You are on page 1of 14

CME ARTICLE

A JH
CME Information: Chronic Myeloid Leukemia: 2016 update
on diagnosis, therapy and monitoring
CME Editor: Ayalew Tefferi, M.D.
Author: Elias Jabbour, M.D. and Hagop Kantarjian, M.D.

If you wish to receive credit for this activity, please refer to the website: www.wileyhealthlearning.com

Accreditation and Designation Statement


Blackwell Futura Media Services is accredited by the Accreditation Council for Continuing Medical Education to provide continuing medical
education for physicians.
Blackwell Futura Media Services designates this journal-based CME for a maximum of 1 AMA PRA Category 1 CreditTM. Physicians should
only claim credit commensurate with the extent of their participation in the activity.

Educational Objectives
Upon completion of this educational activity, participants will be better able to:

 Manage patients with newly diagnosed CML in CP


 Understand the milestones and indications for switch of therapy
 Manage patients with resistant disease
 Differentiate the TKIs available
 Manage patients with advanced phases, including the role of allogeneic stem cell transplantation

Activity Disclosures
No commercial support has been accepted related to the development or publication of this activity.
CME Editor: Ayalew Tefferi, M.D. has no relevant financial relationships to disclose.
Author: Elias Jabbour, M.D. and Hagop Kantarjian, M.D. have no relevant financial relationships to disclose.
This activity underwent peer review in line with the standards of editorial integrity and publication ethics maintained by American Journal of
Hematology. The peer reviewers have no conflicts of interest to disclose. The peer review process for American Journal of Hematology is single
blinded. As such, the identities of the reviewers are not disclosed in line with the standard accepted practices of medical journal peer review.
Conflicts of interest have been identified and resolved in accordance with Blackwell Futura Media Services Policy on Activity Disclosure and
Conflict of Interest. The primary resolution method used was peer review and review by a non-conflicted expert.

Instructions on Receiving Credit


This activity is intended for physicians. For information on applicability and acceptance of continuing medical education credit for this activity,
please consult your professional licensing board.
This activity is designed to be completed within one hour; physicians should claim only those credits that reflect the time actually spent in the
activity. To successfully earn credit, participants must complete the activity during the valid credit period, which is up to two years from initial
publication. Additionally, up to 3 attempts and a score of 70% or better is needed to pass the post test.
Follow these steps to earn credit:

 Log on to www.wileyhealthlearning.com
 Read the target audience, educational objectives, and activity disclosures.
 Read the activity contents in print or online format.
 Reflect on the activity contents.
 Access the CME Exam, and choose the best answer to each question.
 Complete the required evaluation component of the activity.
 Claim your Certificate.

This activity will be available for CME credit for twelve months following its launch date. At that time, it will be reviewed and potentially
updated and extended for an additional twelve months.

C 2016 Wiley Periodicals, Inc.


V

252 American Journal of Hematology, Vol. 91, No. 2, February 2016


ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES
AJH Educational Material A JH
Chronic myeloid leukemia: 2016 update on diagnosis,
therapy, and monitoring
Elias Jabbour* and Hagop Kantarjian

Disease overview: Chronic Myeloid Leukemia (CML) is a myeloproliferative neoplasm with an incidence of 1-2
cases per 100,000 adults. It accounts for approximately 15% of newly diagnosed cases of leukemia in adults.
Diagnosis: CML is characterized by a balanced genetic translocation, t(9;22)(q34;q11.2), involving a fusion of
the Abelson gene (ABL1) from chromosome 9q34 with the breakpoint cluster region (BCR) gene on
chromosome 22q11.2. This rearrangement is known as the Philadelphia chromosome. The molecular
consequence of this translocation is the generation of a BCR-ABL1 fusion oncogene, which in turn translates
into a BCR-ABL oncoprotein. Frontline therapy: Three tyrosine kinase inhibitors (TKIs), imatinib, nilotinib, and
dasatinib are approved by the United States Food and Drug Administration for first-line treatment of patients
with newly diagnosed CML in chronic phase (CML-CP). Clinical trials with 2nd generation TKIs reported
significantly deeper and faster responses; their impact on long-term survival remains to be determined.
Salvage therapy: For patients who fail frontline therapy, second-line options include second and third
generation TKIs. Although second and third generation TKIs are potent and selective TKIs, they exhibit
unique pharmacological profiles and response patterns relative to different patient and disease
characteristics, such as patients comorbidities, disease stage, and BCR-ABL1 mutational status. Patients who
develop the T315I gatekeeper mutation display resistance to all currently available TKIs except ponatinib.
Allogeneic stem cell transplantation remains an important therapeutic option for patients with CML-CP who
have failed at least two TKIs, and for all patients in advanced phase disease.
Am. J. Hematol. 91:253265, 2016. V
C 2015 Wiley Periodicals, Inc.

Disease Overview
Chronic Myeloid Leukemia (CML) is a myeloproliferative neoplasm with an incidence of one to two cases per 100,000 adults. It accounts for
approximately 15% of newly diagnosed cases of leukemia in adults [1]. In 2015, it is estimated about 7,000 new CML cases will be diagnosed in
the United States, and about 1,100 patients will die of CML. Since 2000, the year of introduction of imatinib, the annual mortality in CML has
decreased from 1020% down to 12% [1]. Consequently, the prevalence of CML in the United States, estimated at about 25-30,000 in 2000, has
increased to an estimated 801,00,0001 in 2015, and will reach a plateau of about 1,80,000 cases by 2030 [2].
Central to the pathogenesis of CML is the fusion of the Abelson murine leukemia (ABL1) gene on chromosome 9 with the breakpoint cluster
region (BCR) gene on chromosome 22. This results in expression of an oncoprotein termed BCR-ABL1 [3]. BCR-ABL1 is a constitutively active
tyrosine kinase that promotes growth and replication through downstream pathways such as RAS, RAF, JUN kinase, MYC and STAT [410] This
influences leukemogenesis by creating a cytokine-independent cell cycle with aberrant apoptotic signals in response to cytokine withdrawal.
Until a little more than a decade ago, drug therapy for CML was limited to nonspecific agents such as busulfan, hydroxyurea, and interferon-
alfa (INF-a) [11]. INF-a led to disease regression and improved survival but was hindered by its modest efficacy and a multitude of toxicities. Allo-
geneic stem cell transplantation (allo-SCT) is curative, but carries risks of morbidity and mortality. Further, allo-SCT is an option only for patients
with good performance status and organ functions, and who have an appropriate stem cell donor.
The CML therapeutic landscape changed dramatically with the development of small molecule tyrosine kinase inhibitors (TKIs) that potently inter-
fered with the interaction between the BCR-ABL1 oncoprotein and adenosine triphosphate (ATP), blocking cellular proliferation of the malignant
clone. This targeted approach altered the natural history of CML, improving the 10-year survival rate from approximately 20% to 8090% [1.2,12].
In this review, we will highlight the evidence supporting the use of each of the available TKIs, including how to select an agent in various cir-
cumstances and phases of the disease. Allo-SCT is an important treatment option in CML chronic phase (CP) post TKIs failure and in advanced
CML phases, and its role will be discussed. Cytogenetic and molecular benchmarks for patients on therapy will be discussed. Finally, appropriate
monitoring strategies for patients on various TKIs will be covered.

Department of Leukemia, the University of Texas M. D. Anderson Cancer Center, Houston, Texas
Conflict of interest: Nothing to report
*Correspondence to: Elias Jabbour, MD Anderson Cancer Center, Houston, Texas, USA, Box 428, 1515 Holcombe Blvd, Houston, TX 77030. E-mail: ejab-
bour@mdanderson.org
Received for publication: 7 December 2015; Accepted: 9 December 2015
Am. J. Hematol. 91:253265, 2016.
Published online: in Wiley Online Library (wileyonlinelibrary.com).
DOI: 10.1002/ajh.24275

C 2015 Wiley Periodicals, Inc.


