.e. corticosteroid therapy,24 the physician is surgical.2,25 Microadenomas not causing
should consider all patients, particularly symptomatic hormonal dysfunction may just
pregnant or postpartum young women, with be followed, and some prolactinomas26 and
rapidly developing hypothyroidism and an GH secreting adenomas27 may be successfully

304
ANTERIOR PITUITARY TUMORS

treated with pharmacologic suppression of Diabetes insipidus may also be a complication


hormone hypersecretion (see below). For of surgery (usually transient) but is easily
patients who require surgery, neurosurgeons controlled by appropriate hormonal therapy.
with extensive experience in pituitary surgery Postoperative hypopituitarism is uncommon
have substantially better results than those who and can also be controlled by hormonal
are less experienced. In most patients, the therapy. About 50% of patients who have
tumor can be reached from below through a evidence of hypopituitarism preoperatively
transsphenoidal approach. Transsphenoidal recover some endocrine function.31 Persistent
surgery takes advantage of access to the skull postoperative hormone hypersecretion, even if
base and sella turcica via the nose and air MRI demonstrates complete tumor removal,
sinuses. The original early surgical approach to mandates additional pharmacologic therapy
pituitary tumors via a frontal craniotomy was and/or radiotherapy.
abandoned in the late 1950s because of its
limited exposure and the inability of the Radiation therapy
surgeon to see the anatomy and pathology Radiation therapy (RT) using doses of 4550 Gy
adequately. In the 1960s, the introduction of in 1.8-Gy fractions is also effective at reducing
the operating microscope into neurosurgery, tumor growth.32 Radiation is indicated for
with its powerful illumination and magnifica- persistent or recurrent, medically refractory
tion, made the transsphenoidal approach the hypersecreting adenomas, or administered
procedure of choice for resection of pituitary postoperatively to patients with invasive or
tumors. A few very large tumors that do not large, incompletely removed tumors.33 Late
enlarge the sella turcica to allow good surgical complications include hypopituitarism, and
access, or that spill laterally and cannot be rarely with modern radiation techniques, optic
reached through the sella from below, may nerve, hypothalamic or temporal lobe damage.
require a craniotomy and transfrontal These effects usually develop within a year or
approach. Surgery is both safe and efficacious. two. Years later, as a very late-delayed effect,
It has an additional advantage of establishing secondary brain tumors may appear.34 Excluding
the correct diagnosis. In one study, a preoper- of the above complications, radiation therapy
ative diagnosis of non-functioning pituitary does not cause cognitive dysfunction.29 Unlike
adenoma was incorrect in 4 out of 22 surgery, radiation may take years to normalize
patients.28 Operative mortality and morbidity elevated hormone levels. Radiosurgery may
rates from several series are generally 0.5% produce more rapid hormonal and clinical
and 2.2% respectively. The most common responses with similar or less toxicity than
complication is CSF rhinorrhea predisposing conventional RT,35,36 but it cannot be used for
to meningitis. Cognitive dysfunction has been large tumors or those < 3 mm from the optic
reported to follow both transfrontal and nerve or chiasm. Charged particle beams have
transsphenoidal surgery, with the former been advocated by some.37
associated with more severe impairment.29 Rare
complications include traumatic injury to the Pharmacologic therapy
hypothalamus resulting in coma or death, Some pituitary adenomas have proved sensitive
laceration of the carotid artery and damage to to pharmacologic treatment.38 PRL-secreting
the optic chiasm. These benign tumors may tumors that require therapy can be treated with
rarely seed the meninges after operation.30 dopamine agonists to inhibit the output of

305
PITUITARY AND SELLAR REGION TUMORS

PRL. These drugs substantially reduce the size Table 10.6


of prolactinomas and control their growth. Signs and symptoms of prolactinomas.
Bromocriptine and pergolide were the most
frequently used agents, but cabergoline is more Women Men
effective and better tolerated and is now the
drug of choice.26,39 The drug need be given only Amenorrhea Impotence
Galactorrhea Decreased libido
twice weekly. Somatostatin and its analogs,
Decreased libido Gynecomastia
such as octreotide, have been used in the Infertility
management of tumors associated with
acromegaly.27 However, the degree of tumor
shrinkage is not as dramatic as with dopamine
agonist therapy of prolactinomas. In selected
patients, somatostatin analogs may be primary endocrine symptoms in men may explain why
therapy, but they are usually reserved to tumors in men are generally larger than those
control symptoms if surgery and radiotherapy in women. Fewer than 10% of PRL-secreting
fail. Gene therapy of pituitary tumors, still in microadenomas evolve into macroadenomas;
its infancy, may hold hope for the future.40 the others seem not to grow after they are
identified, or even regress spontaneously.7 The
Treatment complications diagnosis is made on the basis of MR imaging
Aside from the usual complications of surgery and PRL levels. A PRL level in excess of
and RT,41 patients treated for pituitary adeno- 200 ng/ml is, with rare exceptions,43 diagnostic
mas are at increased risk of cerebral infarction, of a prolactinoma, while levels in excess of
probably related to the combination of 1000 ng/ml suggest that the prolactinoma is
endocrine dysfunction and RT, both of which invasive. The treatment of choice is pharmaco-
may accelerate atherosclerosis.42 logic.26,39 PRL levels are controlled with
dopamine agonists in almost all microadeno-
mas;44 about 60% of patients with macroade-
nomas respond with > 80% tumor shrinkage.45
Specific anterior pituitary In women with the amenorrhea-galactorrhea
tumors syndrome, dopamine agonist therapy can
restore appropriate hormonal levels and allow
Prolactinomas (Table 10.6)
for pregnancy. PRL levels can be followed to
PRL-secreting pituitary adenomas are the most determine the effectiveness of therapy. The
common clinically recognized tumor of the drugs are discontinued during pregnancy and
pituitary gland, accounting for about one-third resumed after pregnancy. There is a risk,
of symptomatic tumors. They occur in both however, of accelerated tumor growth during
sexes but are most common in young women pregnancy in patients with macroadenoma, so
and older men. Women usually present with that one might consider surgery before
amenorrhea and galactorrhea as a consequence pregnancy in such a patient.
of PRL excess; some present with infertility. In men with asymptomatic microadenomas,
Men present with headache and visual loss treatment is unnecessary. Macroadenomas can
from the enlarging mass or sometimes with be treated successfully by dopamine agonist
impotence or gynecomastia. The lack of early therapy. The treatment must be maintained for

306
SPECIFIC ANTERIOR PITUITARY TUMORS

life and the lesion tends to scar. Some neuro- Table 10.7
surgeons believe that the scarring makes subse- Signs and symptoms of somatotroph adenomas.
quent surgery more difficult,46 although the
final results of surgery are usually the same Children Adults
whether treated with dopamine agonists or not.
If drugs fail or cannot be tolerated by the Tall Hand, feet and jaw growth
Rapid growth Deep voice
patient, surgery is effective.47 The surgical cure
Joint pain Joint pain
rate for microadenomas is as high as 75%, Increased sweating Increased sweating
higher if PRL levels are < 100 ng/ml and lower Diabetes mellitus
if levels are > 200 ng/ml; it is about 30% for Renal stones
macroadenomas and even lower for invasive Headache
Cardiac disease
tumors. Patients whose prolactin levels fall
10% within 10 min after resection and reach
< 20 ng/ml are probably cured.48 Symptomatic
recurrence is seen in about 25% of patients.
Risk factors for recurrence include basal PRL negative, ectopic acromegaly caused by carci-
levels > 400 ng/ml, macroadenoma, male sex, noids or small cell lung cancer must be consid-
and prolonged preoperative dopamine agonist ered in the differential diagnosis and a search
therapy.49 RT or radiosurgery should be for those lesions undertaken. The treatment of
reserved for patients who fail both pharmaco- choice for most GH-secreting adenomas is
logic therapy and surgery. surgery;2 however, recent experience dictates
that a trial of octreotide or slow release
Growth hormone-secreting lanreotide51 is warranted as primary therapy for
some patients, giving response rates > 60%.27
adenomas (Table 10.7)
Dopamine agonists are effective in some
GH excess from pituitary adenomas results patients, usually at high doses.44 Biochemical
in pituitary gigantism in children50 and cure, defined as normalization of IGF-1 levels
acromegaly in adults. Coarsening of facial or basal or glucose suppressed GH levels of
features, separation of teeth and enlargement of 2 ng/ml or less, can be achieved in almost 90%
the hands and feet characterized by an increase of patients with microadenomas and 55% of
in shoe and glove size are signs of acromegaly. patients with macroadenomas.52 Tumor recur-
Patients may develop diabetes mellitus. With rence is uncommon in adults but more common
further tumor growth, hypopituitarism and in children.50 Risk factors for recurrence include
visual complaints develop. Headache, carpal basal GH levels > 40 ng/ml, macroadenoma,
tunnel syndrome and arthritic pain are common invasive adenoma and an abnormal postopera-
complications of acromegaly. The diagnosis can tive glucose tolerance test.49 Radiation therapy
be made by inspection of the patient and is can be delivered to invasive tumors or those
supported by elevation of GH following a that fail to achieve a biochemical cure. Within
glucose load, and by elevation of insulin-like 10 years of RT, 80% of patients achieve GH
growth factor-1 (IGF-1) in the serum when the levels below 5 ng/L, but IGF-1 levels remain
pituitary tumor cannot be identified on MRI. elevated53 and RT is rarely curative. Long-term
The tumors are sometimes too small to be treatment with a somatostatin analog is often
identified by MRI. If the pituitary MRI is required.

307
PITUITARY AND SELLAR REGION TUMORS

Table 10.8
Weight gain Diabetes mellitus Signs and symptoms of
Abdominal striae (purple) Hypertension corticotrophic adenomas.
Increased hair growth (face, chest, abdomen) Osteoporosis
Depression Proximal muscle weakness
Easy bruising

ACTH-secreting adenomas (Table to verify a pituitary origin of excessive ACTH


10.8) secretion.54 Rarely, the adenoma may be
ectopic, e.g. in the cavernous sinus.55
About 15% of pituitary adenomas secrete Transsphenoidal exploration of the pituitary
ACTH. ACTH secretion leads to two distinct gland can lead to curative resection of more
clinical syndromes: Cushings disease and than 70% of microadenomas and 30% of
Nelsons syndrome. Seventy-five percent of macroadenomas.2 Most failures are a result of
patients with Cushings disease are women, difficulty locating the tumor. Intraoperative
usually in the third and fourth decades. ultrasound often helps.56 However, transsphe-
Patients characteristically develop deposition of noidal surgery for Cushings disease has a
fat in the face (moon facies), supraclavicular higher complication rate than that for other
fossae, abdomen (central obesity) and back of pituitary adenomas, primarily related to more
the neck (buffalo hump). Muscle atrophy leads medical complications, especially deep vein
to thin extremities, accentuating the central thrombosis.57 Prophylactic therapy for throm-
obesity. Hirsutism, thinning and hyperpigmen- bosis in the postoperative period should be
tation of the skin, and vascular fragility, lead considered. Despite surgery, persistence or
to diffuse ecchymoses and purple abdominal recurrence of tumor is common. Pharmacologic
striae. Proximal weakness is first characterized blocking agents such as mitotane, ketoconazole
by difficulty in getting off the toilet seat and and metyrapone have some effect but probably
subsequently in climbing stairs. Patients may should be used only after failure of surgical
develop hypertension, glucose intolerance, and intervention. The same applies to RT, which
osteoporosis. The disorder, untreated, is life- controls more than 80% of patients who fail
threatening with a 5-year mortality of about transsphenoidal surgery.58 However, IGF-1,
50%. The diagnosis is made by: (1) elevated also elevated in patients with Cushings disease
levels of 24-hour urinary free cortisol, (2) loss as well as those with acromegaly, is not
of ACTH suppression by glucocorticoids and normalized by RT.53 In some instances, bilat-
(3) elevated ACTH levels. Because some eral adrenalectomy is necessary to ameliorate
cancers (especially small cell lung cancer) the potentially fatal hyperadrenocorticalism.
secrete ACTH-like substances and cause When patients with Cushings disease are
Cushings syndrome, and because ACTH- treated with bilateral adrenalectomy rather
secreting pituitary adenomas are often too than hypophysectomy, Nelsons syndrome
small to identify on MR scan, it is sometimes occurs in about 1015% of these patients. The
necessary to measure ACTH in each inferior persistent high levels of ACTH stimulate
petrosal sinus which drains the pituitary gland melanocyte-stimulating hormone. The patients

308
SPECIFIC ANTERIOR PITUITARY TUMORS

develop diffuse hyperpigmentation and expan- of a somatostatin analog has reduced TSH
sion of the sella turcica, which can lead to levels,61 but generally it does not reduce the
compression of adjacent neural structures. tumor mass.
Transsphenoidal surgery cures only 25%. RT
may help control tumor growth, but some Gonadotropin-secreting pituitary
patients die of continued local growth and even
metastases.59
adenomas
Gonadotropic adenomas secrete FSH and LH.
Thyroid-stimulating adenomas They account for 515% of pituitary adeno-
mas. They are usually macroadenomas62 and
(Table 10.9) present with visual loss and hypopituitarism.
TSH-secreting adenomas account for about 1% These hormones can cause hypogonadism with
of all pituitary tumors. Most patients who diminished libido in men and amenorrhea in
present with hyperthyroidism have TSH levels premenopausal women, but are usually asymp-
that are not repressed by the elevated thyroid tomatic until they compress normal tissue.
hormone levels. TSH levels are normal or high Microadenomas do not require treatment
and are accompanied by an increase in plasma unless symptomatic, or if a woman wishes to
alpha subunits,60 indicating that the problem become pregnant. Macroadenomas should be
lies in the pituitary and not the thyroid. Except treated surgically. Persistence or recurrence of
for TSH secretion, many tumors are silent so tumors occurs in about 40% of patients.62
that by the time the diagnosis is recognized, Residual tumor may respond to RT.
often after thyroid ablation has been performed
for presumed thyroiditis, the tumor is usually Non-secreting pituitary adenomas
macroscopic in size. About 50% of large
tumors demonstrate mitoses and nuclear atypia Non-functioning pituitary adenomas account for
and have invaded parasellar structures. Most about one-third of clinically recognized pituitary
TSH-secreting tumors also secrete GH or PRL, adenomas and most incidental tumors encoun-
also helping to establish the diagnosis of a tered at autopsy. The tumors present as
pituitary rather than a thyroid lesion. The macroadenomas with headache, visual distur-
treatment is surgical, with adjunctive radio- bances and symptoms of pituitary failure. PRL
therapy in cases of invasion. Surgery cures may be elevated because of pituitary stalk
about 25% of tumors. In some instances, use compression. These tumors generally occur in the
older age group, are diagnosed by MRI and are
treated surgically. Visual abnormalities are
Table 10.9 reversed in most, and preserved if they are normal
Signs and symptoms of thyrotroph adenomas. preoperatively; endocrine function is preserved.
Symptomatic recurrence develops in fewer than
10% of patients after gross total resection.33
Tachycardia Insomnia
Tremor Anxiety Radiotherapy prevents tumor regrowth in about
Weight loss Diarrhea 90% of patients after subtotal resection.63,64 Risk
Increased appetite Enlarged thyroid factors for recurrence include macroadenoma,
Heat intolerance invasive adenoma, plurihormonal adenoma, silent
ACTH adenoma and oncocytoma.49,65

309
PITUITARY AND SELLAR REGION TUMORS

Pituitary carcinoma the anterior pituitary gland, the oral mucosa and
teeth (Fig. 10.6). The tumor has two histologic
Pituitary carcinoma is rare; fewer than 100 cases types. The adamantinomatous type, representing
have been reported.20 The diagnosis is made teeth primordia, is usually a calcified multi-
when tumors of anterior pituitary origin, cystic tumor of childhood with a peak age of
believed to be adenomas, cause distant metas- onset of 59; it rarely occurs in adults. The
tases or disseminate into the subarachnoid space. squamous papillary type, representing oral
The tumors often declare themselves with clini- mucosa primordia, is a tumor of adults and
cal evidence of invasion of the intracranial or is rarely associated with macroscopic cysts,
spinal leptomeninges. Extraneural metastases to although microscopic cysts may sometimes be
the mediastinum and lymph nodes also occur. identified. Xanthogranulomatous change with
The tumors may be hormonally active or cholesterol clefts, inflammatory infiltrates and
hormonally silent. Once distant metastases necrosis is generally considered a variant of the
develop, standard anti-cancer chemotherapy may adamantinomatous type, although some place it
be tried but is usually ineffective. The metastatic in a separate category.66 The incidence of cranio-
disease is usually responsible for death. pharyngioma is 1.3 per million without gender
or race specificity. Thus, approximately 338
cases occur annually in the USA.67 Overall,
Posterior pituitary tumors about half of craniopharyngiomas occur in
children or adolescents. No risk factors, either
Posterior pituitary tumors are rare. The most
genetic or environmental, have been identified
common primary posterior pituitary tumor is
for this tumor. Abnormalities involving chromo-
the granular cell tumor, usually an incidental
somes 2 and 12 have been reported.
finding at autopsy. If the tumor causes
symptoms, diabetes insipidus is usually the first
symptom, a rare finding with pituitary adeno- Pathology
mas. Astrocytomas, usually low-grade, also
The tumors are usually well-circumscribed,
occur in the posterior pituitary.
sometimes surrounded by a pseudocapsule.
The most common tumor of the neuro-
However, within this capsule there may be
hypophysis is a metastasis, usually from breast
microscopic rests of tumor that can lead to recur-
cancer, with lung and prostate cancer being less
rence after what is believed to be a gross total
common. In most, diabetes insipidus is the first
resection. Within the adamantinomatous tumors
symptom. About 20% of patients presenting to
are so-called wet or ghost cell keratins. These
a general hospital with diabetes insipidus have
consist of cells in which the nucleus is missing.
metastatic breast cancer as the cause.
The papillary type is associated with so-called
dry keratin and is often located in the 3rd ventri-
cle rather than the suprasellar cistern. It may be
Craniopharyngiomas difficult to distinguish craniopharyngiomas from
epidermoid cysts based on histology alone.
Introduction
Grossly, adamantinomatous craniopharyngiomas
The craniopharyngioma is a low-grade supra- contain both solid and cystic components. Cystic
sellar tumor believed to arise from remnants of tumors express vascular endothelial growth
Rathkes pouch, the embryologic precursor of factor (VEGF); solid tumors do not.68 The cysts,

310
CRANIOPHARYNGIOMAS

Figure 10.6
Craniopharyngioma. The MRI on the right is a contrast-enhanced scan showing a large suprasellar tumor with
a cystic component (arrow). The lesion was originally misinterpreted as an optic glioma because of the
apparent involvement of the optic tract on the T2-weighted image (left MRI). This is edema extending through
the optic tracts, giving the mustache sign characteristic of craniopharyngiomas. The photomicrograph shows
epithelial sheets containing cysts with peripheral palisading and the typical wet keratin.

which contain cholesterol, have a thick yellow- present in the cyst wall. The papillary cranio-
ish machine oil-like appearance and are filled pharyngioma lacks calcification but has choles-
with debris that may include keratin. terol-filled cysts as well. The squamous cells are
Microscopically, the adamantinomatous type not columnar as they are in the adamantinoma-
consists of stratified squamous epithelium, tous variety.
sometimes flattened when it lines the cyst. Keratins can be identified by immunohisto-
Compact wet keratin is more nodular and can chemistry (high molecular weight in adamanti-
calcify. Fibrosis and chronic inflammation are nomatous types and low molecular weight in

311
PITUITARY AND SELLAR REGION TUMORS

papillary types). Intense gliosis and Rosenthal Unlike with pituitary tumors, diabetes insipidus
fibers are common; the MIB-1 index is usually is a common presenting complaint.
less than 1%, but may be somewhat more The tumor frequently compresses the optic
active in recurrent tumors. chiasm, causing a bitemporal hemianopia,
the characteristic visual abnormality. Neither
adults nor children may be aware of the visual
Clinical findings
field defect. Adults may become aware of it
Symptoms and signs when the failure of peripheral vision leads to
Craniopharyngiomas are located just above the automobile accidents. In one series of children,
sella (rare ectopic craniopharyngiomas have 5 of 75 children required emergency surgical
been reported in the optic nerve, sphenoid decompression because they were virtually
bone, pharynx and posterior fossa). Therefore, blind by the time of referral.69 Other visual
signs and symptoms are similar to those of field abnormalities, including unilateral visual
pituitary tumors and include endocrine deficits loss, are also occasional symptoms.
and visual problems (Table 10.10). Increased intracranial pressure and hydro-
In addition, because the tumors often cephalus occur because the tumor often grows
compress the 3rd ventricle, increased intracra- to a large size in both adults and children
nial pressure and hypothalamic dysfunction are without earlier symptoms or signs having been
also common. Endocrine dysfunction includes recognized. Of 75 children, 25% required an
growth failure in children and impotence in emergency surgical procedure for relief of
adults. About 75% of patients have deficien- hydrocephalus, often presenting with acute
cies of GH, 40% deficiencies of gonadotropins alterations of consciousness.69 About two-
and about 25% deficiencies of ACTH or TSH. thirds of all children present with symptoms
of increased intracranial pressure, including
headache and vomiting. Papilledema occurs in
Table 10.10 20% of children.
Craniopharyngioma: clinical findings. Hypothalamic dysfunction was reported in
23% of children and included weight gain
I. Endocrine deficiency resulting from hypothalamic hyperphagia or
Growth failure (children) weight loss resulting from hypothalamic
Impotence (adults) anorexia, in addition to behavioral, memory
Diabetes insipidus (2030%) and cognitive changes. Deterioration in school-
II. Visual abnormalities
work may be an early symptom in children.
Bitemporal hemianopia
Blindness Personality changes include psychomotor
III. Increased intracranial pressure (children) retardation, emotional immaturity, apathy and
Headache, vomiting short-term memory losses.
Papilledema (20%) In men, decreased libido or impotence occur
Cognitive dysfunction
in about 90% of patients. Premenopausal
Alterations of consciousness
IV. Hypothalamic dysfunction (23%) women often present with amenorrhea (80%).
Weight gain (or loss) Pregnancy has been reported to exacerbate
Behavioral problems symptoms caused by a craniopharyngioma.70
Memory loss Signs of increased intracranial pressure are less
common, although about half of adult patients

312
CRANIOPHARYNGIOMAS

have headaches. In rare instances, rupture of a Treatment


craniopharyngioma may produce severe aseptic
meningitis with severe headaches, stiff neck The tumor is often adherent and difficult
and alterations of consciousness. to separate from vascular structures, optic
apparatus, and hypothalamus. When the tumor
Imaging can be completely removed, surgery is often
Craniopharyngioma is one tumor in which curative,72 with a recurrence rate of 19% at
plain skull films may be useful. Suprasellar 10 years. Rarely, the tumor will seed the
calcification is seen in over 75% of children leptomeninges postoperatively.73 Unfortunately,
and in somewhat less than half of adults. the location of the tumor and its tenacious
Enlargement of the sella with erosion of the adherence to vital structures often makes
anterior clinoids and dorsum sellae is also complete removal either impossible or
common. In the absence of calcification, plain unacceptably dangerous. However, some
skull films may show sellar enlargement, falsely reports suggest that as many as 90% of tumors
suggesting a pituitary rather than a suprasellar can be totally removed using modern micro-
tumor. CT scans are also sometimes useful, surgical techniques. In one series, 90% of the
showing contrast enhancement of the solid 144 patients who underwent microsurgical
portion of the tumor as well as identifying the resection of craniopharyngioma were com-
typical calcifications, which are sometimes pletely resected and only 7% had a recur-
hard to see on MR scan. On MR, the cystic rence.74 Frontal or frontotemporal craniotomy
portion of the tumor may be either hyper- or is the most common exposure used to remove
hypointense on the T1-weighted image (choles- the tumor. Transnasal surgery for certain
terol and keratin are hypointense on T1, tumors that are infradiaphragmatic appears
whereas hemorrhage is hyperintense). On the safe and effective.75 Diabetes insipidus often
T2-weighted image, cholesterol and hemor- follows surgery for craniopharyngioma,
rhage are hyperintense and keratin hypo- although anterior pituitary function may be
intense. The solid portion of the tumor is retained.76 If complete removal is not possible,
usually hypointense on T1, hyperintense on T2, postoperative radiotherapy appears to decrease
and contrast enhances. An MR angiogram may considerably the incidence of recurrence.72
help the surgeon identify the relationship of the Radiosurgery is sometimes useful for the
tumor to major vessels passing near the treatment of small solid craniopharyngiomas,77
suprasellar cistern. An unusual sign of cranio- but most tumors are too large and their
pharyngioma is the so-called mustache sign.71 proximity to the optic tract (< 3 mm) prevents
This sign is related to vasogenic edema caused its use in most instances. Interstitial implanta-
by increased permeability of the bloodbrain tion of radionuclides, including phosphorus-
barrier in the optic tracts and optic radiations 32, yttrium-90, gold-198, and rhenium-186,
(Fig. 10.6). Vasogenic edema is thought to all of which emit -particles, has been recom-
result from leakage of the craniopharyngioma mended for some monocystic craniopharyn-
contents, causing swelling and bloodbrain giomas; the target dose to the inner wall of the
barrier breakdown in the optic pathways. It is cyst is about 200350 Gy. Only 32P is available
usually asymptomatic but may be misinter- in the USA.78
preted as representing an optic nerve tumor Chemotherapy is not widely applied for cranio-
rather than a craniopharyngioma. pharyngiomas. Bleomycin has been directly

313
PITUITARY AND SELLAR REGION TUMORS

injected into the tumor,79 with encouraging surgical removal.83 Visual deterioration,
results, but also sometimes with severe toxicity.80 hydrocephalus and cognitive dysfunction may
Despite multimodality treatment, about 25% of develop during or within 2 months of post-
craniopharyngiomas recur, even some that have operative RT due to cyst enlargement or
been totally resected. At least one investigator hydrocephalus. Early detection and treatment
reports the cumulative recurrence rate at 10 years of the enlarged cyst by drainage or placement
as 33% and that after 15 years as 40%, not of a ventriculoperitoneal (VP) shunt improves
differing by age, radical surgery, or postoperative long-term outcome.84 Late-delayed effects also
radiotherapy.81 Repeat surgery should be the first develop as a consequence of RT, particularly
consideration since total tumor resection can memory dysfunction. Secondary glial tumors
sometimes be accomplished even when the tumor can also appear many years after successful RT
has recurred. If radiation has not been used, it for a craniopharyngioma.
should be applied in the postoperative period.
Other options as outlined above can also be
considered.82
Rathke cleft cyst
Arising from the epithelial remnants of
Prognosis
Rathkes cleft, these cysts, which are found
Total surgical removal results in about 80% either in sellar or suprasellar areas, are usually
cure.72 About half of the patients with subtotal small and asymptomatic. Symptomatic cysts
removal are also cured and many more can be are associated with symptoms similar to those
salvaged after recurrence. However, a small but of pituitary tumors, including pituitary
significant percentage of patients are damaged dysfunction, visual disturbances and headache
by treatment. Surgical mortality ranges from (Fig. 10.7). Sometimes they may grow large
0% to 15% in several surgical series, and surgi- enough to cause symptoms comparable to
cal morbidity, including endocrine dysfunction, those of a craniopharyngioma. The cyst has
visual field abnormalities and chronic hypo- both an intrasellar and suprasellar location in
thalamic dysfunction, varies with preoperative at least 70% of patients, and symptoms are
status. The side effects can be obviated to a usually present for 3 years before the diagno-
substantial degree by planned subtotal resec- sis is established. On MR scan, they may
tion followed by RT. Risk factors for high have either high or low intensity on both T1-
morbidity include lethargy, visual deteriora- and T2-weighted images, depending on cyst
tion, papilledema, hydrocephalus, tumor calci- contents. Some cysts contain intracystic
fication or adhesiveness, adverse intraoperative nodules, hyperintense on T1 and hypointense
events and age < 5 years at presentation.72 on T2.85 Unlike craniopharyngiomas, the
Tumors impinging on the hypothalamus, tumors are not calcified and when small and
patients with hypothalamic disturbance at intrasellar may be confused with a microade-
diagnosis, and attempts to remove adherent noma. They are best treated surgically, often by
tumors from the hypothalamus are likely to a transsphenoidal approach, which usually
have a higher surgical morbidity.69 Others leads to both visual and hormonal recovery
believe that neuropsychological performance and appears to cure the tumor.86 The recur-
and quality of life are not impaired by radical rence rate is low.

314
REFERENCES

Figure 10.7
A Rathke pouch cyst (arrow). The symptoms in this 12-year-old boy were headache and visual loss. The
photomicrograph shows a cyst lined by ciliated epithelium (arrow).

Germinomas References
Germinomas usually occur in the pineal region 1. Anderson JR, Antoun N, Burnet N et al.
in boys, in the suprasellar cistern in girls, and Neurology of the pituitary gland. J Neurol
Neurosurg Psychiatry 1999; 66: 70321.
occasionally within the parenchyma of the 2. Ciric I, Rosenblatt S, Kerr W, Jr et al.
brain, particularly in the thalamus, in either Perspective in pituitary adenomas: an end of the
boys or girls (Chapter 8). When in the suprasel- century review of tumorigenesis, diagnosis, and
lar area, they can produce hormonal or visual treatment. Clin Neurosurg 2000; 47: 99111.
symptoms similar to those of a craniopharyn- 3. Brat DJ, Scheithauer BW, Staugaitis SM et al.
Pituicytoma a distinctive low-grade glioma of
gioma. However, germinomas have a different
the neurohypophysis. Am J Surg Pathol 2000;
radiographic appearance with diffuse contrast 24: 3628.
enhancement and they do not calcify or have 4. Teramoto A, Hirakawa K, Sanno N, Osamura
cysts. Y. Incidental pituitary lesions in 1000

315
PITUITARY AND SELLAR REGION TUMORS

unselected autopsy specimens. Radiology 1994; 17. Nishioka H, Ito H, Haraoka J, Akada K.
193: 1614. Immature teratoma originating from the
5. Sano T, Kovacs KT, Scheithauer BW, Young pituitary gland: Case report. Neurosurgery
WF. Aging and the human pituitary gland. 1999; 44: 6447.
Mayo Clin Proc 1993; 68: 9717. 18. Somali MH, Anastasiou AL, Goulis DG,
6. Tomita T, Gates E. Pituitary adenomas and Polyzoides C, Avramides A. Pituitary abscess
granular cell tumors incidence, cell type, and presenting with cranial nerve paresis. Case
location of tumor in 100 pituitary glands at report and review of literature. J Endocrinol
autopsy. Am J Clin Pathol 1999; 111: 81725. Invest 2001; 24: 4550.
7. Kovacs K, Scheithauer BW, Horvath E, Lloyd 19. Landman RE, Wardlaw SL, McConnell RJ et
RV. The World Health Organization classifi- al. Pituitary lymphoma presenting as fever of
cation of adenohypophysial neoplasms. Cancer unknown origin. J Clin Endocrinol Metab
1996; 78: 50210. 2001; 86: 14706.
8. Blevins LS. Aggressive pituitary tumors. 20. Morita A, Meyer FB, Laws ER, Jr.
Oncology 1998; 12: 130716. Symptomatic pituitary metastases. J Neurosurg
9. Thapar K, Laws ER Jr. Pituitary tumors. In: 1998; 89: 6973.
Kaye AH, Laws ER Jr, eds. Brain tumors: an 21. Honegger J, Fahlbusch R, Bornemann A et al.
encyclopedic approach. New York: Churchill Lymphocytic and granulomatous hypophysitis:
Livingstone, 2001: 80356. Experience with nine cases. Neurosurgery
10. Ohkura N, Kishi M, Tsukada T, Yamaguchi 1997; 40: 71322.
K. Menin, a gene product responsible for 22. Thodou E, Asa SL, Kontogeorgos G et al.
multiple endocrine neoplasia type 1, interacts Clinical case seminar: lymphocytic hypo-
with the putative tumor metastasis suppressor physitis: clinicopathological findings. J Clin
nm23. Biochem Biophys Res Commun 2001; Endocrinol Metab 1995; 80: 230211.
282: 120610. 23. Cooper R, Belilos E, Drexler S et al. Idiopathic
11. Lamberts SW, De Herder WW, Van der Lely giant-cell granulomatous hypophysitis mimick-
AJ. Pituitary insufficiency. Lancet 1998; 352: ing acute meningitis. Am J Med Sci 1999; 318:
12734. 33942.
12. Gsponer J, de Tribolet N, Druaz JP et al. 24. Kristof RA, Van Roost D, Klingmller D,
Diagnosis, treatment, and outcome of pituitary Springer W, Schramm J. Lymphocytic
tumors and other abnormal intrasellar masses hypophysitis: non-invasive diagnosis and treat-
retrospective analysis of 353 patients. ment by high dose methylprednisolone pulse
Medicine 1999; 78: 236-69. therapy? J Neurol Neurosurg Psychiatry 1999;
13. Wiren L, Bengtsson BA, Johannsson G. 67: 398402.
Beneficial effects of long-term GH replacement 25. Jho HD, Alfieri A. Endoscopic transsphenoidal
therapy on quality of life in adults with GH pituitary surgery: various surgical techniques
deficiency. Clin Endocrinol 1998; 48: 61320. and recommended steps for procedural transi-
14. Karanjia N, Jacobson DM. Compression of tion. Br J Neurosurg 2000; 14: 43240.
the prechiasmatic optic nerve produces a 26. Webster J, Piscitelli G, Polli A et al. A compar-
junctional scotoma. Am J Ophthalmol 1999; ison of cabergoline and bromocriptine in the
128: 2568. treatment of hyperprolactinemic amenorrhea.
15. Hanna FWF, Williams OM et al. Pituitary N Engl J Med 1994; 331: 9049.
apoplexy following metastasis of bronchogenic 27. Newman CB, Melmed S, George A et al.
adenocarcinoma to a prolactinoma. Clin Octreotide as primary therapy for acromegaly.
Endocrinol 1999; 51: 37781. J Clin Endocrinol Metab 1998; 83: 303440.
16. Roelandse FW, van der Zwart N, Didden JH, 28. Mindermann T, Staub JJ, Probst A. High rate
van Loon J, Souverijn JH. Detection of CSF of unexpected histology in presumed pituitary
leakage by isoelectric focusing on polyacryl- adenomas. Lancet 1998; 352: 1445.
amide gel, direct immunofixation of transferrins, 29. Peace KA, Orme SM, Padayatty SJ, Godfrey
and silver staining. Clin Chem 1998; 44: 3513. HP, Belchetz PE. Cognitive dysfunction in

316
REFERENCES

patients with pituitary tumour who have been noma: 1H MR spectroscopic findings. Int J
treated with transfrontal or transsphenoidal Radiat Oncol Biol Phys 1999; 45: 699705.
surgery or medication. Clin Endocrinol 1998; 42. Brada M, Burchell L, Ashley S, Traish D. The
49: 3916. incidence of cerebrovascular accidents in
30. Matsuki M, Kaji Y, Matsuo M, Kobashi Y. patients with pituitary adenoma. Int J Radiat
MR findings of subarachnoid dissemination of Oncol Biol Phys 1999; 45: 6938.
a pituitary adenoma. Br J Radiol 2000; 73: 43. Albuquerque FC, Hinton DR, Weiss MH.
7835. Excessively high prolactin level in a patient
31. Webb SM, Rigla M, Wgner A, Oliver B, with a nonprolactin-secreting adenoma case
Bartumeus F. Recovery of hypopituitarism report. J Neurosurg 1998; 89: 10436.
after neurosurgical treatment of pituitary 44. Muratori M, Arosio M, Gambino G et al. Use
adenomas. J Clin Endocrinol Metab 1999; 84: of cabergoline in the long-term treatment of
3696700. hyperprolactinemic and acromegalic patients. J
32. Laws ER, Jr., Vance ML. Conventional radio- Endocrinol Invest 1997; 20: 53746.
therapy for pituitary tumors. Neurosurg Clin 45. Colao A, Di Sarno A, Landi ML et al. Long-
N Am 2000; 11: 61725. term and low-dose treatment with cabergoline
33. Lillehei KO, Kirschman DL, Kleinschmidt- induces macroprolactinoma shrinkage. J Clin
DeMasters BK, Ridgway EC. Reassessment of Endocrinol Metab 1997; 82: 35749.
the role of radiation therapy in the treatment 46. Bevan JS, Adams CBT, Burke CW et al. Factors
of endocrine-inactive pituitary macroadeno- in the outcome of transsphenoidal surgery for
mas. Neurosurgery 1998; 43: 4328. prolactinoma and non-functioning pituitary
34. Simmons NE, Laws ER, Jr. Glioma occurrence tumour, including pre-operative bromocriptine
after sellar irradiation: case report and review. therapy. Clin Endocrinol 1997; 26: 54156.
Neurosurgery 1998; 42: 1728. 47. Tyrrell JB, Lamborn KR, Hannegan LT,
35. Marcou Y, Plowman PN. Stereotactic radio- Applebury CB, Wilson CB. Transsphenoidal
surgery for pituitary adenomas. Trends microsurgical therapy of prolactinomas: Initial
Endocrinol Metab 2000; 11: 1327. outcomes and long-term results. Neurosurgery
36. Izawa M, Hayashi M, Nakaya K et al. Gamma 1999; 44: 25461.
knife radiosurgery for pituitary adenomas. J 48. Guieu R, Dufour H, Grisoli F et al. An ultra-
Neurosurg 2000; 93: 1922. rapid prognostic index in microprolactinoma
37. Levy RP, Schulte RWM, Slater JD, Miller DW, surgery. J Neurosurg 1999; 90: 103741.
Stater JM. Stereotactic radiosurgery the role 49. Cheung AYC, Sligh T, Bauserman S, Schultz G.
of charged particles. Acta Oncol 1999; 38: Evaluation of modern pathologic nomencla-
1659. ture, tumor imaging and treatment of pituitary
38. Colao A, Di Sarno A, Marzullo P et al. New adenomas in a recent surgical series. J
medical approaches in pituitary adenomas. Neurooncol 1998; 37: 14553.
Horm Res 2000; 53, S3: 7687. 50. Abe T, Abou Tara L, Ldecke DK. Growth
39. Cannav S, Curt L, Squadrito S et al. hormone-secreting pituitary adenomas in child-
Cabergoline: a first-choice treatment in hood and adolescence: Features and results of
patients with previously untreated prolactin- transnasal surgery. Neurosurgery 1999; 45: 110.
secreting pituitary adenoma. J Endocrinol 51. Cannav S, Squadrito S, Curt L et al. Results
Invest 1999; 22: 3549. of a two-year treatment with slow release
40. Castro MG, Southgate T, Lowenstein PR. lanreotide in acromegaly. Horm Metab Res
Molecular therapy in a model neuroendocrine 2000; 32: 2249.
disease: developing clinical gene therapy for 52. Kaltsas GA, Isidori AM, Florakis D et al.
pituitary tumours. Trends Endocrinol Metab Predictors of the outcome of surgical treatment
2001; 12: 5864. in acromegaly and the value of the mean
41. Chong VFH, Rumpel H, Aw YS, Ho GL, Fan growth hormone day curve in assessing post-
YF, Chua EJ. Temporal lobe necrosis follow- operative disease activity. J Clin Endocrinol
ing radiation therapy for nasopharyngeal carci- Metab 2001; 86: 164552.