V

doi:10.1002/ajh.24275 American Journal of Hematology, Vol. 91, No. 2, February 2016 253
Jabbour and Kantarjian ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES

Manifestations and Staging The Ph chromosome is usually present in 100% of metaphases, often
as the sole abnormality. Ten to 15% of patients have additional chro-
About 50% of patients with CML diagnosed in the United States mosomal changes (clonal evolution) involving trisomy 8, isochromo-
are asymptomatic. The disease is diagnosed often after a routine some 17, additional loss of material from 22q or double Ph, or others.
physical examination or blood tests. CML can be classified into three Ninety percent of patients have a typical t(9;22); 5% have variant
phases: CP, accelerated phase (AP), and blast phase (BP). Most (90 translocations which can be simple (involving chromosome 22 and a
95%) patients present in CML-CP. Common signs and symptoms of chromosome other than chromosome 9), or complex (involving one
CML-CP, when present, result from anemia and splenomegaly. These or more chromosomes in addition to chromosomes 9 and 22).
include fatigue, weight loss, malaise, easy satiety, and left upper quad- Patients with Ph-variants have response to therapy and prognosis
rant fullness or pain. Rare manifestations include bleeding (associated similar to Ph-positive CML. About 25% of patients present with a
with a low platelet count and/or platelet dysfunction), thrombosis morphologic picture of CML without the Ph-positivity by cytogenetic
(associated with thrombocytosis and/or marked leukocytosis), gouty studies. FISH and PCR document Ph-negative BCR-ABL1 rearranged
arthritis (from elevated uric acid levels), priapism (usually with CML. Such patients have similar response and outcome on TKI ther-
marked leukocytosis or thrombocytosis), retinal hemorrhages, and apy as patients with Ph-positive CML.
upper gastrointestinal ulceration and bleeding (from elevated hista- Bone marrow aspiration is mandatory for all patients in whom
mine levels due to basophilia). Leukostatic symptoms (dyspnea, CML is suspected, as it will confirm the diagnosis (eg, cytogenetic anal-
drowsiness, loss of coordination, confusion) due to leukemic cells ysis), and provide information needed for staging in terms of the blast
sludging in the pulmonary or cerebral vessels, are uncommon in CP and basophil percentages. Baseline reverse transcriptase-polymerase
despite white blood cell (WBC) counts exceeding 100 x 109/L. Spleno- chain reaction is imperative to identify the specific type of rearrange-
megaly is the most consistent physical sign detected in 4050% of ment that can be appropriately followed when assessing for response to
cases. Hepatomegaly is less common (less than 10%). Lymphadenopa- TKI therapy. About 2-5% of patients have b2a3 or b3a3 (not b2a2 or
thy and infiltration of skin or other tissues are rare. When present, b3a2) variants of p210 BCR-ABL1 or p230 transcripts that may yield a
they favor Ph-negative CML or AP or BP of CML. Headaches, bone false negative PCR by routine probes and (if not tested at diagnosis)
pain, arthralgias, pain from splenic infarction, and fever are more fre- would give the false impression that a patient may be in complete
quent with CML transformation. Most patients evolve into AP prior molecular response on TKI.
to BP, but 20% transit into BP without AP warning signals. CML-AP
might be insidious or present with worsening anemia, splenomegaly
and organ infiltration; CML-BP presents as an acute leukemia (mye- Differential Diagnosis
loid in 60%, lymphoid in 30%, megakaryocytic or undifferentiated in CML must be differentiated from leukemoid reactions, which usu-
10%) with worsening constitutional symptoms, bleeding, fever and ally produce WBC counts, lower than 50 3 109/L, toxic granulocytic
infections. vacuolation, Dohles bodies in the granulocytes, absence of basophilia,
and normal or increased LAP levels. The clinical history and physical
Diagnosis examination generally suggest the origin of the leukemoid reaction.
Corticosteroids can rarely cause extreme neutrophilia with a left shift,
The diagnosis of typical CML is simple and consists of document- but this abnormality is transient and of short duration.
ing, in the setting of persistent unexplained leukocytosis (or occasion- CML may be more difficult to differentiate from other myeloproli-
ally thrombocytosis), the presence of the Philadelphia (Ph) ferative or myelodysplastic syndromes. Patients with agnogenic mye-
chromosome abnormality, the t(9;22)(q34;q11), by routine cytogenet- loid metaplasia with or without myelofibrosis frequently have
ics, or the Ph-related molecular BCR-ABL1 abnormalities by fluores- splenomegaly, neutrophilia, and thrombocytosis. Polycythemia vera
cence in situ hybridization (FISH) or by molecular studies [1315]. with associated iron deficiency, which causes normal hemoglobin and
A FISH analysis relies on the co-localization of large genomic hematocrit values, can manifest with leukocytosis and thrombocytosis.
probes specific to the BCR and ABL genes. Comparison of simultane- Such patients usually have a normal or increased LAP score, a WBC
ous marrow and blood samples by FISH analysis shows high con- count less than 25 3 109/L, and no Ph abnormality.
cordance. FISH studies may have a false positive range of 15% The greatest diagnostic difficulty lies with patients who have spleno-
depending on the probes used. megaly and leukocytosis but who do not have the Ph chromosome. In
Reverse transcriptase-polymerase chain reaction (RT-PCR) amplifies some, the BCR-ABL1 hybrid gene can be demonstrated despite a nor-
the region around the splice junction between BCR and ABL1. It is mal or atypical cytogenetic pattern. Patients who are Ph negative and
highly sensitive in detecting minimal residual disease. PCR testing can BCR-ABL1 negative are considered to have Ph-negative CML or chronic
either be qualitative (QPCR), providing information about the presence myelomonocytic leukemia. Rarely, patients have myeloid hyperplasia,
of the BCR-ABL1 transcript, or quantitative, assessing the amount of which involves almost exclusively the neutrophil, eosinophil, or basophil
BCR-ABL1 transcripts. Qualitative PCR is useful for diagnosing CML; cell lineage. These patients are described as having chronic neutrophilic,
quantitative PCR is ideal for monitoring residual disease. Simultaneous eosinophilic, or basophilic leukemia and do not have evidence of the Ph
peripheral blood and marrow QPCR studies show a high level of con- chromosome or the BCR-ABL1 gene. Isolated megakaryocytic hyperpla-
cordance. False-positive and false-negative results can happen with sia can be seen in essential thrombocythemia, with marked thrombocy-
PCR. False-negative results may be from poor-quality RNA or failure tosis and splenomegaly. Some patients who present with clinical
of the reaction; false-positive results can be due to contamination. A characteristics of essential thrombocythemia (with marked thrombocyto-
0.51 log difference in some samples can occur depending on testing sis but without leukocytosis) have CML; cytogenetic and molecular
procedures, sample handling, and laboratory experience [1315]. For studies showing the Ph chromosome, the BCR-ABL1 rearrangement, or
correlative purpose and monitoring without necessarily performing both lead to the appropriate diagnosis and treatment.
repeat marrow studies, a complete cytogenetic response (CCyR; 0%
Ph-positive metaphases by cytogenetic) is equivalent to a negative
FISH test (62%) and BCR-ABL1 transcripts by International Standard Frontline Treatment Options
[IS] <1%. A partial cytogenetic response (Ph-positive metaphases The three commercially available TKIs for the treatment of CML
35%) is equivalent to BCR-ABL1 transcripts 10% [IS]. include imatinib, dasatinib, and nilotinib. Current guidelines endorse