317
PITUITARY AND SELLAR REGION TUMORS

53. Barkan AL, Halasz I, Dornfeld KJ et al. Radiation therapy for pituitary adenoma: treat-
Pituitary irradiation is ineffective in normaliz- ment outcome and prognostic factors. Int J
ing plasma insulin-like growth factor I in Radiat Oncol Biol Phys 1994; 30: 55765.
patients with acromegaly. J Clin Endocrinol 65. Breen P, Flickinger JC, Kondziolka D,
Metab 1997; 82: 318791. Martinez AJ. Radiotherapy for nonfunctional
54. Padayatty SJ, Orme SM, Nelson M, Lamb JT, pituitary adenoma: analysis of long-term tumor
Belchetz PE. Bilateral sequential inferior control. J Neurosurg 1998; 89: 9338.
petrosal sinus sampling with corticotrophin- 66. Paulus W, Honegger J, Keyvani K, Fahlbusch
releasing hormone stimulation in the diagnosis R. Xanthogranuloma of the sellar region: a
of Cushings disease. Eur J Endocrinol 1998; clinicopathological entity different from
139: 1616. adamantinomatous craniopharyngioma. Acta
55. Sanno N, Tahara S, Yoshida Y et al. Ectopic Neuropathol (Berl) 1999; 97: 37782.
corticotroph adenoma in the cavernous sinus: 67. Bunin GR, Surawicz TS, Witman PA et al. The
Case report. Neurosurgery 1999; 45: 9147. descriptive epidemiology of craniopharyn-
56. Watson JC, Shawker TH, Nieman LK et al. gioma. J Neurosurg 1998; 89: 547-51.
Localization of pituitary adenomas by using 68. Vaquero J, Zurita M, De Oya S et al.
intraoperative ultrasound in patients with Expression of vascular permeability factor in
Cushings disease and no demonstrable craniopharyngioma. J Neurosurg 1999; 91:
pituitary tumor on magnetic resonance 8314.
imaging. J Neurosurg 1998; 89: 92732. 69. De Vile CJ, Grant DB, Kendall BE et al.
57. Semple PL, Laws ER, Jr. Complications in a Management of childhood craniopharyngioma:
contemporary series of patients who under- can the morbidity of radical surgery be
went transsphenoidal surgery for Cushings predicted? J Neurosurg 1996; 85: 7381.
disease. J Neurosurg 1999; 91: 1759. 70. Aydin Y, Can SM, Gulkilik A et al. Rapid
58. Estrada J, Boronat M, Mielgo M et al. The long- enlargement and recurrence of a preexisting
term outcome of pituitary irradiation after unsuc- intrasellar craniopharyngioma during the
cessful transsphenoidal surgery in Cushings course of two pregnancies Case report. J
disease. N Engl J Med 1997; 336: 1727. Neurosurg 1999; 91: 3224.
59. Kemink SAG, Wesseling P, Pieters GFFM et al. 71. Kearney D, Tay-Kearney ML, Khangure MS.
Progression of a Nelsons adenoma to pituitary Craniopharyngioma and the moustache sign.
carcinoma; a case report and review of the Australas Radiol 1993; 37: 3701.
literature. J Endocrinol Invest 1999; 22: 705. 72. Duff JM, Meyer FB, Ilstrup DM et al. Long-term
60. Bertholon-Grgoire M, Trouillas J, Guigard MP, outcomes for surgically resected craniopharyn-
Loras B, Tourniaire J. Mono- and plurihor- giomas. Neurosurgery 2000; 46: 291302.
monal thyrotropic pituitary adenomas: patho- 73. Ito M, Jamshidi J, Yamanaka K. Does cranio-
logical, hormonal and clinical studies in 12 pharyngioma metastasize? Case report and
patients. Eur J Endocrinol 1999; 140: 51927. review of the literature. Neurosurgery 2001;
61. Skoric T, Korsic M, Zarkovic K et al. Clinical 48: 9335.
and morphological features of undifferentiated 74. Yasargil MG, Curcic M, Kis M et al. Total
monomorphous GH/TSH-secreting pituitary removal of craniopharyngiomas. Approaches
adenoma. Eur J Endocrinol 1999; 140: 52837. and long-term results in 144 patients. J
62. Young WF Jr, Scheithauer BW, Kovacs KT et Neurosurg 1990; 73: 311.
al. Gonadotroph adenoma of the pituitary 75. Abe T, Ldecke DK. Transnasal surgery for
gland: a clinicopathologic analysis of 100 infradiaphragmatic craniopharyngiomas in
cases. Mayo Clin Proc 1996; 71: 64956. pediatric patients. Neurosurgery 1999; 44:
63. Gittoes NJ, Bates AS, Tse W et al. 95764.
Radiotherapy for non-functional pituitary 76. Honegger J, Buchfelder M, Fahlbusch R.
tumours. Clin Endocrinol 1998; 48: 3317. Surgical treatment of craniopharyngiomas:
64. Tsang RW, Brierley JD, Panzarella T, endocrinological results. J Neurosurg 1999; 90:
Gospodarowicz MK, Sutcliffe SB, Simpson WJ. 2517.

318
REFERENCES

77. Chung WY, Pan DH, Shiau CY, Guo WY, 82. Caldarelli M, Di Rocco C, Papacci F, Colosimo
Wang LW. Gamma knife radiosurgery for CJ. Management of recurrent craniopharyn-
craniopharyngiomas. J Neurosurg 2000; 93, gioma. Acta Neurochir (Wien) 1998; 140:
S3: 47-56. 44754.
78. Pollock BE, Lunsford LD, Kondziolka D, Levine 83. Honegger J, Barocka A, Sadri B, Fahlbusch R.
G, Flickinger JC. Phosphorus-32 intracavitary Neuropsychological results of craniopharyn-
irradiation of cystic craniopharyngiomas: gioma surgery in adults: a prospective study.
current technique and long-term results. Int J Surg Neurol 1998; 50: 1928.
Radiat Oncol Biol Phys 1995; 33: 43746. 84. Rajan B, Ashley S, Thomas DG et al.
79. Savas A, Arasil E, Batay F, Selcuki M, Craniopharyngioma: improving outcome by
Kanpolat Y. Intracavitary chemotherapy of early recognition and treatment of acute
polycystic craniopharyngioma with bleomycin. complications. Int J Radiat Oncol Biol Phys
Acta Neurochir (Wien) 1999; 141: 5478. 1997; 37: 51721.
80. Savas A, Erdem A, Tun K, Kanpolat Y. Fatal toxic 85. Byun WM, Kim OL, Kim D. MR imaging
effect of bleomycin on brain tissue after intracystic findings of Rathkes cleft cysts: significance of
chemotherapy for a craniopharyngioma: case intracystic nodules. Am J Neuroradiol 2000;
report. Neurosurgery 2000; 46: 2136. 21: 4858.
81. Bulow B, Attewell R, Hagmar L et al. 86. el-Mahdy W, Powell M. Transsphenoidal
Postoperative prognosis in craniopharyngioma management of 28 symptomatic Rathkes cleft
with respect to cardiovascular mortality, cysts, with special reference to visual and
survival, and tumor recurrence. J Clin hormonal recovery. Neurosurgery 1998; 42:
Endocrinol Metab 1998; 83: 3897904. 716.

319
11
Primary central nervous system lymphoma and other
hemopoietic tumors

Introduction dural masses (e.g. plasmacytoma and


Hodgkins disease)1 or diffusely seeding the
Leukemias, lymphomas, Hodgkins disease and leptomeninges (e.g. leukemia and lymphoma).
other tumors of hemopoietic origin, when Occasionally, hemopoietic tumors arise within
found in the CNS, usually represent metastases the CNS (Fig. 11.1), with no systemic tumor
of tumor that started systemically. The metas- being identified either at initial workup or, in
tases mostly affect the meninges, causing either most cases, even at autopsy.2,3 The most

(a) (b)

C
A

Figure 11.1
(a) The typical location of primary CNS lymphoma and of some other hemopoietic tumors. Primary CNS
lymphomas tend to occur deep in the brain, usually near the ventricles (A,D). Plasmacytomas and other
hemopoietic tumors are usually durally based (B). All of these lesions may involve the leptomeninges (C).
(b) Histiocytic lesions, especially Langerhans cell histiocytosis, may involve either the hypothalamic pituitary
axis or brain parenchyma.

320
PRIMARY CNS LYMPHOMA

Table 11.1 encountered at the rate of approximately 1


Primary CNS lymphomas and hemopoietic neoplasms. every 24 years. Beginning in the mid-1980s,
the incidence at MSKCC appeared to increase
Malignant lymphomas to the point where approximately 15% of the
B-cell lymphomas primary brain tumors encountered in immuno-
T-cell lymphomas competent hosts were PCNSL.4 Although a
Intravascular lymphoma
few studies question whether an increase in
Plasmacytomas
Granulocytic sarcomas PCNSLs is real,5 most recognize a dramatic
Histiocytic tumors epidemiologic change over the past 2030
Langerhans cell histiocytosis years.6,7 A substantial rise has also been noted
Non-Langerhans cell histiocytosis in ocular lymphoma, a part of the spectrum of
Hemophagocytic lymphohistiocytosis CNS lymphoma.
PCNSL occurs in two settings, the first in
patients with known immune suppression and
the second in immunocompetent patients.8 The
common hemopoietic tumor to arise within the
former are usually children or young adults,
CNS is the primary central nervous system
and the latter older adults and the elderly. Only
lymphoma (PCNSL) (Table 11.1).
PCNSL arising in the immunocompetent host
is discussed in this chapter. Current estimates
Primary CNS lymphoma indicate that 67% of primary intracranial
neoplasms are lymphomas, with an incidence
Introduction
of 0.30.4 per 100 000 in the immunocompe-
Most textbooks indicate that PCNSLs repre- tent population and 4.7 per 1000 person-years
sents 12% of primary brain tumors. Indeed, in the AIDS population about 3600-fold
20 years ago at MSKCC such tumors were higher than in the general population.6,9 In the

Figure 11.2
A patient with a
typical primary CNS
lymphoma. Note the
uniform enhancement,
the paucity of edema,
the fuzzy tumor
margins and the
periventricular
location of these
multiple lesions. The
photomicrograph
shows infiltration of
neoplastic large B-
cells around a blood
vessel.

321
PRIMARY CENTRAL NERVOUS SYSTEM LYMPHOMA AND OTHER HEMOPOIETIC TUMORS

immunocompetent population, the peak disorder is increased in patients with Sjogrens


incidence is between 50 and 70 years,10 with syndrome and rheumatoid arthritis, indepen-
males slightly more likely to be involved than dent of therapy. Other established or putative
females. risk factors include exposure to certain pesti-
PCNSL can arise anywhere in the nervous cides or hair dyes, infection with the HTLV-1
system, including the eyes, spinal cord and virus or EpsteinBarr virus (EBV) (see below)
leptomeninges, but it primarily involves the and old age, possibly a result of decreased
periventricular areas of the brain and brain- immunity with aging. One kind of gastric
stem (Fig. 11.2). About 40% of PCNSLs are lymphoma has been associated with chronic
multiple in the immunocompetent population, infection by Helicobacter pylori. An important
whereas 5070% of immunocompromised risk factor is a history of a previous cancer. A
patients have multiple PCNSL lesions. Twenty previous or concomitant malignant neoplasm
percent of patients have ocular involvement at has been found in up to 15% of so-called
presentation and about 42% of patients have immunocompetent patients with PCNSL.3
demonstrable leptomeningeal involvement at Because of the relatively small number of
diagnosis.11,12 At least an additional 40% of PCNSLs, the environmental factors that play a
patients with PCNSL have an increased role in this disorder have not been established.
number of lymphocytes in the cerebrospinal A casecontrol study of PCNSL has suggested
fluid (CSF), which cannot be identified on that lower education was associated with the
cytologic examinations as malignant, but disease and that PCNSL patients were less
strongly suggest that leptomeningeal disease is likely to have had tonsillectomy or use oral
present. In some patients, focal leptomeningeal contraceptives.13 However, these findings are
infiltration is seen on a surgical specimen even preliminary and need to be confirmed.
with negative cytology on CSF examination, The most important risk factor for PCNSL is
indicating that the incidence of subarachnoid immunosuppression. As many as 1040% of
involvement may be underestimated by CSF patients infected with human immunodeficiency
analysis alone. virus type 1 (HIV-1) have PCNSL at autopsy,
although at least half are missed prior to
death.14,15 In the early days of transplantation,
Etiology PCNSL was a common complication of renal
PCNSL is a typical non-Hodgkins lymphoma. and other organ transplants,16 a result of the
Non-Hodgkins lymphoma has increased in massive immunosuppression necessary to
incidence about 34% per year since the maintain the transplant. Fully half of the
1970s, with the greatest increase seen in extra- lymphomas arising in the post-transplantation
nodal lymphomas, particularly gastric and period arose in the brain, an extraordinarily
CNS. This increase cannot be accounted for by high proportion, given that < 1% of all
the number of AIDS patients who develop lymphomas arise in brain in the immunocom-
lymphoma, even though there is a marked petent population. PCNSL is also known to
overrepresentation of extranodal tumors in the accompany congenital or other acquired immuno-
AIDS population. Among the possible risk deficiency states, such as the WiskottAldrich
factors for the development of non-Hodgkins syndrome and collagenvascular diseases. In
lymphoma in general are immunosuppression most immunosuppressed states, PCNSL appears
including autoimmunity: the incidence of the to be related to latent EBV infection.16 After

322
PRIMARY CNS LYMPHOMA

primary EBV infection, which usually occurs in of an inflammatory process and become trans-
youth, a small number of B-cells retain a latent formed into malignant cells within the nervous
infection, which immortalizes this tiny popula- system. Unfortunately, neither hypothesis is
tion of cells. Growth of these cells is kept under truly satisfying. Against the first is the failure
control by normal suppressor T-cells. Removal to find deposits of lymphoma in other
of T-cell control by immunosuppression allows immunologically privileged sites, such as the
these B-cells to grow, leading to lymphoma testis, in patients with PCNSL. Against the
formation. Viral genome can be found within second hypothesis is the fact that there appears
tumor tissue in 85100% of patients with AIDS- to be no increased incidence of PCNSL in
related PCNSL.17,18 On the contrary, in PCNSL patients with inflammatory diseases of the
arising in immunocompetent hosts, viral genome nervous system that attract lymphocytes,
is rarely found within the tumor. such as multiple sclerosis or encephalitis.
Approximately 15% of immunocompetent Furthermore, almost exclusively, T-cells are
patients with PCNSL have had a prior malig- associated with inflammatory diseases, and
nancy.3 Many of these cancers occurred PCNSL is usually a disease of B-lymphocytes.
decades previously and the patient is cured of Although only a few PCNSLs have been
their original primary. There is no specific genetically analyzed, the genetics do not
tumor type which pre-dates the development of differ from the genetics of non-Hodgkins
PCNSL; a variety of solid and hemopoietic lymphoma occurring elsewhere in the body.19
tumors have been observed. Furthermore, Chromosomal abnormalities with gains of
treatment of the original tumor is not usually chromosomes 12q, 18q and 22q and loss of 6q
of the sort which predisposes to second malig- have been described.20,21 An analysis of cell
nancy formation; for example, abdominal surface markers, such as ICAM and integrins,
resection of a colon cancer without alkylating fails to demonstrate any differences between
agent chemotherapy would not be expected to systemic and cerebral lymphomas.22 Telo-
lead to PCNSL years later. Whether the history merase activity and telomerase-related RNA
of prior cancer reflects an underlying genetic are present in almost all PCNSLs.23
abnormality is unknown.
Why hemopoietic tumors should arise within
Pathology
the nervous system is unknown, given the fact
that the CNS is devoid of lymph nodes and Macroscopically, lymphomas tend to have a
deficient in lymphatics (see Chapter 2). Two fleshy, granular appearance with ill-defined
hypotheses have been advanced. The first is borders; hemorrhage and necrosis are rare
that a hemopoietic tumor develops outside of except in AIDS patients, where they are seen
the CNS. The tumor cells circulate in the blood commonly. Microscopically, the tumors gener-
and seed multiple organs, including the brain. ally exhibit an angiocentric growth pattern with
The immune system has the capacity to find multiple areas of perivascular lymphocytes,
and eliminate tumor cells in most of the body, which eventually become confluent, producing
but the brain, being a relatively immuno- a mass. The tumor may contain a prominent
logically privileged site, gives sanctuary to astrocytic and microglial response and a large
lymphoma cells which grow and form a tumor number of reactive lymphocytes (T-cells) are
in that organ. A second hypothesis suggests common even in the B-cell lymphomas.24
that lymphocytes traffic into the CNS as part Microscopically, PCNSL is indistinguishable

323
PRIMARY CENTRAL NERVOUS SYSTEM LYMPHOMA AND OTHER HEMOPOIETIC TUMORS

from high-grade non-Hodgkins lymphomas findings include headache, personality and


occurring elsewhere in the body. They are cognitive changes, and progressive hemiparesis.
identified both by their microscopic appearance The symptoms usually progress over weeks or
and by their monoclonality, most frequently months, more like glioblastoma multiforme
IgM kappa. PCNSLs are virtually all inter- than lower-grade gliomas. The median time
mediate or high-grade subtypes, with approxi- from symptom onset to diagnosis is 35
mately half of tumors having a diffuse large cell months in immunocompetent patients, and
histology, with diffuse large cell immunoblastic longer when personality change is the only
subtype forming 20%, diffuse small cleaved cell symptom.27 Leptomeningeal involvement is
10%, and others in lesser quantities. In AIDS usually asymptomatic until late in the course of
patients, there is a much higher incidence of the the disease.
diffuse large cell immunoblastic subtype. It is Because ocular involvement is common,28
unknown whether the subtypes carry different patients may present with visual symptoms.
prognoses, because there have been too few Usually the patient complains of floaters or
patients in most categories to analyze. T-cell visual blurring. A misdiagnosis of uveitis is
lymphomas represent about 2% of all PCNSLs;25 common, particularly in those patients who
one Japanese series reports that 8.5% of 466 have isolated ocular disease as the first
patients with PCNSL had T-cell lymphoma.2 manifestation of PCNSL. The ocular findings
The vasculature of PCNSL differs from that on slit-lamp exam resemble vitreitis or uveitis
of gliomas. Like gliomas, the tumors are highly and the patient may be so treated, often
vascular, but endothelial proliferation is absent. initially successfully because of the response of
Electron microscopy reveals two distinct lymphoma to corticosteroids. However, such
populations of cells: electron-dense endothelial patients ultimately become refractory to corti-
cells that give evidence of apoptosis, and costeroid therapy, often precipitating consider-
electron-lucent cells that give evidence of cellu- ation of lymphoma as the diagnosis. The
lar regeneration. These findings are not present diagnosis may then be difficult to establish
in the endothelial cells of gliomas. Investigators because of the chronic steroid treatment (see
interpret these changes to represent apoptosis below). In about half of the patients, ocular
induced by cytokines released from necrotic involvement is asymptomatic and detectable
or apoptotic tumor lymphocytes. If so, the only on slit-lamp examination.
findings may partially explain so-called ghost
tumors, PCNSLs that regress in response to Imaging
corticosteroids (see below).26 MR is far superior to CT scan for delineat-
ing the extent of PCNSL involvement;
however, the contrast-enhanced images of
Clinical findings
either modality reveal similar characteristic
Symptoms and signs findings. The tumors are usually supratento-
As indicated in Chapter 3, the signs and rial and periventricular (Fig. 11.2). They are
symptoms depend on the tumor site. Because hypointense on the MR T1-weighted image
the tumors are often deep, seizures are less and hypo- to isointense relative to gray
common than in other primary brain tumors matter on T2-weighted images with variable
but behavioral changes and focal signs are surrounding edema.29 The T2 signal reflecting
more common than in gliomas.27 Common dense cellularity helps to differentiate

324
PRIMARY CNS LYMPHOMA

lymphomas from gliomas that are hyper- Other diagnostic tests


intense on T2. Lymphomas enhance homoge-
neously after contrast administration and Unlike most intracranial tumors, where cranial
have rather indistinct borders. Ring enhance- imaging will suffice to reveal the full extent of
ment is uncommon. Even more than gliomas, disease, patients with PCNSL should undergo
lymphomas may widely infiltrate the nervous a neurologic staging evaluation including an
system without causing a change in signal ocular examination and slit-lamp, and a
on MR scan. When patients are treated lumbar puncture for CSF analysis (Table 11.2).
with corticosteroids, PCNSL will sometimes If a lumbar puncture is performed before
show a rapid complete disappearance of biopsy in a patient suspected of having a
contrast enhancement. This response has PCNSL, a diagnosis of lymphoma can be made
been called a ghost tumor and may result in up to 15% of patients (see below). In
from cytokine-induced apoptosis as seen in patients whose cranial MRI suggests PCNSL, if
PCNSL endothelial cells, and a direct the ocular examination shows inflammatory
response of lymphoma cells that contain infiltrates in the vitreous, a vitrectomy may be
corticosteroid receptors.26 performed and the material examined for
Hypointense lesions, or T2 hyperintense malignant lymphocytes. If lymphoma is identi-
lesions that do not contrast enhance on MR fied, a brain biopsy can be avoided. However,
scan, are occasionally found in both immuno- if a patient has biopsy-proven PCNSL and cells
competent and immunosuppressed patients, are identified on slit-lamp examination in the
but they are more common among the vitreous, this is presumptive evidence of
immunosuppressed population.16,30 Ring- lymphoma involving the eye, and a vitrectomy
enhancing lesions similar to those of toxoplas- is not essential for pathologic confirmation.
mosis are characteristic of PCNSL in the AIDS When trying to establish the diagnosis of
patient, making the differential diagnosis diffi- PCNSL, the presence of lymphocytes in a CSF
cult, whereas ring enhancement is uncommon or vitreous sample is not always sufficient to
in the immunocompetent population. In distinguish inflammatory infiltrates from
addition, the lesions are more frequently lymphoma. In both CSF and vitreous specimens,
located in the subcortical white matter in immunohistochemical staining may establish the
AIDS patients. Perfusion MRI suggests that diagnosis; if a substantial percentage of the cells
most PCNSLs have a low cerebral blood
volume, similar to lower-grade gliomas and in
contrast to high-grade gliomas.29 PET scanning Table 11.2
Diagnostic evaluation of PCNSL patients.
reveals a hypermetabolic lesion but this is
non-specific and not helpful in the immuno-
competent population; however it may help 1. Cranial MRI
distinguish tumor from toxoplasmosis (hypo- 2. Spinal MRI (enhanced) if spinal symptoms
are present
metabolic) in AIDS patients.31 Single-photon
3. Lumbar puncture
emission tomography may help distinguish 4. Ophthalmologic examination including slit
PCNSL from other lesions.32 In PCNSL lamp
uptake of 123IMP is delayed, whereas it occurs 5. Bone marrow
early in other brain tumors. MRS may also 6. Abdominal-pelvic CT scan
help.33

325
PRIMARY CENTRAL NERVOUS SYSTEM LYMPHOMA AND OTHER HEMOPOIETIC TUMORS

are B-cells, lymphoma is strongly suggested. If underway. When CNS metastases are present at
the cells demonstrate a monoclonal population, initial diagnosis of systemic lymphoma, the
this confirms the malignant nature of the systemic manifestations are usually obvious
lymphocytes even when they appear histologi- clinically. It is extremely rare to have systemic
cally benign. Like many systemic non-Hodgkins non-Hodgkins lymphoma present as an
lymphomas, PCNSL may be accompanied by intracranial mass. Thus, an extensive systemic
an inflammatory infiltrate composed of T-cells. work-up in a patient believed to have PCNSL
These reactive cells can infiltrate tumor tissue in is usually unrewarding. However, an occasional
the brain and can also accompany malignant patient presents with what appears to be
lymphocytes in CSF or the vitreous complicat- PCNSL and is discovered subsequently to have
ing the cytologic diagnosis. systemic lymphoma.34 Because the yield is so
A strong clinical suspicion of PCNSL on low, not all investigators believe a systemic
imaging, or by examination of the vitreous, or workup is necessary in patients with presumed
spinal fluid, should lead one to consider a PCNSL. However, we suggest a limited workup
stereotactic needle biopsy rather than an open to include CT of abdomen and pelvis and bone
craniotomy with attempt at surgical removal. marrow examination for all patients with
As indicated below, corticosteroids should be PCNSL.
withheld until just before or immediately after
the needle biopsy is performed. Optimally CSF,
Differential diagnosis
and, if appropriate, a vitreous specimen should
be obtained prior to the start of corticosteroids Table 11.3 lists the differential diagnosis of
as well. PCNSL in immunocompetent and immunosup-
PCNSL rarely metastasizes outside the pressed patients. In immunocompetent
nervous system, although, as indicated above, patients, lymphoma must be distinguished from
it may spread widely within the nervous other CNS tumors, particularly gliomas and
system, including the eyes, leptomeninges and metastases. As indicated above, clinical
rarely spinal cord. Metastasis of systemic findings and imaging usually suggest the
lymphoma to the CNS usually occurs late in diagnosis, but only biopsy will unequivocally
the course of the disease, after the diagnosis of establish the diagnosis and dictate treatment.
systemic lymphoma is evident and treatment is Rarely, lymphoma may either mimic a benign

Table 11.3
Immunocompetent patients Immunosuppressed patients Differential diagnosis of
PCNSL.
Glioma Toxoplasmosis
Metastases Fungal infection
Other hemopoietic tumors Brain abscess(es)
Schwannoma35 Pituitary adenoma36
Multiple sclerosis Progressive multifocal leukoencephalopathy
Sarcoid Glioma
Inflammatory pseudotumor37,38 Metastases

326
PRIMARY CNS LYMPHOMA

brain tumor35 or even develop within a benign accompanying their PCNSL, the tumor cells
intracranial tumor.36 Other hemopoietic may be gone and the reactive T-lymphocytes
tumors, as indicated later in this chapter, may persist, giving the false impression that this
mimic non-Hodgkins lymphoma, and discrim- was an inflammatory process and not a
inating between these entities may be difficult neoplasm. Because the differential diagnosis
even with biopsy. Especially important is includes a variety of inflammatory disorders
distinguishing non-neoplastic lesions from of the nervous system that can also be
PCNSL. Such lesions include multiple sclerosis, suppressed by corticosteroids (e.g. multiple
sarcoid and inflammatory pseudotumors. sclerosis), the clinician cannot tell whether
Inflammatory pseudotumor or plasma cell the original lesion was tumor or inflamma-
granuloma may be difficult to distinguish from tion. However, corticosteroids are not
lymphoma histologically; genetic studies may curative and the patient usually relapses
be required to demonstrate a clonal origin of within a matter of months, often with more
the plasma or B-cells or identify gene aggressive disease, although we have seen
rearrangements, indicating that the lesion is occasional long-term remissions. One of our
neoplastic rather than inflammatory. patients went 7 years while being treated
In immunosuppressed patients, opportunistic with two courses of corticosteroids alone
infections such as toxoplasmosis, aspergillosis, before developing a recurrence, which proved
progressive multifocal leukoencephalopathy to be PCNSL on biopsy. Spontaneous remis-
and bacterial brain abscesses may mimic sions lasting as long as 4 years have also been
lymphoma. Gliomas, metastases and other reported,40 perhaps representing the ability of
lesions that also occur in immunocompetent the patients immune system to partially
patients may occasionally mimic lymphoma. control the tumor.

Surgery
Treatment The correct surgical approach to PCNSL is a
Corticosteroids stereotactic needle biopsy. Because PCNSL
Unlike their use in other brain tumors, where infiltrates so widely and is usually in a deep
they control edema, corticosteroids also location, surgery does not increase survival and
function as oncolytic agents for PCNSL. often results in worse neurologic deficits. If
Steroid-induced apoptosis of lymphoma cells PCNSL was not suspected prior to craniotomy
does not require wild-type p53 activity or and a diagnosis is made on frozen section,
caspase activation, but the apoptosis is inhib- further resection should be abandoned and the
ited by the bcl-2 oncogene.39 In about 60% patient treated as indicated below. Surgical
of patients treated with corticosteroids alone, resection should be reserved for only those
there is substantial regression of the tumor patients whose tumor mass is producing herni-
within days to the point where the tumor ation leading to imminent death. Unlike most
may completely disappear. A needle biopsy other primary brain tumors, extirpation is an
performed after several days of corticosteroid unfavorable prognostic factor, perhaps because
treatment may reveal either normal brain these patients are in poor neurologic condition
tissue or evidence of necrosis without preoperatively.10 Hemorrhage, sometimes fatal,
lymphocytes being present. In patients who may complicate biopsy in more than 10% of
have a prominent reactive lymphocytosis immunosuppressed patients.16

327
PRIMARY CENTRAL NERVOUS SYSTEM LYMPHOMA AND OTHER HEMOPOIETIC TUMORS

Radiation therapy The eyes are not included in the radiation


For reasons that are unclear, radiation therapy portal unless ocular lymphoma is present, in
(RT) that usually provides excellent local which case both eyes are radiated to a total of
control in systemic non-Hodgkins lymphoma, 3640 Gy. RT is effective in controlling ocular
does not in PCNSL. Whereas non-CNS diffuse lymphoma, although ocular relapse can occur
large cell lymphoma has a 7590% local after RT. In addition, up to 80% of patients
control rate in Stage I, the local control rate who present with isolated ocular lymphoma
is only 39% in the brain, and most recur- eventually develop cerebral involvement if
rences appear in regions of the brain that have followed for up to 10 years, and most develop
received 60 Gy of irradiation.41 A complete CNS disease within 3 years of their ocular
response to RT has been reported in most diagnosis. Because there is occult involvement
patients with PCNSL, over 80% in one of the leptomeninges in virtually all PCNSL
series.42 However, relapse is generally early patients, craniospinal RT has been used in a
and the median survival is only 1218 small number of patients.45 However, neuraxis
months, with fewer than 5% of patients RT did not improve survival over whole-brain
surviving 5 years after RT. Whole-brain RT is RT alone and it is associated with significant
the treatment of choice, because of the morbidity, particularly myelosuppression,
widespread nature of PCNSL, even in those which can compromise the subsequent admin-
patients who have a single lesion on MRI. A istration of chemotherapy. Radiation toxicity,
number of RT protocols have been proposed. especially dementia, is common in those who
The Radiation Therapy Oncology Group are over 60 (Fig. 11.3).46 Leukoencephalopathy
(RTOG) conducted a prospective study using on MRI without evident dementia is frequent
40-Gy whole-brain RT plus a 20-Gy boost to in younger patients (Fig. 11.4).
the tumor site. Median survival was only 12.2
months.43 Age and Karnofsky performance Chemotherapy
status (KPS) were important prognostic The initial treatment of choice for PCNSL is
factors. Patients over 60 had a median chemotherapy.47,48 Whether RT is required after
survival of 7.6 months, compared to 23.1 successful chemotherapy is not established. The
months for those younger than 60. Those standard chemotherapeutic approach to the treat-
with a KPS * 70 had a median survival of ment of systemic non-Hodgkins lymphoma,
21.1 months versus 5.6 months for those with cyclophosphamide, doxorubicin, vincristine and
a KPS of 4060. Increasing the dose to 60 Gy prednisone (CHOP), is not successful in treating
in the boosted field did not improve survival PCNSL.4951 Complete remissions are achieved
or local control. We have also found that, in only a few patients, and early relapse is
despite a whole-brain dose of 40 Gy and a common. The reported median survival is only
14.40-Gy boost, recurrences developed with 9.516 months when CHOP is combined with
equal frequency both within and outside the whole-brain RT, no improvement over that seen
boosted area.44 Therefore, there was no added with RT alone. These agents, which effectively
benefit with dose escalation beyond 40 treat systemic lymphoma, prednisone excepted,
50 Gy. Our approach now is to use 45-Gy cross the bloodbrain barrier poorly. Because
whole-brain RT without a boost, recognizing PCNSL widely and microscopically infiltrates
that cerebral, leptomeningeal or even spinal the brain, infiltrative disease in every patient is
recurrences are possible. protected by the bloodbrain barrier. Although

328
PRIMARY CNS LYMPHOMA

Figure 11.3
Complications of radiation therapy for the treatment of primary CNS lymphoma. This 84-year-old woman
developed a lesion deep in the left parietal lobe in December 1996. The lesion responded completely to 50-Gy
whole-brain radiation and did not recur. However, as scans in December 1996 (left), April 1997 (middle), and
September 1998 (right) show, there is progressive hyperintensity in the white matter associated with loss of
normal cognitive function.

Figure 11.4
Younger people
tolerate radiation
better than older
people. This scan
is of a 35-year-old
man 8 years post-
radiation and
chemotherapy for
primary CNS
lymphoma. The
tumor has not
recurred. There is
a great deal of
white matter
hyperintensity but
he functions at a
very high level
without evident
cognitive difficulty.

329
PRIMARY CENTRAL NERVOUS SYSTEM LYMPHOMA AND OTHER HEMOPOIETIC TUMORS

quantity to treat the tumor. However, if high


doses (> 1 g/m2) are used, adequate concentra-
tion of drug reach and kill tumor cells protected
by the bloodbrain barrier. The rest of the body
is protected by leucovorin, a folate analog that
reverses the effect of methotrexate but does not
reach the CNS. Accordingly, most protocols
now call for high-dose methotrexate with
leucovorin rescue, sometimes in association
with other drugs given in high enough doses to
cross the bloodbrain barrier, e.g. cytarabine, or
lipid-soluble agents such as procarbazine. Rapid
infusion of methotrexate over 3 h, rather than
the conventional 624 h, significantly increases
levels of the drug in CSF and presumably
brain.54 Some investigators have used intra-
arterial hyperosmolar agents to open the
bloodbrain barrier, followed by intra-arterial
methotrexate,55 but there is no evidence that
this technique is better than forcing water-
soluble agents across the bloodbrain barrier by
using high intravenous doses. Moreover, the
intra-arterial method is associated with unique
acute toxicities including seizures, strokes,
Figure 11.5
arterial dissections and cerebral swelling.56
Response of primary CNS lymphoma to
methotrexate. The left scan show the typical lesion Several protocols based on high-dose metho-
prior to treatment. The right scans are after two trexate have reported complete remission rates
courses of high-dose methotrexate. The mass has of 8090% and event-free survival rates of
reduced (arrow), and ventricular compression and greater than 5 years. Our long-term follow-up
shift has returned to normal. reveals a 22% 5-year survival rate with some
patients 12 years from diagnosis; these patients
are probably cured of their PCNSL.57
enhancing lesions may respond to CHOP Age and performance status are important
because of focal disruption of the barrier, micro- prognostic factors.58 Not only do patients over
scopic tumor, which is protected by an intact 60 years old have a poor prognosis, regardless
bloodbrain barrier, continues to grow. When of treatment, but they are also more likely to
drugs which cross the bloodbrain barrier and suffer neurotoxicity, usually dementia, from
are effective against lymphoma are used the combined modality therapy if they survive more
outcome is much better. than a year.57 Accordingly, several protocols
High-dose methotrexate is recognized as the call for treatment with chemotherapy alone in
single most active agent against PCNSL (Fig. patients over 60, reserving RT for relapse.
11.5).52,53 Methotrexate in standard doses does Although overall survival for patients treated in
not cross the bloodbrain barrier in sufficient this fashion is comparable to those treated with

330
PRIMARY CNS LYMPHOMA

up-front chemotherapy and RT, CNS relapse younger, and is much poorer in patients who
occurs earlier after chemotherapy alone.47,59 are immunocompromised than in those who
Several other chemotherapy regimens, some of are immunocompetent. Patients in poor condi-
which incorporate high-dose methotrexate, have tion at diagnosis also fare worse than those in
been tried but none has been shown to be good condition. Telomerase activity predicts a
superior.60,61 In some relapsed patients who shorter interval to relapse and shorter
achieve a complete remission from induction survival.23
chemotherapy, high-dose chemotherapy with Median survival is only 12 months with
bone marrow or stem cell rescue without cranial supportive care alone. With surgery alone, the
RT is being tried as experimental treatment.62 A median survival is about 3 months, little longer
major effort in the design of new regimens is not than with no treatment at all (Table 11.4).
only to improve efficacy but also to reduce the Patients treated with RT alone have a
risk of delayed cognitive impairment. median survival of about 1218 months with
Salvage therapy, given when patients fail relapse usually occurring 1214 months after
first-line treatment, significantly improves treatment. The high-dose methotrexate-based
survival and probably improves quality of life. chemotherapeutic regimens yield an overall
Injection of methotrexate and thiotepa directly survival of about 4060 months. Some patients
into the vitreous can successfully treat recur- may be secondarily salvaged by additional
rent ocular lymphoma.63 Biological treatment chemotherapy or, if they have not received it,
of non-CNS lymphomas is becoming increas- RT. Approximately 50% of patients can
ingly successful.64 Such approaches include the achieve a second remission with salvage treat-
anti-CD-20 monoclonal antibody (Rutuximab), ment and 25% have prolonged survival for
radioimmune conjugates and patient-specific many years after reinduction.
vaccination. Preliminary evidence with Rutux- Late neurotoxicity can limit survival and
imab suggests that it may have activity against quality of life for some patients after success-
PCNSL.65 Whether these techniques will prove ful treatment of their PCNSL. Progressive
useful in PCNSL is unknown. memory impairment, ataxia and eventually
urinary incontinence are the major clinical
manifestations of leukoencephalopathy. While
Prognosis
elderly patients are particularly vulnerable,
Regardless of treatment, the prognosis is worse even young patients can occasionally develop
in patients over 60 than in those who are this irreversible complication. The symptoms

Table 11.4
Treatment Median survival (months) Treatment and median
survival rates.
Surgery alone 34
Whole-brain RT 1218
CHOP + Whole-brain RT 9.516
HD-MTXa + Whole-brain RT 4060

aHD-MTX = high-dose methotrexate

331
PRIMARY CENTRAL NERVOUS SYSTEM LYMPHOMA AND OTHER HEMOPOIETIC TUMORS

often appear months after completion of treat- isolated site of disease, particularly with
ment. There is no good treatment for delayed plasmacytomas or granulocytic sarcomas.
neurotoxicity although ventriculoperitoneal Neurologic symptoms and signs are related to
shunt can sometimes ameliorate symptoms, tumor location. These tumors often invade or
especially ataxia and urinary incontinence.66 compress cerebral cortex. Therefore, seizures
The main objective is to prevent the sequelae and lateralizing signs, such as hemiparesis
of therapy by designing effective but less toxic and confusion, are common at presentation.
regimens. Neuroimaging, either CT or MRI, typically
reveals a dural-based, prominently enhancing
mass. It may be difficult to distinguish such
lesions from a meningioma except that signifi-
Other hemopoietic tumors cant edema of the underlying brain is more
Plasmacytomas67,68 (Fig. 11.6), Hodgkins common with hemopoietic tumors. A dural tail
disease69,70 and granulocytic sarcomas71 may also be evident with these lymphoid
(myelogenous leukemia chloroma) occasionally tumors. CT usually reveals any accompanying
arise in the CNS. Most are dural-based, underlying bone destruction better than MRI.
although a few are parenchymal.72,73 When Magnetic resonance venography (MRV) should
these tumors develop in the absence of systemic be performed for any lesion located close to the
disease, they are difficult to distinguish from superior sagittal sinus to assess its patency.
non-hemopoietic tumors. Their diagnosis Surgical resection is often the principal treat-
depends on biopsy, and treatment is similar to ment, and also enables accurate histologic
that of their systemic counterparts. diagnosis. Unless a patient has known active
Intracranial disease may be a manifestation systemic disease, the identification of one of
of widespread metastases, or can occur as an these intracranial lesions should prompt a

Figure 11.6
Plasmacytoma involving the
dura. This 65-year-old
woman presented with
headache and some changes
in behavior. A scan
revealed an enhancing
dural-based mass (arrow)
that on biopsy was a
plasmacytoma. Local
radiation therapy was
delivered. The mass
disappeared and she has
been well in the ensuing 5
years. The photomicrograph
shows dense sheets of
abnormal plasma cells,
some with intranuclear
inclusion bodies.

332
HISTIOCYTIC TUMORS

comprehensive evaluation to determine post-operatively unless done prior to surgery


whether there is evidence of a systemic malig- to avoid confusion with surgically-related
nancy. Post-surgical therapy is often deter- shedding of tumor cells in the CSF. Demon-
mined by the need for generalized systemic stration of leptomeningeal metastases is rare
treatment, but focal RT may be used to control but would necessitate intrathecal chemotherapy
CNS disease. If such a lesion is identified radio- as well as cranial RT.
graphically in a patient with known or active
systemic tumor, such as a dural granulocytic
sarcoma in a patient with documented acute
myelogenous leukemia, then focal RT alone
Histiocytic disorders
may be used since surgery is not essential for A heterogeneous group of tumor and tumor-
diagnosis and these lesions are usually like masses composed of histiocytes, including
radiosensitive. In addition, patients with these Langerhans cell histiocytosis and non-
lesions should have a lumbar puncture to assess Langerhans cell histiocytosis, sometimes occur
whether there is also involvement of the in isolation within the nervous system (Fig.
subarachnoid space. In operated patients, the 11.7). These lesions are usually not malignant
lumbar puncture should be done a few weeks and all are very rare (Table 11.5).

Figure 11.7
Langerhans cell histiocytosis. This 25-year-old man developed diabetes insipidus followed by pituitary failure.
He had no neurologic complaints but an MR scan of the head revealed multiple contrast enhancing masses in
the cerebellum. Cerebellar biopsy was consistent with Langerhans cell histiocytosis. After endocrine
replacement, he had no neurologic complaints nor has he developed any in the past 7 years. The
photomicrograph shows a mixed cellular infiltrate composed of eosinophils, plasma cells, and the characteristic
Langerhans cells.

333
PRIMARY CENTRAL NERVOUS SYSTEM LYMPHOMA AND OTHER HEMOPOIETIC TUMORS

Table 11.5 accurate classification for disorders previously


Histiocytic disorders affecting the CNS. called histiocytosis X, eosinophilic granuloma,
HandSchullerChristian disease and Abt
Dendritic cell-related disorders LettererSiwe disease. Neurologic dysfunction
Langerhans cell histiocytosis occurs in about 16% of patients with
Solitary histiocytomas of various dendritic Langerhans cell histiocytosis. The hypothala-
cell phenotypes
micpituitary axis, the hypothalamus and the
Macrophage-related disorders
Hemophagocytic lymphohistiocytosis74 cerebellum are the sites most commonly
RosaiDorfman disease (sinus histiocytosis involved. Occasionally, mass lesions are found
with massive lymphadenopathy)75 elsewhere in the brain. CNS lesions may occur
ErdheimChester syndrome76 either in the presence or the absence of the
Malignant histiocytosis77
characteristic bony lesions in the skull. Because
of its predilection for the hypothalamic
pituitary axis, diabetes insipidus is the most
common neurologic manifestation. Diabetes
Langerhans cell histiocytosis (from Greek insipidus may precede other neurologic or
histio meaning web or tissue and kytos systemic symptoms by months or years (Table
meaning cell) is a rare disorder characterized 11.6).
by proliferation of cells similar to the MR images generally reveal infundibular
Langerhans cell, a dendritic cell of the epider- thickening or an enhancing suprasellar or
mis.78 Langerhans cell histiocytosis is the hypothalamic mass lesion. Occasionally, space

Table 11.6
Langerhans cell histiocytosis: 38 patients at diagnosis.

Systemic involvement Per cent CNS involvement/MRI Per cent

Multisystem disease 72 White matter lesions, no enhancement 55


Single-system bone disease 18 White matter lesions with enhancement 24
Single-system skin, lymph node 0 Gray matter lesions, no enhancement 50
Primary CNS disease 10 Gray matter lesions with enhancement 8
Bone 84 Extraparenchymal, dural based 32
Skull 74 Extraparenchymal, arachnoidal based 5
Temporal bone 34 Extraparenchymal, choroid plexus based 8
Skin 58 Infundibular thickening 21
Diabetes insipidus 31 (Partial) empty sella 37
Orbits 24 Hypothalamic mass lesions 10
Endocrinopathies 18 Atrophy, diffuse 26
Lungs 16 Atrophy, localized 16
Gastrointestinal tract 10 Therapy-related enhancement and localized atrophy 15
Liver 10
Spleen 10

Modified from Grois et al,79 with permission.

334
REFERENCES

occupying lesions are found elsewhere in the is affected in about 75% of patients. The
brain. The symptoms are location specific. disorder can appear either as an isolated
MRI usually reveals a contrast enhancing mass lymphocytic meningitis or with parenchymal
that can be in the parenchyma of the brain or symptoms including seizures, ataxia74 and
can involve the meninges or choroid plexus. In brainstem abnormalities. When the brain itself
the absence of other systemic lesions the is involved, CT scan may show calcifications.
diagnosis can be made only by biopsy. On MR scan, white matter hyperintensity
The cerebellum and brainstem can be is evident on T2-weighted images; lesions
involved in one of two ways. The patient may occasionally contrast enhance. Although CNS
present with a contrast-enhancing mass in the signs may appear early, most patients have
posterior fossa similar to those seen in the evidence of systemic disease. The diagnosis is
suprasellar or hypothalamic regions. Other suggested by the presence of hemophagocytic
patients present with progressive cerebellar cells within the spinal fluid. Although the
signs associated with a normal MR scan or disease is usually lethal, some patients respond
with non-enhancing white matter hyper- to bone marrow transplantation.74
intensity on the T2-weighted image. This Other non-Langerhans cell histiocytoses
phenomenon appears to be a reaction to that may present with CNS involvement
systemic Langerhans cell histiocytosis and has include the RosaiDorfman syndrome75 and
been described in several patients as a para- the ErdheimChester syndrome.76 The diag-
neoplastic syndrome.80 nosis of these disorders can be made only by
The pathology of Langerhans cell histio- biopsy. These syndromes may present with
cytosis in the brain differs from that elsewhere dural or brain masses. Diagnosis is established
in the body and often lacks features that are radiographically if systemic disease is known;
specifically diagnostic of the disease, making however, in those instances where no systemic
isolated CNS disease difficult to diagnose. The disease is present, CNS biopsy may be
paraneoplastic cerebellar and brainstem required. The ErdheimChester disease, like
syndrome may show only destruction of Langerhans cell histiocytosis, may present
Purkinje cells and demyelination with an with cerebellar signs, apparently as a parane-
absence of histiocytes. oplastic effect of the disorder on the nervous
Single-mass lesions respond to surgery.81 RT system.
is useful in some instances, although it does
not appear to reverse diabetes insipidus.
Some patients have responded to systemic References
chemotherapy.81
1. Recht L, Straus DJ, Cirrincione C et al. Central
Several non-Langerhans cell histiocytic nervous system metastases from non-
lesions can also involve the nervous system Hodgkins lymphoma: treatment and prophy-
(Table 11.5). Hematophagocytic lymphohistio- laxis. Am J Med 1988; 84: 42535.
cytosis is generally a lethal disease character- 2. Hayabuchi N, Shibamoto Y, Onizuka Y et al.
ized by a hemophagocytic syndrome including Primary central nervous system lymphoma in
Japan: a nationwide survey. Int J Radiat Oncol
high fever, hemophagocytic cells, pancytopenia,
Biol Phys 1999; 44: 26572.
low fibrinogen level, hemodilution, elevation of 3. DeAngelis LM. Primary central nervous system
liver enzymes and hypertriglyceridemia. The lymphoma. J Neurol Neurosurg Psychiatry
cause of the disorder is unknown, but the CNS 1999; 66: 699701.

335
PRIMARY CENTRAL NERVOUS SYSTEM LYMPHOMA AND OTHER HEMOPOIETIC TUMORS

4. DeAngelis LM. Primary central nervous system 16. Phan TG, ONeill BP, Kurtin PJ. Posttransplant
lymphoma: a new clinical challenge. Neurology primary CNS lymphoma. Neuro-Oncol 2000;
1991; 411: 61921. 2: 22938.
5. Hao D, DiFrancesco LM, Brasher PM et al. 17. MacMahon EME, Glass JD, Hayward SD et al.
Is primary CNS lymphoma really becoming EpsteinBarr virus in AIDS-related primary
more common? A population-based study of central nervous system lymphoma. Lancet
incidence, clinicopathological features and out- 1991; 338: 96973.
comes in Alberta from 1975 to 1996. Ann 18. Rouach E, Rogers BB, Wilson DR, Kirkpatrick
Oncol 1999; 10: 6570. JB, Byffone GJ. Demonstration of Epstein
6. Corn BW, Marcus SM, Topham A, Hauck W, Barr virus in primary central nervous system
Curran WJ Jr. Will primary central nervous lymphomas by the polymerase chain reaction
system lymphoma be the most frequent brain and in situ hybridization. Hum Pathol 1990;
tumor diagnosed in the year 2000? Cancer 21: 54550.
1997; 79: 240913. 19. Paulus W. Classification, pathogenesis and
7. ONeill BP, Janney CA, Olson J et al. The molecular pathology of primary CNS
continuing increase in primary central nervous lymphomas. J Neurooncol 1999; 43: 2038.
system non-Hodgkins lymphoma (PCNSL): a 20. Weber T, Weber RG, Kaulich K et al. Character-
surveillance epidemiology, and end results istic chromosomal imbalances in primary central
(SEER) analysis. Proc Am Soc Clin Oncol nervous system lymphomas of the diffuse large
2001; 2: 53. B-cell type. Brain Pathol 2000; 10: 7384.
8. Schabet M. Epidemiology of primary CNS 21. Harada K, Nishizaki T, Kubota H et al.
lymphoma. J Neurooncol 1999; 43: 199201. Distinct primary central nervous system
9. CBTRUS (2000). Statistical report: primary lymphoma defined by comparative genomic
brain tumors in the United States, 19921997. hybridization and laser scanning cytometry.
Central Brain Tumor Registry of the United Cancer Genet Cytogenet 2001; 125: 14750.
States, 2000. 22. Bashir R, Coakham H, Hochberg F. Expression
10. Bataille B, Delwail V, Menet E et al. Primary of LFA-1/ICAM-1 in CNS lymphomas: possible
intracerebral malignant lymphoma: report of mechanism for lymphoma homing into the
248 cases. J Neurosurg 2000; 92: 2616. brain. J Neurooncol 1992; 12: 10310.
11. Peterson K, Gordon KB, Heinemann M-H, 23. Harada K, Kurisu K, Arita K et al. Telomerase
DeAngelis LM. The clinical spectrum of ocular activity in central nervous system malignant
lymphoma. Cancer 1993; 72: 8439. lymphoma. Cancer 1999; 86: 10505.
12. Balmaceda C, Gaynor JJ, Sun M, Gluck JT, 24. Bashir R, Chamberlain M, Ruby E, Hochberg
DeAngelis LM. Leptomeningeal tumor in FH. T-cell infiltration of primary CNS
primary central nervous system lymphoma: lymphoma. Neurology 1996; 46: 4404.
recognition, significance, and implications. 25. Gijtenbeek JM, Rosenblum MK, DeAngelis
Ann Neurol 1995; 38: 2029. LM. Primary central nervous system lymphoma.
13. Schiff D, Suman VJ, Yang P, Rocca WA, Neurology 2001; 57(4): 7168.
ONeill BP. Risk factor for primary central 26. Molnar PP, ONeill BP, Scheithauer BW,
nervous system lymphoma. Cancer 1998; 82: Groothuis DR. The bloodbrain barrier in
97582. primary CNS lymphomas: ultrastructural
14. Loureiro C, Gill PS, Meyer PR et al. Autopsy evidence of endothelial cell death. Neuro-
findings in AIDS-related lymphoma. Cancer Oncol 1999; 1: 89100.
1988; 62: 7359. 27. Herrlinger U, Schabet M, Bitzer M, Petersen D,
15. Cornford ME, Holden JK, Boyd MC, Berry K, Krauseneck P. Primary central nervous system
Vinters HV. Neuropathology of the acquired lymphoma: From clinical presentation to
immune deficiency syndrome (AIDS): report diagnosis. J Neurooncol 1999; 43: 21926.
of 39 autopsies from Vancouver, British 28. Herrlinger U. Primary CNS lymphoma:
Columbia. Can J Neurol Sci 1992; 19: Findings outside the brain. J Neurooncol 1999;
44252. 43: 22730.

336
REFERENCES

29. Sugahara T, Korogi Y, Shigematsu Y et al. 41. Nelson DF. Radiotherapy in the treatment of
Perfusion-sensitive MRI of cerebral primary central nervous system lymphoma
lymphomas: a preliminary report. J Comput (PCNSL). J Neurooncol 1999; 43: 2417.
Assist Tomogr 1999; 23: 2327. 42. Corn BW, Dolinskas C, Scott C et al. Strong
30. DeAngelis LM. Cerebral lymphoma presenting correlation between imaging response and
as a nonenhancing lesion on computed survival among patients with primary central
tomographic/magnetic resonance scan. Ann nervous system lymphoma: A secondary analy-
Neurol 1993; 33: 30811. sis of RTOG studies 83-15 and 88-06. Int J
31. Roelcke U, Leenders KL. Positron emission Radiat Oncol Biol Phys 2000; 47: 299303.
tomography in patients with primary CNS 43. Nelson DF, Martz KL, Bonner H et al. Non-
lymphomas. J Neurooncol 1999; 43: 2316. Hodgkins lymphoma of the brain: can high
32. Yoshikai T, Fukahori T, Ishimaru J et al.123I- dose, large volume radiation therapy improve
IMP SPET in the diagnosis of primary central survival? Report on a prospective trial by the
nervous system lymphoma. Eur J Nucl Med Radiation Therapy Oncology Group (RTOG):
2001; 28: 2532. RTOG 8315 Int J Radiat Oncol Biol Phys
33. Kuhlmann T, Schrter A, Dechent P et al. 1992; 23: 917.
Diagnosis of a multifocal B cell lymphoma 44. DeAngelis LM, Yahalom J, Thaler HT, Kher
with preceding demyelinating central nervous U. Combined modality therapy for primary
system lesions by single voxel proton MR CNS lymphoma. J Clin Oncol 1992; 10:
spectroscopy. J Neurol Neurosurg Psychiatry 63543.
2001; 70: 25962. 45. Brada M, Dearnaley D, Horwich A, Bloom
34. Schwarz S, Schwab S, Harms W, Hacke W. HJG. Management of primary cerebral
Systemic lymphoma in patients with the initial lymphoma with initial chemotherapy: prelimi-
diagnosis of primary central nervous system nary results and comparison with patients
lymphoma: a report of two cases. J Neurol treated with radiotherapy alone. Int J Radiat
1999; 246: 8557. Oncol Biol Phys 1990; 18: 78792.
35. Aziz KMA, van Loveren HR. Primary 46. Schlegel U, Pels H, Oehring R, Blmcke I.
lymphoma of Meckels cave mimicking trigem- Neurologic sequelae of treatment of primary
inal schwannoma: case report. Neurosurgery CNS lymphomas. J Neurooncol 1999; 43:
1999; 44: 85962. 27786.
36. Kuhn D, Buchfelder M, Brabletz T, Paulus W. 47. Abrey LE, Yahalom J, DeAngelis LM.
Intrasellar malignant lymphoma developing Treatment for primary CNS lymphoma: the
within pituitary adenoma. Acta Neuropathol next step. J Clin Oncol 2000; 18: 314450.
(Berl) 1999; 97: 3116. 48. Schlegel U, Schmidt-Wolf IGH, Deckert M.
37. Postler E, Bornemann A, Skalej M et al. Primary CNS lymphoma: clinical presentation,
Intracranial inflammatory tumors: A survey of pathological classification, molecular pathogene-
their various etiologies by presentation of 5 sis and treatment. J Neurol Sci 2000; 181: 112.
cases. J Neurooncol 1999; 43: 20917. 49. ONeill BP, Wang CH, OFallon JR et al.
38. Makino K, Murakami M, Kitano I, Ushio Y. Primary central nervous system non-Hodgkins
Primary intracranial plasma-cell granuloma: a lymphoma (PCNSL): Survival advantages with
case report and review of the literature. Surg combined initial therapy? A final report of
Neurol 1995; 43: 3748. the North Central Cancer Treatment Group
39. Weller M. Glucocorticoid treatment of primary (NCCTG) study 86-72-52. Int J Radiat Oncol
CNS lymphoma. J Neurooncol 1999; 43: Biol Phys 1999; 43: 55963.
2379. 50. Schultz C, Scott C, Sherman W et al. Preirradia-
40. Al-Yamany M, Lozano A, Nag S, Laperriere tion chemotherapy with cyclophosphamide,
N, Bernstein M. Spontaneous remission of doxorubicin, vincristine and dexamethasone for
primary central nervous system lymphoma: primary CNS lymphomas: initial report of
report of 3 cases and discussion of patho- Radiation Therapy Oncology Group protocol
physiology. J Neurooncol 1999; 42: 1519. 88-06. J Clin Oncol 1996; 14: 55664.

337
PRIMARY CENTRAL NERVOUS SYSTEM LYMPHOMA AND OTHER HEMOPOIETIC TUMORS

51. Mead GM, Bleehen NM, Gregor A et al. A nisolone following radiotherapy for primary
medical research council randomized trial in central nervous system lymphoma: a phase II
patients with primary cerebral non-Hodgkin study. J Neurooncol 1999; 42: 1617.
lymphoma cerebral radiotherapy with and 61. Cheng AL, Yeh KH, Uen WC, Hung RL, Liu
without cyclophosphamide, doxorubicin, MY, Wang CH. Systemic chemotherapy alone
vincristine, and prednisone chemotherapy. for patients with non-acquired immunodefi-
Cancer 2000; 89: 135970. ciency syndrome-related central nervous system
52. Reni M, Ferreri AJM, Garancini MP, Villa E. lymphoma a pilot study of the BOMES
Therapeutic management of primary central protocol. Cancer 1998; 82: 194651.
nervous system lymphoma in immunocompe- 62. Soussain C, Suzan F, Hoang-Xuan K et al.
tent patients: results of a critical review of the Results of intensive chemotherapy followed by
literature. Ann Oncol 1997; 8: 22734. hematopoietic stem-cell rescue in 22 patients
53. Blay JY, Conroy T, Chevreau C et al. High- with refractory or recurrent primary CNS
dose methotrexate for the treatment of primary lymphoma or intraocular lymphoma. J Clin
cerebral lymphomas: analysis of survival and Oncol 2001; 19: 7429.
late neurologic toxicity in a retrospective series. 63. De Smet MD, Vancs VS, Kohler D, Solomon
J Clin Oncol 1998; 16: 86471. D, Chan CC. Intravitreal chemotherapy for the
54. Hiraga S, Arita N, Ohnishi T et al. Rapid treatment of recurrent intraocular lymphoma.
infusion of high-dose methotrexate resulting in Br J Ophthalmol 1999; 83: 44851.
enhanced penetration into cerebrospinal fluid 64. Bendandi M, Longo DL. Biologic therapy for
and intensified tumor response in primary lymphoma. Curr Opin Oncol 1999; 11: 34350.
central nervous system lymphomas. J 65. Raizer JJ, DeAngelis LM, Zelenetz AD, Abrey
Neurosurg 1999; 91: 22130. LE. Activity of Rituximab in primary central
55. Kraemer DF, Fortin D, Doolittle ND, Neuwelt nervous system lymphoma (PCNSL). Proc Am
EA. Association of total dose intensity of Soc Clin Oncol 2000;19: 166a.
chemotherapy in primary central nervous 66. Thiessen B, DeAngelis LM. Hydrocephalus in
system lymphoma (Human non-acquired radiation leukoencephalopathy results of ven-
immunodeficiency syndrome) and survival. triculoperitoneal shunting. Arch Neurol 1998;
Neurosurgery 2001; 48: 103340. 55: 70510.
56. Doolittle ND, Miner ME, Hall WA et al. 67. Vaicys C, Schulder M, Wolansky LJ,
Safety and efficacy of a multicenter study using Fromowitz FB. Falcotentorial plasmacytoma
intraarterial chemotherapy in conjunction with case report. J Neurosurg 1999; 91: 1325.
osmotic opening of the bloodbrain barrier for 68. Vujovic O, Fisher BJ, Munoz DG. Solitary
the treatment of patients with malignant brain intracranial plasmacytoma: case report and
tumors. Cancer 2000; 88: 63747. review of management. J Neurooncol 1998; 39:
57. Abrey LE, DeAngelis LM, Yahalom J. Long- 4750.
term survival in primary CNS lymphoma. J 69. Herrlinger U, Klingel K, Meyermann R et al.
Clin Oncol 1998; 16: 85963. Central nervous system Hodgkins lymphoma
58. Corry J, Smith JG, Wirth A, Quong G, Liew without systemic manifestation: case report
KH. Primary central nervous system and review of the literature. Acta Neuropathol
lymphoma: Age and performance status are (Berl) 2000; 99: 70914.
more important than treatment modality. Int J 70. Biagi J, MacKenzie RG, Lim MS, Sapp M,
Radiat Oncol Biol Phys 1998; 41: 61520. Berinstein N. Primary Hodgkins disease of the
59. Sandor V, Stark-Vancs V, Pearson D et al. CNS in an immunocompetent patient: a case
Phase II trial of chemotherapy alone for study and review of the literature. Neuro-oncol
primary CNS and intraocular lymphoma. J 2000; 2: 23943.
Clin Oncol 1998; 16: 30006. 71. Byrd C, Edenfield JW, Shields DJ, et al.
60. Shibamoto Y, Sasai K, Oya N, Hiraoka M. Extramedullary myeloid tumours in acute
Systemic chemotherapy with vincristine, nonlymphocytic leukaemia: a clinical review. J
cyclophosphamide, doxorubicin and pred- Clin Oncol 1995; 13: 180016.

338
REFERENCES

72. Klein R, Mullges W, Bendszus M et al. Primary 77. Gogusev J, Nezelof C. Malignant histiocytosis.
intracerebral Hodgkins disease: report of a Histologic, cytochemical, chromosomal, and
case with EpsteinBarr virus association and molecular data with a nosologic discussion.
review of the literature. Am J Surg Pathol Hematol Oncol Clin N Am 1998; 12: 44563.
1999; 23: 47781. 78. Howarth DM, Gilchrist GS, Mullan BP et al.
73. Clark WC, Callihan T, Schwartzberg L, Langerhans cell histiocytosis Diagnosis,
Fontanesi J. Primary intracranial Hodgkins natural history, management, and outcome.
lymphoma without dural attachment. Case Cancer 1999; 85: 227890.
report. J Neurosurg 1992; 76: 6925. 79. Grois NG, Favara BE, Mostbeck GH, Prayer
74. Haddad E, Sulis ML, Jabado N et al. D. Central nervous system disease in
Frequency and severity of central nervous Langerhans cell histiocytosis. Hematol Oncol
system lesions in hemophagocytic lymphohisti- Clin N Am 1998; 12: 287305.
ocytosis. Blood 1997; 89: 794800. 80. Goldberg-Stern H, Weitz R, Zaizov R, Gornish
75. Andriko JA, Morrison A, Colegial CH, Davis M, Gadoth N. Progressive spinocerebellar de-
BJ, Jones RV. Rosai-Dorfman disease isolated generation plus associated with Langerhans cell
to the central nervous system: a report of 11 histiocytosis: a new paraneoplastic syndrome? J
cases. Mod Pathol 2001; 14: 1728. Neurol Neurosurg Psychiatry 1995; 58: 1803.
76. Wright RA, Hermann RC, Parisi JE. 81. Hund E, Steiner HH, Jansen O, Sieverts H,
Neurological manifestations of Erdheim- Sohl G, Essig M. Treatment of cerebral
Chester disease. J Neurol Neurosurg Psychiatry Langerhans cell histiocytosis. J Neurol Sci
1999; 66: 725. 1999; 171: 14552.

339
12
Miscellaneous central nervous system neoplasms and
tumors

Introduction Table 12.1


Miscellaneous CNS neoplasms and tumors
In this chapter, we discuss a number of
intracranial mass lesions not considered Hereditary syndromes
elsewhere in this book (Table 12.1). Included Neurofibromatosis-1 (NF-1)
are familial intracranial tumors, cystic tumors Neurofibromatosis-2 (NF-2)
and some other rare tumors. von HippelLindau
Tuberous sclerosis
LiFraumeni
Cowden
Turcot
Nevoid basal cell carcinoma syndrome
Familial central nervous (Gorlin)
system tumors Retinoblastoma
Bloom syndrome
Introduction Fanconi anemia
Familial melanoma
Table 1.7 lists some of the hereditary Rhabdoid predisposition syndrome
syndromes associated with brain tumors and Cysts and tumor-like lesions
the genetic defects that underlie each Rathke cleft cyst (Chapter 10)
Epidermoid cyst
syndrome. In this chapter, we will emphasize Dermoid cyst
the associated non-central nervous system Colloid cyst of the 3rd ventricle
(CNS) findings that should lead one to suspect Enterogenous cyst
the possibility of a hereditary brain tumor Neuroglial cyst
syndrome. Most of these syndromes are Arachnoid cyst
Granular cell tumor (Chapter 10)
autosomal dominant, indicating that a careful
(choristoma, pituicytoma)
family history will often identify the patient at Hypothalamic neuronal hamartoma
risk. However, in many the penetrance is Nasal glial heterotopia
incomplete, and family members affected with Plasma cell granuloma (Chapter 11)
the gene may not show clinical stigmata. In
some, such as neurofibromatosis-1 (NF-1),
tuberous sclerosis and retinoblastoma, the
mutation rate is so high that the disorder often A careful search for associated non-CNS
occurs in the absence of family history as a findings may assist in making the diagnosis, and
sporadic new mutation. also identify lesions outside the nervous system

340
FAMILIAL CENTRAL NERVOUS SYSTEM TUMORS

Table 12.2
Skin and other organ involvement in familial brain tumor syndromes.

Syndrome Skin Other tissues

Neurofibromatosis-1 Caf-au-lait spots, axillary Iris hamartomas, osseous lesions,


freckling, (cutaneous) pheochromocytoma, leukemia
neurofibromas
Neurofibromatosis-2 None Posterior lens opacities, retinal
hamartoma
von HippelLindau None Retinal hemangioblastomas, renal cell
carcinoma, pheochromocytoma,
visceral cysts, endolymphatic sac
tumor
Tuberous sclerosis Cutaneous angiofibroma (adenoma Cardiac rhabdomyomas, adenomatous
sebaceum), peau chagrin, polyps of the duodenum and small
subungual fibromas intestine, cysts of the lung and
kidney, lymphangioleiomyomatosis,
renal angiomyolipoma
LiFraumeni None Breast carcinoma, bone and soft
tissue sarcomas, adrenocortical,
lung and GI carcinomas, leukemia
Cowden Multiple trichilemmomas, fibromas Hamartomatous polyps of the eye,
colon, and thyroid; breast
carcinoma and thyroid cancer
Turcot syndrome Caf-au-lait spots Colorectal polyps, colon carcinoma
Nevoid basal cell Multiple basal cell carcinomas, Jaw cysts, ovarian fibromas, skeletal
carcinoma syndrome palmar and plantar pits abnormalities
(Gorlin)
Bloom Sun sensitivity, patches of hyper- Characteristic face and voice, gonadal
and hypopigmentation failure, diabetes, immunodeficiency
Fanconi anemia Caf au-lait spots, hyper- and Anemia, skeletal malformations,
hypopigmentation enlarged cerebral ventricles,
gastrointestinal malformations
Multiple endocrine Facial angiofibroma, lipomas, Hyperparathyroidism, gastrinoma,
neoplasia (MEN-1) collagenomas insulinoma, thyroid/bronchial
carcinoid
Retinoblastoma None Retinal tumors, osteosarcomas and
other tumors
Familial melanoma Patches of hyperpigmentation None
Rhabdoid predisposition None Renal tumors, extrarenal malignant
syndrome rhabdoid tumors

that can be treated before they become Most hereditary neoplasms arise from abnor-
symptomatic. These findings are outlined in malities of tumor suppressor genes. The two-
Table 12.2 and detailed in the paragraphs below. hit theory proposed by Knudson to explain the

341
MISCELLANEOUS CENTRAL NERVOUS SYSTEM NEOPLASMS AND TUMORS

different clinical patterns of hereditary and factors inactivate the second allele leading to
sporadic retinoblastomas can be applied to a tumor development. In sporadic cancers, both
number of other hereditary syndromes. That alleles must be inactivated by environmental
theory posits that the patient is born with a factors for a tumor to develop. Examples of
germline mutation deleting one allele of a gatekeeper genes causing familial brain tumors
tumor suppressor gene. When an environmen- are NF-1 and NF-2.
tal event deletes the second allele in a single Caretaker gene inactivation does not directly
cell, both tumor suppressor genes are then non- promote tumor growth; instead, it leads to
functional, the cell is released from normal genetic instability that promotes other somatic
growth controls, and a tumor develops. In gene mutations that in turn lead to cancer
hereditary retinoblastoma, there is a much growth. In familial cancers, caretaker mutations
higher likelihood that an environmental event in the germ line occur in two different forms.
will delete the remaining normal allele in each In the autosomal dominant syndromes, one
eye, explaining why most hereditary retinoblas- mutant allele of the caretaker is inherited and
tomas appear bilaterally. Conversely, the low the remaining allele is mutated by environmen-
likelihood that an environmental event will tal factors. In other cases, both alleles of the
delete both alleles in multiple retinal cells gene must be inherited in mutant form to cause
explains why most sporadic retinoblastomas susceptibility. An example of a caretaker gene
are unilateral. is the gene causing Fanconi anemia, associated
Tumor suppressor gene abnormalities can with medulloblastomas and astrocytomas. The
cause tumors even in the absence of deletion of genetic defect results in hypersensitivity to DNA
both alleles. Mutations in tumor suppressor cross-linking agents and defects in the repair of
genes may lead the mutated gene to have a the cross-linked DNA. A second abnormality is
dominant negative effect; that is, the protein that associated with hereditary non-polyposis
product of the mutated gene preferentially colorectal cancer. The gene hMSH2 is involved
binds its receptor but does not function in repair of DNA mismatches. Loss of both
normally, thus leading to unrestrained growth. alleles of the gene results in complete loss of
Some p53 mutations in brain tumors function DNA mismatch repair activity and in subse-
in this way. quent hypermutability of the cell.
Most familial or hereditary syndromes Genetic testing is available for all the famil-
associated with brain tumors result from loss ial syndromes associated with brain tumors.
of tumor suppressor genes along the lines of This provides specific information for each
Knudsons two-hit hypothesis (Chapter 1). family and allows for the identification of new
There are two different kinds of tumor mutations in a family. Genetic counseling is
suppressor genes that can be implicated in essential for all patients with familial germline
familial cancers.1 These have been termed syndromes, to enable them to understand the
gatekeepers and caretakers. Gatekeepers are complexities of these inherited conditions.2
genes that directly inhibit tumor growth, i.e.
tumor suppressor genes. In most instances,
Neurofibromatosis-1
these genes are rate-limiting for cell growth and
both copies must be inactivated for a tumor to NF-1 (Fig. 12.1)35 is an autosomal dominant
develop. In familial cancers, one gene is inacti- disorder4 caused by an abnormality of the NF-
vated in the germ line and environmental 1 gene at chromosome 17q 11.2. The disorder

342
FAMILIAL CENTRAL NERVOUS SYSTEM TUMORS

Imprinting may play an important role in other


familial cancer syndromes as well.
The NF-1 gene is large, spanning at least 350
kilobases. The gene for oligodendrocyte myelin
glycoprotein is embedded within intron 27b
of the NF-1 gene, perhaps explaining the
occasional occurrence of multiple sclerosis in
NF-1 patients.6 Genetic inactivation of the NF-
C 1 gene can be accomplished by single base pair
A
mutations, deletions of the entire gene, single
or multiple exon deletions, insertions or
B chromosome rearrangement. The protein
product of the gene, neurofibromin, is a 220-
kDa protein consisting of 2818 amino acids.
D Neurofibromin is found in high levels in
neurons, non-myelinated Schwann cells and
testis, and at low levels in adult astrocytes and
myelinated Schwann cells. A developmentally
Figure 12.1 regulated isoform found in forebrain neurons
The common sites of intracranial lesions in patients may be relevant to learning disabilities found
with NF-1. (A) Optic nerve. (B) Optic chiasm. (C) in some NF-1 children.7 Neurofibromin is a
Hypothalamus. Brainstem gliomas (D) are more Ras guanidine triphosphate-activating protein
common in patients with NF-1 and have a more
(GTPase) that promotes the conversion of the
benign prognosis.
Ras oncoprotein from the active to the inactive
form. Inactivation of neurofibromin may lead
to stimulation of the Ras signal transduction
pathway which controls cell division, resulting
in unrestrained cell proliferation. Hyperactivity
of the Ras pathway is characteristic of, and
possibly necessary for, glioma formation. It is
is also called von Recklinghausens disease. The observed in all sporadic malignant gliomas as
disorder is frequent, with an incidence of about well as those found in patients with NF-1. The
1 in 3000 people, 3050% of whom represent mutation rate of the gene may be 100 times
new mutations, predominantly in the paternal higher than the usual mutation rate for a single
germ line. Genomic imprinting may also play locus. The reason for the high mutation rate is
a role in this disease. Genomic imprinting is a not clear. Other genes may also play a modify-
phenomenon where only one of the two alleles, ing role in NF-1, explaining why the phenotype
either the maternal or paternal allele, is is so variable from patient to patient even in
expressed in the cells of the offspring; the other the same family. Mutations of hMLH DNA
allele is suppressed. If the expressed allele is mismatch repair genes, the genetic defect in one
mutated, the heterozygotic cell will behave like form of Turcots syndrome (see below), predis-
it has a homozygous mutation because of pose patients to hematologic malignancies, i.e.
suppression of the normal non-mutated allele. leukemia or lymphoma, and to NF-1. These

343
MISCELLANEOUS CENTRAL NERVOUS SYSTEM NEOPLASMS AND TUMORS

Table 12.3 the presence of six is only suggestive of the


Clinical diagnostic criteria for NF-1. disorder. Axillary freckling and small raised
pigmented hamartomas of the iris, Lisch
The presence of two or more of the following nodules, which originate from melanocytes, are
is diagnostic: a more specific sign. They generally appear late
in childhood or in adulthood, and, by age 60,
1. Six or more caf-au-lait spots, greater than
all patients have these nodules. Axillary and
5 mm in diameter in prepubertal and over
15 mm in postpubertal individuals inguinal freckling usually develop during late
2. Two or more neurofibromas of any type, or childhood or puberty.
one plexiform neurofibroma The common tumors of NF-1 are circum-
3. Axillary and/or inguinal freckling scribed and plexiform neurofibromas arising
4. Optic nerve glioma
from Schwann cells of the peripheral nervous
5. Two or more small elevated hamartomas of
the iris (Lisch nodules) system (Fig. 12.2). The circumscribed neuro-
6. A distinctive osseous lesion, such as fibromas can present either cutaneously (Fig.
sphenoid wing dysplasia or thinning of long 12.3) or subcutaneously and are not a specific
bone cortex, with or without sign of NF-1. They are, as their name indicates,
pseudoarthrosis well circumscribed and, if symptomatic, can be
7. A first-degree relative (parent, sibling, or
resected. Plexiform neurofibromas are irregu-
offspring) with NF-1 according to the above
criteria lar, thickened and non-circumscribed, often
extending long distances along the nerve. NF-
1 expression is reduced or absent10 in these
tumors. These can involve the orbit and extend
from the orbit into the brain or into the spinal
abnormalities of DNA mismatch repair appar- cord from a paraspinal lesion, but otherwise do
ently leave uncorrected mutations in neuro- not affect the CNS. More commonly, they can
fibromin, leading to an NF-1 gene phenotype.8 involve the brachial or lumbosacral plexuses or
Table 12.3 outlines the diagnostic criteria for viscera. In a small percentage of patients (less
NF-1 as delineated by the NIH Consensus than 15%), the tumors may undergo malignant
Conference on Neurofibromatosis in 1987. degeneration and become neurofibrosarcomas.
Minor disease features, including macrocephaly Patients may also have neuroendocrine tumors,
(* 97th percentile), short stature () 3rd including pheochromocytomas and carcinoids,
percentile), hypertelorism (clinical impression) and they are at increased risk for leukemia. A
and thoracic abnormalities such as pectus number of osseous, vascular and nervous
excavatum, assist in the diagnosis in young system lesions, as indicated in Table 12.4, can
children.9 A wide variety of tumors and other also be found.
lesions occur in NF-1. Furthermore, the pheno- Most CNS tumors associated with NF-1 are
type of the disease can vary widely; some pilocytic astrocytomas (Chapter 5). They are
patients are asymptomatic throughout life and located in the optic nerve,11 hypothalamus or
others die from malignant neural tumors. Caf- brainstem of children. There is also an
au-lait spots are generally the earliest manifes- increased frequency of diffuse astrocytomas
tation and are present in almost all adults with and glioblastomas but these are much rarer.
the disease. However, many normal individuals Brainstem gliomas are associated with
have two or three caf-au-lait spots and even neurofibromatosis. These tumors generally run

344
FAMILIAL CENTRAL NERVOUS SYSTEM TUMORS

Figure 12.2
Multiple neurofibromas
in a patient with NF-1.
This CT scan shows
bilateral orbital lesions
(arrows). The lesion on
the right is causing
proptosis and deviation
of the eye. The patients
complaint was diplopia.
The photomicrograph
shows bland spindle-
shaped cells in a loose
mucoid interstitial
matrix.

lesions are rarely biopsied, those associated


with NF-1 could be hamartomas, which do not
require treatment, rather than neoplasms.
Furthermore, patients with NF-1 are at
increased risk for leukemia,14 making
chemotherapy with potentially leukemogenic
agents, such as alkylating agents, risky. MRS
can help distinguish benign pontine lesions
associated with NF-1 from brainstem
gliomas.15 However, asymptomatic brainstem
gliomas should be followed and do not require
immediate treatment. The lesions most likely to
progress are those that are focal rather than
Figure 12.3 diffuse and non-enhancing. If treatment is
Multiple neurofibromas in a 55-year-old man with required, one might consider chemotherapy,
NF-1. The lesions began appearing at age 14. There particularly in young children;16 focal radiation
was no family history of NF-1. therapy (RT) is also useful, particularly for
older children or adults.
Optic gliomas are also associated with NF-1
a more benign course than in patients without and most but not all reports suggest that the
NF-1 and have even been known to regress prognosis is better in NF-1 patients.17,18
spontaneously.13 Neuroimaging cannot defini- However, once optic gliomas progress, survival
tively distinguish malignant from benign rates of NF-1 patients do not differ from those
lesions in the brainstem and because brainstem of others. Almost all symptomatic optic