254 American Journal of Hematology, Vol. 91, No. 2, February 2016 doi:10.1002/ajh.24275
ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES Chronic myeloid leukemia

TABLE I. Summary of Pivotal Phase III Trials of Approved Tyrosine Kinase Inhibitors for the Frontline Treatment of Chronic Myeloid Leukemia

BCR-ABL < 10% Longest


Trial Treatment CCyR (%) MMR (%) at 3 months (%) EFS/PFS (%) OS (%) follow-up (years)

At 6 years At 6 years
IRIS Imatinib (n 5 304) 83 86 NR 81 85 8
At 2 years At 5 years At 5 years
DASISION Dasatinib (n 5 259) 86 76 84 85 91 5
Imatinib (n 5 260) 82 64 64 86 90
At 2 years At 5 years At 5 years At 6 years
ENESTnd Nilotinib 300 mg (n 5 282) 87 77 91 95 92 6
Nilotinib 400 mg (n 5 281) 85 77 89 97 96
Imatinib (n 5 283) 77 60 67 93 91

NR5Not reported

all three as options for the initial management of CML in the chronic arms (imatinib 400 mg once daily, imatinib 600 mg once daily, imati-
phase (CML-CP) (Table I). nib 400 mg once daily plus peginterferon alfa-2a, or imatinib 400 mg
once daily plus subcutaneous cytarabine) [19]. Patients were initially
Imatinib assigned to receive peginterferon alfa-2a at a dose of 90 mcg once
Imatinib mesylate was the first TKI to receive approval by the weekly. Because of a high rate of discontinuation due to toxicity, the
Food and Drug Administration (FDA) for the treatment of patients dose was later modified to 45 mcg once weekly. At 12 months, rates
with CML-CP. It acts via competitive inhibition at the ATP-binding of CCyR were similar among the four groups. The imatinib plus
site of the BCR-ABL1 oncoprotein, which results in the inhibition of peginterferon alfa-2a treated group obtained higher rates of MMR
phosphorylation of proteins involved in cell signal transduction. It and deeper molecular responses, but follow-up was not sufficient to
efficiently inhibits the BCR-ABL1 kinase, but also blocks the platelet- define the impact on long-term outcomes.
derived growth factor receptor (PGDFR) and the C-KIT tyrosine
kinase [16]. Dasatinib
The International Randomized Study of Interferon and STI571 Dasatinib is an oral, second generation TKI that is 350 times more
(IRIS) study is considered a landmark clinical trial for TKIs and potent than imatinib in vitro [2022]. It also inhibits the Src family
CML [17]. Investigators randomized 1,106 patients in CML-CP to of kinases, which may be important in blunting critical cell signaling
receive imatinib 400 mg/day or INF-a plus low-dose cytarabine. After pathways [23]. Though initially evaluated in patients in the salvage
a median follow-up of 19 months, outcomes for patients receiving setting, it was later compared in frontline CML to imatinib to test the
imatinib were significantly better than in those treated with INF-a possibility that frontline use of the more potent TKIs might improve
plus cytarabine, notably the rates of CCyR rate (74% vs. 9%, outcomes compared to imatinib.
P < 0.001), and freedom-from-progression to AP or BP at 12 months The DASISION trial was a phase III randomized study comparing
(99% vs. 93%, P < 0.001). Further highlighting the challenge of using imatinib 400 mg once daily to dasatinib 100 mg once daily in newly
IFN-a was the high crossover rate to imatinib due to intolerance. The diagnosed patients with CML [24]. The primary outcome was con-
responses to imatinib were also durable: in an 8-year follow-up of the firmed CCyR (cCCyR) at 12 months. A total of 519 patients were
IRIS study [12], the estimated event-free survival rate was 81%, and randomized (1:1). Patients assigned to dasatinib achieved cCCyR at
the overall survival (OS) rate was 93% when only CML-related deaths 12 months more often than those on imatinib (77% vs. 66%,
were considered. P 5 0.007). Many of the secondary endpoints of interest also favored
While the results using imatinib are impressive, only 55% of the dasatinib arm. A five-year follow-up showed that dasatinib
patients enrolled in the IRIS study remained on therapy at the 8-year induced more rapid and deeper responses at early time points com-
follow-up time. This underscored the need for additional treatment pared to imatinib [25]. At 3 months, a higher proportion of patients
options for patients who had failed or were intolerant to imatinib. treated with dasatinib achieved BCR-ABL1 transcripts [IS]  10%
This led to the rational development of second generation TKIs that (84% vs. 64%, P < 0.0001). Meeting this threshold in either arm pre-
would effectively treat patients unable to continue on imatinib dicted for better progression-free survival and OS. Transformations to
therapy. CML-AP or CML-BP were fewer in patients treated with dasatinib
versus imatinib (4.6% vs. 7.3%).
High-dose imatinib and imatinib-based combination In another multicenter trial, several North American cooperative
Other strategies for frontline therapy include using higher doses of groups randomized patients with newly diagnosed CML-CP to either
imatinib or combining a TKI with an additional agent, such as inter- dasatinib 100 mg once daily or imatinib 400 mg once daily [26]. Sim-
feron. In the Tyrosine Kinase Inhibitor Optimization and Selectivity ilar to the results of the DASISION study, dasatinib treated patients
(TOPS) study, patients were randomized to receive imatinib 400 mg achieved higher rate of CCyR compared to patients receiving imatinib
once daily or twice daily (800 mg) [18]. Major molecular response (84% vs. 69%, P 5 0.04). There was more toxicity experienced in the
(MMR) rate at 12 months was the study primary endpoint. with cyto- dasatinib armGrades 3 or 4 adverse events 58% with dasatinib and
genetic response and time to such responses collected as secondary 35% imatinib), Mostly hematologic toxicity [26].
outcomes. Patients in the high-dose group achieved faster CCyR and A third Phase 3 randomized study, SPIRIT 2, compared imatinib
MMR, but rates were not significantly different at 12 months. 400 mg daily with dasatinib 100 mg daily [27]. The primary endpoint
Interferon has re-emerged as an interesting therapeutic option in of this trial is EFS at 5 years, with the rate of achievement of MMR,
CML with the advent of pegylated formulations requiring less fre- a key secondary endpoint. The interim results showed the 12-month
quent administration and improved tolerability. In a phase III MMR rates with dasatinib versus imatinib to be 58% versus 43% (P
randomized study, patients were assigned to one of four treatment <0.001). The 12-month CCyR rates were 51% and 40% (P <0.002),

doi:10.1002/ajh.24275 American Journal of Hematology, Vol. 91, No. 2, February 2016 255
Jabbour and Kantarjian ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES

TABLE II. Frontline TKIs Therapy: MDACC Experience limited elevation of indirect bilirubin (10%), elevations of blood sugar
(1020%), and rare pancreatitis (12%).
Imatinib Imatinib
% Cumulative 400 mg 800 Nilotinib Dasatinib
The MD Anderson Cancer Center (MDACC)
experience
CCyR 85 90 99 97
MR4.5 56 74 80 78 We recently published the long-term responses and outcomes of
patients with CML-CP and provided a comparison of four commonly
used TKI modalities (Table II) [32]. Unlike previous reports that
compared only imatinib 400 mg with either imatinib 800 mg, dasati-
respectively. Progression rate to CML-AP was 0.7% with imatinib and nib, or nilotinib in randomised trials, our study provided a compara-
0.5% with dasatinib. The progression rate to CML-BP was 1.7% ver- tive analysis of four TKI modalities after a long follow-up. The
sus 1%. analysis included 482 patients treated (July 2000 - September 2013)
Pleural effusions occurred more frequently on dasatinib (19% vs. in consecutive or parallel clinical trials, with imatinib 400 mg daily
<1%). Other side effects of dasatinib include myelosuppression (n 5 68), imatinib 800 mg daily (n 5 200), dasatinib 50 mg twice
(20%), and rare pulmonary hypertension (12%). daily or 100 mg daily (n 5 106), or nilotinib 400 mg twice daily
(n 5 108). More patients receiving imatinib 800 mg or second-
Nilotinib generation TKIs (i.e., dasatinib or nilotinib) achieved CCyR (58
Nilotinib is a structural analog of imatinib. Its affinity for the ATP- [87%] of 67 for imatinib 400 mg vs. 180 [90%] of 199 for imatinib
binding site on BCR-ABL1 is 30-50 times more in vitro [28]. Like 800 mg, vs. 100 [96%] of 104 for dasatinib vs. 99 [93%] of 107 for
dasatinib, nilotinib initially demonstrated the ability to induce hemato- nilotinib), MMR (51 [76%] vs. 171 [86%] vs. 93 [90%] vs. 97 [91%]),
logic and cytogenetic responses in patients who had failed imatinib. and 45 log or higher reduction in BCR-ABL1 transcripts (MR4.5
Similar to the data with dasatinib, nilotinib was also compared to response 38 [57%] vs. 148 [74%] vs. 76 [71%] vs. 76 [71%]). This
imatinib in a large, international, randomized study (ENEST-nd). In finding was consistent over time (360 months); where at any time
ENEST-nd, two doses of nilotinib (300 mg or 400 mg twice daily) point, imatinib induced lower rates of cytogenetic and molecular
were compared to imatinib 400 mg once daily [29]. The primary end- responses. This landmark assessment contrasts with previous reports
point was the rate of MMR at 12 months. This endpoint was where results are reported cumulatively (by not at) and can be
achieved at statistically significantly higher rates for both doses of misleading. The 5-year event-free survival significantly differed
nilotinib compared to imatinib (44% and 43% vs. 22%, P < 0.001). between the imatinib 400 mg group and the other TKI groups (imati-
The cumulative incidence of CCyR by 24 months was 87% with nilo- nib 800 mg P 5 0029, dasatinib P 5 0003, nilotinib P 5 0.031). How-
tinib 300 mg twice daily, 85% with nilotinib 400 mg twice daily, and ever, there was no significant difference in the 5-year failure-free
77% with imatinib 400 mg daily (P < 0.001) [29]. survival (P 5 0.32, P 5 0.075, P 5 0.332), transformation-free survival
With a minimum follow-up of 5 years, the two arms of nilotinib (P 5 0.053, P 5 0.038, P 5 0.493), or OS (P 5 0.563, P 5 0.162,
demonstrated better early results compared with imatinib [30]. The P 5 0.981). We also compared event-free and OS according to
cumulative incidences of MMR by 60 months were 77%, 77%, and whether or not MMR or MR4.5 response was achieved in addition to
60%, respectively (P <0.0001%). The incidences of BCR-ABL1 tran- CCyR. As expected, in patients who achieved CCyR, no difference in
scripts [IS] < 0.0032% (roughly equivalent to a 4.5 log reduction of outcomes was observed whether MMR or MR4.5 response was
disease) by 72 months were 56%, 55%, and 33%, respectively (P achieved or not [32].
<0.0001%). The incidences of transformation to AP or BP were 3.9%, Current guidelines recommend any of the three TKIs, imatinib, das-
2.1%, and 7.4%, respectively (P 5 0.06 and 0.003, respectively). The tinib or nilotinib as good therapeutic options with a category 1 recom-
estimated 5-year EFS rates were 95%, 97%, and 93%, respectively. mendation for initial treatment of CML-CP [33]. Second generation
The estimated 5-year survival rates were 94%, 96%, and 92%, respec- TKIs produced higher rate of early optimal responses, but have so far
tively. While nilotinib was superior to imatinib across all Sokal score noimpact on long-term survival (probably because of available effective
categories in inducing higher rates of CCyR and MMR, the advantage salvage therapies). The main advantage of second generation TKI is
in reducing the rates of transformation was more pronounced in obtained in patients with high-risk disease; a relevant decrease in the
patients with intermediate- and high-Sokal risk CML. The rates of rate of transformation to AP and a BP was achieved with nilotinib
transformations were 1% and 1% and 0% among patients with low- and dasatinib. As such, second generation TKIs in the frontline setting
Sokal risk treated with nilotinib 300 mg orally twice daily, nilotinib could be reserved to patients with higher risk disease. Higher dose ima-
400 mg orally twice daily, or imatinib 400 mg orally once. The rates tinib and combination approaches in the frontline setting are investiga-
were 2%, 1%, and 10% among patients with intermediate-Sokal risk tional due to conflicting results of various studies to date. These
and 9%, 5%, and 11% among patients with high-Sokal risk. strategies are also not benign interventions, as they add to the eco-
In a second randomized trial with the same design that enrolled 267 nomic and toxicity burden of the overall treatment plan. Allo-SCT or
Chinese patients, the MMR rate was 52% at 12 months with nilotinib other chemotherapy agents are not recommended as upfront treatments
compared with 28% with imatinib [31]. However, the rates of both for CML-CP given the excellent outcomes and long-term survival
CCyR (84% vs. 87%) and progression-free survival (95% each) were achieved with the TKIs. An exception may be in emerging nations
similar at 24 months. Overall, six patients in each arm progressed to where allo-SCT is a one-time procedure costing $14-20,000, accessible
AP/BP. In both arms, the estimated 2-year survival rate was 98%. to most patients. On the other hand generic imatinib in such geogra-
While nilotinib therapy was overall well tolerated, there was an phies (e.g. India) costs only less than $400 per year of therapy. At
increased risk of accumulated vascular events on therapy. The 6-year MDACC, patients with low-risk disease are offered frontline therapy
cumulative cardiovascular event rates were 9.9%, 15.9%, and 2.5%, with imatinib. Patients with high-risk disease are candidates for
among patients treated with nilotinib 300 mg twice daily, nilotinib second-generation TKIs for the time being; this practice may change
400 mg twice daily, and imatinib 400 mg daily, respectively [30]. once the generic formulation of imatinib is available in the United
Other notable side effects are headache and skin rashes (common States. Because of the higher rates of durable complete molecular
220-30%- but mild to moderate; alleviated by dose reduction), self- responses with second generation TKIs (which could lead to