345
MISCELLANEOUS CENTRAL NERVOUS SYSTEM NEOPLASMS AND TUMORS

Table 12.4
Tumors Major manifestations of
Neurofibromas Dermal NF-1.
Nodular/circumscribed
Plexiform
Gliomas Optic nerve glioma
Pilocytic astrocytoma
Astrocytoma
Glioblastoma multiforme
Sarcomas Malignant peripheral nerve sheath tumor
Triton tumor
Rhabdomyosarcoma
Neuroendocrine tumors Pheochromocytoma
Carcinoid tumor
Hemopoietic tumors Juvenile chronic myeloid leukemia

Other features
Osseous lesions Scoliosis
Height reduction
Macrocephaly
Pseudoarthrosis
Sphenoid wing dysplasia
Limb hypertrophy
Nervous system Intellectual handicap
Epilepsy
Neuropathy
Hydrocephalus (aqueductal stenosis)
Choroidal abnormalities12
Vascular lesions Fibromuscular hyperplasia

gliomas develop before age 6. Optic nerve abnormalities. Since many optic gliomas
gliomas are found in about 15% of patients involve the optic chiasm and hypothalamus,
with NF-1, but at least one-half are asymp- making surgery difficult, it is often wise to
tomatic. Similarly, about 15% of optic gliomas follow without therapy if the patient is not
in most series occur in patients with NF-1. showing progressive symptoms.19 If symptoms
However, a recent series of 21 children with develop and the tumor is surgically inaccessi-
visual pathway gliomas found NF-1 in 62%. ble, RT and/or chemotherapy are useful.16,19
The tumor usually develops by the time the Radiotherapy is effective but the response may
child is 10, but it may be asymptomatic for a take several years.20 RT complications are more
long period. When symptoms develop, they common and more severe in patients with NF-
include decreased visual acuity in one eye if the 1,21 including radiation-induced vascular dis-
tumor is in the optic nerve or in both eyes if orders and cognitive dysfunction.22
the lesion is chiasmal. The tumor may produce If the patient is known to have neurofibro-
proptosis and, if it involves the hypothalamus, matosis and develops CNS symptoms, MR
precocious puberty as well as other endocrine scan generally establishes the diagnosis. When

346
FAMILIAL CENTRAL NERVOUS SYSTEM TUMORS

the patient is not known to have neurofibro- Table 12.5


matosis and the MR scan reveals an optic Diagnostic criteria for NF-2.
glioma, a careful search for the stigmata of NF-
1 in both the patient and first degree relatives The following are diagnostic:
is warranted. Brainstem gliomas are less likely
to be caused by NF-1, but because of the 1. Bilateral vestibular schwannomas; or
2. A first-degree relative with NF-2, and either
hereditary and prognostic implications, a
(a) a unilateral vestibular schwannoma, or
search is warranted under these conditions as (b) two of the following: meningioma,
well. NF-1 should be identified early in life and schwannoma, glioma, neurofibroma,
the patient followed lifelong in order to detect posterior subcapsular lens opacity, or
and treat complications in their early stages. cerebral calcification; or
Guidelines for surveillance at various ages have 3. Two of the following
(a) Unilateral vestibular schwannoma
been published.23,24 One caution: headache,
(b) Multiple meningiomas
particularly tension-type headache in children (c) Either schwannoma, glioma,
under age 10, is more common in patients with neurofibroma, posterior subcapsular lens
NF-1 than in the general population.25 opacity, or cerebral calcification
Headache alone is not an indication for MRI.
Malignant neoplasms and hypertension are the
major causes of the decreased life expectancy
of NF-1 patients.26
form of the disease with very slow growing
Neurofibromatosis-2 vestibular schwannomas with a late onset.30
Table 12.5 outlines the diagnostic criteria
NF-2 is an autosomal dominant disorder charac- from the 1987 NIH Consensus Conference on
terized by bilateral vestibular schwannomas, Neurofibromatosis. About 95% of NF-2
meningiomas and gliomas.5,27 The disorder is also patients have bilateral vestibular schwannomas
called central neurofibromatosis and is entirely (Chapter 9). Other tumors involving the CNS
different from NF-1, von Recklinghausens include meningiomas, gliomas, schwannomas
disease. The incidence is much lower than that of other than on the vestibular nerve, and glial
NF-1, about 1 per 40 000, but new mutations are microhamartomas. Glial microhamartomas,
so frequent that about half of the patients have also called hamartias, are circumscribed
no family history. clusters of cells with large atypical nuclei that
The NF-2 gene has been mapped to chromo- react strongly for S-100 protein. These lesions
some 22q11. The gene product, called merlin, found within the cortex by microscopic exami-
binds to components of the cytoskeleton and is nation are asymptomatic, but are patho-
believed to help organize membrane proteins. gnomonic of NF-2.
The protein is involved in cell proliferation and Approximately 5% of patients with unilat-
motility,28 perhaps by interacting with CD44.29 eral vestibular schwannomas have NF-2.
The most frequent germline mutations are point Multiple meningiomas are also common in NF-
mutations that can be found in many parts of 2 (Fig. 12.4). Meningioangiomatosis, a prolif-
the gene and probably result in a truncated eration of meningothelial cells surrounding
protein. A mutation at intron 15 splice donor small vessels, occurs as a single intracortical
site results in a mutation that causes a mild lesion in some patients with NF-2 and may

347
MISCELLANEOUS CENTRAL NERVOUS SYSTEM NEOPLASMS AND TUMORS

Figure 12.4
A patient with NF-2.
This 45-year-old woman
with known hearing loss
in the left ear developed
headache. A scan
revealed bilateral
acoustic schwannomas
as well as a large
meningioma both above
and below the tentorium
(arrows left). The
meningioma was
successfully removed
(right). She continues to
have bilateral hearing
loss although non-
progressive.

occur independently of a meningioma;31 the von HippelLindau disease


disorder is usually asymptomatic. Intracerebral
calcifications are also relatively frequent. They von HippelLindau (VHL) disease is an auto-
occur in the cerebral and cerebellar cortices, somal dominant illness characterized by
in paraventricular areas and in the choroid hemangioblastomas of the brain and spinal
plexus. They are usually asymptomatic. cord, retinal angiomas, renal cell carcinomas,
Peripheral neuropathy, not due to detectable pheochromocytoma, and cysts and tumors of
tumor, may be a rare manifestation of NF-2.32 other viscera, including the endolymphatic
Patients with NF-2 can present either with sac.34 Only about 25% of cerebellar heman-
unilateral or bilateral hearing impairment or gioblastomas occur in VHL patients, the others
other symptoms anywhere along the neuraxis being sporadic (Fig. 6.10). The incidence of
related to meningiomas or schwannomas VHL is about 1 in 36 000, similar to NF-2 and
(Chapters 6 and 9). The diagnosis should be much less common than NF-1. Unlike NF-1
suspected in any patient with bilateral vestibu- and NF-2, new VHL mutations are rare, as is
lar schwannomas or with multiple menin- incomplete penetrance. Thus, in most patients
giomas. Patients at risk for NF-2 should have a family history or examination of the parents
audiologic or MR evaluation every year or two establishes the familial nature of the illness.
during late adolescence and early adulthood. While penetrance is complete, expression is
Early detection of vestibular schwannomas is highly variable, ranging from a mild symptom
important, because treatment of small lesions (e.g. retinal angioma) to a lethal illness (renal
may preserve hearing.33 The treatment of cancer).
vestibular schwannomas is outlined in Chapter The mean age of VHL patients with cerebel-
9. Criteria for long-term surveillance and lar hemangioblastomas is significantly lower
management of NF-2 patients have been than that of sporadic patients. This finding,
published.23 along with the functional loss of both alleles

348
FAMILIAL CENTRAL NERVOUS SYSTEM TUMORS

at the VHL locus, is consistent with the two- Table 12.6


hit hypothesis. The locus for the VHL gene is Manifestations of VHL disease.
on chromosome 3p2526. The protein
encoded by the gene is a 213 amino acid Organ Lesion
protein that binds to the protein elongen,
which regulates elongation of an mRNA Retina Angioma
Kidney Cysts
transcript. The protein is a component of a
Renal cell carcinoma
ubiquitin ligase, necessary for the oxygen- Pancreas Cysts
dependent degradation of hypoxia inducible Adenomas
factor (HIF); thus, when the VHL protein is Islet cell tumors
absent, vascular endothelial growth factor Adrenal gland Pheochromocytomas
Epididymis Cysts
(VEGF) is unregulated.35 Germline mutations
Cystadenomas
include large deletions, or frame shifts, as well Inner ear Endolymphatic sac tumor
as a variety of missense mutations and other Other organs Visceral cysts
small intragenic mutations. Most families with Adenomas
pheochromocytomas have missense mutations CNS Hemangioblastoma of
whereas those without pheochromocytomas spinal cord, cerebellum
have deletions or premature termination
mutations.34
The disorder is usually recognized when a
patient develops cerebellar signs and a typical
hemangioblastoma is identified on MR scan. described in Chapter 6, and are no different in
When a hemangioblastoma is identified, one VHL patients.
should look for other CNS and extracranial
manifestations of VHL disease36 (Table 12.6).
Early identification of spinal hemangioblast-
Tuberous sclerosis
omas, renal cell carcinomas or pheochromo- The tuberous sclerosis complex (TSC) is an
cytomas may allow one to cure a disease autosomal dominant disorder characterized by
which could become lethal if untreated. benign tumors of the nervous system, including
Retinal angiomas will be found in about 60% hamartomas and subependymal giant cell astro-
of VHL patients. Although they occur cytomas.37 Although usually an inherited
throughout life, they are typically the earliest disease, in many patients the disorder arises
finding of VHL disease, and are sometimes from a spontaneous mutation. The disorder is
present in infancy. The angiomas are bilateral relatively common with an incidence between 1
in about 50% of patients and may cause in 5000 and 1 in 10 000. There are two distinct
profound visual loss due to hemorrhage, tuberous sclerosis genes, both of which function
retinal detachment, glaucoma and cataract. as tumor suppressor genes; tuberous sclerosis
Cerebellar hemangioblastomas occur in complex 1 (TSC1) on chromosome 9q34 and
5070% of patients but may be symptomatic tuberous sclerosis complex 2 (TSC2) on
in only half. Although they can occur in child- chromosome 16p13.38 The protein product of
hood, they usually occur in early middle age, the TSC1 gene is called hamartin. Its function
between 30 and 40 years. The diagnosis and is not known; it has been localized to cytoplas-
treatment of cerebellar hemangioblastomas are mic vesicles and it interacts with tubulin, the

349
MISCELLANEOUS CENTRAL NERVOUS SYSTEM NEOPLASMS AND TUMORS

protein product of the TSC2 gene. Germ line order, but it may be a potential cause of failure
mutations of either gene are inactivating, and of molecular diagnosis.40 The clinical syndrome
loss of heterozygosity at either region occurs in of the TSC is identical regardless of which gene
tuberous sclerosis tumors, indicating that both is dysfunctional. Most patients with TSC are
genes are tumor suppressor genes. NF-1 and mentally retarded. Those with TSC1 mutations
TSC, although distinctive phenotypically, have are less likely to be retarded than those with
molecular similarities. The genes of both TSC2 mutations. Cortical tubers can be identi-
syndromes are hypothesized to function as fied on MR scan and are thought to be the
growth regulators by modulating the activity of cause of epilepsy, including infantile spasm (Fig.
small GTPase molecules.39 Tuberin, which has 12.5).41 Hamartin and tuberin are coexpressed
a GTPase activating domain is localized to the in the tubers.42 Most patients develop facial
Golgi apparatus and may be involved in vesic- angiofibromas, so-called adenoma sebaceum.
ular transport. Mosaicism, a phenomenon in These characteristic lesions, along with others
which a fraction of germline and somatic cells outlined in Table 12.7, allow one to identify
contain a mutation or chromosomal abnormal- the syndrome. Imaging generally reveals
ity, occurs in a number of genetic disorders, subependymal nodules in the walls of the lateral
including TSC. Somatic mosaicism, in which ventricle. In neonates the lesions are hyper-
only some somatic cells are affected, may lead intense on T1 and hypointense on T2; the
to a milder phenotype. Germ cell mosaicism opposite findings occur in older children and
may cause the disease in children of apparently adults. The hamartomas often calcify and rarely
unaffected parents.37 Mosaicism is usually may hemorrhage. The tumors may grow large
associated with a less severe form of the dis- enough to obstruct the ventricular system,

Figure 12.5
Tubers in a
patient with
tuberous sclerosis.
The face was
marked with
typical adenoma
sebaceum
(arrows). Note the
candle-guttering
of the ventricular
system (left
arrow). This
patient was
asymptomatic
with respect to
the tubers.

350
FAMILIAL CENTRAL NERVOUS SYSTEM TUMORS

Table 12.7 conferences have outlined recommendations for


Major manifestations of the tuberous sclerosis diagnostic criteria and evaluation.37 These
complex (TSC).
include cranial MR scans every 13 years
during childhood.
Manifestation Frequency
(%)
Other hereditary syndromes
CNS
Cortical tuber 90100 The LiFraumeni syndrome is a rare autosomal
Subependymal nodule 90100 dominant disorder leading to multiple different
White matter hamartoma 90100 tumors in affected patients, often children and
Subependymal giant cell 616 young adults.2 The common tumors are breast
astrocytoma cancer, osteosarcoma and brain tumors. Other
Skin
tumors include soft tissue sarcomas, leukemia
Facial angiofibroma 8090
(adenoma sebaceum) and lung, adrenal, gastric and colon cancers.
Hypomelanotic macule 8090 Approximately 10% of patients suffer gliomas,
Shagreen patch 2040 most of which occur in young adulthood,
Forehead plaque 2030 usually before age 45. Some develop medul-
Peri- and subungual fibroma 2030
loblastomas and supratentorial PNETs.45
Eyes
Retinal hamartoma 50 Overall, penetrance of the gene is about 50%
Retinal giant cell astrocytoma 2030 by age 30 and 90% by age 60. The disorder is
Hypopigmented iris spot 1020 usually caused by germline mutations in the
Kidney p53 gene,46 most of which are basepair substi-
Angiomyolipoma 50 tutions that result in missense mutations in the
Heart
conserved domain and are similar to p53
Cardiac rhabdomyoma 50
Digestive system mutations occurring in sporadic tumors. Some
Microhamartomatous rectal 7080 families with the clinical syndrome do not have
polyp p53 germline mutations and their genetic defect
Liver hamartoma 4050 is unknown (see below, rhabdoid predisposi-
tion syndrome). Brain tumors cluster in
individual families, suggesting that some
specific p53 mutations may be relatively organ
leading to hydrocephalus. Tumors over 5 mm in specific. In addition to gliomas, medulloblas-
diameter that are incompletely calcified and tomas and PNETs, choroid plexus tumors,
enhance on MRI are at higher risk of contin- ependymomas and schwannomas have been
ued growth.43 If they cause symptoms, the reported. Except for the generally younger age
tumors can be resected, and total resection of patients with brain tumors and the slightly
usually cures. Rare tumors undergo malignant higher male/female ratio compared to sporadic
degeneration and may recur. In addition to tumors, the brain tumors in these patients
benign tumors of the brain, children and adults do not differ clinically from their sporadic
with TSC are at risk for developing a number counterparts.
of malignant tumors including renal cell carci- The diagnosis is suspected in a patient with
nomas, malignant angiomyolipomas and a brain tumor who either has a previous
glioblastoma multiforme.44 Recent consensus history of extraneural tumors, especially an

351
MISCELLANEOUS CENTRAL NERVOUS SYSTEM NEOPLASMS AND TUMORS

osteosarcoma, or has a strong family history of differentiation. PTEN mutations have also been
brain or extraneural tumors. The treatment is reported in sporadic glioblastomas (Chapter 5).
the same as that for sporadic brain tumors but In Cowdens disease men and women are
patients with LiFraumeni syndrome success- affected equally. The LhermitteDuclos lesion
fully treated for a brain tumor should be is characterized by diffuse enlargement of
carefully followed for the development of other cerebellar folia containing large ganglion cells
cancers. that are probably Purkinje cells which expand
and replace the granular and molecular layers.
LhermitteDuclos disease is a rare autosomal The lesion develops slowly, producing ataxia
dominant disorder usually presenting in early and symptoms of increased intracranial
adulthood as a cerebellar hemispheric mass.47 pressure. On MR scan, a characteristic sign is
The estimated gene frequency is about 1 per the presence of tiger stripes or parallel linear
million, although others believe it is under- striations on the T2-weighted image. The
recognized and thus substantially more tumors do not enhance.
frequent. LhermitteDuclos disease was thought
to be a sporadic, isolated condition, but Turcots syndrome refers to several different
recently many cases have been reported in disorders which have in common multiple
association with Cowdens disease,48 an colorectal neoplasms, either polyps or carcino-
autosomal dominant disorder also called the mas, and neuroepithelial tumors including
multiple hamartoma syndrome which includes glioblastomas, medulloblastomas, astrocy-
multiple facial trichilemmomas (a benign tomas and ependymomas.45,51 Some cases are
tumor of the outer root sheath of hair follicles), variants of the familial adenomatous polyposis
oral mucosal papillomas, palmoplantar kerato- syndrome or hereditary non-polyposis colorec-
sis, dysmorphic anomalies and hamartomas of tal carcinoma syndrome.52 The brain tumors do
the thyroid, breast, gastrointestinal tract, and not differ from the sporadic variety, except that
eye. The hamartomas may become malignant, the gliomas generally occur before age 30 and
leading to fibroadenomatous or fibrocystic the medulloblastomas often occur after age 10.
disease in 75% of patients, and breast cancer The diagnosis and treatment are the same as
in 2550% of patients. Thyroid abnormalities for sporadic brain tumors, except that younger
occur in more than half of patients, but thyroid patients with a strong family history of colon
cancer in less than 10%. The diagnosis is made polyps or carcinoma should undergo examina-
by pathognomonic mucocutaneous lesions, tion of the colon.
trichilemmomas and papillomatous papules, Several genetic abnormalities have been
which are present in all individuals by age 30.49 identified. In those patients with the familial
Other neurologic disorders in addition to the adenomatosis polyposis syndrome, germline
LhermitteDuclos lesion include megencephaly, mutations in the APC gene located on chromo-
a bridged sella turcica, mental retardation, some 5q21 lead to failure of function of the
gliomas, meningiomas and neuromas. APC gene product. This protein interacts with
Cowdens disease has been mapped to -catenin and appears to modify its association
chromosome 10q23. The gene involved is the with the E-cadherin cell adhesion molecule. Its
PTEN gene50 which has a tyrosine phosphatase exact role in the production of either colon
domain as well as a tensin-homology domain; cancers or brain tumors is not clear. Other
it is involved in regulation of cell growth and families do not have germline mutations of the

352
FAMILIAL CENTRAL NERVOUS SYSTEM TUMORS

APC gene but appear to have DNA replication transcriptional repressor which, when absent,
errors in their tumors similar to those found in leads to uncontrolled transcription. Similar
patients with hereditary non-polyposis colo- mutations are infrequently found in sporadic
rectal cancer. These patients carry mutations of medulloblastomas.54
hMLH-1 or hPMS-253 genes. The hMLH-1
gene on chromosome 3p21 encodes a protein Fanconi anemia (FA) is an autosomal recessive
responsible for strand-specific DNA mismatch disorder that confers a predisposition to bone
repair. The hPMS-2 gene at chromosome 7p22 marrow failure and leukemia. Several other
interacts with the hMLH-1 protein in forming congenital abnormalities are present as well, as
a mismatch protein complex. These gene indicated in Table 12.2. The disorder is found
mutations are associated with microsatellite in all races and ethnic groups and has a carrier
instability in tumors. This abnormality is rare frequency of about 1 in 300. This may be a low
in brain tumors in the absence of Turcots estimate because of poor ascertainment of cases.
syndrome. As many as 0.5% of all people may be heterozy-
gous for an FA gene.56 There are at least 7
Gorlin syndrome, also called the nevoid basal cell distinct FA genes. They are believed to be
carcinoma syndrome, is a rare autosomal caretaker genes but their exact function is
dominant disorder in which patients develop unknown.57. The basic defect in FA is believed
multiple basal cell carcinomas at an early age. The to result in abnormalities in DNA repair, growth
incidence is about 1 in 50 000. Additional extra- factor homeostasis and cell cycle regulation. It is
neural lesions include jaw keratocysts, pitting of probably the abnormality of DNA repair that
the palms and soles, and skeletal deformities. leads to malignancy. The most common malig-
Intracranial lesions include falx calcifications, nancy is leukemia, particularly acute myeloge-
hydrocephalus, dysgenesis of the corpus callosum nous leukemia, but other cancers including
and medulloblastomas.45 Medulloblastoma occurs medulloblastomas and astrocytomas have been
in 510% of patients with the syndrome. reported.58
Conversely, among patients with medullo-
blastoma, only 12% have Gorlin syndrome. Bloom syndrome is an autosomal recessive
The medulloblastoma develops around 2 disorder characterized by dwarfism, sun sensi-
years of age, younger than for sporadic medullo- tivity and a characteristic facial appearance.59
blastomas, and frequently has a desmoplastic A variety of tumors have been recorded in
phenotype. Treatment may lead to a more favor- patients with Bloom syndrome, including
able outcome in these patients than in patients medulloblastoma, meningioma and retinoblas-
with sporadic medulloblastomas. However, toma. The disorder is common in Ashkenazi
basal cell carcinomas are likely to develop in the Jews and is a result of mutation in a single
irradiated skin, and patients should be followed locus, BLM, which maps to chromosome
carefully for the late development of these skin 15q26. The gene product is homologous to
cancers. The genetic abnormality is in the PTCH other proteins that function as DNA
gene on chromosome 9q31.54 The gene encodes helicases.60 A DNA helicase (helix is from the
a protein necessary for the intracellular signal- Greek for coil) is a repair enzyme which
ing pathway of the Sonic hedgehog gene, neces- uncoils DNA. The disorder leads to unstable
sary for normal development of several organs DNA in the somatic cells of those who inherit
including the brain.55 The protein is probably a the disease, leading in turn to spontaneous

353
MISCELLANEOUS CENTRAL NERVOUS SYSTEM NEOPLASMS AND TUMORS

hypermutability which is probably responsible ated with pineal region or suprasellar tumors,
for the development of neoplasms. The disease so-called trilateral retinoblastoma.45,63
is rare and only a few patients with brain
tumors have been reported. Rhabdoid predisposition syndrome is a recently
described autosomal dominant disease charac-
Familial melanoma. About 51 000 people terized by malignant rhabdoid tumors involv-
developed melanoma in 200161 in the USA, an ing the kidney and other organs including the
impressive increase in frequency in melanoma brain. In addition, a number of patients have
in recent decades, probably related to sun choroid plexus carcinomas, medulloblastomas
exposure. Perhaps as many as 10% of all cases and central PNETs. The disorder is associated
of melanoma are familial. Individuals who have with a germline mutation of the hSNF5/INI1
a first-degree relative who has developed gene localized on chromosome 22. The
melanoma under the age of 50 have a relative penetrance of the syndrome is high and most
risk of 6.5 fold of developing melanoma tumors occur in children below the age
compared to the general population. Germ line of 3. The gene encodes a member of the
mutations in the locus that encodes p16 SW1/SNF ATP-dependent chromatin-remodel-
(INK4A) and p14 ARF are associated with ing complex. Its exact role in the causation of
melanoma susceptibility in familial melanoma.62 the renal and extrarenal tumors is unknown.
Other genetic abnormalities may exist as well. Because some of the multiple tumors are
similar to those of the LiFraumeni syndrome,
Multiple endocrine neoplasia type I (MEN-1) it is possible that some cases of apparent
is an autosomal dominant disorder that leads LiFraumeni syndrome without p53 germline
to a variety of endocrine tumors, including mutations belong to this syndrome.64
pituitary adenomas. About 15% of patients
have prolactinomas, 5% other hormone- Other familial brain tumors are undoubtedly
producing pituitary adenomas, and 2% non- more common than can be encompassed by the
secreting tumors. The tumors are benign. known genetic syndromes. A population-based
Associated endocrine disorders include primary study indicates that about 5% of gliomas are
hyperparathyroidism, gastrinoma and insuli- familial.65 Further investigation of the genetics
noma. The MEN-1 gene maps to chromosome of brain tumors may lead to the identification
11q13; its protein product is called menin. of more familial syndromes.66 Some clearly
represent failure to diagnose the syndromes
Retinoblastoma is the prototypic example of a described above, making each of them more
familial cancer. It is the most common intra- common than currently recognized. Others are
ocular malignancy in children, with a world- a result of hereditary gene defects not yet
wide incidence of about 1 in 20 000 live births. identified. Careful attention to the possible
Sixty percent of the tumors are non-hereditary familial occurrence of such tumors will not
and unilateral, and 40% are hereditary, most only allow for appropriate genetic counseling
of which are bilateral. The gene maps to but may identify new genetic defects that may
chromosome 13q14 and is called RB-1. Its be a site for therapeutic intervention. Figure
protein product is 110 kDa in size and is a 12.6 shows an algorithm for patients with CNS
classic tumor suppressor. The tumors rarely tumors that might be potentially familial in
involve the CNS but occasionally are associ- origin.

354
FAMILIAL CENTRAL NERVOUS SYSTEM TUMORS

Patients and/or family members with diagnosis of a known hereditary


cancer syndrome, patients with positive family history, or patients with
young onset (< 50 years) of cancer

Initial risk and educational counseling with patient and family; review
genetics of cancer, increased cancer risks, and preventive intervention options.

Complete family history obtained and Molecular diagnostic studies of index


diagnoses of cancer in family members patient (replication error studies of tumors
confirmed by medical records, if possible. or genetic testing of lymphocytes).

DIAGNOSIS OF HEREDITARY CANCER If a specific causal gene cannot be


SYNDROME IN THE FAMILY IS identified, then genetic testing of at-risk
CONFIRMED family members should not be pursued.
Conventional screening guidelines and
interventions as reviewed in the initial
counseling session apply.

Predictive genetic testing may be offered to


at-risk family members

Gene-positive individuals will be Gene-negative individuals will be


encouraged to adhere to preventive encouraged to adhere to screening
screening recommendations and counseled guidelines for the general population.
regarding other options (i.e. prophylactic
surgery), if applicable.

Figure 12.6
Cancer risk assessment and gene testing algorithm from Peterson and Cordori94 with permission.

355
MISCELLANEOUS CENTRAL NERVOUS SYSTEM NEOPLASMS AND TUMORS

Neoplastic cysts and cyst-like discussed in this book except in the context of
differential diagnosis. The cysts discussed here
tumors fall into two large categories, those lined by
In some patients, mass lesions of the brain cause neuroepithelium which secretes CSF and those
their symptoms not by growth of tumor cells that have a non-neuroepithelial lining secreting
but by enlargement of a fluid-filled cyst. Some a fluid different from CSF; some of these latter
of these cysts are associated with neoplasms; lesions occasionally become true neoplasms.
others are not but must be considered in the Lesions in these two categories can often be
differential diagnosis of neoplasm. Table 12.8 distinguished by MR scan (see below).
classifies these cysts. Many such cysts that
accompany brain tumors, such as pilocytic Dermoids and epidermoids
astrocytomas and hemangioblastomas, are
discussed in the chapters on those primary These tumors account for only about 1% of all
neoplasms. Cysts of infectious origin, such as intracranial neoplasms, the epidermoid being
cysticercosis and ex-vacuo cysts, i.e. those that three or four times more frequent than the
result from loss of brain substance, are not dermoid.68,69 Both tumors are probably malfor-
mations originating from incomplete cleavage
of neural from fetal cutaneous ectoderm during
Table 12.8 the fourth week of gestation. Thus, fetal
Intracranial cysts. epiblasts are not excluded from the nervous
system during development when the neural
Cysts with CSF-like fluid tube closes. The tumors usually occur in the
(a) Ex vacuo midline along the base of the brain and the
Leptomeningeal cysts spinal canal, including fourth ventricle,
Cysts after surgical resection
suprasellar and parasellar cisterns. They may
Cysts after cerebral infarction or trauma
Porencephalic cysts or may not communicate with the skin surface
VirchowRobin spaces through a midline dermal tract. The epider-
(b) Cysts with fluid-secreting walls moid cyst is lined by keratin-producing
Arachnoid cysts squamous epithelial cells and may occur later-
Neuroepithelial cysts ally in the skull or cerebellopontine angle.
Choroid plexus cyst
When these tumors are found in the middle ear,
(c) Midline cysts of dysgenetic origin
Cysts arising from cavum septi pellucidi, they are called cholesteatomas. Although
cavum vergae and cavum veli interpositi congenital, epidermoids generally make their
DandyWalker cysts appearance in adulthood. Spinal epidermoids
Cysts with non-neuroepithelial lining are sometimes associated with lumbar
Pineal cysts
puncture, particularly those done early in life.70
Rathkes cleft cysts (Chapter 10)
Colloid cysts The dermoid cyst is lined by squamous
Epidermoid cysts epithelium and underlying dermal appendages
Dermoid cysts including hair follicles and adnexa. The cystic
Cysts associated with brain neoplasms contents may include hair or primordial teeth.
Cysts associated with infections Dermoid cysts are commonly found within the
Modified from Mooij and Go63 with permission. midline of the cerebellum, usually in associa-
tion with a dermal sinus, and usually present

356
NEOPLASTIC CYSTS AND CYST-LIKE TUMORS

in childhood. Suprasellar dermoids in connec- The clinical presentation depends on the


tion with dermal sinuses can also occur. location of the tumor. They expand slowly,
Dermoids are common in the spinal canal usually through desquamation of epithelial
where they may be associated with a sinus tract cells, and may become quite large before
and a bony malformation. signs and symptoms develop (Fig. 12.7). The

Figure 12.7
An epidermoid cyst of the posterior fossa. This 50-year-old man presented with mild headache. A scan revealed
a large epidermoid cyst in the posterior fossa (arrow left upper). The cyst was believed to be asymptomatic. A
year elapsed without any significant change in his symptoms, when he developed severe headache, diplopia,
difficulty swallowing and some weakness in one lower extremity. A scan of the head revealed hydrocephalus
but no change in the size of the epidermoid cyst. A scan of the spine revealed contrast enhancing nodules on
lumbar roots (arrows, right). Although the CSF did not show abnormal cells, biopsy of the lumbar meninges
revealed squamous cell carcinoma. The patient died and an autopsy revealed that the epidermoid contained
squamous cell carcinoma both within it and involving the meninges. The photomicrograph shows the benign
keratin-filled cyst lined by well-differentiated squamous epithelium.

357
MISCELLANEOUS CENTRAL NERVOUS SYSTEM NEOPLASMS AND TUMORS

supratentorial lesions may present with The diagnosis is made radiographically.73


headaches, seizures or visual field defects. Epidermoids are hypointense on T1-weighted
Tumors in the cerebellopontine angle present images but more intense than CSF, and iso- to
with hearing loss. Other posterior fossa hyperintense on T2-weighted images but isoin-
symptoms include trigeminal neuralgia and tense relative to CSF. There may be rim
hemifacial spasm. Pineal region epidermoids enhancement. Dermoids are often hyperintense
present with a headache, diplopia and on T1-weighted images and relatively
vertigo.71 Occasionally, rupture of the cyst hypointense on T2-weighted images (Fig. 12.8).
structure may spill irritating contents into the Signal void may represent some calcification
subarachnoid space, causing an aseptic and hyperintensity may represent fat deposi-
meningitis. Repetitive episodes of aseptic tion. The difference between signal intensity in
meningitis, sometimes associated with focal dermoids and that in epidermoids is probably
neurologic signs, have been reported as the explained by cholesterol, which, in its solid,
presenting manifestation of these lesions.72 At crystalline state, is hypointense on T1, but in
least one of our patients carried a diagnosis the liquid state appears hyperintense. CT scans
of multiple sclerosis for many years before of either epidermoids or dermoids show a
the cyst was found. Recurrent infectious lesion without enhancement. In some patients
meningitis may be a presenting symptom if with recurrent aseptic meningitis, the ruptured
the dermoid communicates with the surface. cyst may not be seen on scan during an acute

Figure 12.8
A dermoid cyst. This 56-year-old woman presented with partial complex seizures and was found to have this
intracranial lesion that was hyperintense before contrast and did not enhance. Note the absence of edema
around this large extraaxial mass. The photomicrograph shows the cyst wall is lined by squamous epithelium
with an underlying stroma containing a sebaceous gland (arrow).

358
NEOPLASTIC CYSTS AND CYST-LIKE TUMORS

episode; subsequent imaging may be required treatment of choice. Using microsurgical


to establish the diagnosis. Rupture may lead to techniques, total resection and cure is possible
subarachnoid seeding of the cyst contents and in some instances, but total resection is often
cells.74 Rarely, the cysts may undergo malignant difficult because the tumor capsule adheres to
degeneration to squamous cell carcinoma; in cranial nerves, blood vessels and other vital
one of our patients carcinoma seeded the structures. However, even subtotal resection
leptomeninges and was the presenting manifes- may yield a prolonged remission of symptoms.
tation of the epidermoid.75 Mortality is quite low, but morbidity, including
Both dermoids and epidermoids are grossly aseptic meningitis and transient cranial nerve
well-defined masses with smooth, lobulated, palsies as well as hydrocephalus, may occur,
somewhat irregular surfaces and a pearly particularly in surgery of posterior fossa
appearance. Dermoids are filled with hair and masses. Sometimes the cyst wall cannot be
keratinized debris. Microscopically, the tumors totally eradicated. Such patients should be
are lined by simple stratified squamous epithe- followed by serial MR scans. Radiotherapy and
lium with a variable degree of keratinization. chemotherapy are not useful.
The presence of dermal appendages in the cyst
wall, such as sebaceous glands, sweat glands
Colloid cysts
and hair follicles or fatty tissue, defines the
tissue as a dermoid (Fig. 12.8). These non-neoplastic lesions located in the 3rd
When the tumors are found incidentally, they ventricle and attached to the choroid plexus
may be followed without specific therapy produce their symptoms by obstructing the
unless they become symptomatic. In foramen of Monro and causing hydrocephalus
symptomatic patients, surgical removal is the (Fig. 12.9).76 Colloid cysts are so named

Figure 12.9
A colloid cyst of
the 3rd ventricle
(arrow). Note the
hydrocephalus. The
photomicrograph
shows amorphous
cyst contents with
a lining of
columnar ciliated
epithelium.

359
MISCELLANEOUS CENTRAL NERVOUS SYSTEM NEOPLASMS AND TUMORS

because of the glue-like nature of the cyst attacking the tumor, but shunt malfunction,
contents (colloid from the Greek for glue and leading to sudden recurrence of hydrocephalus,
appearance). The cyst is lined by cuboidal to and the slit ventricle syndrome causing severe
columnar ciliated epithelial and/or goblet cells, headache80 make this a problematic treatment.
thought to be endodermal in origin. Surgery through either a transcallosal or
Characteristically, the patient suffers from transcortical approach is the treatment of
headaches due to the hydrocephalus. At one choice, but it may be associated with morbid-
time it was believed that colloid cysts would ity. Many neurosurgeons accomplish this by
intermittently obstruct the ventricular system endoscopy.81 Transient memory deficits from
by a ball valve mechanism, producing sudden traction on the fornix have been noted in 26%
increased intracranial pressure (ICP), occasion- of patients after a transcallosal approach. In a
ally leading to herniation and death.76 The few, there was permanent memory difficulty.82
choroidal attachment of the colloid cyst Patients fare better when the operation is done
renders the mass sessile and makes a ball valve by a surgeon experienced in approaching third
mechanism impossible. Sudden symptoms in ventricular tumors.76 Removal of the cyst cures
patients with colloid cysts are caused by the patient, but asymptomatic patients can
pressure waves (Chapter 3) developing in a safely be followed without surgery.83 RT and
setting of chronically elevated ICP. chemotherapy have no role to play.
The diagnosis is made by MR scan, showing
a cyst within the 3rd ventricle at the level of
Intracranial arachnoid cysts
the foramen of Monro. The cyst is iso- to
hyperintense on the T1-weighted image and These are CSF-filled cystic structures lined with
hypo- to isointense on T2.77 Colloid cysts can a transparent membrane resembling the arach-
range from a few millimeters to several noid. They are a congenital malformation and
centimeters in diameter. They are spherical or not a response to cerebral atrophy. They are
ovoid and have a smooth, semi-translucent often found in the middle cranial fossa84 (Fig.
wall. The viscosity of the cyst content varies 12.10) compressing the temporal lobe, at the
from fluid to solid. The mucin-like cyst cerebral convexity, the suprasellar area, the
contents contain sulfonated carbohydrate quadrigeminal area and the posterior fossa,
epitopes. The carbohydrate epitopes are similar particularly the cerebellopontine angle. They
to those reported for mucins of the salivary are occasionally intraventricular, arising from
gland and other peripheral cysts believed to be the arachnoid in the choroidal fissure.85 Most
of endodermal origin.78 are asymptomatic but some may cause
In some asymptomatic patients, particularly headache, dizziness or seizures; some may grow
older patients without hydrocephalus, the cysts large enough to produce substantial compres-
do not grow and need no treatment.79 Two sion of the brain and elevate intracranial
types of treatment have been recommended for pressure. The cyst may bleed into itself or
symptomatic patients. One is drainage of the rupture into the subdural space, causing an
cyst via stereotactic needle placement. Although effusion that is symptomatic and requires
this relieves symptoms temporarily, the cyst drainage.86,87
usually reaccumulates. The best approach is The diagnosis is made on MR scan. The
surgical removal. Some surgeons have recom- lesion has signal characteristics similar to those
mended shunting both ventricles and not of CSF, sometimes deeply indenting but not

360
NEOPLASTIC CYSTS AND CYST-LIKE TUMORS

Figure 12.10
Two symptomatic arachnoid cysts. Left MRI: a 25-year-old university teacher complained of new-onset
headache and difficulty in concentrating. Several physicians believed that the cyst of the cavum velum
interpositum was coincidental and not responsible for the symptoms. However, after a year of symptoms,
endoscopic evacuation of the cyst abolished all symptoms. Right MRI: a 30-year-old woman presented with
headache. A scan revealed a large arachnoid cyst. Note that the cyst and the spinal fluid have the same signal
intensity and that the gyri and sulci of the brain have a normal configuration, indicating that they are not
compressed. The headache was relieved by analgesics and eventually disappeared. The photomicrograph shows
an arachnoid cyst with a delicate fibrous connective tissue membrane lined by meningothelial cells.

invading brain parenchyma. Intrathecal injection grow suggests that there is enough loss of fluid
of contrast material usually demonstrates slow from the cyst into the subarachnoid space to
if any communication between the cyst and the balance fluid production within the cyst.
subarachnoid space. However, the fact that the The problem in diagnosis is to determine
membrane secretes fluid but the cyst does not which cysts are symptomatic and which are

361
MISCELLANEOUS CENTRAL NERVOUS SYSTEM NEOPLASMS AND TUMORS

discovered incidentally in the workup of a


patient for headache or seizures. Most patients
with seizure disorders do improve after treatment
of an arachnoid cyst, suggesting that the cortical
gliosis caused by the cyst may be a source of
seizures and may improve after the cyst is
treated. Many headaches improve as well.
Nonetheless, in some instances, the headache is
due to another cause and treatment of the cyst
will not help. As with brain tumors, new onset
headaches, change in headache frequency or
intensity and focal headache at the site of the cyst
suggest that the cyst is symptomatic. Arachnoid
cysts that are asymptomatic should just be Figure 12.11
followed. Symptomatic arachnoid cysts should A hypothalamic hamartoma in a young boy with
be drained by creating a communication between gelastic seizures (arrow) (see text).
the cyst and the ventricular system or subarach-
noid space, depending on the location of the cyst.
In some instances, communication can be estab- also common. Mental retardation is common.
lished by puncturing the wall of the cyst directly MR scan reveals a non-enhancing mass lesion
or endoscopically. In some instances, communi- involving the hypothalamus, isointense with
cation requires a cystperitoneal shunt. Stereo- brain. On the T2-weighted images, the tumors
tactic intracavitary irradiation by injection of 32P are isointense or slightly hyperintense.
has proved safe and effective.88 Differential diagnosis includes hypothalamic
astrocytoma, optic glioma and germinoma. In
Hypothalamic neuronal the asymptomatic patient, no treatment is
hamartomas necessary. Hormonal abnormalities can often
be corrected with appropriate agents. Surgery92
These neuroepithelial masses are composed of and RT93 have been reported to control
mature and often clustered neurons and glial seizures in some patients; radiofrequency
stroma. Most occur at the base of the brain, ablation helped one patient.94
attached to the floor of the fourth ventricle, or
within the hypothalamus (Fig. 12.11). The
tumors are hamartomas but sometimes they References
expand. They are usually asymptomatic but
1. Vogelstein B, Kinzler KW. The genetic basis of
may cause endocrine disorders or seizures, human cancer. New York: McGrawHill, 1998.
particularly gelastic seizures89,90 (inappropriate 2. Tomlinson GE. Familial cancer syndromes and
sudden laughter, from Greek to laugh) or ictal genetic counseling. Cancer Treat Res 1997; 92:
autonomic dysfunction e.g. hypertension, 63-97.
tachycardia, bradycardia.91 They can cause 3. Pollack IF, Mulvihill JJ. Neurofibromatosis 1
and 2. Brain Pathol 1997; 7: 82336.
precocious puberty in both sexes. Children may 4. Feldkamp MM, Gutmann DH, Guha A.
be larger than normal with advanced bone age; Neurofibromatosis type 1: piecing the puzzle
hormonal dysfunction on laboratory tests is together. Can J Neurol Sci 1998; 25: 18191.

362
REFERENCES

5. Karnes PS. Neurofibromatosis: a common imaging and spectroscopy in the distinction


neurocutaneous disorder. Mayo Clin Proc from diffuse pontine glioma. Neurosurgery
1998; 73: 10716. 1997; 40: 3317.
6. Johnson MR, Ferner RE, Bobrow M, Hughes 16. Packer RJ, Ater J, Allen J et al. Carboplatin
RA. Detailed analysis of the oligodendrocyte and vincristine chemotherapy for children
myelin glycoprotein gene in four patients with with newly diagnosed progressive low-grade
neurofibromatosis 1 and primary progressive gliomas. J Neurosurg 1997; 86: 74754.
multiple sclerosis. J Neurol Neurosurg 17. Deliganis AV, Geyer JR, Berger MS. Prognostic
Psychiatry 2000; 68: 6436. significance of type 1 neurofibromatosis (von
7. Gutmann DH, Zhang YJ, Hirbe A. Recklinghausen disease) in childhood optic
Developmental regulation of a neuron-specific glioma. Neurosurgery 1996; 38: 11148.
neurofibromatosis-1 isoform. Ann Neurol 18. Chan MY, Foong AP, Heisey DM et al.
1999; 46: 77782. Potential prognostic factors of relapse-free
8. Wang Q, Lasset C, Desseigne F et al. survival in childhood optic pathway glioma: a
Neurofibromatosis and early onset of cancers multivariate analysis. Pediatr Neurosurg 1998;
in hMLH1-deficient children. Cancer Res 29: 238.
1999; 59: 2947. 19. Shuper A, Horev G, Kornreich L et al. Visual
9. Cnossen MH, Moons KG, Garssen MP et al. pathway glioma: an erratic tumour with thera-
Minor disease features in neurofibromatosis peutic dilemmas. Arch Dis Child 1997; 76:
type 1 (NF1) and their possible value in 25963.
diagnosis of NF1 in children < or = 6 years 20. Tao ML, Barnes PD, Billett AL et al.
and clinically suspected of having NF1. Childhood optic chiasm gliomas: Radiographic
Neurofibromatosis team of Sophia Childrens response following radiotherapy and long-term
Hospital. J Med Genet 1998; 35: 6247. clinical outcome. Int J Radiat Oncol Biol Phys
10. Peters N, Waha A, Wellenreuther R et al. 1997; 39: 57987.
Quantitative analysis of NF1 and OMGP gene 21. Grill J, Couanet D, Cappelli C et al. Radiation-
transcripts in sporadic gliomas, sporadic induced cerebral vasculopathy in children with
meningiomas and neurofibromatosis type 1- neurofibromatosis and optic pathway glioma.
associated plexiform neurofibromas. Acta Ann Neurol 1999; 45: 3936.
Neuropathol (Berl ) 1999; 97: 54751. 22. Cappelli C, Grill J, Raquin M et al. Long term
11. Listernick R, Louis DN, Packer RJ, Gutmann follow up of 69 patients treated for optic
DH. Optic pathway gliomas in children with pathway tumours before the chemotherapy era.
neurofibromatosis 1. Consensus statement Arch Dis Child 1998; 79: 3348.
from the NF1 optic pathway glioma task force. 23. Gutmann DH, Aylsworth A, Carey JC et al.
Ann Neurol 1997; 41: 1439. The diagnostic evaluation and multidisciplinary
12. Yasunari T, Shiraki K, Hattori H, Miki T. management of neurofibromatosis 1 and
Frequency of choroidal abnormalities in neurofibromatosis 2. JAMA 1997; 278: 517.
neurofibromatosis type 1. Lancet 2000; 356: 24. Huson SM. What level of care for the neuro-
98892. fibromatoses? Lancet 1999; 353: 11146.
13. Perilongo G, Moras P, Carollo C et al. 25. DiMario FJ Jr, Langshur S. Headaches in
Spontaneous partial regression of low-grade patients with neurofibromatosis-1. J Child
glioma in children with neurofibromatosis-1: a Neurol 2000; 15: 2358.
real possibility. J Child Neurol 1999; 14: 3526. 26. Zoller ME, Rembeck B, Oden A, Samuelsson
14. Stiller CA, Chessells JM, Fitchett M. M, Angervall L. Malignant and benign tumors
Neurofibromatosis and childhood leukaemia/ in patients with neurofibromatosis type 1 in a
lymphoma: a population-based UKCCSG defined Swedish population. Cancer 1997; 79:
study. Br J Cancer 1994; 70: 96972. 212531.
15. Broniscer A, Gajjar A, Bhargava R et al. Brain 27. Bondy M, Ligon BL. Epidemiology and etiol-
stem involvement in children with neurofibro- ogy of intracranial meningiomas: a review. J
matosis type 1: role of magnetic resonance Neurooncology 1996; 29: 197205.

363
MISCELLANEOUS CENTRAL NERVOUS SYSTEM NEOPLASMS AND TUMORS

28. Reed N, Gutmann DH. Tumorigenesis in 41. Hancock E, Osborne JP. Vigabatrin in the
neurofibromatosis: new insights and potential treatment of infantile spasms in tuberous
therapies. Trends Mol Med 2001; 7: 15762. sclerosis: literature review. J Child Neurol
29. Morrison H, Sherman LS, Legg J et al. The 1999; 14: 714.
NF2 tumor suppressor gene product, merlin, 42. Johnson MW, Emelin JK, Park SH, Vinters
mediates contact inhibition of growth through HV. Co-localization of TSC1 and TSG2 gene
interactions with CD44. Genes Dev 2001; 15: products in tubers of patients with tuberous
96880. sclerosis. Brain Pathol 1999; 9: 4554.
30. Sainio M, Jaaskelainen J, Pihlaja H, Carpen O. 43. Nabbout R, Santos M, Rolland Y et al. Early
Mild familial neurofibromatosis 2 associates diagnosis of subependymal giant cell astro-
with expression of merlin with altered COOH- cytoma in children with tuberous sclerosis. J
terminus. Neurology 2000; 54: 11328. Neurol Neurosurg Psychiatry 1999; 66: 3705.
31. Wiebe S, Munoz DG, Smith S, Lee DH. 44. Al-Saleem T, Wessner LL, Scheithauer BW et
Meningioangiomatosis A comprehensive al. Malignant tumors of the kidney, brain, and
analysis of clinical and laboratory features. soft tissues in children and young adults with
Brain 1999; 122: 70926. the tuberous sclerosis complex. Cancer 1998;
32. Trivedi R, Byrne J, Huson SM, Donaghy M. 83: 220816.
Focal amyotrophy in neurofibromatosis 2. J 45. Taylor MD, Mainprize TG, Rutka JT.
Neurol Neurosurg Psychiatry 2000; 69: 25761. Molecular insight into medulloblastoma and
33. Subach BR, Kondziolka D, Lunsford LD et al. central nervous system primitive neuroectoder-
Stereotactic radiosurgery in the management mal tumor biology from hereditary syndromes:
of acoustic neuromas associated with neuro- a review. Neurosurgery 2000; 47: 888901.
fibromatosis Type 2. J Neurosurg 1999; 90: 46. Akashi M, Koeffler HP. LiFraumeni
81522. syndrome and the role of the p53 tumor
34. Friedrich CA. Von HippelLindau syndrome suppressor gene in cancer susceptibility. Clin
a pleomorphic condition. Cancer 1999; 86: Obstet Gynecol 1998; 41: 17299.
1658-62. 47. Tuli S, Provias JP, Bernstein M. Lhermitte
35. Kondo K, Kaelin WG, Jr. The von Duclos disease: literature review and novel
HippelLindau tumor suppressor gene. Exp treatment strategy. Can J Neurol Sci 1997; 24:
Cell Res 2001; 264: 11725. 15560.
36. Richard S, Campello C, Taillandier L, Parker 48. Robinson S, Cohen AR. Cowden disease and
F, Resche F. Haemangioblastoma of the central LhermitteDuclos disease: Characterization of
nervous system in von HippelLindau disease. a new phakomatosis. Neurosurgery 2000; 46:
French VHL Study Group. J Intern Med 1998; 37183.
243: 54753. 49. Chapman MS, Perry AE, Baughman RD.
37. Sparagana SP, Roach ES. Tuberous sclerosis Cowdens syndrome, LhermitteDuclos
complex. Curr Opin Neurol 2000; 13: 1159. disease, and sclerotic fibroma. Am J
38. Cheadle JP, Reeve MP, Sampson JR, Dermatopathol 1998; 20: 4136.
Kwiatkowski DJ. Molecular genetic advances 50. Eng C. Genetics of Cowden syndrome: through
in tuberous sclerosis. Hum Genet 2000; 107: the looking glass of oncology. Int J Oncol
97114. 1998; 12: 70110.
39. Gutmann DH. Parallels between tuberous 51. McLaughlin MR, Gollin SM, Lese CM,
sclerosis complex and neurofibromatosis 1: Albright AL. Medulloblastoma and glio-
common threads in the same tapestry. Semin blastoma multiforme in a patient with Turcot
Pediatr Neurol 1998; 5: 27686. syndrome: a case report. Surg Neurol 1998;
40. Kwiatkowska J, Wigowska-Sowinska J, 49: 295301.
Napierala D, Slomski R, Kwiatkowski DJ. 52. Foulkes WD. A tale of four syndromes: famil-
Mosaicism in tuberous sclerosis as a potential ial adenomatous polyposis, Gardner syndrome,
cause of the failure of molecular diagnosis. attenuated APC and Turcot syndrome. QJM
N Engl J Med 1999; 340: 7037. 1995; 88: 85363.

364
REFERENCES

53. Taylor MD, Perry J, Zlatescu MC et al. The 67. Mooij JJ, Go KG. Cysts, cyst-like tumors and
hPMS2 exon 5 mutation and malignant other maldevelopmental tumors. In: Vecht C,
glioma. J Neurosurg 1999; 90: 94650. ed. Handbook of clinical neurology vol 68
54. Vortmeyer AO, Stavrou T, Selby D et al. Neuro-oncology part 2. Amsterdam: Elsevier,
Deletion analysis of the adenomatous polypo- 1997: 30942.
sis coli and PTCH gene loci in patients with 68. Gormley WB, Tomecek FJ, Qureshi N, Malik
sporadic and nevoid basal cell carcinoma GM. Craniocerebral epidermoid and dermoid
syndrome-associated medulloblastoma. Cancer tumours: a review of 32 cases. Acta Neurochir
1999; 85: 26627. (Wien) 1994; 128: 11521.
55. Ming JE, Roessler E, Muenke M. Human 69. Iaconetta G, Carvalho GA, Vorkapic P, Samii
developmental disorders and the Sonic hedge- M. Intracerebral epidermoid tumor: a case
hog pathway. Mol Med Today 1998; 4: 3439. report and review of the literature. Surg Neurol
56. Garcia-Higuera I, Kuang Y, DAndrea AD. The 2001; 55: 21822.
molecular and cellular biology of Fanconi 70. Potgieter S, Dimin S, Lagae L et al. Epidermoid
anemia. Curr Opin Hematol 1999; 6: 838. tumours associated with lumbar punctures
57. Joenje H, Patel KJ. The emerging genetic and performed in early neonatal life. Dev Med
molecular basis of Fanconi anaemia. Nat Rev Child Neurol 1998; 40: 2669.
Genet 2001; 2: 44657. 71. Konovalov AN, Spallone A, Pitzkhelauri DI.
58. Alter BP, Tenner MS. Brain tumors in patients Pineal epidermoid cysts: diagnosis and manage-
with Fanconis anemia. Arch Pediatr Adolesc ment. J Neurosurg 1999; 91: 3704.
Med 1994; 148: 6613. 72. Aristegui FJ, Delgado RA, Oleaga ZL,
59. German J. Bloom syndrome: a mendelian Hermosa CC. Mollarets recurrent aseptic
prototype of somatic mutational disease. meningitis and cerebral epidermoid cyst.
Medicine (Baltimore) 1993; 72: 393406. Pediatr Neurol 1998; 18: 1569.
60. Neff NF, Ellis NA, Ye TZ et al. The DNA 73. Kallmes DF, Provenzale JM, Cloft HJ,
helicase activity of BLM is necessary for the McClendon RE. Typical and atypical MR
correction of the genomic instability of bloom imaging features of intracranial epidermoid
syndrome cells. Mol Biol Cell 1999; 10: 66576. tumors. Am J Roentgenol 1997; 169: 8837.
61. Greenlee RT, Hill-Harmon MB, Taylor M, 74. Miyagi Y, Suzuki SO, Iwaki T et al. Magnetic
Thum M. Cancer Statistics, 2001. Ca Cancer J resonance appearance of multiple intracranial
Clin 2001; 51: 1536. epidermoid cysts: intrathecal seeding of the
62. Kaufman DK, Kimmel DW, Parisi JE, Michels cysts? Case report. J Neurosurg 2000; 92:
VV. A familial syndrome with cutaneous 7114.
malignant melanoma and cerebral astro- 75. Khan RB, Giri DD, Rosenblum MK, Petito FA,
cytoma. Neurology 1993; 43: 172831. DeAngelis LM. Leptomeningeal metastasis
63. Paulino AC. Trilateral retinoblastoma is the from an intracranial epidermoid cyst.
location of the intracranial tumor important? Neurology 2001; 56: 141920.
Cancer 1999; 86: 13541. 76. Mathiesen T, Grane P, Lindgren L, Lindquist
64. Svenet N, Sheridan E, Amram D et al. C. Third ventricle colloid cysts: a consecutive
Constitutional mutations of the hSNF5/INI1 12-year series. J Neurosurg 1997; 86: 512.
gene predispose to a variety of cancers. Am J 77. Urso JA, Ross GJ, Parker RK, Patrizi JD,
Hum Genet 1999; 65: 13428. Stewart B. Colloid cyst of the third ventricle:
65. Malmer B, Iselius L, Holmberg E et al. Genetic radiologicpathologic correlation. J Comput
epidemiology of glioma. Br J Cancer 2001; 84: Assist Tomogr 1998; 22: 5247.
42934. 78. Veerman EC, Go KG, Molenaar WM,
66. Chne P, Ory K, Redi D, Soussi T, Hegi ME. Amerongen AV, Vissink A. On the chemical
Functional analyses of a unique p53 germline characterization of colloid cyst contents. Acta
mutant (Y236Delta) associated with a familial Neurochir (Wien) 1998; 140: 3036.
brain tumor syndrome. Int J Cancer 1999; 82: 79. Pollock BE, Schreiner SA, Huston J III. A
1722. theory on the natural history of colloid cysts

365
MISCELLANEOUS CENTRAL NERVOUS SYSTEM NEOPLASMS AND TUMORS

of the third ventricle. Neurosurgery 2000; 46: 88. Thompson TP, Lunsford LD, Kondziolka D.
107781. Successful management of sellar and suprasellar
80. Stoeckli SJ, Bohmer A. Persistent bilateral arachnoid cysts with stereotactic intracavitary
hearing loss after shunt placement for hydro- irradiation: An expanded report of four cases.
cephalus. Case report. J Neurosurg 1999; 90: Neurosurgery 2000; 46: 151822.
7735. 89. Striano S, Meo R, Bilo L et al. Gelastic
81. Rodziewicz GS, Smith MV, Hodge CJ, Jr. epilepsy: Symptomatic and cryptogenic cases.
Endoscopic colloid cyst surgery. Neurosurgery Epilepsia 1999; 40: 294302.
2000; 46: 65560. 90. Sturm JW, Andermann F, Berkovic SF.
82. Aggleton JP, McMackin D, Carpenter K et al. Pressure to laugh: an unusual epileptic
Differential cognitive effects of colloid cysts in symptom associated with small hypothalamic
the third ventricle that spare or compromise hamartomas. Neurology 2000; 54: 9713.
the fornix. Brain 2000; 123: 80015. 91. Kahane P, Di Leo M, Hoffmann D, Munari C.
83. Pollock BE, Houston J, III. Natural history of Ictal bradycardia in a patient with a hypo-
asymptomatic colloid cysts of the third thalamic hamartoma: a stereo-EEG study.
ventricle. J Neurosurg 1999; 91: 3649. Epilepsia 1999; 40: 5227.
84. Wester K. Peculiarities of intracranial arach- 92. Rosenfeld JV, Harvey AS, Wrennall J,
noid cysts: Location, sidedness, and sex Zacharin M, Berkovic SF. Transcallosal resec-
distribution in 126 consecutive patients. tion of hypothalamic hamartomas, with
Neurosurgery 1999; 45: 7759. control of seizures, in children with gelastic
85. Maiuri F, Iaconetta G, Gangemi M. Arachnoid epilepsy. Neurosurgery 2001; 48: 10818.
cyst of the lateral ventricle. Surg Neurol 1997; 93. Rgis J, Bartolomei F, de Toffol B et al.
48: 4014. Gamma knife surgery for epilepsy related to
86. Albuquerque FC, Giannotta SL. Arachnoid hypothalamic hamartomas. Neurosurgery
cyst rupture producing subdural hygroma 2000; 47: 134351.
and intracranial hypertension: case reports. 94. Parrent AG. Stereotactic radiofrequency
Neurosurgery 1997; 41: 9515. ablation for the treatment of gelastic seizures
87. Parsch CS, Krauss J, Hofmann E, associated with hypothalamic hamartoma
Meixensberger J, Roosen K. Arachnoid cysts case report. J Neurosurg 1999; 91: 8814.
associated with subdural hematomas and 95. Petersen GM, Codori A-M. Genetic testing for
hygromas: analysis of 16 cases, long-term familial cancer. In: Vogelstein B, Kinzler KW,
follow-up, and review of the literature. eds. The genetic basis of human cancer. New
Neurosurgery 1997; 40: 48390. York: McGraw-Hill, 1998: 591-9.

366
13
Intracranial metastases

Introduction
Patients with intracranial metastases far A B
outnumber those who suffer from primary
intracranial tumors. In the USA new
symptomatic primary intracranial tumors do
not exceed 30 000 per year, whereas C
symptomatic intracranial metastases number
greater than 100 000 per year. The diagnosis of
F
a brain metastasis may be difficult for several
reasons. (1) Metastases can affect any central
nervous system (CNS) location, mimicking
both the clinical and imaging findings of
primary intracranial tumors (Fig. 13.1). (2)
Metastases are increasing in frequency as the
first site of relapse in patients whose cancers
have apparently been cured1 or are under
good systemic control.2 (3) Symptoms and E D
signs of brain metastases may appear before
the primary tumor has been discovered.3 In
some instances, a patient may suffer multiple Figure 13.1
symptomatic intracranial metastases but the Cranial metastases. Most metastases affect the brain
primary cancer is never found even after an directly by hematogenous spread to the white matter
extensive search.3 (4) Primary brain tumors are of the cerebral hemispheres (A) (see also Fig. 1.2).
The brain may be affected secondarily by a skull
more common in patients who have suffered metastasis that invades the epidural space and
other cancers than they are in the general compresses the brain. The skull metastasis may also
population. Examples include meningiomas in compress the sagittal sinus (B). The tumor may
patients with breast cancer (Chapter 6) and involve the cranial leptomeninges and invade the
gliomas in patients with LiFraumeni brain by growing down the VirchowRobin spaces
(C). A metastasis to the base of the skull may affect
syndrome (Chapter 12). (5) Other CNS
the pituitary gland (D) or cranial nerves (E) as they
processes, such as brain abscesses, can mimic exit from the skull. Subdural metastasis may cause
brain metastases and occur with increased effusions (F) that compress the brain. (Redrawn from
frequency in patients with cancer. Radiographic Posner4 with permission).

367
INTRACRANIAL METASTASES

Table 13.1
Frequency of symptomatic intracranial metastases from systemic cancers.

Primary tumors New cases No. of deaths Percentage Total no. of


USA 2001 USA 2001 with estimated deaths with
intracranial symptomatic
tumor at intracranial
autopsy, MSKCC metastases

Lung 169 500 157 400 34 37 461


Breast 193 700 40 600 30 8 526
Colon and rectum 135 400 56 700 7 2 778
Urinary organs 87 500 25 000 23 4 025
Melanoma 51 400 7 800 72 3 931
Prostate 198 100 31 500 31a 6 835
Pancreas 29 200 28 900 7 1 416
Leukemia 31 500 21 500 23b 3 461
Lymphoma (non-Hodgkins) 63 600 26 300 16b 2 945
Female genital tract 80 300 26 300 7 1 288
Brain and CNS 17 200 13 100 100 13 100
All sites 1 268 800 553 400 24 92 971

Largely skull and dura.


a

Largely leptomeningeal.
b

CNS, central nervous system; MSKCC, Memorial Sloan-Kettering Cancer Center.

images often do not differentiate metastases intracranial metastases, excluding calvarial


from infection and diagnosis must be based tumors that do not invade or compress under-
upon the clinical situation and occasionally lying brain, are found at autopsy.6 Clinical data
requires biopsy. For all these reasons, diagnoses from Memorial Sloan-Kettering Cancer Center
other than metastases must be considered in (MSKCC) indicate that patients with an
cancer patients who develop intracranial mass intracranial metastasis identified at autopsy
lesions and, conversely, metastases must be had an approximately 70% chance of having
considered in patients without known cancer suffered neurologic symptoms during life.7
who develop intracranial mass lesions. A Using the 70% factor, the final column
comprehensive review of neurologic complica- indicates the number of patients with each
tions of cancer is available.4 primary tumor likely to suffer neurologic
Table 13.1 uses extant data to estimate the symptoms from an intracranial metastasis
frequency of symptomatic intracranial metas- during their lifetime. Overall, we estimate that
tases in the USA. The first two columns of over 70 000 of the patients who die of cancer
figures are taken from American Cancer in 2001 will have had neurologic symptoms
Society 2001 data, indicating the number of from intracranial metastases during life.
new patients with specific cancers and the No epidemiologic studies describe the
number of deaths expected.5 The third column lifetime risk of either symptomatic or asymp-
estimates the percentage of patients in whom tomatic intracranial metastases in patients with

368
INTRODUCTION

Table 13.2 spreading to the CNS increases with closer


Site of primary tumors in patients with CNS proximity of the primary to the cranium.
metastases.
The age at which patients develop brain
metastases is similar to the average age of the
Primary tumor Percentage of primary cancer with the exception that for a
brain metastases given cancer, young people are more likely to
develop intracranial metastases than the
Lung 38
Non-small cell 34 elderly. Whether this is due to ascertainment or
Small cell 4 is a real finding is uncertain.
Breast 19 The numbers from Tables 13.1 and 13.2 are
Melanoma 13 clinically important for several reasons:
Renal 4
Unknown primary 1
(1) When one encounters a patient with an
Other 25
Total 100 intracranial mass, it is statistically more
likely to be metastatic than primary. The
exact likelihood is difficult to calculate.
Because 50% of metastatic lesions are
multiple (the figures vary depending on
cancer. Furthermore, the incidence varies with the primary tumor), and 5% of gliomas
the location and histology of the primary are multiple, the presence of more than
neoplasm, for example CNS metastases from one lesion suggests but does not prove
lung cancer are common; while those from metastases. An exception is lymphoma,
ovarian cancer are rare. The most common where about 40% of lesions are multiple
source of a brain metastasis is lung cancer, (Chapter 11). When one encounters one or
particularly from non-small cell lung cancer. more brain lesions in a patient with no
However, small cell lung cancer, a less common history of cancer, the likelihood of metas-
tumor, is more likely to metastasize to the CNS tasis is still relatively high. For example,
than non-small cell lung cancer (Table 13.2). as many as 10% of patients with lung
The second most common source of CNS cancer come to initial medical attention
metastasis is breast cancer,8 both because it is with neurologic symptoms from a brain
a common tumor and it often metastasizes to metastasis; this phenomenon occurs less
the brain. Melanoma is an uncommon cancer frequently with other primary cancers. In
that very often metastasizes to the brain. one series, a single supratentorial brain
Intracranial metastases differ by sex. Lung lesion in a patient not known to have
cancer is now the most common primary cancer was metastatic in 15% of patients.9
tumor in both men and women. Only about Conversely, 5% of patients with a single
1% of breast cancers occur in men. Thus, supratentorial brain tumor and a history
intracranial metastases from breast cancer in of cancer had a new primary brain tumor.
men are rare. Melanoma is more likely to Accordingly, even in the absence of a
metastasize to the brain in men than women, history of cancer, if the clinical evaluation
probably due to the greater incidence of truncal suggests the possibility of metastasis, e.g.
melanomas in men and appendicular either multiple lesions or a single spherical
melanomas in women. The risk of melanoma lesion with a regular rim of contrast

369
INTRACRANIAL METASTASES

enhancement, a focused search for a symptomatic metastatic brain tumors


primary tumor with body CT scan is because in patients with extensive systemic
warranted before directly attacking the cancer, neurologic symptoms are often
brain lesion. ignored and not evaluated. The incidence
(2) Intracranial metastases often cause devas- of brain metastasis at autopsy is also often
tating symptoms even when the patients underestimated because careful
systemic cancer is asymptomatic. Thus, neuropathologic studies were not carried
the development of a symptomatic intra- out in many series. Although the data
cranial metastasis substantially diminishes from Table 13.1 were published in 1978,
the patients quality of life, regardless of they are not likely to be replicated because
its effect on the duration of life. of the current low autopsy rate.
(3) Brain metastases are treatable. Even in (6) Brain metastases can appear at any time in
patients with far advanced cancer, early the course of a systemic cancer. In some
detection and effective treatment of patients, the brain metastasis is the
intracranial metastases may restore quality presenting symptom and may even be
of life even if survival is not increased. responsible for death before the primary
(4) The biology of metastases differs from tumor can be identified. In others, an
that of primary brain tumors: most isolated metastasis may appear in the
primary brain tumors are diffusely infil- brain 20 or more years after treatment and
trating, whereas metastases are usually apparent cure of the primary tumor. The
well-circumscribed. Consequently, metas- first situation is particularly characteristic
tases are more likely to respond to focal of lung cancer, and the second of
therapy than are primary brain tumors. In melanoma and occasionally breast cancer.
addition, brain tumors rarely metastasize Routine brain scans of newly diagnosed
so that organ transplantation from a patients with lung cancer demonstrate an
donor with a primary brain tumor, benign asymptomatic brain metastasis in 35% of
or malignant, has an extremely low risk of patients.13 One series reports a figure of
that tumor developing in the immunosup- 22% occult brain metastases at initial
pressed recipient. However, glioblastoma evaluation for lung cancer.14 Usually, brain
has rarely been transmitted from a liver metastases in the setting of known cancer
graft.10 Thus, some investigators believe typically occur 6 months to 2 years after
that patients with primary brain tumors diagnosis and are usually associated with
can serve as organ donors, unlike patients systemic tumor progression.
with other systemic cancers.11
(5) The figures in Table 13.1 come from
autopsy data at a cancer hospital where Pathophysiology of the
the ascertainment of both symptomatic
and asymptomatic brain metastases is
metastatic process
complete. In epidemiologic studies To reach the brain, a tumor that arises
reported in the literature, primary CNS elsewhere in the body must undergo a compli-
tumors equal or exceed intracranial cated and arduous series of steps.15,16 This is so
metastases.12 However, these studies difficult that only a small number of tumor
grossly underestimate the incidence of cells ever complete the process illustrated in

370
PATHOPHYSIOLOGY OF THE METASTATIC PROCESS

C F'
G
D
F
E'

D
H
D'

Figure 13.2
Pathophysiology of the metastatic process. Metastasis is a multistep process. In this schematic illustration: (A) a
malignant neoplasm arises in an organ distant from the CNS and as it grows, it develops its own vascular
supply. (B) Clones of malignant cells with metastatic potential enter blood or lymph channels and eventually
reach the venous circulation. (C) The malignant cells enter the right heart with the venous circulation and
either exit through the pulmonary artery to the lung (D) or cross a patent foramen ovale (D') to enter the
systemic circulation. Most tumors that enter the lung either arrest in the pulmonary capillary bed, grow as
pulmonary metastases and subsequently seed the pulmonary venous circulation, or (E) alternatively transverse
the pulmonary vascular bed without arresting (E') to enter the pulmonary venous circulation. Malignant clones
in the pulmonary venous circulation then enter the left heart and exit into the systemic circulation (F), along
with those cells that may have crossed a patent foramen ovale (F'). Once in the systemic circulation, the
likelihood of entering the cerebral circulation is high because, in the resting state, 1520% of cardiac output
supplies the CNS. (G, H) Tumor cells entering the cerebral circulation must then arrest in brain capillaries or
venules, cross the vessel wall, and grow within the brain. (Redrawn from Posner4 with permission.)

Fig. 13.2. A systemic cancer begins in the body lymph channels that eventually reach the
by a series of genetic steps not dissimilar from venous circulation. Because systemic tumors
those that occur in primary brain tumors enter the venous circulation and ultimately the
(Chapter 1). Once established, the tumor right side of the heart, the first capillary bed
grows, develops its own blood supply (angio- they encounter is in the lung. Accordingly,
genesis Chapter 2), invades local tissues and many patients with brain metastases either
enters the circulation by invading venules or have primary lung tumors or lung metastases

371
INTRACRANIAL METASTASES

at the time the brain lesions become


symptomatic. To reach the arterial circulation,
the tumor must either: (1) grow in the lung and
31
seed the pulmonary venous circulation, (2) 7%
47
traverse the lung capillary bed to enter the left 94 10.5%
21%
side of the heart, or (3) cross a patent foramen 12
3% 84
ovale to enter the left heart directly where 19%
tumor cells can then enter the arterial circula- 47
10.5% 66
tion (an extraordinary case report illustrates 15%
a tumor embolus that occluded the middle
cerebral artery and a second embolus within an
open foramen ovale).17
Two factors promote intracranial metastases:
(1) In the resting state, the brain receives Figure 13.3
1520% of the bodys blood flow, thus making Schematic drawing showing the distribution of single
metastases to the brain. The posterior fossa is
it likely that circulating tumor cells will reach significantly overrepresented in patients with pelvic
the brain. (2) Certain tumor cells find the brain (prostate or uterus) tumors and gastrointestinal
a propitious place for arrest and growth: the primary tumors (left), compared with patients with
seed and soil hypothesis posits that tumor cells other primary tumors (right) (p < 0.001). (Redrawn
(the seed) must find an organ (the soil) that from Delattre et al18 with permission.)
supports their growth in order to become a
metastasis. This is one of the reasons that the
probability of brain metastasis varies among itself but dura, leptomeninges, pituitary and
tumor types and that the site of a brain metas- pineal glands can also develop metastases.
tasis may vary depending on the histology of Although, as indicated above, certain primary
the primary tumor. For example, certain tumors have a predilection to metastasize to
primary tumors such as those from the kidney certain portions of the nervous system, the
and colon are more likely to metastasize to the overall distribution of brain metastasis is also
cerebellum than are lung cancers or those determined by the size of the region and its
arising elsewhere in the body18 (Fig. 13.3), vasculature. Thus, about 85% of brain metas-
although other reports disagree with this tases are found in the cerebral hemispheres,
reported preferential distribution.19 Once in the usually in the posterior portion of the
intracranial cavity, the tumor must arrest hemispheres at the watershed between the
within the capillary bed, cross the capillary middle and posterior cerebral arteries. There is
bed, grow within the organ, vascularize itself also an overrepresentation in the anterior
through the process of angiogenesis, and then border zone between the anterior and middle
grow large enough to cause symptoms. At each cerebral arteries. About 1015% of metastases
step in the metastatic process, the tumor cells are found in the cerebellum, a number
may fail, so that only 0.01% of cells that reach somewhat larger than might be expected on the
the circulation ever become metastases.16 basis of blood supply and probably represent-
Tumors may metastasize to virtually any ing the predilection for some pelvic tumors to
portion of the intracranial cavity (Fig. 13.1). metastasize to the cerebellum. Only about 3%
The most common site is the brain parenchyma of metastases are found in the brainstem.

372
PATHOPHYSIOLOGY OF THE METASTATIC PROCESS

Several unusual properties of the metastatic from uncontrolled systemic leukemia. As


process can confuse and confound the physi- chemotherapeutic agents became effective in
cian. For example: (1) The intracranial tumor controlling systemic disease, the incidence of
may be large but the primary tumor small CNS involvement began to rise to the point
or undetectable. Several reports describe where it reached almost 50% in patients with
metastatic brain tumors whose primary was acute lymphoblastic leukemia. The chemo-
undetectable even at autopsy.3 The pathogene- therapeutic agents used to treat leukemia were
sis is believed to be secretion by the primary largely water-soluble and did not cross the
tumor of both angiogenic and anti-angiogenic bloodbrain barrier; the few leukemic cells that
factors (Chapter 2). Antiangiogenic factors in reached the CNS were protected behind the
the primary tumor may control its growth bloodbrain barrier where they could prolifer-
whereas angiogenic factors secreted by the ate until they produced neurologic symptoms.
tumor may accelerate the growth of metastases. Prophylactic treatment of the CNS by radia-
(2) Metastases may be biologically different tion therapy and/or intrathecal drugs has again
from the primary tumor. Although metastatic decreased the incidence of CNS metastases
tumors are usually histologically and biologi- from leukemia to under 10%. This does not
cally similar to the primary, this is not always mean that leptomeningeal tumor, once estab-
true. Even two metastases to the same organ lished, cannot be treated by systemic water-
may differ somewhat in their biologic proper- soluble agents. Once tumor is established, the
ties. For example, markers such as estrogen bloodCSF barrier is disrupted (Chapter 2) and
receptors may be present in the primary tumor the CNS becomes accessible to treatment with
but not the metastasis. Genetic differences agents that do not normally cross the
between a primary tumor and its metastasis bloodbrain barrier. Furthermore, some
often exist.2022 For example, transforming systemic agents can penetrate an intact
growth factor (TGF) inhibits metastatic bloodbrain barrier when given in high doses.
ovarian cancer more than it does the primary Accordingly, high-dose methotrexate delivered
tumor.23 The explanation is that metastases systemically for prophylaxis can, in patients
represent clones of the primary tumor that with acute leukemia, prevent the development
may differ from the bulk of the primary. of CNS metastatic disease.
Telomerase, an enzyme present in most primary The CNS as a sanctuary site for microscopic
cancers, is also expressed in most brain metas- disease now appears to apply to breast cancer,
tases, but the concentration varies markedly. small cell lung and perhaps other cancers.2527
No correlation exists between telomerase The CNS is becoming an important site of
concentration and survival.24 (3) In patients isolated relapse in patients with these tumors.2
whose systemic tumor is otherwise controlled, When a chemosensitive tumor has relapsed in
the brain seems to be a more common site for the CNS, it does not necessarily mean that the
isolated metastatic disease. This cannot be CNS disease is resistant to the chemotherapeu-
explained by either blood flow or the nature of tic agents that controlled the systemic tumor.
the CNS microenvironment. The bloodbrain The bloodbrain barrier may have prevented
barrier probably explains this phenomenon: in those tumor cells from ever seeing a significant
the first half of the 20th century, when no concentration of the drug, thus preserving the
treatment was available for acute leukemia, tumor cells intrinsic chemosensitivity to that
CNS involvement was rare as patients died agent.28

373
INTRACRANIAL METASTASES

Table 13.3
Pathology Signs and symptoms of brain metastases at
Most metastases differ from diffuse gliomas in presentation.
that they form discrete, well-circumscribed
spherical masses. The center of large metastases Percentage
is often necrotic. The tumors may be cystic or of patients
hemorrhagic and occasionally even calcify.
Headache 24
Intraparenchymal brain metastases usually arise Hemiparesis 20
just below the cortex at the gray matterwhite Cognitive and behavioral disturbances 14
matter junction and expand by pushing normal Seizures (focal or generalized) 12
brain aside rather than invading it. A pseudo- Ataxia 7
capsule can be identified in some tumors. Other 16
No symptoms 7
Leptomeningeal tumors may appear only as
thickening or as a decrease in translucency of Data from Posner29 with permission.
the arachnoid membrane. Dural tumors may
form dural plaques or nodules.
Histopathologically, metastatic intracranial
tumors recapitulate the phenotype of the
primary from which they arose. Mitoses are relatively small metastatic lesions. Table 13.3
usually present and may be more striking in the lists the major symptoms of brain metastases at
metastasis than in the primary tumor. In a few presentation.29
instances, immunohistochemistry and genetic The major difference between the clinical
changes in the metastasis may differ somewhat signs of primary and metastatic brain tumors
from the primary. The capillaries of brain is that metastases usually grow more rapidly
metastases resemble those of the primary than even malignant primary brain tumors,
tumor rather than those of the brain. They causing subacute symptoms that evolve over a
have gap rather than tight junctions and fenes- few weeks rather than months. Sometimes, as
trated rather than continuous endothelial cell in primary brain tumors, the symptoms are
membranes. These vessels form in tumors only acute in onset and then either improve or grow
a few millimeters in diameter, disrupting the progressively worse. Acute symptoms may be
bloodbrain barrier and making even small caused by hemorrhage into the metastasis or by
lesions visible on contrast MR scans. a seizure with a prolonged postictal state. Any
intracranial metastatic tumor can bleed, but
certain primary tumors, such as melanoma,
Clinical findings thyroid, renal and choriocarcinoma, have a
propensity to bleed. Because the most common
Signs and symptoms source of brain metastasis is lung cancer, the
The symptoms and signs of brain metastasis do most common hemorrhagic metastases are
not differ significantly from those of most from lung primaries. When multiple metastases
primary brain tumors (Chapter 3). There is a are present, it is common to see simultaneous
proportionately greater amount of edema hemorrhage into many of the lesions; the
surrounding brain metastases, which often mechanism for this is unclear. When a patient
causes increased intracranial pressure despite presents with what appears to be a simple

374
CLINICAL FINDINGS

cerebral hemorrhage, one can suspect an toxic/metabolic encephalopathy and can only
underlying tumor when prominent edema be distinguished by neuroimaging. Conversely,
surrounds the hemorrhagic mass early in its some patients with brain metastases are
course, an unusual finding in primary hemor- completely asymptomatic. Because of the
rhages. Likewise, prominent contrast enhance- propensity for asymptomatic metastases,
ment immediately after the onset of patients with lung cancer or melanoma should
hemorrhage strongly indicates an underlying be evaluated with a brain MRI prior to defini-
metastasis. However, in some instances, even at tive surgery of the primary tumor.13,14 When
surgery, the hemorrhage has destroyed the tumor is found only in the brain and one
tumor so that one cannot identify the original suspects that it is a metastasis, whole-body
hemorrhagic nidus. FDG PET scans may aid in locating a small
On occasion, focal seizures can cause neuro- primary.31
logic signs that do not resolve. The seizure
probably causes ischemia in the brain immedi-
Imaging
ately surrounding the tumor by stealing its
blood supply. Metastatic tumors, primarily The most important and usually the only neces-
cause their symptoms by compressing sary diagnostic test is an MRI (Fig. 13.4).
surrounding brain. Consequently, symptoms Although the scan cannot unequivocally differ-
caused by metastasis usually respond better to entiate metastases from other lesions, certain
corticosteroids than those caused by primary abnormalities suggest metastases. Metastases
brain tumors. are usually spherical and have more regular
One rare but unique presentation of a brain margins than primary tumors. They are usually
metastasis is a three-phase presentation: a found at graywhite matter junctions in water-
patient has sudden neurologic symptoms that shed areas of brain.18 When small, they
quickly resolve, as in a transient ischemic uniformly contrast enhance, and when larger,
attack. The MR scan is usually normal. Several they may ring enhance. The ring is usually
months elapse without symptoms, then the more regular than in a primary tumor but
patient, in a subacute or gradual fashion, thicker than in an abscess. The tumor is usually
develops symptoms recapitulating the so-called surrounded by a substantial amount of edema,
transient ischemic attack. A scan now reveals a frequently more than one sees with compara-
metastasis in the area. The pathophysiology is bly sized primary brain tumors. Very small
believed to be a tumor embolus transiently metastases may appear as small dots of
occluding a brain vessel, causing reversible contrast enhancement with or without hyper-
ischemia. The embolus breaks up and enters intensity on the T2-weighted image; they
the capillary bed, where some tumor cells are usually lack surrounding edema.
able to extravasate into the brain and grow as One-half of patients have a single identifi-
a metastasis. The phenomenon is common in able brain metastasis, 20% two metastases
cardiac myxoma.30 and 13% three metastases.18 Thus, approxi-
Multiple bilateral brain metastases can have mately 80% of patients are potential candi-
a unique presentation. These patients can dates for focal therapy. There is some
develop a subacute confusional state without variation in the number of intracranial metas-
any evidence of lateralizing signs. Clinically, tases by primary tumor type. For example,
these patients are identical to those with a melanomas are more likely to cause multiple

375
INTRACRANIAL METASTASES

Figure 13.4
Multiple metastases
from lung
adenocarcinoma. Notice
the ring-like
enhancement of the
metastasis in the
cerebellum (lower right,
arrow). A massive
amount of edema
appears as hypointensity
on T1 (upper left) and
hyperintensity on FLAIR
(upper right)
surrounding the
contrast-enhancing
lesion in the right
frontal lobe. A small,
contrast-enhancing
lesion more anteriorly in
the right frontal lobe is
not surrounded by
edema (arrow).