256 American Journal of Hematology, Vol. 91, No. 2, February 2016 doi:10.1002/ajh.24275
ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES Chronic myeloid leukemia

discontinuation of TKI therapy and potential molecular cures discussed therapy, the annual price of imatinib in 2014 had quadrupled to
later), considerations of second generation TKIs in younger patients $132,000. This price is in the same range as dasatinib and nilotinib,
with CML (e.g. age <50 years) vs. older patients, may be entertained. both priced above $130,000/year of therapy.
Today, the prices of TKIs are all fairly comparable. However, in
Selecting a Frontline Therapy February 2016 imatinib will be available as a generic formulation.
Patients age and comorbidities and TKI toxicity profile. While The price will eventually fall dramatically below $5-8,000/year (as in
multiple TKIs are available for patients with newly diagnosed patient Canada; the annual price of generics in India is only $400). Physi-
with CML in CP, each has a distinct toxicity profile to consider when cians will then have to assess the treatment value of second genera-
deciding on a therapy. Most TKIs are reasonably well tolerated with tion TKI (dasatinib or nilotinib) in the frontline setting against the
adequate monitoring and supportive care. For patients at risk of develop- generic imatinib in relation to benefits versus cost. Second generation
ing pleural effusions (existing lung injuries), TKI other than dasatinib TKIs may be offered for patients with high-risk disease, while imati-
should be selected. This might be relevant for patients with a history of nib and/or its generic formulation will be offered for patients with
lung disease (e.g., chronic obstructive pulmonary disease), cardiac disease low-risk disease. Experts recently assessed, following a Markov model,
(e.g., congestive heart failure), or uncontrolled hypertension. Pulmonary the most cost-effective strategy for treating newly diagnosed CML-CP
arterial hypertension (PAH) is also a rare important complication of after imatinib loses patent exclusivity [45]. They found that initiating
dasatinib [34], and patients with preexisting PAH may be considered for imatinib as frontline treatment and treating in a stepwise approach,
alternative TKIs in the frontline setting. Dasatinib also inhibits platelets compared to physician choice, costs less and offered clinically equiva-
function [35], and patients taking concomitant anticoagulants may be at lent utility. In this analysis, the stepwise therapy was found to have
an increased risk of hemorrhagic complications [36]. an incremental cost-effectiveness ratio (ICER) of $227,136/Quality-
Nilotinib has been associated with hyperglycemia; caution should adjusted life years (QALY). The ICER was favorable for stepwise ther-
be exercised in patients with uncontrolled diabetes when initiating apy for each Sokal risk group. Furthermore, the efficacy and safety of
therapy, and avoided or prescribed with caution in patients with dia- generic imatinib was compared to the patented drug. In a series of
betes or history of pancreatitis. During preclinical development, nilo- 972 patients treated with either patented (n 5 671) or generic imati-
tinib was shown to potentially prolong the QT interval, and nib (n 5 237) at Tata Memorial Hospital in Mumbai, the rates of
parameters were put in place to monitor for this complication after CCyR were 72% and 75%, respectively; the 5-year survival rate was
the drug was approved; potassium and magnesium should be repleted 86% [43,44].
to appropriate serum levels before starting nilotinib or determining Disease characteristics. For patients with CP-CML it is common to
an individual patients QT interval. Patients should always be coun- use one of the available risk stratification scores, such as Sokal [47] or
seled to take nilotinib in a fasting state to avoid excess drug exposure. Hasford [48] to help predict outcomes. Patients with low-risk disease
Nilotinib has also been associated with vasospastic and vaso-occlusive are expected to have optimal responses when imatinib, dasatinib, or
vascular events, such as ischemic heart disease, ischemic cerebrovas- nilotinib. However, selecting a second generation TKI as frontline ther-
cular events, and peripheral artery occlusive disease (PAOD) [3740]. apy (i.e. dasatinib or nilotinib) based upon the Sokal or Hasford scores
In the 6-year follow-up on the ENEST-nd trial [30], approximately was proven more beneficial in patients with intermediate or high risk
10% of patients experienced vascular events. Nilotinib use should be disease, as shown in the DASISION and ENEST-nd trials [24,25,29,30].
limited in patients with risk factors such as diabetes mellitus or coro- Patients with higher risk disease have a lower likelihood of achieving
nary or cerebrovascular artery disease. Avoiding nilotinib in patients the early milestones of CCyR and MMR and, particularly, higher chan-
with significant past vascular histories is warranted with the availabil- ces of disease transformation to AP-CML or BP-CML.
ity of other viable options. We recently reported that the expression of the b2a2 transcript
Imatinib causes bothersome quality-of-life side-effects including type correlates with an inferior response rate and long-term outcome,
weight gain, fatigue, peripheral and periorbital edema, bone and muscle and these associations are independent of other prognostic factors
aches, nausea and others. However, most are mild to moderate. Less [49]. Our analysis suggested that the inferior outcome observed for
than 510% experience elevations in creatinine with long-term therapy. patients with no major cytogenetic response (MCyR) at 3 months are
Finally, patients age plays an important role in treatment decision. largely restricted to patients with b2a2 transcripts. The inferior out-
Patients younger than 50 years are expected to live 301 more years. come for patients treated with standard-dose imatinib might be
Therefore, inducing a durable CMR may potentially lead to therapy
mostly restricted to patients with b2a2. These observations merit pro-
discontinuation. Second generation TKIs induce a significantly higher
spective validation, but they suggest that the transcript type might be
rate of CMR compared with imatinib. The issue of durable CMR and
a biomarker that may help select therapy and course of action for
potential therapy discontinuation plays a less important role in
patients with CML treated with TKI [49].
elderly patients where the expected survival is shorter and discontinu-
Any of the thTKIs currently approved for frontline CML therapy
ing therapy is less relevant.
may be selected. These include imatinib, dasatinib, or nilotinib. While
Cost. The cost of cancer drugs has risen exponentially over the
past decade [41]. All anticancer agents approved in 2014 were priced dasatinib and nilotinib have demonstrated superiority over imatinib
at over $120,000 annually [41]. Unlike TKIs in CML, most of these when early surrogate markers are used, imatinib is still highly effective
therapies do not provide a substantial improvement in survival or in a large number of patients with CML. At MDACC, when choosing
other objective measures to justify the cost. An international group of an agent, we consider issues such as comorbidities, patients age,
CML experts called attention to the high prices of TKIs [42]. The adverse event profile, risk stratification score, transcripts type, and
case of imatinib was used to illustrate the problem with the high can- cost. Kinase domain mutation profile plays no role in selecting an ini-
cer drug prices. When first approved, the annual price for imatinib tial TKI, but becomes relevant in the relapse setting.
was less than $30,000 in the United States. This was the price set to
make the development and commercialization of imatinib profitable Monitoring Treatment Response:
when it was expected that most patients would be treated for perhaps Surrogate Endpoints and Milestones
510 years. Patients now remain on imatinib and live a normal life-
span as long as they are compliant with the medication [43,44]. Para- Because patients with CML on TKI therapy are expected to live for
doxically, with a higher number of patients and a longer duration of a long period of time, surrogate markers of outcome are important.

doi:10.1002/ajh.24275 American Journal of Hematology, Vol. 91, No. 2, February 2016 257
Jabbour and Kantarjian ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES

In general, achieving a deeper response faster has been associated [25,29,55,56]. The important question is what to advise a patient who
with improved outcome, though the result of molecular testing is does not meet the 3-month benchmark? One option is to switch
dependent on the laboratory and their techniques. Advances in tech- TKIs early, but there are no data that this will alter long-term out-
nology have made available tests that avoid the need for traditional come. Several experts suggested that a follow-up measurement at 6
bone marrow examinations for treatment monitoring (except when months will help define patients clearly in need of a change in ther-
changing TKI; or in unusual situations like unexpected myelosuppres- apy [49]. This strategy has been retrospectively analyzed by several
sion in order to exclude transformation or the development of myelo- large groups with conflicting results [5759]. The results of two inde-
dysplastic syndrome or other marrow conditions). pendent study groups have suggested that all patients with BCR-ABL1
transcripts > 10% at 3 months do not necessarily have an inferior
Important points for monitoring and determining outcome [58,59]. Patients who continued on therapy and achieved
treatment failure transcripts levels less than 10% by 6 months had the same long-term
At baseline, all patients should undergo a bone marrow examination favorable outcome as those with optimal molecular responses at 3
to establish the diagnosis, assess percentage of blasts and basophils, months. Patients with BCR-ABL1 transcripts [IS] >10% after 6
and perform cytogenetic analysis to confirm the presence of the Phila- months have a worse outcome, still the 4-year survival with BCR-
delphia chromosome and to exclude clonal evolution. The current rec- ABL1 transcripts [IS] >10% vs. <10% are 100% vs. 74%. Thus it may
ommendation that patients have a follow-up bone marrow study at 3, be reasonable to change the TKI from imatinib to second generation
6, and 12 months after starting therapy may not be necessary anymore TKI for BCR-ABL1 transcripts [IS] >10% after 6 months [58]. How-
[50]. An alternative method to determine cytogenetic response is with ever, a similar situation in a patient on second generation TKI (dasa-
the use of FISH and PCR on peripheral blood. If a patient is respond- tinib, nilotinib) does not necessarily imply consideration of allo-SCT.
ing optimally, and the FISH study is negative at 6 or 12 months and
When to switch therapy
or BCR-ABL1 transcripts [IS] <1%, it may be reasonable to omit mar-
row exams, as the patient is likely to be in CCyR [51,52] The aim of therapy in CML should be the achievement of CCyR
For patients in durable CCyR receiving TKI therapy, periodic within 12 months and to continue to be in CCyR at any time beyond
molecular monitoring using RT-Q-PCR is acceptable and useful, but 12 months, especially for standard dose imatinib therapy. For second
may lead to erroneous changes in treatment due to discordant results generation TKIs, CCyR may need to be achieved sooner for an opti-
between laboratories or even within the same laboratory. This leads mal outcome, for example, within 6 months [60]. Patients who do
to potentially discontinuing a useful therapy that the patient may not achieve a complete hematologic response by 3 months should be
have been tolerating well. One strategy to minimize this is to use considered for a change in therapy (Tables IV and V).
interphase FISH as a complementary diagnostic test along with the One question is whether to change therapy at 3 months based on
molecular test to detect possible false positive or negative results gen- the level of BCR-ABL1 transcripts [IS]. For patients on imatinib ther-
erated by either assay [52]. For patients in CCyR, the achievement apy, if the 3-month transcript level is greater than 10%, we suggest
and maintenance of a MMR is of debatable significance. Several stud- an approach similar to the ELN guidelines [49]: perform serial molec-
ies evaluating patients receiving imatinib or second generation TKIs ular monitoring between 3 and 6 months to determine the response
have found that patients in CCyR have similar survival whether they precisely. If patients still have greater than 10% BCR-ABL1 transcripts
achieved MMR or not [53,54]. [IS] at 6 months, the chances of attaining CCyR are low, and a
Early molecular response has been shown in a number of studies change of therapy might be in indicated. Furthermore, a 3-month
to have strong prognostic value (Table III). This has been shown with value of 10% [IS] may not be accurate: in a recent report in 1 sample
each of the 3 TKIs used in the frontline setting. A BCR-ABL1 tran- collected at 3 months and tested 96 times, the mean value was 11%
scripts [IS] < 10% at 3 months separates patients into high and low (range, 5-16%), with only 31% of tests being 10% [61]. This
risk categories for long-term outcomes (i.e., progression, survival) approach also applies to patients on second generation TKIs, because,
as mentioned earlier, very early switching has not yet shown to influ-
ence long-term outcome [62]. Another study randomized patients in
TABLE III. Percentage of Survival by Early Molecular Response CCyR on imatinib for at least 2 years to continue imatinib or switch
to nilotinib [63]. Switching to nilotinib induced deeper molecular
responses, but did not translate into an improvement in progression-
Study Q-PCR<10% Q-PCR>10%
free survival or in other meaningful outcomes.
UK (8-year) 93 54 Patients who meet all the relevant benchmarks in the first 12
MDACC (10-year) 98 94 months are monitored periodically using FISH and molecular testing
ENEST-nd 97 87 (molecular testing only if BCR-ABL1 [IS] transcripts consistently
DASISION 97 86
BELA 98 86
below 0.1%). If there are clear signs of possible failure, patients
should undergo a bone marrow examination with cytogenetics, and
molecular testing including analysis for mutations. Any degree of

TABLE IV. Response Evaluation to TKIs Used as First-Line Therapy (ELN 2013)

Optimal Warning Failure

Baseline CCA in Ph1 cells High-risk Sokal or


Hasford score
3 months Ph1 < 35% And/or BCR-ABL  10% Ph1 36 95% And/or BCR-ABL > 10% Ph1 > 95% And/or No CHR
6 months Ph1 0% And/or BCR-ABL < 1% Ph1 1 35% And/or BCR-ABL 1 10% Ph1 > 35% And/or BCR-ABL > 10%
12 months BCR-ABL  0.1% BCR-ABL 0.1 1% BCR-ABL > 1% And/or Ph1 > 0%
Any time BCR-ABL  0.1% CCA in Ph- cells (27 or 7q-) Loss of CHR Loss or CCyR
Confirmed loss of MMR CCA in Ph1 cells

258 American Journal of Hematology, Vol. 91, No. 2, February 2016 doi:10.1002/ajh.24275
ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES Chronic myeloid leukemia

TABLE V. MDACC Criteria for Response/Failure and Change of Therapy TABLE VI. Important response categories in CML

Time (months) Imatinib Second generation TKIs Response Translates into:

3-6 MCyR; PCR10% [IS] CCyR; PCR  1% [IS] BCR-ABL1  10% at Significantly improved survival
12 CCyR; PCR  1% [IS] CCyR; PCR  1% [IS] 6 months; CCyR later
Later CCyR; PCR  1% [IS] CCyR; PCR  1% [IS] MMR Modest improvement in EFS;
possible longer duration
CCyR; no survival benefit
CMR Possibility of therapy
cytogenetic relapse calls for a change in therapy. Fluctuating molecu- discontinuation (clinical trials only)
lar levels during continuous CCyR would only prompt closer moni-
toring and a compliance assessment.
In several studies, the achievement of a CCyR (Ph-positive meta-
Second and third generation TKIs
phases 0%; BCR-ABL1 transcripts [IS]  1%) at 12 months or later on
TKI therapy was associated with significant survival benefit compared Before their approval to treat first-line CML-CP, both nilotinib
with achievement of lesser degrees of response. Achievement of CCyR is and dasatinib were approved for use in second-line CML-CP follow-
the primary endpoint of TKI therapy. Achievement of BCR-ABL1 tran- ing prior therapy including imatinib [67,68]. Clinical studies of
scripts [IS]  0.1% (MMR) was associated with modest improvements second-line and third line TKIs are summarized in Table VII. Based
in event-free survival rates, possible longer durations of CCyR, but not on these studies, several noteworthy ideas have emerged. First,
with a survival benefit. The achievement of CMR (non-measurable second-line treatment with nilotinib, dasatinib, or bosutinib can yield
BCR-ABL1 transcripts) offers the possibility of treatment discontinua- high rates of response in patients who have inadequate response to
tion in clinical trials only (Table VI). Lack of achievement of MMR or imatinib, including high rates of MMR. Second, dose escalation of
of CMR should not be interpreted as a need to change TKI therapy or imatinib can improve response rates in patients with inadequate
to consider allo-SCT. Response assessments at earlier times on frontline response to standard-dose imatinib [69], but switching to second-line
TKI therapy (36 months) have shown better outcomes with achieve- TKI can be more effective [70]. Several studies that evaluated second-
ment of a major cytogenetic response by 36 months on imatinib ther- line nilotinib [71,72], dasatinib [70,72], or bosutinib [73], and high-
apy (Ph-positive metaphases  35%; BCR-ABL1 transcripts [IS]  10%). dose imatinib (400 mg BID) have demonstrated significantly higher
While this is interpreted to mean that a change to second TKI therapy rates of CHR, CCyR, and MMR with the newer TKIs than with high-
may be considered if such outcome is not obtained, no studies have dose imatinib. Moreover, PFS in these studies was better with the
shown that changing therapy from imatinib to second TKIs has newer TKIs than with high-dose imatinib. Earlier switch to second-
improved patients outcomes. When nilotinib or dasatinib are used in line TKI may be more effective than later switch. In the TIDEL-II
front-line therapy, achievement of complete cytogenetic response by 36 study, patients who had suboptimal response to imatinib and were
months of TKIs therapy has been associated with improved outcomes. switched to nilotinib had a higher rate of CMR at 12 months than
At MDACC, our major treatment milestones are at 6 and 12 patients who had dose escalation of imatinib prior to being switched
months. Patients with lack of PCyR (BCR-ABL1 transcripts [IS] > 10%) to nilotinib [74]. In a retrospective pooled analysis of three clinical
at 6 months, or without CCyR (BCR-ABL1 transcripts [IS]  1%) at 12 studies of second-line dasatinib for patients resistant to or intolerant
months, or with loss of response at any time are candidates for a of imatinib, patients who were switched to dasatinib after the loss of
switch of therapy. The choice of TKIs is based on the mutation profiles MCyR (early intervention group) had higher rates of CHR, CCyR,
and patients comorbidities. We do not consider a change of TKI ther- and MMR, as well as 24-month EFS, TFS, and OS, than patients who
apy in patients in CCyR but without MMR. were switched after the loss of both MCyR and CHR (late interven-
tion group) [75]. Although this analysis included studies with distinct
Management of TKI Resistance study designs and various dosing schedules of dasatinib, the essential
finding that earlier switch to dasatinib was associated with better
A remaining problem with the widespread use of all commercially outcomes.
available TKIs is increased drug resistance. A common mechanism of Bosutinib was initially studied in patients who had resistance to or
resistance involves point mutations in the kinase domain of BCR- intolerance of imatinib [73]. After a dose escalation period, 500 mg
ABL1, which impairs the activity of the available TKIs. Second gener- once daily was selected as the phase II dose, with the potential for
ation TKIs overcome most of the mutations that confer resistance to dose escalation to 600 mg once daily for patients not meeting prespe-
imatinib, though novel mutations rendering the leukemia resistant to cified benchmarks. A total of 288 patients were enrolled in the pivotal
dasatinib and/or nilotinib have emerged. One important mutation, phase II trial; more than two thirds had imatinib-resistant disease.
T315I known as the gatekeeper mutation, displays resistance to all The primary endpoint of MCyR at 6 months was achieved in 31%;
currently available TKIs except ponatinib. 41% achieved a CCyR. Bosutinib appeared to retain activity across
Before defining a patient as having TKI resistance and modifying most known mutations that confer imatinib resistance, except for
therapy, treatment compliance and drug-drug interactions should be T315I. Responses were independent of whether patients had resist-
assessed. Rates of imatinib adherence have been estimated to range ance to or intolerance of imatinib. The most common toxicities were
from 75% to 90%, and lower adherence rates correlated with worse diarrhea, nausea, vomiting, and rash. Diarrhea occurred in 84% of
outcome [6466]. In a study of 87 patients with CML-CP treated patients, with 9% experiencing grade 3 diarrhea (there were no grade
with imatinib 400mg daily, an adherence rate of 90% or less resulted 4 events documented). Other notable adverse events included myelo-
in MMR rate of 28% compared with 94% with greater than 90% suppression and liver function test abnormalities.
adherence rate (P <0.001) [64]. CMR rates were 0% vs. 44% Ponatinib is a third generation TKI, and the first TKI in class to
(P 5 0.002), and no molecular responses were observed when adher- exhibit activity against CML with T315I mutation [76]. It is 500 times
ence rates were 80% or lower. Lower adherence rates have been as potent than imatinib at inhibiting BCR-ABL1 [77]. The approval
described in younger patients, those with adverse effects of therapy, of ponatinib was based on the phase II PACE trial, where 449
and those who have required dose escalations [64]. patients with heavily pretreated CML or Ph-positive acute

doi:10.1002/ajh.24275 American Journal of Hematology, Vol. 91, No. 2, February 2016 259
Jabbour and Kantarjian ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES

lymphoblastic leukemia (ALL) were treated [78]. Patients were con-

CP: chronic phase; AP: accelerated phase; MyBP: myeloid blast phase; LyBP: lymphoid blast phase; BP: blast phase; MaHR: major hematologic response; CHR: complete hematologic response; NEL: no
Ma HR: 34
sidered for this trial if they had resistance to or intolerance of dasati-

N 5 94
nib or nilotinib, or if they had CML with T315I mutation. The dose

NR

NR
NR

NR
BP

36

36

9
-
of ponatinib was 45 mg once daily, and patients were stratified by
disease phase and presence or absence of a T315I mutation. Of the
267 patients who received ponatinib in CML-CP, 56% achieved a

At 3 years
Ponatinib

Ma HR: 57
MCyR by 12 months, which included 45/64 (70%) patients with a
N 5 79

NR

NR
NR

NR
AP

96
36

59
55
T315I mutation. Patients responded more favorably if they had
received fewer TKIs. After a median follow-up of 3.5 years, 59% of
patients achieved MCyR (primary endpoint) at any time; 83% of
those remained in MCyR at 3 years [79]. Furthermore, 39% of
N 5 270

patients achieved a MMR or better. The 3-years PFS and OS rates


NR
NR
CP

30
36

53
75

81
-
-
were 60% and 81%, respectively [79]. Arterial occlusive events
occurred in 28% of patients (23% serious). The most common all-
grade treatment-emergent adverse events occurring in  40% of CP-
N 5 38

NR

NR
NR
BP

50
36
36

35
18
CML patients were abdominal pain (46%), rash (46%), thrombocyto-
3

penia (45%), headache (43%), constipation (41%), and dry skin (41%)
[79]. Other notable toxicities include severe skin rashes (4-7%), pan-
At 4 years
Bosutinib

N 5 51

creatitis (7%), and severe hypertension (20%).


NR

NR

60
AP

20
54
54

27
0
6

As of early 2014, ponatinib labeling included a revised warning


regarding the risk of thrombotic events (13% per year), vascular
occlusions, heart failure, and hepatotoxicity, revised dosing informa-
N 5 200
CP

tion and indications limited to adults with T315I mutation and those
69

98
46
85
24

81

12

-

for whom no other TKI is indicated [80]. Vascular occlusion adverse


events were more frequent with increasing age and in patients with
Percent response

prior history of ischemia, hypertension, diabetes, or hyperlipidemia


N 5 31
LyBP

NR
82

32

42
19

16
13
0
3

[79]. Factors associated with increased risk of vascular occlusion


events include older age, higher dose, history of myocardial infarction
or prior vascular events, and longer duration of CML [79]. Studies
N 5 105
MyBP

assessing lower dose schedule of ponatinib are ongoing. In commu-


NR
82

29

42
22

10
11
3

nity practice, it may be safe to use ponatinib 30 mg daily (and lower


At 4 years
Nilotinib

the dose to 15 mg daily for toxicities) rather than use the FDA
TABLE VII. Summary of Important Phase II Trials of 2nd and 3rd Generation TKIs After Prior TKI Failure

approved dose of 45 mg daily.