Figure 13.5
A dural metastasis
(arrow) compressing the
brain in a patient with
prostate cancer.

brain metastases than are renal cancers. 13.5). Most are asymptomatic but some may
However, these data are not consistent enough cause symptoms from sagittal sinus compres-
to allow reliable predictions. sion or subdural effusion. Dural metastasis
Skull and dural metastases are particularly may be difficult to distinguish from a menin-
common in breast and prostate cancers (Fig. gioma (Chapter 6), particularly in women with

376
CLINICAL FINDINGS

breast cancer who have an increased incidence Table 13.4


of meningiomas.32 Other tumors, particularly Differential diagnosis of brain metastases.
subtypes of breast cancer and acute leukemia,
have a predilection to seed the leptomeninges. Primary intracranial tumor
Leptomeningeal (LM) tumor is identified by Glioma
leptomeningeal enhancement of the cerebral Meningioma
Lymphoma
and spinal meninges.33 Sometimes LM metas-
Infection
tases are suggested on MRI in patients whose Brain abscess
brain metastases are multiple, small and Bacterial
located immediately adjacent to CSF, such as at Fungal
the base of sulci or in a subependymal location. Parasitic (e.g. toxoplasmosis)
Viral encephalitis (e.g. progressive multifocal
However, even in patients with predominantly
leukoencephalopathy, herpes zoster)
cerebral symptoms from leptomeningeal Demyelinating disease
metastases, enhancement of the cerebral Multiple sclerosis
leptomeninges may be sparse. It is often useful Chemotherapy-induced
in such patients to scan the spine looking for Cerebral infarction
nodules within the spinal subarachnoid space, Cerebral hemorrhage
particularly on the cauda equina. In addition,
many such patients suffer hydrocephalus from
obstruction of CSF pathways by tumor within
the subarachnoid space and this is also visual- attacked directly. A note of caution: secretory
ized on cranial MRI. meningiomas can synthesize CEA and raise
serum levels.34
In patients with a cancer that is known to
cause immunosuppression but rarely metasta-
Differential diagnosis sizes to the nervous system, e.g. Hodgkins
Table 13.4 lists some considerations in the disease, one should suspect infection with an
differential diagnosis of a metastatic intra- opportunistic organism such as Nocardia or
cranial tumor. Toxoplasma rather than cancer metastasis (Fig.
In the appropriate setting, i.e. a patient with 13.6). Lymphoreticular neoplasms, including
a cancer known to metastasize to the brain and Hodgkins disease, some leukemias and
one or more spherically shaped contrast- lymphomas, characteristically cause immune
enhancing lesions surrounded by edema, the suppression, particularly depression of cell-
diagnosis is virtually certain and requires little mediated immunity, leading to CNS infection.
further workup. In a patient without known Patients with solid tumors do not become
cancer, but the typical appearance of a metas- immunosuppressed until after treatment with
tasis on MR of the brain, a CT of the chest, either corticosteroids or immunosuppressing
abdomen and pelvis or a PET scan may reveal chemotherapeutic agents. In patients with solid
a primary lesion. If this test, along with tumors being treated with chemotherapy, brain
biochemical markers of systemic cancer, e.g. infections with fungi, Gram-negative bacteria
CEA, PSA and CA-125,3 fail to suggest a and viruses must be considered. Viral infec-
primary tumor, further search is likely to be tions, particularly herpes simplex encephalitis,
fruitless and the brain lesion should be appear to be more common in patients with

377
INTRACRANIAL METASTASES

Figure 13.6
A brain abscess mistaken
for a metastasis. This man
with esophageal cancer had
received immunosuppressive
chemotherapy. He
developed a headache and
was found to have a
contrast-enhancing mass in
the frontal lobe. The lesion
was believed to be a
metastasis. Craniotomy
revealed a bacterial abscess.
No source was found.

Figure 13.7
A primary tumor mistaken for a metastases. A 60-year-old man with a known history of renal cell
carcinoma but without prior evidence of metastatic disease presented with headache and was found
to have a ring-enhancing lesion in the tectum of the midbrain compressing the aqueduct (arrow).
The hyperintensity surrounding the anterior horns of the ventricles (right MRI) is believed to
represent transependymal absorption of spinal fluid resulting from the hydrocephalus. A needle
biopsy of the tumor revealed not a metastatic renal cancer but a glioblastoma multiforme.

brain tumors and systemic cancer than in the Herpes zoster may cause an encephalitic
general population and must always be consid- disorder associated with immunosuppressing
ered in the differential diagnosis of an acute cancers and characterized by multiple areas of
neurologic disorder in a cancer patient.35 contrast-enhancing leukoencephalopathy. The

378
DIFFERENTIAL DIAGNOSIS

disorder may follow herpetic skin infection by usually be established by following the MR
months or years and may even occur in scan. A metastatic tumor will grow, whereas a
patients who did not have prior skin lesions or multiple sclerosis lesion will disappear in
in whom skin lesions were unrecognized.36 several weeks. Figure 13.8 illustrates the
Demyelinating disease may occasionally problem. This patient with known breast
mimic cerebral metastases. Although there are cancer treated previously with chemotherapy
isolated reports of multiple sclerosis and presented with neurologic symptoms and a
primary brain tumor coexisting,37 there is no contrast-enhancing lesion in the brainstem. The
evidence of an increased incidence of multiple lesion could not be distinguished from a breast
sclerosis in patients with systemic cancer. metastasis. However, the patient had a past
However, because acute demyelinating lesions history of neurologic episodes, and a number
may contrast enhance, a patient who has multi- of non-enhancing lesions were found in the
ple sclerosis and a coincidental cancer may brain consistent with demyelinating disease but
occasionally present with neurologic signs and not with metastases. The question then was
an enhancing lesion(s) in the brain. If the radio- whether she had two diseases, multiple sclero-
graphic diagnosis is unclear, the diagnosis can sis and metastatic cancer. Her symptoms did

Figure 13.8
Multiple sclerosis
mistaken for a
metastasis. A patient
with known breast
cancer developed
brainstem signs. An MR
showed an enhancing
lesion in the brainstem
(top left) but there were
multiple non-enhancing
lesions in the cerebral
hemispheres. Because of
the possibility that the
brainstem lesion was
due to multiple sclerosis
rather than metastatic
tumor, the patient was
followed. Two months
later, the enhancement
had largely disappeared
(bottom left).

379
INTRACRANIAL METASTASES

Figure 13.9
Encephalopathy caused by 5-FUlevamisole. This patient with Dukes B colon cancer developed a
mild hemiparesis and underwent an MR scan (left) that showed a contrast enhancing lesion in the
left hemisphere. Several other lesions did not contrast-enhance but were apparent as hyperintensity
on the T2-weighted image (right). After a diagnosis of metastatic tumor, the patient received
3000 cGy to the whole brain. She became demented and bedridden. The needle biopsy specimen
of the lesion following the radiation therapy revealed the typical demyelination reported as a side-
effect of 5-FU-levamisole therapy. Metastatic disease was not present.

not progress and a repeat scan showed disap- despite the fact that the lesions themselves
pearance of the enhancement, indicating that disappeared when the chemotherapy was
the entire process was related to demyelination. discontinued. A stereotactic needle biopsy done
Another problem occurs in patients receiving after the fact revealed demyelination and no
chemotherapy with 5-FU and levamisole, or metastatic tumor. Therefore, this distinction is
sometimes with levamisole alone.38 This critical since premature diagnosis and treat-
regimen, once commonly used for colon cancer, ment of brain metastasis can result in severe
can cause multiple contrast enhancing demyeli- neurologic damage.39
nating lesions in the brain which resolve with Overall, cerebrovascular disease is much
discontinuation of the drugs (Fig. 13.9). more common than brain tumors, either
Patients with demyelinating disease, from primary or metastatic. Accordingly, patients
either multiple sclerosis or chemotherapy, who present with acute lesions are often
appear to be more susceptible to radiation thought to have cerebral infarcts or hemor-
neurotoxicity.39 One of our patients with rhages. These disorders are more likely to
chemotherapy-induced demyelination received occur in patients with cancer, for several
standard whole-brain RT for presumed metas- reasons. Patients with cancer usually suffer
tases. She rapidly became severely demented, from a hypercoagulable state. Deep vein

380
TREATMENT

thromboses, extremely common in patients Treatment


with systemic cancer, can cause brain infarction
by paradoxical embolization.40 The most General considerations
common symptomatic complication of cancer-
The diagnostic and therapeutic approach to
induced nonbacterial thrombotic endocarditis
patients with brain metastases depends on the
is brain infarction.41 Lung cancer itself or
number and location of metastases, on the
pulmonary metastases can embolize to brain,
biology of the primary tumor, and on the extent
producing infarction and later brain metas-
of systemic disease.19,44 Obviously, different
tases.42 RT for head and neck tumors may
approaches are required in patients known to
accelerate arteriosclerosis and can lead to
have cancer from those not known to have
carotid occlusion and stroke.
cancer. Algorithms outlining the diagnostic
The diagnosis of infarction is generally not
and therapeutic approaches in some of these
difficult. A scan during the acute episode of
situations are presented in Figs 13.10 and
cerebral infarction does not contrast enhance
13.11. The therapeutic approach to patients
and the lesion may be visible only on diffusion-
with brain metastases is similar to that of
weighted MR images, a pattern very sensitive
primary brain tumors, and is divided into
for the diagnosis of stroke. The lesion itself is
supportive and definitive treatments (Table
far more diffuse than one sees with well-
13.5) (Chapter 4).
circumscribed brain metastases. Cerebral
hemorrhage may be more difficult. Patients
receiving chemotherapy for cancer are often Supportive therapy
thrombocytopenic and susceptible to brain
As indicated in Chapter 4, supportive therapy
hemorrhage. Others may be hypercoagulable
may include corticosteroids, anticonvulsants,
and suffer venous sinus occlusion, leading to
psychotropic drugs and anticoagulants.
hemorrhagic infarction.
Corticosteroids are necessary only for
If there is any question about the diagnosis,
symptomatic metastases, where they usually
a biopsy is essential. The most telling finding
produce a dramatic response, and are also
comes from the controlled trial of brain metas-
tases conducted by Patchell et al.43 A series of
patients with known cancer and a single Table 13.5
contrast-enhancing lesion of the brain were Treatment of brain metastases.
randomized to receive either surgical resection
or stereotactic biopsy followed by RT. Despite
Supportive Definitive
the presence of cancer (usually lung cancer)
and careful examination by experienced Corticosteroids Surgery
neuro-oncologists, 11% of the patients were Antibiotics Radiation
discovered not to have metastatic lesions on Anticonvulsants Whole-brain RT (WBRT)
Anticoagulants Radiosurgery
pathologic examination. These lesions included
Antidepressants Whole-brain
high-grade and low-grade primary brain RT + radiosurgery
tumors and inflammatory or immune-mediated Chemotherapy
lesions. Accordingly, when there is any Systemic
question about the diagnosis, a biopsy is Local
essential.

381
INTRACRANIAL METASTASES

Single lesion

Known cancer No known cancer

Cancer controlled CT chest, abdomen,


or uncontrolled pelvis, biochemical markers

Surgically accessible Surgically inaccessible Cancer found No cancer

Resect Consider biopsy Biopsy primary Surgically Not surgically


accessible accessible

? Post-Op WBRT Chemosensitive Not chemosensitive Resect Biopsy


(see text) or asymptomatic or not asymptomatic

Systemic chemo WBRT Metastasis No metastasis

Response No response Lesion resected Lesion not resected Treat appropriately

+/ WBRT RS +/ WBRT Consider WBRT >3 cm <3 cm


+/ RS boost

Extensive WBRT +/ RS +/ WBRT


systemic disease local boost

Chemosensitive Not chemosensitive

Figure 13.10
Approach to the patient with a single lesion. RS, Radiosurgery; WBRT, whole-brain radiation therapy.

indicated during the course of definitive treat- Pneumocystis carinii infection are indicated.
ment with surgery or radiation. Corticosteroids However, these drugs often cause side-effects,
are not required for asymptomatic patients particularly platelet suppression. Also, as
with small metastases unless RT or surgery is indicated in Chapter 4, anticonvulsants are not
imminent. We do not routinely use gastric used prophylactically. We recommend them
protection for patients receiving corticos- only if the patient has had a seizure. Deep vein
teroids. In patients expected to be on corticos- thrombosis is a common complication of
teroids for a prolonged period, e.g. more than patients with cancer, particularly those with
6 weeks, prophylactic antibiotics to prevent brain metastases. Prophylactic treatment is

382
TREATMENT

Multiple lesions

Known cancer No known cancer

Primary controlled Extensive systemic CT chest, abdomen,


or controllable disease pelvis biochemical markers

4 or more 2 or 3 WBRT +/ Cancer No cancer


lesions lesions systemic chemo found found

WBRT Resect RS to inaccessible Biopsy 1 large No resectable


+/ chemo accessible <3 cm +/ WBRT primary accessible lesion lesion

WBRT +/ Resect Biopsy


systemic chemo

Metastasis No metastasis

Treat
appropriately

Figure 13.11
Approach to the patient with multiple lesions. RS, Radiosurgery; WBRT, whole-brain radiation therapy; chemo,
chemotherapy.

probably not indicated, but patients must be is superior to whole-brain RT alone, both in
followed carefully for symptoms and treated preventing brain relapse and in improving
promptly. Psychologic complications of meta- quality of life43,45 (Fig. 13.12). A third trial46
static cancer are common; antidepressants are found no benefit. In our opinion, in patients
effective in relieving depression. Even when the whose systemic disease is quiescent or control-
physician believes that the depression is ratio- lable, surgical removal of a single brain metas-
nal and justified, these drugs should be utilized tasis substantially improves survival. Two-year
as they effectively ameliorate reactive depres- survivals are 1520%, depending on the
sion. primary tumor; 5-year survivals are about
10%, and occasional cures are reported.47
There are now retrospective data to support
Definitive treatment
the resection of two or even three metastases,
Surgery with the outcome comparable to a surgically
Two controlled trials clearly indicate that for treated single brain metastasis. The operative
patients with single brain metastases, surgical mortality is 13%; neurologic worsening, often
removal followed by whole-brain radiotherapy transient, occurs in 510%. Patients with

383
INTRACRANIAL METASTASES

Figure 13.12
A single brain
metastasis with both
cyst and calcification
in a patient with
ovarian cancer. The
tumor responded to
surgical resection.
Note that the
calcium is hyperdense
on CT and
hypointense on MRI.

cerebellar lesions have a worse prognosis than Radiation therapy


those with hemispheral lesions,48 and are at
increased risk for subsequent leptomeningeal The time-honored treatment of brain metas-
metastases. tases has been whole-brain radiation, deliver-
A recent controlled trial indicates that ing 30 Gy in 10 fractions. For patients with
patients with a single metastasis who receive extensive systemic disease or multiple brain
postoperative radiotherapy have fewer recur- metastases, this still remains the best option.
rences of cancer in the brain and are less likely Most patients have at least a transient
to die of neurologic causes than similar patients response to radiation; they symptomatically
treated with surgical resection alone.49 improve with steroids and radiotherapy, and
However, overall survival was comparable are less likely to die of their neurologic
between the groups because patients with disease than are patients who do not receive
controlled cerebral disease died of progressive this treatment (Fig. 13.13). Survival remains
systemic tumor. Accordingly, the best extant short, with a median of 46 months, because
data suggest that a patient with a single, surgi- most patients die of uncontrolled systemic
cally accessible brain metastasis, whose tumor. Even those patients who respond to
systemic disease is not imminently terminal, RT and whose tumors are controlled,
should have that metastasis removed and however, are at risk for reseeding of the brain
receive postoperative whole-brain RT. from the systemic cancer. In occasional
However, because of the toxicity of whole- patients, RT actually sterilizes the brain
brain RT, particularly in the older age group, metastases and they do not recur. Because of
many physicians do not use it in the immedi- late-delayed radiation toxicity in those
ate postoperative period, but reserve radiation patients whose systemic prognosis is more
until relapse. than a year, particularly after resection of a

384
TREATMENT

single brain metastasis, we recommend a dose comes from two sources. First, prophylactic
of 4050 Gy given in 1.82.0-Gy fractions.50 irradiation of the brain in patients with small cell
The lower dose per fraction diminishes radia- lung cancer substantially decreases the subse-
tion toxicity. Cognitive deficits from RT are quent development of CNS metastases. This is
more severe in the very young, the elderly because micrometastases were present at the time
and those who receive extensive chemother- of radiation and were eliminated by it. Second,
apy. Nevertheless, even young adults often a controlled trial demonstrated that postopera-
complain of memory loss after whole-brain tive whole-brain RT decreases relapse in the
RT. brain not only at the surgical site but through-
The reason for using whole-brain RT in brain out the brain, indicating that small metastatic
metastases as opposed to focal RT is because deposits were eliminated by the radiation.49
micrometastases may be present elsewhere in the
brain, even when one can visualize only one or Radiosurgery
two metastatic tumors. Whole-brain radiation Radiosurgery is increasingly being used instead
will prevent the growth of these microscopic as of surgery for the treatment of single or even
well as macroscopic tumors. Evidence for this multiple brain metastases.5153 The treatment

Figure 13.13
Response of a small
cell lung cancer to
whole-brain radiation
therapy. The patient
had multiple
metastases, one of
which was
surrounded by a
massive amount of
edema (top panels).
Two months after
radiation therapy, the
tumors had
disappeared and the
edema had largely
cleared (bottom
panels).

385
INTRACRANIAL METASTASES

appears to be most effective against those contradicts the controlled trial of RT after
tumors that are relatively resistant to conven- surgery.49 A randomized trial is necessary to
tional external beam radiation therapy, such as resolve this question. Until randomized trials
melanoma54 and renal cell carcinoma.55,56 The prove the superiority of one treatment over the
reason is unclear, but the higher dose per other, we believe that surgery is the preferred
fraction may eliminate otherwise resistant treatment for accessible lesions in good-
tumor cells. Some report 78% tumor control prognosis patients. Surgery not only treats the
with < 10% complications.52 Tumor control is lesion, but it also establishes the diagnosis,
defined as disappearance, decrease in size or which has a 10% error rate43 based on
stability of the lesion. Other reports indicate neuroimaging alone. However, there are situa-
that local control is maintained over 2 years in tions in which radiosurgery is the therapy of
61% of patients.57 MRI may show a transient first choice, e.g. elderly patients with multiple
increase in tumor enhancement which does lesions which cannot all be resected (e.g. three
not necessarily indicate tumor growth.57 or four), or patients with brainstem metas-
Radiosurgery has advantages over surgery in tases.60
that it is relatively non-invasive, not even
requiring hospitalization in most instances, and Chemotherapy
can often reach areas that are surgically Chemotherapy is increasingly being recog-
inaccessible. Radiosurgery has the disadvantage nized as efficacious for brain metastases from
that it is not useful for tumors larger than chemosensitive systemic cancers (Fig. 13.14).
34 cm. Theoretically, radiosurgery should be These include germ cell tumors, breast cancer,
a much better technique for brain metastases small cell lung cancer and some others.61,62
than gliomas, because metastases are well Chemotherapy can be applied either systemi-
circumscribed and do not infiltrate widely into cally or locally into the brain tumor bed.63
normal brain. At least one non-randomized The latter approach is still highly experimen-
comparative study of surgery versus radio- tal and its efficacy is unclear. In appropriate
surgery suggests that survival is equal.58 patients, one should probably use chemother-
However, a case-control retrospective trial of apy to treat brain metastases that have
the two modalities found that patients lived disrupted the bloodbrain barrier, i.e. contrast
longer and did better with surgical resection. enhance, before RT is applied. In addition,
This was partially attributed to the propensity systemic chemotherapy is probably best used
for radiosurgery to cause brain necrosis that in in patients who do not require corticosteroids,
itself is symptomatic and may require surgery since those drugs restore bloodbrain barrier
as treatment. A recent retrospective study function and decrease the amount of water-
comparing radiosurgery with surgery plus soluble drug that can reach the tumor. There
whole-brain RT yielded similar periprocedure are several reasons for administering
morbidity (7.7% versus 8.9%), mortality chemotherapy before radiation: (1) it is useful
(1.6% versus 1.2%) and 1-year survival (53% to be able to judge tumor response to
versus 43% not statistically significant) rates. chemotherapy before initiating RT if the
Local control rates were also similar at 1 year tumor is responsive, the chemotherapy can be
(75% versus 83%).58 The report suggests that continued even after radiation is given. (2)
whole-brain RT may not be necessary after After RT the blood supply to the tumor is
radiosurgery. Others agree,58,59 although this disrupted and may decrease the access of

386
TREATMENT

Figure 13.14
Response to
chemotherapy. A
patient with known
ovarian cancer
developed a brain
metastasis with only
mild headache as the
symptom, but
because she was
about to undergo
systemic
chemotherapy, it was
elected to follow her
on treatment. The
pre-chemotherapy
(left panels) and
post-chemotherapy
images show a clear
response of the
tumor. The tumor
was well controlled
for about 2 years
before her systemic
disease and brain
metastasis escaped
treatment.

chemotherapy to the metastases. (3) Some patients with measurable non-small cell lung
evidence suggests that chemotherapy delivered cancer, 3 (7%) had a complete response and
before radiation is less neurotoxic than the 10 (25%) a partial response. None of 8
converse. The evidence is best for methotrex- melanomas responded.64
ate but may apply to other drugs as well. Chemotherapy is not without its neurologic
Usually, the chemotherapeutic agent is complications. A recent study of women receiv-
chosen based on the underlying primary. ing adjuvant chemotherapy for breast cancer
However, there are circumstances in which indicated that there was a risk of late cognitive
there appears to be a good response of a impairment when compared with patients not
variety of metastatic tumor types to a single receiving such chemotherapy. The cognitive
protocol. Among 56 patients with brain metas- impairment was not caused by anxiety, depres-
tases from breast cancer, cisplatin and etopo- sion or fatigue or related to time since treat-
side gave a complete response in 7 (13%) and ment; only some patients were aware of their
a partial response in 14 (21%). Among 43 neuropsychological disorder.65

387
INTRACRANIAL METASTASES

Specific treatment situations ganglia) lesions. Such patients are reported


to have excellent local control of tumor
The number of variables both within the CNS with radiosurgery. Current data would
and outside the CNS is so great that each suggest that if local control is achieved
patient is unique. Nevertheless, a number of with radiosurgery, patients should also
common scenarios are outlined and guidelines receive whole-brain RT to help prevent the
suggested for each. What follows are tentative appearance of tumors in other areas of the
recommendations based on the current state of brain. However, whole-brain RT is often
knowledge. withheld in older patients because of the
high incidence of cognitive impairment.
(1) The patients systemic tumor has been For asymptomatic patients, particularly
successfully treated or has a relatively good those with evidence of disease elsewhere in
prognosis (more than 1 year). The patient the body, and with relatively chemosensi-
has either a single, surgically accessible tive tumors (e.g. breast cancer, small cell
brain metastasis or, in especially good-risk lung cancer), systemic chemotherapy may
patients, two or rarely three accessible be useful as initial treatment. Systemic
metastatic lesions. Such patients have the chemotherapy may achieve control of
best prognosis for preserved nervous the brain and systemic tumors without
system function and survival if focal resorting to radiation. If chemotherapy
therapy is delivered to the brain metas- succeeds, it can be continued. If it fails,
tases. No controlled trials have yet radiosurgery can be substituted. We do
addressed whether surgical removal of the not use chemotherapy as the first treat-
tumor or radiosurgery is the superior ment in symptomatic brain metastases,
modality. Surgery is more costly and the because surgery and radiotherapy control
morbidity is similar; however, most compli- symptoms much faster than chemotherapy
cations occur early after surgery and later and with greater reliability.
after radiosurgery.66 If the lesion is greater (3) The patient has poor-prognosis systemic
than 34 cm in diameter, it should proba- disease and multiple brain metastases. In
bly be removed. One report indicates that this situation, whole-brain RT, 30 Gy in
in patients with one large metastasis and 10 fractions, along with corticosteroids,
one or more small lesions (< 2 cm), surgery will usually control brain symptoms for
of the large lesion followed by whole-brain the duration of the patients life. If one of
RT yields results equal to surgery of a the multiple brain lesions is large and
single metastasis followed by RT.67 In one symptomatic, removal of the mass may
randomized trial, postoperative whole- improve the patients quality of life even
brain RT improved control of the CNS though it may not extend the duration of
disease, but did not improve overall survival.
survival.49 However, not all patients relapse (4) A patient previously treated for a brain
after surgery and the cognitive side-effects metastasis improves and then relapses
of whole-brain RT require one to give each after several months to a year. All of the
patient individual consideration. treatment modalities and considerations
(2) A good-risk systemic patient with 13 that apply to the initial treatment of a
small but surgically inaccessible (i.e. basal CNS metastasis also apply to recurrence.

388
TREATMENT

In general, treatment after recurrence is cancer substantially diminishes the likeli-


less effective than initial treatment, but hood of subsequent brain metastases.
nonetheless may improve both quality and Some studies suggest that, despite this,
length of life. A patient in good systemic there is no increase in overall survival;
condition with a single surgically accessi- other studies demonstrate that survival is
ble brain metastasis that recurs either at prolonged.68 Unfortunately, prophylactic
the site of initial craniotomy or at a brain irradiation comes at a cost, particu-
distance should be considered for a second larly in patients who have received or will
craniotomy. In rare instances, even a third receive systemic chemotherapy, which
operation for a locally recurrent tumor includes all patients with small cell lung
may help the patient. However, long-term cancer. Many patients suffer significant
survival after a second resection is rare. In cognitive dysfunction, although not
patients with surgically inaccessible lesions always to a disabling degree. The longer
who are poor candidates for surgery, the patient survives, the more likely it is
radiosurgery can be used for lesions not that cognitive dysfunction will become
previously treated with radiosurgery. Prior manifest since this is often a delayed
whole-brain RT does not preclude the use complication of treatment.69
of radiosurgery in selected instances. A (6) Malignant germ cell tumors may present
second course of whole-brain RT, if brain with brain metastases, although other
metastases have been stable for 6 months disease, especially bulky thoracic disease,
or longer and then recur, may also be is always present.70 Chemotherapy may be
beneficial and usually can be administered effective and sufficient treatment for the
safely. In some patients who fail brain metastasis.
chemotherapy, retreatment with different
chemotherapeutic agents may give a
Complications of treatment
response. With a repeat course of whole-
brain RT, 2025 Gy in 1.82.0-Gy The neurologic complications that result from
fractions, most patients improve at least treatment of intracranial metastases are in
transiently, and occasional patients survive general the same as those resulting from treat-
6 months to a year. The likelihood of ment of primary intracranial tumors (Chapter
patients developing cognitive dysfunction 4). Operative mortality from craniotomy for
or other signs of radiation toxicity are treatment of brain metastases is under 3%,
increased by the second course of radia- usually occurring only in patients debilitated
tion, but most patients do not live long from their primary cancer. Many patients with
enough to suffer this side effect. CNS metastases have advanced systemic
(5) The patient has small cell lung cancer or disease, are immunosuppressed from prior
another cancer with a high propensity for chemotherapy and may be poorly nourished.
brain metastases. A staging cranial MR Thus, wound healing and postoperative infec-
scan is negative. A number of studies have tions are somewhat more common following
addressed the question of prophylactic surgery for CNS metastases than for primary
brain RT in this situation. Strong evidence tumors. Furthermore, hypercoagulability
suggests that prophylactic radiation of the caused by the underlying cancer or hypocoag-
brain of patients with small cell lung ulability caused by prior treatment predisposes

389
INTRACRANIAL METASTASES

such patients to clotting disorders, e.g. venous patients for cognitive impairment. Accordingly,
thrombosis, and bleeding from thrombo- we divide patients who are to receive whole-
cytopenia. The operative morbidity, including brain RT into prognostic groups: those with a
worsening of neurologic symptoms (mostly good systemic prognosis, i.e. likely to live more
transient) is about 510%. Because the than 1 year, are treated in 1.82.0-Gy fractions
majority of tumors are well demarcated from to a total dose of 4050 Gy; those who have a
surrounding normal brain, neurologic worsen- poor prognosis (less than 6 months) are treated
ing is less commonly encountered after surgery in 3-Gy fractions to a total of 30 Gy. However,
for metastasis than for glioma. Thus, most even the smaller dose fractions may cause
patients tolerate craniotomy well and leave the cognitive dysfunction in some patients.
hospital within 4 or 5 days. Even patients
debilitated with widespread systemic disease
often respond well to craniotomy. However,
the local recurrence rate of 30% without
Prognosis
postoperative whole-brain RT suggests that, Untreated symptomatic brain metastases
despite gross total resection, microscopic tumor usually cause death within 1 or 2 months.
often remains after surgery.49 Corticosteroids can control symptoms for
Radiosurgery also has its complications. several weeks to a few months. Whole-brain
Headache, nausea, vomiting, seizures and even RT yields a median survival of 46 months
transient worsening of neurologic symptoms with 80% or more having neurologic improve-
may follow within hours of radiosurgery. The ment. Patients with a single brain metastasis
symptoms are usually easily controlled with treated with surgery and whole-brain RT have
steroids. Local tumor control is defined by a median survival of 40 weeks and some live
radiation oncologists as cessation of tumor longer than a year. Prolonged survival can also
growth; tumor shrinkage is not required. be seen after radiosurgery. Rarely, patients have
Accordingly, the residual tumor, or the long survivals and appear to be cured.
necrosis produced by radiosurgery, may lead The prognosis depends in part on the
to edema with symptoms that are not easily histology of the primary tumor, on the stage
controlled. Radionecrosis typically becomes of systemic disease, and on the number and
symptomatic several weeks to months follow- location of CNS metastases. Chemo- and
ing radiosurgery. This may require further radiosensitive tumors have a better prognosis
surgery to alleviate symptoms or chronic corti- than chemo- and radioresistant tumors.
costeroid use with its own attendant compli- Patients with single brain metastases that are
cations. This is one of the disadvantages of surgically removed and whose systemic disease
radiosurgery when compared with surgical is well controlled have a 1015% 5-year
removal, where there is no gross tumor tissue survival and an occasional apparent cure.
left behind. Whether radiosurgery can also give such long
Whole-brain RT has few immediate side- survival is unclear. Accordingly, good-risk
effects, but in patients who survive more than patients should be approached aggressively in
a year or in those who have been heavily an attempt to eliminate the focal disease and
pretreated with chemotherapy, dementia is a prevent recurrence elsewhere in the brain.
common delayed side-effect. In addition, older In a recent study of over 1000 patients with
patients are at greater risk than younger brain metastases,71 the overall median survival

390
REFERENCES

was 3.4 months, with a 1-year survival of 12% 7. Cairncross JG, Kim J-H, Posner JB. Radiation
and a 2-year survival of 4%. Steroids only therapy of brain metastases. Ann Neurol 1980;
7: 52941.
yielded a median survival of 1.3 months, which
8. Lentzsch S, Reichardt P, Weber F, Budach V,
increased to 3.6 months in patients treated Drken B. Brain metastases in breast cancer:
with RT and to 8.9 months in patients treated Prognostic factors and management. Eur J
with surgery followed by radiotherapy. In Cancer [A] 1999; 35: 5805.
addition to treatment, performance status, 9. Voorhies RM, Sundaresan N, Thaler HT. The
response to steroids and systemic tumor single supratentorial lesion. An evaluation of
preoperative diagnostic tests. J Neurosurg
activity were important prognostic factors. 1980; 53: 3648.
Somewhat less important were the site of the 10. Jonas S, Bechstein WO, Lemmens HP et al.
primary tumor, the age of the patient and the Liver graft-transmitted glioblastoma multi-
number of brain metastases. Treatment data forme. A case report and experience with 13
must be interpreted carefully, particularly with multiorgan donors suffering from primary
cerebral neoplasia. Transpl Int 1996; 9: 4269.
respect to the poor-prognosis patients who
11. Chui AK, Herbertt K, Wang LS et al. Risk of
receive less aggressive treatment. Many patients tumor transmission in transplantation from
receive less aggressive treatment because they donors with primary brain tumors: an
are substantially less well than those patients Australian and New Zealand registry report.
who are treated vigorously. Sometimes, statis- Transplant Proc 1999; 31: 12667.
tical analysis does not fully account for these 12. Counsell CE, Grant R. Incidence studies of
primary and secondary intracranial tumors: a
differences. systematic review of their methodology and
results. J Neurooncol 1998; 37: 24150.
13. Yokoi K, Kamiya N, Matsuguma H et al.
References Detection of brain metastasis in potentially
operable non-small-cell lung cancer a
1. Boogerd W, Hart AAM, Tjahja IS. Treatment comparison of CT and MRI. Chest 1999; 115:
and outcome of brain metastasis as first site 7149.
of distant metastasis from breast cancer. 14. Earnest F, Ryu JH, Miller GM et al. Suspected
J Neurooncol 1997; 35: 1617. non-small cell lung cancer: Incidence of occult
2. Robnett TJ, Machtay M, Stevenson JP, brain and skeletal metastases and effectiveness
Algazy KM, Hahn SM. Factors affecting the of imaging for detection pilot study.
risk of brain metastases after definitive Radiology 1999; 211: 13745.
chemoradiation for locally advanced non- 15. Fidler IJ. The PagetEwing Award Lecture
small-cell lung carcinoma. J Clin Oncol 2001; The seed and soil hypothesis at the millen-
19: 13449. nium: Recent advances in the pathogenesis of
3. Giordana MT, Cordera S, Boghi A. Cerebral cancer metastasis. Clin Exp Metastasis 1999;
metastases as first symptom of cancer: a 17: 731.
clinico-pathologic study. J Neurooncol 2000; 16. Liotta LA, Kohn EC. Invasion and Metastases.
50: 26573. In: Bast RC, Kufe DW, Pollock RE et al, eds.
4. Posner JB. Neurologic Complications of Cancer Medicine. Hamilton: BC Decker, 2000:
Cancer. Philadelphia: FA Davis, 1995. 12131.
5. Greenlee RT, Hill-Harmon MB, Taylor M, 17. Kearsley JH, Tattersall MH. Cerebral
Thum M. Cancer Statistics, 2001. Ca Cancer J embolism in cancer patients. Q J Med 1982;
Clin 2001; 51: 1536. 51: 27991.
6. Posner JB, Chernik NL. Intracranial metastases 18. Delattre J-Y, Krol G, Thaler HT, Posner JB.
from systemic cancer. Adv Neurol 1978; 19: Distribution of brain metastases. Arch Neurol
57587. 1988; 45: 7414.

391
INTRACRANIAL METASTASES

19. Graf AH, Buchberger W, Langmayr H, Schmid 30. Alvarez-Sabn J, Lozano M, Sastre-Garriga J et
KW. Site preference of metastatic tumours al. Transient ischaemic attack: A common
of the brain. Virchows Arch A Pathol Anat initial manifestation of cardiac myxomas. Eur
Histopathol 1988; 412: 4938. Neurol 2001; 45: 16570.
20. Morita R, Fujimoto A, Hatta N, Takehara K, 31. Lassen U, Daugaard G, Eigtved A, Damgaard
Takata M. Comparison of genetic profiles K, Friberg L. 18F-FDG whole body positron
between primary melanomas and their metas- emission tomography (PET) in patients with
tases reveals genetic alterations and clonal unknown primary tumours (UPT). Eur J
evolution during progression. J Invest Cancer [A] 1999; 35: 107682.
Dermatol 1998; 111: 91924. 32. Markopoulos C, Sampalis F, Givalos N,
21. Ruan S, Fuller G, Levin V, Bruner JM, Zhang Gogas H. Association of breast cancer with
W. Detection of p21WAF1/Cip1 in brain metas- meningioma. Eur J Surg Oncol 1998; 24:
tases. J Neurooncol 1998; 37: 2238. 3324.
22. Hampl M, Hampl JA, Schwarz P et al. 33. Freilich RJ, Krol G, DeAngelis LM.
Accumulation of genetic alterations in brain Neuroimaging and cerebrospinal fluid cytology
metastases of sporadic breast carcinomas is in the diagnosis of leptomeningeal metastasis.
associated with reduced survival after metasta- Ann Neurol 1995; 38: 517.
sis. Invasion Metastasis 1999; 18: 8195. 34. Buhl R, Hugo HH, Mihajlovic Z, Mehdorn
23. Hurteau JA, Allison B, Sutton GP et al. HM. Secretory meningiomas: clinical and
Transforming growth factor- differentially immunohistochemical observations. Neuro-
inhibits epithelial ovarian carcinoma cells from surgery 2001; 48: 297301.
primary and metastatic isolates without up- 35. Schiff D, Rosenblum MK. Herpes simplex
regulation of p21WAF1. Cancer 1999; 85: 18105. encephalitis (HSE) and the immunocompro-
24. Kleinschmidt-DeMasters BK, Shroyer AL, mised: a clinical and autopsy study of HSE in
Hashizumi TL et al. Part I. Telomerase levels the settings of cancer and human immunodefi-
in human metastatic brain tumors show four- ciency virus-type 1 infection. Hum Pathol
fold logarithmic variability but no correlation 1998; 29: 21522.
with tumor type or interval to patient demise. 36. Weaver S, Rosenblum MK, DeAngelis LM.
J Neurol Sci 1998; 161: 11623. Herpes varicella-zoster encephalitis in immuno-
25. Freilich RJ, Seidman AD, DeAngelis LM. compromised patients. Neurology 1999; 52:
Central nervous system progression of 1935.
metastatic breast cancer in patients treated 37. Khan OA, Bauserman SC, Rothman MI,
with paclitaxel. Cancer 1995; 76: 2326. Aldrich EF, Panitch HS. Concurrence of
26. Dhote R, Beuzeboc P, Thiounn N et al. High multiple sclerosis and brain tumor: clinical
incidence of brain metastases in patients considerations. Neurology 1997; 48: 13303.
treated with an MVAC regimen for advanced 38. Figueredo AT, Fawcet SE, Molloy DW,
bladder cancer. Eur Urol 1998; 33: 3925. Dobranowski J, Paulseth JE. Disabling
27. Kramer K, Kushner B, Heller G, Cheung NKV. encephalopathy during 5-fluorouracil and
Neuroblastoma metastatic to the central levamisole adjuvant therapy for resected
nervous system The Memorial Sloan- colorectal cancer: A report of two cases.
Kettering Cancer Center experience and a liter- Cancer Invest 1995; 13: 60811.
ature review. Cancer 2001; 91: 15109. 39. Peterson K, Rosenblum MK, Powers JM,
28. Castiglione-Gertsch M, Tattersall M, Hacking Alvord EC, Walker RW, Posner JB. Effect of
A et al and the International Breast Cancer brain irradiation on demyelinating lesions.
Study Group. Retreating recurrent breast Neurology 1993; 43: 210512.
cancer with the same CMF-containing regimen 40. Yeung M, Khan KA, Shuaib A. Transcranial
used as adjuvant therapy. Eur J Cancer [A] Doppler ultrasonography in the detection of
1997; 33A: 23215. venous to arterial shunting in acute stroke
29. Posner JB. Brain metastases: 1995. A brief and transient ischaemic attacks. J Neurol
review. J Neurooncol 1996; 27: 28793. Neurosurg Psychiatry 1996; 61: 4459.

392
REFERENCES

41. Rogers LR, Cho E-S, Kempin S, Posner JB. alone for patients with multiple brain metas-
Cerebral infarction from non-bacterial throm- tases. Int J Radiat Oncol Biol Phys 1999; 45:
botic endocarditis. Clinical and pathological 42734.
study including the effects of anticoagulation. 54. Lavine SD, Petrovich Z, Cohen-Gadol AA et
Am J Med 1987; 83: 74656. al. Gamma knife radiosurgery for metastatic
42. Graus F, Rogers LR, Posner JB. Cerebro- melanoma: An analysis of survival, outcome,
vascular complications in patients with cancer. and complications. Neurosurgery 1999; 44:
Medicine 1985; 64: 1635. 5964.
43. Patchell RA, Tibbs PA, Walsh JW. A random- 55. Mori Y, Kondziolka D, Flickinger JC, Logan
ized trial of surgery in the treatment of single T, Lunsford LD. Stereotactic radiosurgery for
metastases to the brain. N Engl J Med 1990; brain metastasis from renal cell carcinoma.
322: 494500. Cancer 1998; 83: 34453.
44. Nieder C, Nestle U, Motaref B et al. Prognostic 56. Becker G, Duffner F, Kortmann R et al.
factors in brain metastases: Should patients be Radiosurgery for the treatment of brain
selected for aggressive treatment according to metastases in renal cell carcinoma. Anticancer
recursive partitioning analysis (RPA) classes? Res 1999; 19: 16117.
Int J Radiat Oncol Biol Phys 2000; 46: 57. Peterson AM, Meltzer CC, Evanson EJ,
297302. Flickinger JC, Kondziolka D. MR imaging
45. Vecht CJ, Haaxma-Reiche H, Noordijk EM, response of brain metastases after gamma knife
et al. Treatment of single brain metastasis: stereotactic radiosurgery. Radiology 1999;
Radiotherapy alone or combined with neuro- 211: 80714.
surgery? Ann Neurol 1993; 33: 58390. 58. Muacevic A, Kreth FW, Horstmann GA et al.
46. Mintz AH, Kestle J, Rathbone MP et al. A Surgery and radiotherapy compared with
randomized trial to assess the efficacy of gamma knife radiosurgery in the treatment of
surgery in addition to radiotherapy in patients solitary cerebral metastases of small diameter.
with a single cerebral metastasis. Cancer 1996; J Neurosurg 1999; 91: 3543.
78: 14706. 59. Sneed PK, Lamborn KR, Forstner JM et al.
47. Arbit E, Wronski M. The treatment of brain Radiosurgery for brain metastases: Is whole
metastases. Neurosurgery Quarterly 1995; 5: brain radiotherapy necessary? In: J Radiat
117. Oncol Biol Phys 1999; 43(3): 54958.
48. Wronski M, Arbit E. Resection of brain metas- 60. Huang CF, Kondziolka D, Flickinger JC,
tases from colorectal carcinoma in 73 patients. Lunsford LD. Stereotactic radiosurgery for
Cancer 1999; 85: 167785. brainstem metastases. J Neurosurg 1999; 91:
49. Patchell RA, Tibbs PA, Regine WF et al. 5638.
Postoperative radiotherapy in the treatment of 61. Cormio G, Gabriele A, Maneo A et al.
single metastases to the brain: a randomized Complete remission of brain metastases from
trial. JAMA 1998; 280: 14859. ovarian carcinoma with carboplatin. Eur J
50. DeAngelis LM. Management of brain metas- Obstet Gynecol Reprod Biol 1998; 78: 913.
tases. Cancer Invest 1994; 12: 15665. 62. Franciosi V, Cocconi G, Michiara M et al.
51. Seung SK, Sneed PK, McDermott MW et al. Frontline chemotherapy with cisplatin and
Gamma knife radiosurgery for malignant etoposide for patients with brain metastases
melanoma brain metastases. Cancer J Sci AM from breast carcinoma, non small cell lung
1998; 4: 1039. carcinoma, or malignant melanoma. A
52. Loeffler JS, Barker FG, Chapman PH. Role of prospective study. Cancer 1999; 85(7):
radiosurgery in the management of central 1599605.
nervous system metastases. Cancer Chemother 63. Ewend MG, Sampath P, Williams JA, Tyler
Pharmacol 1999; 43: S1114. BM, Brem H. Local delivery of chemotherapy
53. Kondziolka D, Patel A, Lunsford LD, Kassam prolongs survival in experimental brain metas-
A, Flickinger JC. Stereotactic radiosurgery plus tases from breast carcinoma. Neurosurgery
whole brain radiotherapy versus radiotherapy 1998; 43: 118593.

393
INTRACRANIAL METASTASES

64. Franciosi V, Cocconi G, Michiara M et al. 68. Auperin A, Arriagada R, Pignon JP et al.
Front-line chemotherapy with cisplatin and Prophylactic cranial irradiation for patients with
etoposide for patients with brain metastases small-cell lung cancer in complete remission.
from breast carcinoma, non-small cell lung Prophylactic Cranial Irradiation Overview
carcinoma, or malignant melanoma: a prospec- Collaborative Group. N Engl J Med 1999; 341:
tive study. Cancer 1999; 85: 1599605. 47684.
65. Schagen SB, Van Dam FSAM, Muller MJ et al. 69. Fonseca R, ONeill BP, Foote RL et al.
Cognitive deficits after postoperative adjuvant Cerebral toxicity in patients treated for small
chemotherapy for breast carcinoma. Cancer cell carcinoma of the lung. Mayo Clin Proc
1999; 85: 64050. 1999; 74: 4615.
66. Mehta M, Noyes W, Craig B et al. A cost- 70. Mahalati K, Bilen CY, zen H, Aki FT, Kendi
effectiveness and cost-utility analysis of radio- S. The management of brain metastasis in
surgery vs. resection for single-brain nonseminomatous germ cell tumours. Br J Urol
metastases. Int J Radiat Oncol Biol Phys 1997; 1999; 83: 45761.
39: 44554. 71. Lagerwaard FJ, Levendag PC, Nowak PJ et al.
67. Iwadate Y, Namba H, Yamaura A. Identification of prognostic factors in patients
Significance of surgical resection for the treat- with brain metastases: a review of 1292
ment of multiple brain metastases. Anticancer patients. Int J Radiat Oncol Biol Phys 1999;
Res 2000; 20: 5737. 43: 795803.

394
Index

Note: vs indicates the Alkylating agents, 124, 1268 299304


differentiation of two or more Alpha-fetoprotein, germ cell etiology, 2989
conditions. Abbreviation: RT, tumors, 260 specific types, 30610
radiation therapy Alternative therapy, 1378 treatment, 3046
American Cancer Society, CNS malignant tumors, see Malignant
Accelerated fractionation (RT), 110 tumor incidence, 7 tumors
N-Acetyl aspartate (in MRS), 85, 93 Amnesia (memory loss) Anti-angiogenic drugs, 27, 434,
Acoustic neuroma/neurinoma, see callosal tumors, 75, 767 45, 1312
Vestibular schwannoma episodic transient global, 78 Anti-angiogenic factors
ACTH, see Adrenocorticotrophic radiation-related, 1212 (endogenous), 43, 44
hormone Anaplastic astrocytoma, treatment, metastases and, 373
Adamantinomatous 1625 Antibiotics
craniopharyngioma, 310, Anaplastic ependymomas, 181 antimicrobial, prophylactic, 131
31011 Anaplastic meningiomas, 196 antineoplastic, 124, 131
Adenohypophysis, see Anterior Anaplastic oligodendroglioma, 172 Anticoagulants, 137, 3823
pituitary Anemia, Fanconi, 341, 342, 353 Anticonvulsants, 1334, 3823
Adenoma, pituitary, see Anterior Angiofibroma in tuberous sclerosis, first-line/standard, 1334
pituitary, benign tumors 350 newer, 1345
Adenoma sebaceum, tuberous Angiogenesis, 22, 36, 425, see prophylactic, 133
sclerosis, 350 also Anti-angiogenic factors preoperative, 1001
Adenomatous polyposis coli, 341, bloodbrain barrier disruption Antidepressants, 383
3523, see also APC gene associated with, 4850 Anti-epileptics, see Anticonvulsants
Adrenalectomy, bilateral, 3089 brain edema and, 52 Antimetabolites, 124, 12931
Adrenocorticotrophic hormone factors implicated in, 425, 50 Antisense oligonucleotides, 132
(ACTH) secretion metastases and, 373 APC gene, 341, 3523
normal, 298 VEGF and, 22, 424, 45, 50 Apoptosis, 1921
tumor, 3089 glioma, 43, 157 Aquaporins, 47
Age steps in, 45 Ara-C, 1301
incidence increase related to as therapeutic target (angiogenesis Arachnoid, 189
change in (in population), inhibitors), 27, 434, 45, cysts, 3601
1314 1312 Arterial circulation, metastases in,
incidence related to, 1011 Angiogram 372
germ cell tumor, 256 MR, see Magnetic resonance Arterial injection, cytotoxic drugs,
metastases (intracranial) and, 369 angiogram 1256
neuronal/neuronalglial tumors preoperative, 100 Aspartame, 17
and age at presentation, 223 Angiomatous meningioma, 192 Astrocytic tumors, 15068, 17481
prognosis and, 2930 Anosmia, 79 diffuse, 15068
lymphomas, 328, 3301 Anterior pituitary focal, 17481
AIDS/HIV disease-related tumors, 16 (adenohypophysis), 297 incidence, 1502
lymphomas, 322, 325 benign tumors (predominantly types, 6
Alkaline phosphatase, placental, adenoma), 297309 Astrocytoma, 15065, see also
germ cell tumors, 260 classification, 2978 Oligoastrocytoma;
Alkaloids, plant, 124, 1289 clinical findings/diagnosis, Xanthoastrocytomas

395
INDEX

Astrocytoma, (Contd) Blood vessels/vasculature nasopharyngeal, 291


anaplastic, treatment, 1625 (peritumoral) paranasal sinus, 291
diffuse, 15065 lymphomas, 324 pituitary, 310
biopsy, 159 metastatic dissemination differentiation from benign
clinical findings, 1579 (hematogenous spread) via, tumors, 303
differential diagnosis, 15960 367, 3712 Caretaker tumor suppressor
etiology, 1523 morphologic and permeability genes, 342
genetic alterations, 1535 changes, 47, 50 Carmustine (BCNU)
incidence, 1502 new, see Angiogenesis implants/wafers, 126, 165
pathology, 1557 Bloom syndrome, 341, 3534 resistance, 28
prognosis, 156, 165 Bony (osseous lesions), singe-agent, 123
treatment, 1615 neurofibromatosis-1, 346 gliomas, 1634, 166
pilocytic, 1747 Boron neutron capture therapy, Cause, see Etiology
pituitary, 310 11415 Cavernous sinus meningioma,
subependymal giant cell, 1678 Brachytherapy, interstitial, 11314 199
Ataxia, 76, 778 Brain tumor, definition, 4 CCNU, see Lomustine
medulloblastoma, 234 Brain Tumor Study Group CD20, monoclonal antibodies to,
Atrophy, radiation-induced, 118, chemotherapy, combination vs 331
1212 single-agent, 123 CDKs, see Cyclin-dependent
Autonomic neuropathy, vincristine, oligodendroglioma prognosis, kinases
129 173 CEA, meningioma, 196
AZQ (diazaquine), 128 Brainstem tumors, 1667 Cell(s), nervous system, radiation
glioma, 1667 damage, 117
BAI-1, 44 in neurofibromatosis-1, 3445, Cell cycle alterations, 267
Basal cell nevus syndrome, 18, 346 diffuse astrocytoma and, 1534,
341, 353 Langerhans cell histiocytosis, 155
Basal ganglia tumors 335 Cell death, programmed, 1920
germinoma, 255 pilocytic astrocytoma, 174, 175 Cellular phones, 17
signs and symptoms, 77 symptomatology, 68 diffuse glial tumors and, 1523
BCNU, see Carmustine Breast cancer Central Brain Tumor Registry of
Behavioural changes, see Mental meningioma and, 195 US (CBRTUS)
abnormalities metastases (intracranial), 369, 373 incidence data, 89
Benign tumors, concept/meaning, 5 demyelinating disease vs, sex and, 10
Biological tumor markers, germ 37980 survival data, 301
cell tumors, 260 Bromodeoxyuridine, 115 Central neurocytoma, 223, 2236
Biology, 1929 Busulfan, 128 Cerebellar mutism syndrome, 239
metastases, 370 Cerebellar tumors
Biopsy (stereotactic needle), 867, Cadherins, 39 dysplastic Ganz gliocytoma, 223
978, see also Pathology Caf-au-lait spots, 341, 344 glioblastoma, 41
astrocytoma (diffuse), 159 Callosal lesions, amnesia, 75, 767 hemangioblastoma, 208, 209,
germ cell tumors, 262 Camptothecins, 131 210, 211
metastases, 381 Cancer, see Malignant tumors von HippelLindau disease
Bleeding, see Hemorrhage Capillaries, brain and, 208, 209, 210, 211,
Bleomycin, 131 endothelial cells, 46, 47 3489
Blindness, 74 radiation damage, 117 Langerhans cell histiocytosis,
cortical, 71, 7980 leakiness, 54 333, 335
BLM gene, 353 Carboplatin, 127 medulloblastoma, 232
BloodCNS barriers, 4550 Carcinoembryonic antigen (CEA), secondary (from renal cell
bloodbrain barrier, 467 meningioma, 196 carcinoma), 21011
chemotherapy and, see Carcinogens, chemical, 16, 17 secondary (metastases), 372
Chemotherapy Carcinomas, see also signs and symptoms, 77
bloodCSF barrier, 456, 478 Choriocarcinoma Cerebellopontine angle tumors
disruption, 4850, 50 choroid plexus, 181 acoustic tumor vs meningioma,
consequences, 55 embryonal, see Embryonal 276
imaging, 80 carcinoma signs and symptoms, 77

396
INDEX

Cerebral disorders, see Herniation; medulloblastoma, 237 primitive neuroectodermal


Infarction; Infections melanocytic tumors, 213 tumor, 243
Cerebral hemispheres, metastases meningioma, 205 location of genes (associated
in, 372 metastases, 3867 with brain tumors)
Cerebrospinal fluid and bloodbrain barrier, 373, hereditary syndromes
as barrier to blood, 456, 478 386 associated-genes, 18
diagnostic examination (e.g. via oligodendroglioma/anaplastic proto-oncogenes, 23
lumbar puncture), 901 oligodendrogliomas, 172 tumor suppressor genes, 23
lymphomas, 90, 322, 3256 pilocytic astrocytoma, 177 Cisplatin, 127
medulloblastoma, 235 pineal germ cell tumors, 264, lomustinevincristine and,
melanocytic tumors, 212 2645 medulloblastoma, 237
pineal germ cell tumors, 260, pineal parenchymal tumors, 254 Classification/types, 57
2623 skull base tumors, 283 astrocytoma, 151
pineal parenchymal tumors, chondrosarcoma, 290 cranial nerve tumors, 6, 270,
2534 esthesioneuroblastoma, 285 271
leaks Chemotherapy + RT, relative embryonal tumors, 6, 222,
pituitary tumors, 302 timing, 126, 164 2312
postoperative, with vestibular metastatic disease, 3867 germ cell tumors, 6, 256
schwannoma, 277 Children, see also Infants hemopoietic tumors, 321
obstruction, 701 incidence rise in, 14 meningeal tumors, 6, 190
Cerebrovascular disease, see medulloblastoma, see neuronal/neuronalglial tumors,
Hemorrhage; Infarction Medulloblastoma 6, 222
Charged particle beams, 114 Chloroma, myelogenous leukemia pineal tumors, see Pineal tumors
chondrosarcoma, 289 (granulocytic sarcoma), 332, sellar/pituitary tumors, 6, 2978
chordoma, 288 333 skull base tumors, 270
Chemical carcinogens, 16, 17 Chondroid chordoma, 2878 Clear cell lesions of CNS, 209
Chemical modifiers of radiation Chondromas, 214 differential diagnosis, 209
response, 11516 Chondrosarcomas, 28890 meningioma, 192
Chemotherapy (with cytotoxic CHOP, lymphomas, 32830 Clinical features, see Signs and
drugs), 34, 12231 Chordoma, 2868 symptoms
agents used, 12631 Choriocarcinoma, 256 Clival chordoma, 286, 287
classification, 123, 124 pathology, 258 Cognitive abnormalities
bloodbrain barrier, 46, 1235 prognosis, 265 chemotherapy-related, 387
lymphoma and, 32830 Chorionic gonadotropin, human radiation therapy-related, 390
metastases and, 373, 386 (HCG), germinoma, 2578, prophylactic, 389
neurotoxicity, see Neurotoxicity 260 tumor-related, 745
resistance to, 289, 125 Choroid plexus tumors, 1812 Cold, feeling, post-RT, 122
multi-drug, see MDR; P- types, 6 Colloid cysts, 106, 35960
glycoprotein Chromosomes Colorectal cancer, hereditary non-
routes of administration, 125 abnormalities, 246, see also polyposis, 18, 27, 341
timing, 126 Genetic changes Complementary therapy, 1378
Chemotherapy (with cytotoxic central neurocytoma, 223 Compression
drugs) of specific tumors ependymoma, 17980 cranial nerve, see Cranial nerve
anaplastic astrocytoma and esthesioneuroblastoma, 2845 disorders
glioblastoma, 163 gangliogliomas and symptoms caused by, 70
craniopharyngioma, 31314 gangliocytomas, 229 craniopharyngioma, 312
ependymoma, 180 hemangiopericytoma, 206 meningioma, 197
gangliogliomas, 231 lymphomas, 323 pituitary tumors, 299, 300
gliomatosis cerebri, 166 medulloblastoma, 232 Computed tomography, 69
hemangiopericytoma, 208 meningioma, 192, 193 craniopharyngioma, 313
Langerhans cell histiocytosis, oligodendroglioma, 16970 dermoids/epidermoids, 3589
335 pilocytic astrocytoma, 1756 in differential diagnosis, 88
leukemia, and CNS involvement, pineal germ cell tumors, 256 glial tumor
373 pineal parenchymal tumors, cerebrovascular disease vs, 160
lymphomas, 125, 32831 252 ependymoma, 180

397
INDEX

Computed tomography, (Contd) types, 6, 270, 271 Desmoplastic infantile


hemangiopericytoma, 207 VIIIth, see Vestibular ganglioglioma, 223
oligodendroglioma, 171 schwannoma Dexamethasone, 136
hemopoietic tumors, 332 Craniopharyngiomas, 256, 31014 in brain edema, 57
medulloblastoma, 235 Craniotomy with brain metastases, in cerebral herniation (and raised
meningioma, 201 deaths, 389 ICP), 56
MRI compared with, 80, 81 Cushings disease, 308 Diagnosis, 6596
neuronal/neuronalglial tumors Cutaneous involvement, familial differential, see Differential
central neurocytoma, 225 brain tumor syndromes, 341 diagnosis
dysembryoplastic Cyclin(s), 26 elderly, missed (on imaging),
neuroepithelial tumor, Cyclin-dependent kinases (CDKs), 1213
228 26 Diazaquine, 128
pituitary tumors, 301 gliomas and, 155 Diet, see Nutritional factors;
preoperative, 100 Cyclophosphamide, 1278 Nutritional management
skull base tumors and Differential diagnosis, 889
chordoma, 288 vincristinedoxorubicinpred central neurocytoma, 225
esthesioneuroblastoma, 285 nisone (CHOP), lymphoma, clear cell lesions, 209
nasopharyngeal/paranasal 32830 glial tumors
tumors, 291 Cyst(s), 340, 35662 diffuse astrocytoma, 15960
Conformal RT planning, 112 colloid, 106, 35960 oligodendrogliomas, 209
Consciousness, impairment, 75 dermoid/epidermoid, 3569 pilocytic astrocytoma, 177
Contrast-enhanced MR, 80 pineal, 254 lymphomas, 3267
Convulsions, see Seizures Rathkes cleft, 314 melanocytic tumors, 21213
Corpus callosum tumors, signs and Cysticercosis, 17 meningiomas, 2012, 209
symptoms, 77 Cytarabine, 1301 metastases, 37781
amnesia, 75, 767 Cytochrome P-450 enzymes, 17 pituitary tumors, 3034
Cortical blindness, 71, 7980 Cytogenetics, see Chromosomes, vestibular schwannoma, 2756
Cortical mapping, 1023 abnormalities Diffusion-weighted MRI, 82
Corticosteroids, 579, 1357 Cytokeratins, germ cell tumors, Diplopia, 79
in brain edema, 57 260 Dissemination, see also Invasion
mechanisms of action, 589 Cytokine-secreting tumors, in CNS, pathways, 40, 41, 367
taper, 578 symptomatology, 69 hematogenous, 367, 3712
in cerebral herniation (and raised Cytotoxic drug therapy, see Distribution
ICP), 56 Chemotherapy in brain, see Site
lymphoma, 327 geographical, see Geographical
metastases, 3812 Dacarbazine, 128 distribution
side-effects, 1367 DCC gene, 154, 155 Dizziness, see Vertigo
Corticotroph, 298 Deafness, vestibular schwannoma, DNA damage, radiation causing,
failure, 299 2724 108
tumor, 3089 Death (cell), programmed, 1920 DNA repair
Corticotropin-releasing factor use Death (people) with intracranial MGMT in repair of
in brain edema, 57 tumors chemotherapeutic drug
Cowdens disease/syndrome, 18, metastases, 368 damage, 28
341, 352 operative, 38990 mismatch repair genes,
Cranial base tumors, see Skull base primary tumors, 7 mutations, 278
tumors Deep vein thrombosis, 137 p53 and, 24
Cranial nerve Deleted in colon cancer (DCC) Dopamine agonists
disorders/neuropathies (incl. gene, 154, 155 GH-secreting tumor, 307
compressive neuropathies) Deletions, chromosome, 25 prolactinoma, 3056, 306,
skull base tumors, 270 Demyelination 3067
vestibular schwannoma, 274 radiation-induced, 120 Doxorubicin, 131
vincristine-related, 129 tumor vs, 88, 1601 and
Cranial nerve tumors (nerve sheath metastatic, 37980 vincristinecyclophosphamid
tumors), 27082 Depression, 383 eprednisone (CHOP),
malignant, 2812 Dermoids, 3569 lymphoma, 32830

398
INDEX

Drugs Encephalopathy, 11921 Ethnic group and incidence, 9


cytotoxic, see Chemotherapy acute, 118, 119 Etiology, 1419
experimental, 27, 1312, 132 early-delayed, 118, 11920 astrocytomas
anti-angiogenic drugs, 27, late-delayed, 118, 1201 diffuse, 1523
434, 45, 1312 Endocrine function, pituitary focal (pilocytic), 1756
glioma, 165 adenoma, see Hormones central neurocytoma, 227
lymphomas, 331 Endocrinopathy gangliogliomas and
pituitary adenoma medulloblastoma, 239 gangliocytomas, 229
GH-secreting tumor, 307 pituitary adenoma-related, hemangioblastoma, 2089
prolactinoma, 3056, 3067 299300 hemangiopericytoma, 206
preoperative, 1001 radiation-related, 122 medulloblastoma, 232
Dura mater, 189 Endodermal sinus tumor, see Yolk meningioma, 1925
metastases, 376 sac tumor oligodendrogliomas, 169
plasmacytomas, 332 Endoscopic surgery, 103, 106 pineal germ cell tumors, 256
Dysembryoplastic neuroepithelial colloid cyst, 106, 279, 360 pineal parenchymal tumors,
tumors, 223, 2268 Endothelium (and endothelial cells) 252
Dysplastic Ganz gliocytoma of angiogenesis and, 434, 44 pituitary tumors, 2989
cerebellum, 223 bloodbrain barrier and, 46, 47 Etoposide, 129
radiation damage, 117 European Organization for
EBV, see EpsteinBarr virus Environmental risk factors, 1517, Research and Treatment of
Edema, brain, 505 19 Cancer (EORTC), RT, glioma,
consequences, 55 astrocytoma, 1523 162
craniopharyngioma, 313 EORTC study, RT, glioma, 162 Extracellular matrix
imaging, 80, 82 Eosinophilic granuloma, 334 bloodbrain barrier and, 47
mediators, 52 Ependymal cells, 149 tumor invasion and, 3940
meningioma, 53, 198, 200 tumors arising, see Eye involvement, see also Visual
postoperative, 203 Ependymomas; disturbances
treatment, 579 Subependymomas lymphomas, 324
Edema, optic disk (papilledema), Ependymoblastoma, 231 tuberous sclerosis, 351
74 Ependymomas, 17881
Eflornithine, 131 anaplastic, 181 Facial nerve schwannoma, 280
EGFR, see Epidermal growth Epidemiologic studies, 714, see Facial weakness, vestibular
factor receptor also Incidence schwannoma, 274
Elderly metastases (intracranial), 3689 Familial CNS tumor syndromes,
imaging, missed diagnosis, risk factors, 15 see Syndromes
1213 Epidermal growth factor receptor Fanconi anemia, 341, 342, 353
lymphoma prognosis, 328, 330 (EGFR), 20, 21 Fast fluid-attenuated inversion
Electromagnetic radiation, 17 dominant-negative, 115 recovery, 82
diffuse glial tumors and, 1523 glioblastoma multiforme and, FDG in PET studies, 84
Electron microscopy, diagnostic 20, 27, 154 FGF and angiogenesis, 44
value, 90 monoclonal antibodies, 132 Fibroblast growth factor and
Eloquent areas, symptomatology Epidermoids, 3569 angiogenesis, 44
of tumors in, 68 Epilepsy, see Seizures FLAIR, 82
Embolization, preoperative, 100 Episodic symptoms, 756, 78 Fluorodeoxyglucose (FDG) in PET
Embryonal carcinoma, 255 seizures, 76 studies, 84
pathology, 258 Epithelial membrane antigen 5Fluorouracil, 131
prognosis, 265 (EMA), meningioma, 196 demyelinating lesions, 380
Embryonal tumors, 67, 222, EpsteinBarr virus (EBV) Folate analog, lymphomas, 330
23143 lymphomas and, 3223 Follicle-stimulating hormone (FSH)
classification, 6, 222, 2312 nasopharyngeal carcinoma and, secretion
prognosis, 222 291 normal, 298
Embryonic cell rests, germ cell ErdheimChester disease, 335 tumor, 309
tumors arising from, 256 Erectile dysfunction (impotence), Fossa, posterior,
Emesis, see Vomiting post-RT, 122 dermoid/epidermoid cyst, 257,
Encephalitis, herpes simplex, 161 Esthesioneuroblastoma, 2845 358

399
INDEX

Fractionation (RT), 10810 syndromes, 342, 354 varicella-zoster protecting against


altered schedules, 10910 Genetic tests, familial cancer occurrence, 17
conventional schedules, 1089 syndromes, 342, 354, 355 Glial microhamartomas, 347
radiosurgery, 113 Genomic imprinting and Glioblastoma (multiforme), 1625
Frameless stereotaxy, 102 neurofibromatosis-1, 343 cerebellar, 41
Free radicals and brain edema, 52 Geographical distribution, 11 demyelination vs, 88, 1601
Frontal lobe tumors germ cell tumors, 256 edema absent with, 53
meningioma, 197, 200 Germ cell tumors EGFR abnormalities, 20, 27, 154
prognosis, 29 primary, 7, 25566 evaluation leading to diagnosis,
signs and symptoms, 77, 7980 clinical findings, 25860 93
Frozen sections, 90, 103 etiology, 256 genetic defects and, 154, 155
FSH, see Follicle-stimulating non-germinomatous invasion/metastases, 37, 41
hormone (NGGCTs), 255, 256, MRI
Functional MRI, 80, 86 262, 264, 265 functional MRI, 93
glial tumors pathology, 2578 missed on, 13
diffuse astrocytoma, 159 pineal region, 250, 25566 parietal lobe, presentation, 78
glioblastoma, 93 prognosis, 2656 photomicrographs, 151
preoperative, 100, 101 treatment, 2601 prognosis, 165
types, 6, 256 TGF- and immune suppression
Gadolinium, motexafin, 115 secondary, treatment, 389 with, 22
Gadolinium-enhanced MRI, 80 Germinoma, 255, 315 treatment, 1625
postoperative, 104 clinical findings, 260 genetic change affecting, 27
Gait abnormalities, 778 differential diagnosis, 209 Gliocytoma of cerebellum,
dysplastic Ganz, 223
medulloblastoma, 234 pathology, 2578
Glioma, see Glial cell tumors
Gamma ray(s), 106 prognosis, 2656
Gliomatosis cerebri, 38, 1656
Gamma (ray) knife, 113, see also treatment, 2624
treatment, 166
Radiosurgery Giant cell astrocytoma,
Glomus jugulare tumors, 2856
Gangliocytoma, 22831 subependymal, 1678 Glucocorticoids, see Corticosteroids
characteristics, 223 Glial cell(s), 14950 Gonadotroph, 298
Ganglioglioma, 22831 radiation damage, 118 failure, 299
characteristics, 223 types, 14950 tumor, 309
desmoplastic infantile, 223 Glial cell tumors (gliomas), 14988 Gonadotropins, see Follicle-
Ganglioneuroblastoma, 241 angiogenesis, 43, 157 stimulating hormone; Human
Gatekeeper tumor suppressor brainstem glioma, 1667 chorionic gonadotropin
genes, 342 differential diagnosis, see (HCG); Luteinizing hormone
Gemistocyte, 156 Differential diagnosis Gorlins syndrome (nevoid basal
Gender, see Sex diffuse, 15073 cell carcinoma syndrome), 18,
Gene(s) (incl. mutated genes), locations, 149 341, 353
1929, see specific genes dissemination pathways, 41 Grading of tumors
hereditary syndromes, 18 epidemiologic data, 8, 12 astrocytoma (diffuse), 156
Gene therapy, 132 focal, 150, 17382 upgrading, 162
Genetic cancer syndromes, see imaging, 82, 83, 158, 160, immunohistochemistry in, 91
Syndromes 1712, 1767, 180 pineal region tumors, prognostic
Genetic changes/alterations, 1819, MR, see Magnetic resonance value, 2523
see also Chromosomes, imaging Granular cell tumors, pituitary,
abnormalities mixed gliomas, 6 297, 310
diffuse astrocytoma, 1535 mixed neuronal and, see Granulocytic sarcoma, 332, 333
lymphomas, 323 Neuronalglial tumors, Granuloma, eosinophilic, 334
medulloblastoma, 232 mixed Growth, brain tumor
meningioma, 1923 in neurofibromatosis-1, 3446, 347 rate
multiple endocrine neoplasia (and treatment impact on symptomatology,
pituitary adenoma), 299 chemotherapy, 125, 129 66, 68
oligodendroglioma, 16970 heterogeneity affecting, 27 vestibular schwannoma, 272,
pilocytic astrocytoma, 1756 types, 6, 150, see also specific 274, 278
Genetic counselling, familial cancer types sequential steps, 36

400
INDEX

Growth factors (and their Hemopoietic tumors, 32039 HMPAO, Tc-99m-labelled, 84


receptors), 201, 212, see metastatic, 320 hMSH2, 342
also specific growth factors primary, 32039 Hodgkins disease, 332
diffuse astrocytoma and, 153, in neurofibromatosis-1, 346 Hormones, pituitary, 297
154 types, 322 measurement, diagnostic value,
meningioma and, 194 Hemorrhage/bleeding, intracranial 301
therapeutic targeting, 132 as biopsy complication, 87 non-secretion (non-functioning
Growth hormone metastases, 3745 tumors), 298, 309
deficiency, 299 tumor differentiated from, secretion/hypersecretion (by
secretion diffuse astrocytoma, 160 tumors), 298, 3069
normal, 298 tumor-related symptomatology, 68
tumor, 307 imaging, 812 hPMS-2, 353
metastases, 381 hSNFS5/INI1, 18, 354
Halogenated pyrimidine analogs, Heparin, 137 HSV, see Herpes simplex virus
115 Hepatic metastases, meningioma, Human chorionic gonadotropin
Hamartias, 347 196 (HCG), germinoma, 2578,
Hamartin, see TSC1 Hepatocyte growth factor, 445 260
Hamartomas Heredity, see Genes; Genetic Human immunodeficiency virus,
hypothalamic, 362 changes; Syndromes, cancer see AIDS
LhermitteDuclos disease, 352 Herniation, cerebral, 55 Human placental lactogen, germ
tuberous sclerosis, 3501 symptoms, 71 cell tumors, 260
HandSchullerSiwe disease, 334 treatment, 556 Hydrocephalus
Head trauma, 1617 Herpes simplex virus radiation-related, 118, 1212
meningioma, 195 encephalitis, 161 tumor-related, 701
Headache, 716 thymidine kinase gene, 115, craniopharyngioma, 312
colloid cysts, 362 132 medulloblastoma, 235, 239
medulloblastoma, 234 Herpes zoster, see Varicella-zoster meningioma, 2001
meningioma, 198, 1989 Hexamethylpropyleneamine oxime Hydrostatic pressure and brain
neurofibromatosis-1, 347 (HMPAO), Tc-99m-labelled, edema, 52
pituitary adenoma, 300 84 Hydroxyurea, 130
plateau waves and, 75 Histiocytic disorders, 3335 Hyperfractionated irradiation,
vestibular schwannoma, Histologic diagnosis, 8991, see 10910
postoperative, 2767 also Biopsy; Pathology Hyperosmolar agents, 56
vomiting and, 73 Histologic types, site of tumor by, Hypersensitivity reaction,
Hearing loss, vestibular 5 radiation-related, 118
schwannoma, 2724 Histology/histopathology, see also Hyperventilation technique, 56
Heavy particle irradiation, 114 Pathology Hypoglossal schwannoma, 281
Hemangioblastoma, 189, 20811 central neurocytoma, 224 Hypoglycemia,
von HippelLindau disease, 208, glial cell tumors (incl. hemangiopericytoma, 207
209, 210, 211, 3489 astrocytoma), 156 Hypophysis, see Pituitary
Hemangiopericytomas, 189, astrocytoma classification Hypothalamus
2068 criteria based on craniopharyngioma affecting
Hematogenous spread of histology, 151 function, 312
metastases, 367, 3712 ependymoma, 180 hamartomas, 362
Hematoma, postoperative, 105 meningioma, 196
Hematophagocytic metastases, 374
lymphohistiocytosis, 335 pineal region tumors, 252 Ifosfamide, 128
Hemiparesis pituitary adenoma classified by, IGF-1 elevation, 307
contralateral, 76 298 Imaging (neuroimaging), 66, 69,
ipsilateral, 79 RT based on, 111 806, see also specific
Hemispheres, cerebral, metastases History-taking, 65 methods
in, 372 HIV disease, see AIDS elderly, missed diagnosis,
Hemogenic meningitis, post- hMLH1/2, 18 1213
medulloblastoma treatment, neurofibromatosis-1 and, 343 new methods, 3
2389 Turcot syndrome and, 18, 353 preoperative, 100

401
INDEX

Imaging (neuroimaging), (Contd) Incidence, 714 Iododeoxyuridine, 115


specific tumors/cysts/tumor-like astrocytoma, 1502 Ionizing radiation, see Radiation
lesions factors affecting, 912 Iridium-192, interstitial, 114
astrocytoma, diffuse, 158, 160 increasing (possible), 1214 Ischemic infarction, see Infarction
astrocytoma, pilocytic, 1767 primary lymphomas, 14, 321
central neurocytoma, 224, 225 meningiomas, 192 Jugular foramen schwannoma,
chordoma, 288 Industrial/occupational carcinogens, 2801
craniopharyngioma, 313 16, 17
dermoid/epidermoid cysts, 358 Infant(s), medulloblastoma Karyotypic changes, see
dysembryoplastic chemotherapy, 237 Chromosomes, abnormalities
neuroepithelial tumor, Infantile ganglioglioma, Kidney cancer, metastases, 21011
227, 228 desmoplastic, 223 Knudsen two-hit theory of
ependymoma, 180 Infarction, cerebral, tumors oncogenesis, 24, 3412
esthesioneuroblastoma, 285 differentiated from, 88
facial nerve schwannoma, 280 diffuse astrocytomas, 160 Laboratory examination/diagnosis,
gangliogliomas and metastatic, 3801 66, 8091, see also specific
gangliocytomas, 230 Infections, see also Antibiotics and tests
hemangioblastoma, 21011 specific infections/pathogens Lactate dehydrogenase, germ cell
hemangiopericytoma, 207 in etiology, 17 tumors, 260
hemopoietic tumors (other lymphomas, 3223 Lactotroph, 298
than non-Hodgkins nasopharyngeal carcinoma tumors, see Prolactin, tumors
lymphomas), 332 and, 291 secreting
histiocytic disorders, 332, 334 postoperative, 105 Langerhans cell histiocytosis,
lymphomas, 82, 3245 tumor vs, 889 3335
medulloblastoma, 235 metastatic, 3779 Laser, operating, 103
melanocytic tumors, 212 Inflammatory conditions, pituitary, Leptomeninges, see Meninges
meningioma, 201, 259 3034 LettererSiwe disease, 334
metastases, 82, 3757 Infratentorial tumors, Leucovorin rescue, lymphomas,
nasopharyngeal/paranasal symptomatology, 68 330
tumors, 291 Inheritance, see Genes; Genetic Leukemia
oligodendroglioma, 1712 changes; Syndromes, cancer chemotherapy, and CNS
paraganglioma, 286 INI1/hSNFS5, 18, 354 involvement, 373
pineal tumors, 253 Injury, see Head trauma myelogenous/granulocytic,
pituitary tumors, 301, 303 Insulin-like growth factor-1 chloroma associated with (=
trigeminal schwannoma, 279 elevation, 307 granulocytic sarcoma), 332,
vestibular schwannoma, Integrins, 39 333
2745 Intensity-modulated RT, 112 prophylactic cranial irradiation
symptoms and, 69 Interleukin-2, therapeutic use, 132 in, and incidence of second
Immune response, radiation Interstitial brachytherapy, 11314 tumors, 15, 16
damage-related, 118 Intra-arterial chemotherapy, 1256 Levamisole, demyelinating lesions,
Immune suppression, acquired, as lymphomas, 330 380
risk factor, 16 Intracranial pressure, increased LH, see Luteinizing hormone
Immunoglobulin superfamily and (intracranial hypertension), 54, LhermitteDuclos disease, 223, 352
tumor invasion, 39 55 LiFraumeni syndrome, 18, 341,
Immunohistochemistry, 91 craniopharyngioma, 312 3512
neuronal/neuronalglial tumors, meningioma, 201 rhabdoid predisposition
222 sudden transient, see syndrome and, 354
Immunosuppression and Plateau/pressure waves Libido, loss, post-RT, 122
lymphomas, 3223, 326, 327 treatment, 556 Lipomas, meningeal, 213
Immunotherapy, 132 Intrathecal chemotherapy, 126 Liver, meningioma metastases, 196
Implants, cytotoxic drugs (in Invasion/spread, 3642, 6970, see Location, see Site
wafers), 126 also Dissemination; Metastases Lomustine (CCNU)vincristine
gliomas, 126, 165 factors required, 39 and cisplatin, medulloblastoma,
Implants, radioisotopes, 11314 symptoms related to, 6970 237
Impotence, post-RT, 122 Iodine-125, interstitial, 114 and procarbazine, see PCV

402
INDEX

Lumbar puncture, see craniopharyngioma, 313 Magnetic resonance spectroscopy


Cerebrospinal fluid in differential diagnosis, 88 (MRS), 80, 856
Lung cancer metastases, 369 familial cancer syndromes gangliogliomas and
MRI, 376 Cowdens gangliocytomas, 230
pathophysiology of metastasis, disease/LhermitteDuclos glial tumors
3712 disease, 352 diffuse astrocytoma, 1589
treatment, 385, 389 neurofibromatosis-1, 3467 glioblastoma, 93
Luteinizing hormone (LH) functional, see Functional MRI oligodendroglioma, 1712
secretion glial tumors, 82, 158 hemangiopericytoma, 207
normal, 298 differentiation from meningioma, 201
tumor, 309 cerebrovascular disease, Malignant tumors (extracranial)
Lymphatic system 160 frequency of symptomatic
CSF serving as, 48 ependymoma, 180 intracranial metastases,
deficiency (in brain), 545 glioblastoma missed by MR, 368
chemotherapy/radiotherapy 13 from specific organs/tissues, see
and impact of, 125 gliomatosis cerebri, 166 Breast cancer; Lung cancer;
Lymphohistiocytosis, low-grade glioma, 83, 158 Ovarian cancer; Renal cell
hematophagocytic, 335 oligodendroglioma, 171 cancer
Lymphomas, Hodgkins, 332 pilocytic astrocytoma, 176 Malignant tumors (intracranial)
Lymphomas, primary non- hemangioblastoma, 21011 cranial nerves, 2812
Hodgkins, 7, 32132 hemangiopericytoma, 207 meningeal, 190, 213, 214
clinical findings/diagnostic tests, hematophagocytic meningioma, 196
3246 lymphohistiocytosis, 335 pituitary, 310
CSF examination, 90, 322, hemopoietic tumors, 332 differentiation from benign,
3256 lymphomas, 82, 3245 303
differential diagnosis, 3267 Langerhans cell histiocytosis, secondary, 310
epidemiology, 321 3345 Mannitol, 56
increasing incidence and medulloblastoma, 235 Matrix metalloproteinases, 40
relative frequency, 14, postoperative, 236, 2378 gliomas and, 155
321 meningioma, 201, 259 Mayo Clinic, incidence data, 8, see
imaging, 82, 3245 metastases, 82, 3757 also St Anne/Mayo
pathology, 3234 neuronal/neuronalglial tumors classification of astrocytomas
prognosis, 328, 330, 3301 central neurocytoma, 224, MDR gene, 47, see also P-
recurrence, 40, 331 225 glycoprotein
treatment, 32731 dysembryoplastic Medical conditions as risk factors,
chemotherapy, 125, 32831 neuroepithelial tumor, 17
Lymphoreticular neoplasms, 227, 228 Medication, preoperative, 1001
32132 gangliogliomas and Medullary tumors, signs and
metastases, 377 gangliocytomas, 230 symptoms, 77
primary, 32132 pineal germ cell tumors, Medulloblastoma, 23240
25960 bloodbrain barrier with, 50
Macroadenomas, pituitary, 2978 preoperative, 262 classification, 2312
treatment, 3067 pineocytoma, 251, 253 clinical findings, 2345
Maffucci syndrome, 289 pituitary tumors, 301, 303 etiology, 232
Magnetic resonance angiogram Rathkes cleft cyst, 314 pathology, 2334
preoperative, 100 skull base tumors prognosis, 240
tumor blush, 42 chordoma, 288 treatment, 23540
Magnetic resonance imaging, 66, esthesioneuroblastoma, 285 complications, 23840
69, 803 nasopharyngeal/paranasal Melanocytic tumors/lesions,
callosal tumor, 75 tumors, 291 21113
colloid cysts, 360 paraganglioma, 286 primary, 190
cranial nerve tumors in surgery Melanocytoma, 21112
facial nerve schwannoma, 280 intraoperative, 106 clinical findings, 211, 212, 213
trigeminal schwannoma, 279 postoperative, 104 differential diagnosis, 213
vestibular schwannoma, 2745 preoperative, 100 treatment and prognosis, 213

403
INDEX

Melanoma Meningitis treatment, 370, 38190


familial, 354 with craniopharyngioma, aseptic, complications, 38990
metastatic, 369 313 specific situations, 3889
differential diagnosis, 213 with dermoids/epidermoids, Methotrexate, 130
sex and, 369 aseptic and infective, 358 lymphomas, 125, 328, 330
primary, 21112 with medulloblastoma, O6Methylguanine-DNA
clinical findings, 212, 213 postoperative methytransferase (MGMT), 28
differential diagnosis, 213 hemogenic/aseptic, 2389 MIB-1 index, astrocytoma, 156
treatment and prognosis, infectious/bacterial, 238, 239 Microadenomas, pituitary, 2978
213 Meningoepithelial origin, 190 treatment, 306, 307
Melanosis and melanocytosis, Mental (incl. Microglial, 149
diffuse, 211 behavioural/personality) Microhamartomas, glial, 347
Melanotic tumors, see Melanocytic abnormalities, 745, 78 Microsatellite instability, 27
tumors medulloblastoma, 23940 Microscope
Memory loss, see Amnesia meningioma, 200 electron, 90
MEN-1 (disease), see Multiple 6Mercaptopurine, 131 operating, 102
endocrine neoplasia type-1 Merlin, see NF-2 gene Midbrain tumors
MEN-1 (menin) gene, 18, 354 Mesenchymal chondrosarcoma, glioma, 1667
Menin gene, 18, 354 289 signs and symptoms, 77
Meninges (leptomeninges), Mesenchymal tumors, meningeal, Mismatch repair genes, mutations,
189220 190 278
anatomy/components, 189, see Metallothioneins, 289 Mitochondria, endothelial cell,
also Arachnoid; Dura mater; Metastases (of brain tumor 467
Pia mater seeding), see also Mixed tumors
tumors of, 7, 189205 Dissemination; Invasion germ cell tumors, 258
types, 6, 190 biology, 370 gliomas, 6
tumors seeding/metastasizing to choroid plexus tumors, 181, 182 neuronalglial, see
choroid plexus tumors, 181, germ cell tumors, 266 Neuronalglial tumors
182 glioblastoma, 41 MLH1/2, see hMLH1/2
extracranial tumors, 377 lymphoma, 326 Mobile phones, see Cellular
medulloblastoma, 234 meningioma, outside brain, 196 phones
meningioma distinguished Metastases (to brain), 4, 36794 Monoclonal antibodies
from, 202 biology, 370 to CD20, 331
Meningioangiomatosis, 3478 diagnostic difficulties, 367 to EGF receptor, 132
Meningiomas, 190206 differential diagnosis, 37781 Mortality, see Death
cerebellopontine angle, 276 hemopoietic tumors, 320 Mosaicism, tuberous sclerosis,
clinical findings, 197201 imaging, 82, 3757 350
bloodbrain barrier disruption, incidence, 78 Motexafin gadolinium, 115
4850 lymphoma, 326 MR studies, see Magnetic
edema, see Edema meningeal, see Meninges resonance
imaging, 201, 259 pathology, 374 hMSH2, 342
differential diagnosis, 2012, pathophysiology, 3704 Multi-drug resistance gene, 47, see
209 pituitary also P-glycoprotein
etiology, 1925 anterior, 303 Multiple endocrine neoplasia type-
neurofibromatosis-2, 192, 194, posterior, 310 1, 18, 341, 354
347 prognosis, see Prognosis pituitary adenomas and, 2989
incidence, 192 relapse/recurrence within year, Multiple sclerosis vs metastases,
invasion, 39 management, 3889 37980
pathology, 1957 renal cell cancer, 21011 Muscular weakness, generalized,
anaplastic/malignant, 196 signs and symptoms, 3745 75
atypical, 196 before discovery of primary, Mustache sign, 313
pineal region, 259 367, 370 Mutations, see Chromosomes,
prognosis, 2056 frequency of symptomatic abnormalities; Genes
treatment, 2026 metastases, 368 Mutism, cerebellar, syndrome of,
variants, 192, 209 site, 5, 367, 369, 372 239

404
INDEX

Myelogenous leukemia chloroma Neurofibromatosis-2, 18, 341, NF-1 (neurofibromatosis-1), see


(= granulocytic sarcoma), 332, 3478, see also NF-2 gene Neurofibromatosis-1
333 meningioma, 192, 194, 347 NF-1 (neurofibromin) gene, 18,
Myopathy, vincristine, 129 nerve sheath tumors (incl. 342, 343
Myxoid chondrosarcoma, 288 vestibular schwannoma), product, 343
271, 347, 348 NF-2 (merlin) gene, 18, 271, 347
NAA (N-acetyl aspartate) in MRS, Neurofibromin and its gene, see product, 347
85, 93 NF-1 gene NF-2 (neurofibromatosis-2), see
Nasopharyngeal tumors, 2901 Neurohypophysis, see Posterior Neurofibromatosis-2
National Cancer Data Base, pituitary Nitroimidazoles, 115
epidemiologic data, 9, 30 Neuroimaging, see Imaging N-Nitroso compounds,
National Cancer Institutes SEER Neurologic complications of carcinogenicity, 16, 19
registry, incidence data, 12 procedures Nitrosoureas
pediatric, 14 biopsy, 87 therapeutic use
sex and, 10 RT, 122 gliomas, 125
N-CAM, 39 surgery, 105 resistance, 28
Necrosis (radiation-related), 118, Neurologic dysfunction and toxicity, 127
1201, 125 symptoms, see Signs and carcinogenicity, 16
radiosurgery, 390 symptoms Non-polyposis colorectal cancer,
Needle biopsy, see Biopsy Neurologic examination, 656 hereditary, 18, 27, 341
Nelsons syndrome, 308 Neuroma, cranial nerve, see Nutritional factors (in etiology),
Nerve sheath tumors, see Cranial Schwannoma 16, 153
nerve tumors Neuronalglial tumors, mixed, 6, Nutritional management, 138
Nervous system cells, radiation 2212, 22331 Nystagmus, 76
damage, 117 classification, 222
Neural cell adhesion molecule, 39 diagnosis Occipital lobe tumors, signs and
Neurilemmoma, see Schwannoma advances, 2212 symptoms, 77
Neurinoma, see Schwannoma pitfalls, 241 visual field defect, 76
Neuroblastoma, 241, 2423 prognosis, see Prognosis Occupational/industrial
olfactory, 2845 Neuronal hamartomas, carcinogens, 16, 17
Neurocytoma, central, 223, hypothalamic, 362 Octreotide, see Somatostatin analogs
2236 Neuronal tumors, 2212, 22331 Ocular involvement, see Eye;
Neuroectodermal tumors, primitive classification, 6, 222 Visual disturbances
(PNET), 231, 232, 241, 243 diagnostic advances, 2212 Olfactory disturbances, 79
Neuroendocrine tumors in prognosis, 222 Olfactory groove meningioma, 191
neurofibromatosis-1, 346 Neuropathies with vincristine, 129 Olfactory neuroblastoma, 2845
Neuroendoscopic surgery, see Neurosurgery, see Surgery Oligoastrocytoma, 173
Endoscopic surgery Neurotoxicity Oligodendroglial cells (and
Neuroepithelial tumors cytotoxic drugs, 387 precursors), genetic alterations,
classification, 6 5FU and levamisole, 380 169
dysembryoplastic, 223, 2268 methotrexate, 130 Oligodendrogliomas, 16873
Neurofibroma(s), 281 taxanes, 129 differential diagnosis, 209
neurofibromatosis-1 and, 281, vincristine, 129 epidemiology, 169
344, 345, 346 lymphoma treatment, 3312 increasing relative incidence, 14
Neurofibromatosis-1 (von radiation-related, 328, 329 etiology, 169
Recklinghausens disease), 18, radiation, 11622, 125, 390 genetic changes, 16970
341, 3427, see also NF-1 lymphoma treatment, 328, imaging, 1712
gene 329 pathology, 170
diagnostic criteria, 344 medulloblastoma treatment, prognosis, 173
tumors of, 18, 3446 239 signs and symptoms, 171
central neurocytoma, 227 prophylactic, 389 factors impacting on, 66, 67
cranial nerve tumors, 271, steroids, 136 frontal lobe, 7980
282 Neutrons (therapeutic use), 114 treatment, 1723
pilocytic astrocytoma, 175, Nevoid basal cell carcinoma types, 6
176, 344 syndrome, 18, 341, 353 anaplastic, 172

405
INDEX

Oligonucleotides, antisense, 132 Paraneoplastic syndromes, Perfusion MRI, 82


Olliers disease, 289 hemangiopericytoma, 207 lymphomas, 325
Olmstead County, epidemiologic Parietal lobe tumors, signs and preoperative, 100
data, 8 symptoms, 77, 78 Peripheral neuropathy, vincristine,
Oncogenes/proto-oncogenes, 203 Paroxysmal symptoms from 129
amplification, 26 plateau waves, 75 Personality changes, see Mental
Operating microscope, 102 Pathology, 8991, see also Biopsy; abnormalities
Ophthalmological involvement, see Histology PET, see Positron emission
Eye; Visual disturbances colloid cysts, 360 tomography
Opisthotonus, 75 craniopharyngioma, 31012 Pharmacology/pharmacotherapy,
Optic chiasm compression dermoids/epidermoids, 359 see Drugs
craniopharyngioma, 312 diagnostic, 8991 Phosphorus MRS, 86
pituitary adenoma, 3001 glial tumors Pia mater, 189
Optic disk edema (papilledema), diffuse astrocytoma, 1557 Pilocytic astrocytoma, 1747
74 oligodendroglioma, 170 neurofibromatosis-1, 175, 176,
Optic nerve tumors pilocytic astrocytoma, 176 344
gliomas, in neurofibromatosis-1, hemangioblastoma, 20910 Pineal non-neoplastic lesions,
3456 hemangiopericytoma, 206 249
signs and symptoms, 77 intraoperative, 103 Pineal tumors (pineal gland/region),
Ornithine decarboxylase inhibitor, Langerhans cell histiocytosis, 335 24969
131 lymphomas, 3234 parenchymal, 2514
Osmolar therapy, 56 medulloblastoma, 2334 signs and symptoms, 68, 77,
Osseous lesions, neurofibromatosis- melanocytic tumors, 212 2534, 2589
1, 346 meningioma, 1957 types, 6, 250
Osteoblastoma, skull base, 289 metastases, 374 relative frequency, 250
Osteomalacia, neuronal/neuronalglial tumors, Pineoblastoma, 251, 252
hemangiopericytoma, 207 222 imaging, 253
Ovarian cancer metastases, 369 central neurocytoma, 2235, pathology, 252, 253
surgery, 384 227 treatment, 254
Oxygen free radicals and brain dysembryoplastic Pineocytoma, 251, 252
edema, 52 neuroepithelial tumors, MRI, 251, 253
2278 pathology, 252
P-glycoprotein (product of MDR gangliogliomas and treatment, 254
gene), 28, 47 gangliocytomas, 229 Pinocytic vesicles (of capillary
gene (= MDR), 47 pineal region tumors endothelial cells), 46
p53/TP53 (gene and protein), 24, germ cell tumors, 2578 Pituitary apoplexy, 301
25, 27 parenchymal tumors, 2523 Pituitary gland, 296310
angiogenesis and, 44 skull base tumors anatomy, 2967
cytotoxic drug resistance and, 29 chordoma, 2878 anterior, see Anterior pituitary
esthesioneuroblastoma, 285 esthesioneuroblastoma, 285 posterior, see Posterior
glial tumors and, 155, 175 paraganglioma, 286 pituitary
immunohistochemical staining of vestibular schwannoma, 272 Pituitary tumors, 296310
protein, 91 PCV anterior, see Anterior pituitary
LiFraumeni syndrome and, 351 (procarbazinevincristineCCN benign, 297309, 310
therapeutic attempts using p53 U [lomustine]), 123, 129, malignant, see Malignant tumors
gene, 132 1634 posterior, 310
Pain anaplastic astrocytoma, 163 signs and symptoms, 77
head, see Headache anaplastic oligodendroglioma, Placental alkaline phosphatase,
radicular, 80 172 germ cell tumors, 260
Papillary craniopharyngioma, 310, PDGF and PDGFR, see Platelet- Placental growth factor and
311 derived growth factor angiogenesis, 44
Papilledema, 74 Pediatric tumors, see Children Placental lactogen, human, germ
Papillomas, choroid plexus, 181 Performance status, prognostic cell tumors, 260
Paraganglioma, 2856 factor in primary lymphoma, Plant alkaloids, 124, 1289
Paranasal sinus tumors, 2901 328, 3301 Plasmacytomas, 332

406
INDEX

Plateau/pressure waves, 55 intracranial, 4 PTCH gene, 18, 353


symptoms from, 75 classification, 6 PTEN (MMAC1) gene, 18, 23, 352
colloid cysts and, 360 epidemiologic data, 7, 8, 89, glial tumors and, 24, 154, 155
Platelet-derived growth factor 9 Puberty, precocious, germ cell
(PDGF) and its receptor, 21, site, 5 tumors, 2589
22 Primitive neuroectodermal tumors Pyrimidine analogs, halogenated,
diffuse astrocytomas and, 154 (PNET), 231, 232, 241, 243 115
meningiomas and, 194 Procarbazine, 127
Platelet endothelial growth factor, and vincristineCCNU, see PCV Quality of life, 138
45 Progesterone receptors and
Platinum compounds, 127 meningiomas, 193 Race and incidence, 9
hPMS-2, 353 Prognosis, 2931 Radiation
Podophyllins, 129 astrocytoma electromagnetic, see
Point mutations, 25 diffuse, 156, 165 Electromagnetic radiation
Pontine tumors, signs and pilocytic, 177 ionizing, 1516
symptoms, 77 craniopharyngioma, 314 meningioma and, 194
Positron emission tomography embryonal tumors, 222 physics, 10610
(PET), 80, 834 ependymoma, 1801 Radiation therapy (RT), 3, 10622
glial tumors hemangioblastoma, 211 chemotherapy and, see
diffuse astrocytoma, 158, 162 hemangiopericytoma, 208 Chemotherapy + RT
glioblastoma, 93 lymphomas, 328, 330, 3301 craniopharyngioma, 314
low-grade glioma, 83 medulloblastoma, 240 postoperative, 313, 314
oligodendroglioma, 171 melanocytic tumors, 213 dose, 10810
pilocytic astrocytoma, 1767 meningioma, 2056 evolution, 106, 107
hemangiopericytoma, 207 metastases, 3901 experimental methods, 110
lymphoma, 325 with poor prognosis systemic fractionation, see Fractionation
meningioma, 201 cancer, management, 388 glial tumors
neuronal/neuronalglial tumors neuronal/neuronalglial tumors, anaplastic astrocytoma, 163
dysembryoplastic 222 diffuse astrocytoma,
neuroepithelial tumor, 228 central neurocytoma, 226 postoperative, 1612
gangliogliomas and gangliogliomas and ependymoma, 180
gangliocytomas, 230 gangliocytomas, 231 gliomatosis cerebri, 166
preoperative, 100 oligodendroglioma, 173 oligodendrogliomas/anaplastic
Posterior fossa, pathologic diagnosis in, 89 oligodendrogliomas, 172,
dermoid/epidermoid cyst, 257, pineal germ cell tumors, 2656 1723
358 pineal parenchymal tumors, pilocytic astrocytoma, 177
Posterior pituitary grade and, 2523 hemangioblastoma, 211
(neurohypophysis), 297 skull base tumors hemangiopericytoma, 207
failure, 299 chordoma, 288 hemopoietic tumors, 333
tumors, 310 esthesioneuroblastoma, 285 lymphomas, 328, 331
Postoperative care, 1035 Prolactin Langerhans cell histiocytosis,
Precocious puberty, germ cell compression affecting levels, 300 335
tumors, 2589 normal secretion, 298 mechanism of action, 108
Prednisone, tumors secreting (prolactinomas), medulloblastoma, 2367
vincristinecyclophosphamide 300, 302, 3067 complications, 239, 240
doxorubicin and (CHOP), treatment, 3056, 3067 meningioma, 204, 2045
lymphoma, 32830 Proliferation rate (MIB-1 index), metastases, 3846, 388, 389
Preoperative evaluation, 1001 astrocytoma, 156 postoperative, 384
Primary tumors Prosopagnosia, 7980 prophylactic, lung cancer, 389
extracranial (giving intracranial Proto-oncogenes, see Oncogenes neuronal/neuronalglial tumors
metastases) Proton therapy, 114 central neurocytoma, 225
site, 368, 369 chondrosarcoma, 28990 gangliogliomas, 231
symptoms and signs of chordoma, 288 pineal region tumors
metastases before Pseudotumor cerebri, meningioma germ cell tumors, 263, 264
discovery of, 367, 370 mimicking, 201 parenchymal tumors, 254

407
INDEX

Radiation therapy (RT), (Contd) Rhabdoid tumors, 241, see also tumors identified in surgery for,
pituitary tumors, 305 Teratoid/rhabdoid tumors, 2212, 226
ACTH-secreting tumors, 308 atypical Selectins, 39
complications, 306 malignant, 214 Sellar region, 296319
planning and delivery, 11016 predisposition to, 341, 354 anatomy, 2967
second brain tumors after, Risk factors tumors, 297319
1516, 240 astrocytoma, 1523 signs and symptoms, 77
skull base tumors, 283 environmental, see types, 6, 297
chondrosarcoma, 28990 Environmental risk factors Sex, occurrence/incidence related
chordoma, 288 genetic, see Genetic changes; to, 910
esthesioneuroblastoma, 285 Syndromes germ cell tumors, 256
nasopharyngeal/paranasal Robotic surgical equipment, 106 metastases (intracranial), 369
tumors, 291 RTOG study, lymphomas, 328 Sex hormones and meningiomas,
paraganglioma, 286 Rutuximab, 331 1934
steroid dose decreased during, Sexual dysfunction, post-RT, 122
58 S100 protein, 91 Signs and symptoms (clinical
toxicity, see Neurotoxicity St Anne/Mayo classification of features predominantly
Radiation Therapy Oncology astrocytomas, 151 neurologic), 6680
Group, lymphomas, 328 Salvage therapy, lymphomas, 331 approaches to patient, 91, 92
Radicular pain, 80 Sarcoma, see also Chondrosarcoma of bloodbrain barrier
Radioisotopes in brachytherapy, granulocytic, 332, 333 disruption, 55
11314 in neurofibromatosis-1, 346 of brain edema, 54, 55
Radiology, see Imaging Scatter factor, 445 factors determining, 669
Radionuclide imaging, Schwannomas (neurilemmoma; false localizing, 72, 7880
esthesioneuroblastoma, 285 neurinoma; neuroma), 27181 focal, 72, 768
Radioprotectors, 11516 facial nerve, 280 generalized, 716
Radiosensitizers, 115 hypoglossal, 281 in history-taking, 65
Radiosurgery jugular foramen, 2801 pathophysiology, 6971
chordoma, 288 malignant, 2812 specific familial cancer
cranial nerve tumors trigeminal, 27880 syndromes
facial nerve schwannoma, vestibular, see Vestibular neurofibromatosis-1, 344, 346
280 schwannoma neurofibromatosis-2, 348
malignant tumors, 282 Secondary deposits, see Metastases tuberous sclerosis, 350, 351
vestibular schwannoma, 277 Second(ary) tumors (i.e. unrelated specific tumors/cysts/tumor-like
fractionated, 113 to original tumor type), lesions
germ cell tumors, 263 radiation leading to, 1516, astrocytoma, diffuse, 1578
hemangiopericytoma, 2078 240 astrocytoma, pilocytic, 176
melanocytic tumors, 213 Secretory tumors (and their central neurocytoma, 225
meningioma, 2045 symptomatology), 689 chordoma, 288
metastases, 3856, 389 meningioma, 192, 200 colloid cysts, 360, 3612
complications, 390 pituitary, see Hormones, craniopharyngioma, 31213
pituitary tumors, 305 pituitary dermoids/epidermoids, 3578
stereotactic, 11213 Seizures/convulsions (epileptic) esthesioneuroblastoma, 285
Ras and neurofibromin, 343 causes other than tumors, 160 facial nerve schwannoma, 280
Rathkes pouch/cleft drug management, see gangliogliomas and
cyst, 314 Anticonvulsants gangliocytomas, 230
tumor (craniopharyngioma), 256, presentation with, 76 hemangioblastoma, 21011
31014 approaches to patient, 91, 92 hemangiopericytoma, 2067
RB-1 gene, 18, 354 dysembryoplastic hemopoietic tumors (other
Renal cell cancer, metastases, neuroepithelial tumor, 228 than non-Hodgkins
21011 gangliogliomas and lymphomas), 332
Retinoblastoma, 18, 341, 342, gangliocytomas, 230 hypoglossal schwannoma,
354 meningioma, 197 281
Rhabdoid predisposition syndrome, metastases, 375 lymphomas, 324
18, 341, 354 oligodendroglioma, 171 medulloblastoma, 2345

408
INDEX

melanocytic tumors, 21213 Size, tumor craniopharyngioma, 313


meningioma, 197201 chemotherapy response and, 125 dermoids/epidermoids, 359
metastases, see Metastases (to pituitary, in classification, 2978 developments
brain) symptomatology and, 66, 68 future, 1056
nasopharyngeal/paranasal Skin involvement, familial brain past to present, 99100
tumors, 291 tumor syndromes, 341 epilepsy, tumor identified in,
oligodendroglioma, see tuberous sclerosis, 351 2212, 226
Oligodendroglioma Skull base tumors, 28291 extent, as prognostic factor, 30
paraganglioma, 286 chondromas, 214 glial tumors
pineal region tumors, 68, 77, classification, 270 anaplastic astrocytoma, 163
2534, 2589 cranial nerve compression, 270 anaplastic oligodendroglioma,
pituitary adenoma, 299301, treatment, 283 172
306, 307, 308, 309 Smell (sense of), loss, 79 brainstem glioma, see
trigeminal schwannoma, Social class and incidence, 12 Brainstem tumors
2789 Somatostatin analogs (incl. choroid plexus tumors, 182
vestibular schwannoma, 734, octreotide) ependymoma, 180
2723 GH-secreting tumor, 307 pilocytic astrocytoma, 177
surgery and worsening of, 390 prolactinoma, 306 pleomorphic
Simulator, RT, 112 Somatotroph, 298 xanthoastrocytomas, 178
Single photon emission computed adenoma, 307 goals/aims, 978, 98
tomography (SPECT), 80, failure, 299 hemangioblastoma, 211
845 SPECT, see Single photon emission hemangiopericytoma, 207
central neurocytoma, 225 computed tomography hemopoietic tumors, 3323
gangliogliomas and Spread, see Dissemination; Invasion lymphomas, 327, 331
gangliocytomas, 230 Staining, 91 invasion extent impacting on
Site/location/distribution of tumors, Stereotactic needle biopsy, see success, 40
5 Biopsy Langerhans cell histiocytosis,
cranial nerve tumors, 270 Stereotactic radiosurgery, 11213 335
glial cell tumors, 149, 150, 174, Stereotactic radiotherapy, 113 medulloblastoma, 2356
179, 180 Stereotactic surgery, frameless, 102 complications, 2389, 239
hemangioblastoma, 208 Steroids, see Corticosteroids; Sex melanocytic tumors, 213
hemangiopericytomas, 206 hormones meningioma, 2034
hemopoietic tumors, 320 StevensJohnson syndrome, metastases, 3834, 388
lymphomas, 320, 322 anticonvulsants, 133, 134 deaths, 389
histiocytic disorders, 334 Subependymal giant cell inaccessible to surgery, 388
melanocytic tumors, 211 astrocytoma, 1678 neuronal/neuronalglial tumors
meningioma, 190, 191, 259 Subependymomas, 181 central neurocytoma, 225
metastases Supportive therapy, 1338 dysembryoplastic
in brain, 5, 367, 372 metastatic disease, 3813 neuroepithelial tumor,
primary source, 368, 369 Suprasellar germ cell tumors 228
neuronal/neuronalglial tumors, prognosis, 266 gangliogliomas and
223 signs and symptoms, 258, 259 gangliocytomas, 2301
central neurocytoma, 223 Supratentorial tumors, pineal region tumors
dysembryoplastic symptomatology, 68, 72 germ cell tumors, 262, 264
neuroepithelial tumor, Surgery, 3, 97106, see also parenchymal tumors, 254
226 Radiosurgery pituitary tumors, 3045
gangliogliomas and cerebellar, mutism syndrome in ACTH-secreting tumors, 308,
gangliocytomas, 228 children, 239 3089
medulloblastoma, 232 colloid cysts, 360 GH-secreting tumors, 307
pineal region tumors, 250 cranial nerve tumors gonadotrophin-secreting
skull base tumors, 282, 283 facial nerve schwannoma, 280 tumor, 309
esthesioneuroblastoma, 284 malignant tumors, 282 prolactinoma, 307
paraganglioma, 2856 trigeminal schwannoma, 280 TSH-secreting tumor, 309
symptomatology related to, 66, vestibular schwannoma, 106, postoperative care, 1035
68, 77 2767, 277 preoperative evaluation, 1001

409
INDEX

Surgery, (Contd) Thrombosis, deep vein, 137 Tumor suppressor genes, 237
procedures, 1023 Thymidine kinase gene, HSV, 115, familial/hereditary syndromes
skull base, 283 132 and, 342
chondrosarcoma, 289 Thyroid dysfunction, Turcots syndrome, 18, 231, 343,
chordoma, 288 medulloblastoma, 239 3523
esthesioneuroblastoma, 285 Thyroid-stimulating hormone Tyrosine kinases, 22
nasopharyngeal/paranasal (TSH) secretion
tumors, 291 normal, 298 Ultrasonic aspirator, 103
paraganglioma, 286 tumor, 309 Ultrasound, intraoperative, 102
SV40, 17 Thyrotroph, 298
Symptoms, see Signs and failure, 299 Vaccines, antitumor, 132
symptoms tumor, 309 Varicella-zoster (herpes zoster)
Syndromes, cancer, Tight junctions of capillary metastases vs encephalitis caused
hereditary/familial/genetic, 18, endothelial cells, 46 by, 3789
340 Tinnitus, 79 protecting against glioma
astrocytoma, 18 paraganglioma, 286 occurrence, 17
medulloblastoma, 232 Tissue inhibitors of matrix Vascular endothelial growth factor
meningioma, 194 metalloproteinases (TIMPs), 40 (VEGF), 22
Systemic route, chemotherapy, 125 Topoisomerase inhibitors, 131 angiogenesis and, 22, 424, 45,
Topotecan, 131 50
Taenia spp., (cysticercosis), 17 Toxoplasmosis, SPECT brain edema and, 501
Tamoxifen, 131 differentiating tumor from, 85 hemangioblastoma and, 208
gliomas, 126, 165 TP53 gene, see p53 meningioma and, 200
Vascular endothelium, see
Taxanes, 129 Transforming growth factor-_, 21
Endothelium
Technetium-99m in SPECT, 84 Transforming growth factor-, 22
Vascular supply/vasculature, see
Telomerase, 28 Transsphenoidal surgery for
Blood vessels
metastasis and, 373 Cushings disease, 308
Vasculopathy, post-irradiation, 122
Temozolamide, 128 Trauma, see Head trauma medulloblastoma and, 240
Temporal lobe tumors Treatment/therapy, 97145, see Vasogenic edema, 52
epilepsy surgery identifying, 221, also specific tumors/methods craniopharyngioma, 313
226 genetic instability as problem in, Vasopressin deficiency in posterior
Signs and symptoms, 77 27 pituitary failure, 299
Tenascin-C and angiogenesis, 44 imaging assessing effectiveness, VEGF, see Vascular endothelial
Teniposide, 129 823 growth factor
Tentorium novel/experimental methods, 27, Venous circulation, metastases in,
tumors above, symptomatology, 110, 1312 371
68, 72 gliomas, 165 Venous thrombosis, deep, 137
tumors below, symptomatology, lymphomas, 331 Ventriculoperitoneal shunt,
68 Trigeminal nerve, 72 medulloblastoma, 235
Teratoid/rhabdoid tumors, atypical, Trigeminal schwannomas, 27880 Vertigo/dizziness, 734
2412 Trismus, 75 vestibular schwannoma, 734,
Teratoma (mature and immature), Truncal ataxia, 77 274
2556 TSC1 (hamartin) gene, 18, 349, 350 Vestibular function/dysfunction,
pathology, 258 product, 349, 350 734
treatment, 265 TSC2 (tubulin) gene, 34950, 350 vestibular schwannomas, 734,
TGF, see Transforming growth TSC2 gene, 18 274
factor TSH, see Thyroid-stimulating Vestibular schwannoma (acoustic
Thalamic tumors, Signs and hormone neuroma/neurinoma), 271,
symptoms, 77 Tuberous sclerosis, 18, 341, 2718
Thallium-201 in SPECT, 84 34950 clinical findings, 2725
Therapy, see Treatment Tubulin (TSC2 gene product), symptoms, 734, 2723
6Thioguanine, 131 34950, 350 differential diagnosis, 2756
Thiotepa, 128 Tumor-like lesions, 340 neurofibromatosis-2 and, 271,
Three-dimensional conformal RT, Tumor markers, germ cell tumors, 347, 348
112 260 treatment, 106, 2767

410
INDEX

VHL (disease), see von metastases vs, 3779 astrocytoma, 151


HippelLindau disease Visual cortex lesions (causing cranial nerve tumors, 271
VHL gene/gene product/mutations, blindness), 71, 7980 embryonal tumors, 6, 222,
18, 209, 348, 349 Visual disturbances, 74, 79, see 2312
Vinca alkaloids, 222 also Blindness neuronal/neuronalglial
Vincristine, 129 lymphomas, 324 tumors, 6, 222
and occipital tumor, 76 grading, astrocytoma, 156
doxorubicincyclophospham pituitary adenoma, 3001 Will Rogers effect, 152
ideprednisone (CHOP), Vomiting/emesis, tumors causing, World Health Organization, see
lymphoma, 32830 73 WHO
and lomustine (CCNU) medulloblastoma, 234
and cisplatin, von HippelLindau disease (and X-rays, 106
medulloblastoma, 237 VHL gene), 18, 341, 3489 Xanthoastrocytomas, pleomorphic,
and procarbazine, see PCV hemangioblastoma, 208, 209, 1778
toxicity, 129 210, 211, 3489 Xanthomatous lesions, differential
Viral infections, see also specific von Recklinghausens disease, see diagnosis, 209
viruses Neurofibromatosis-1
in etiology, 17 Yolk sac tumor (endodermal sinus
lymphomas, 3223 Wafers, see Implants tumor), 255
nasopharyngeal carcinoma WHO pathology, 258
and, 291 classification, 5, 6 prognosis, 265

411

You might also like