N 5 137

NR
80
AP

20
56

67
31
12

12
9

How to select a second or third line option


At the time of treatment failure, patients should undergo bone mar-
row examination to allow proper determination of the CML phase and
N 5 321

NR
CP

48

94
70

45

87
76

14

documentation of any clonal evolution. All patients should have CML


cells tested for BCR-ABL1 kinase domain mutations, as this will help
guide the selection of the TKI [81,82]. When selecting between dasati-
N 5 48

nib, bosutinib, and nilotinib, in vitro and in vivo data have identified
LyBP

40

50
88

46
29

52
12

distinct mutations that exhibit decreased sensitivity to each of the


agents [83,84]. Physician may favor dasatinib or bosutinib if the patient
has the following mutations: Y253H, E255K/V, or F359C/V. Alterna-
N 5 109
MyBP

tively, nilotinib may be favored in the presence of the V299L and


50

50
36
26
27
91
12

7
At 6 years

F317L mutations. For patients lacking these mutations, the choice


Dasatinib

should be on preexisting conditions, toxicity profiles, and cost.


Bosutinib can be used for patients with most known mutations
N 5 174
AP

93

44

82
45

32
79

19
14

that lead to imatinib failure [73]. Like dasatinib and nilotinib, bosuti-
7

nib lacks activity against T315I. Bosutinib may be a reasonable choice


for patients who fail imatinib who are not good candidates for dasati-
N 5 167

nib or nilotinib. Bosutinib has a relatively distinct toxicity profile


evidence of leukemia; NR: not reported
CP

50
92
24
74

91
13

-

from the other TKIs, with the predominant problem being diarrhea
and other gastrointestinal complaints. Recently, an analysis was con-
ducted to closely characterize the toxicity of bosutinib and the man-
% Hematologic Response

% Cytogenetic Response

agement strategy [85]. Diarrhea was documented in 82% of patients


% Resistant to imatinib
Median follow-up (mo)

(n 5 570), most being grades 1/2 in severity. Myelosuppression and


liver function test abnormalities were also common. With appropriate
supportive care, monitoring, and dose reductions for persistent diar-
rhea, most patients are able to continue therapy with periodic dose
% Survival
Complete

interruptions or adjustment.
Partial

Ponatinib should be considered the agent of choice in patient with


CHR
NEL

CML and T315I mutation, and in instances where other TKIs are not

260 American Journal of Hematology, Vol. 91, No. 2, February 2016 doi:10.1002/ajh.24275
ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES Chronic myeloid leukemia

indicated. No other commercially available TKIs have activity against Treatment Duration and
this mutation. The risk for serious toxicities (vaso-occlusive disease,
pancreatitis, hypertension, severe skin rashes) and of thrombotic Discontinuation
events with ponatinib is significant, but the benefits outweigh the The Stop Imatinib (STIM) trial investigated the risk of relapse in
risks for most patients with a T315I mutation, as there are no other patients on imatinib with ongoing complete molecular response
options for disease control. These side effects are lower with ponati- (CMR) for greater than two years who stopped treatment [91,92]. In
nib 15-30 mg daily. the most recent update, 100 patients had a median follow-up of 50
Second and third generation TKIs have not been compared head-to- months and were monitored closely for evidence of molecular relapse.
head. Selection of one or the other is based on the side-effect profiles, Overall, 61% experienced a molecular relapse, with 95% of the events
mutations profile, drug interactions, compliance issues and the patients occurring within 7 months of stopping imatinib. Almost all patients
preexisting medical conditions. Mutational analysis are required in were able to achieve CMR once imatinib therapy was restarted. Hav-
patients who are failing imatinib or second generation TKIs, or those ing a low-risk Sokal score and duration of imatinib therapy greater
who progress to AP/BP. Baseline mutational analysis on patients with than 60 months predicted for continued CMR after therapy cessation.
newly diagnosed CML-CP are not done, as this has not proven to pre- These results were confirmed in other large studies of CML. The
dict treatment outcome. TWISTER study followed 40 patients who stopped imatinib after
At MDACC, post imatinib failure, the choice of second or third gen- being without detectable minimal residual disease for greater than
eration TKI is based on the disease phase, mutation profile, and two years [93]. Patients were followed for a minimum of 15 months
patients comorbidities. In advanced phases, we favor a combination of (median 43 months) from the time they stopped imatinib; 22 of 40
chemotherapy and TKI (mainly ponatinib or dasatinib). In patients patients became molecularly positive; nearly 70% of the molecular
with CML-CP with T315I mutation, ponatinib will be the first choice relapses occurred within the first 6 months of treatment cessation;
followed by allo-SCT if a donor is available and an optimal response is and patients who resumed TKIs were able to recapture deep molecu-
not achieved. Outside the context of T315I mutations, the type of lar responses. Interestingly, highly sensitive patient-specific PCR was
mutation will dictate the choice of therapy. Of note, patients with poor able to detect the original CML clone in several patients who
response to imatinib and compound mutations may not respond well remained off imatinib for several years. This indicates that it may not
to second generation TKI. A close monitoring should be offered [86]. If be necessary to completely eradicate the disease to allow patients to
an optimal response is not achieved, a switch of therapy to a third gen- enjoy a functional cure.
eration TKI and/or allo-SCT is warranted. In patients with no muta- The previous studies included heterogeneous groups of patients,
tion or a mutation sensitive to all second generation TKIs, the choice particularly ones exposed to interferon prior to the imatinib era. The
will be based on comorbidities. Patients with vascular risk factors and French group conducted a follow-up study to STIM enrolling patients
metabolic dysfunctions are not candidates for nilotinib, while patients who have only been exposed to imatinib as CML therapy (STIM2)
with lung injuries are not candidates for dasatinib. Bosutinib may be [94]. The inclusion criteria were similar to those used in STIM1.
the best choice for patients with cardiac and rhythmic problems. There were 124 patients identified who stopped imatinib therapy.
With a median follow-up of 12 months, 48 patients had molecular
Allogeneic stem cell transplantation (Allo-SCT) relapse; 94% of relapses occurred within 6 months of TKI withdrawal.
The number of patients undergoing allo-SCT for CML-CP has All patients remained sensitive to imatinib or a second generation
decreased significantly since TKIs were introduced, but will start to TKI upon re-challenge. In this study and in others, patients with low
increase again as the prevalence of CML increases, as about 2% of level positivity for BCR-ABL1 transcripts may be able to remain off
therapy with close monitoring. This was recently addressed systemati-
patients become resistant to many TKIs every year and require allo-
cally by the French investigators, who observed that is safe and effec-
SCT. Allo-SCT has a more important role when patients evolve into
tive to resume patients on TKI therapy if their transcript level
AP/BP (see below). Allo-SCT remains an important therapeutic
became greater than 0.1% (i.e., loss of MMR) [95].
option for patients in CML-CP who fail at least 2 TKIs or are poten-
A pan-European Stop Tyrosine Kinase Inhibitor trial (The EURO-
tially harboring the T315I mutation (after a trial of ponatinib ther-
SKI study) aimed to define factors associated with durable deep
apy) [87]. Prior exposure to TKIs does not impact transplant
molecular response (MR) after stopping TKI. An interim analysis on
outcome negatively; patients referred to transplant may have a better
200 patients with 6-month follow-up of molecular events was
outcome if entering the transplant with a better response (lower CML
reported [96]. Adult CML patients in CML-CP on TKI treatment in
burden) [88].
confirmed deep MR (MR4, BCR-ABL1 transcripts [IS] <0.01%) for
Finally, allo-SCT cost versus TKIs cost and availability should be
at least 1 year (>4 log reduction on TKI therapy for >12 months
considered. Allo-SCT, a curative one-time procedure costs $500,000 confirmed by three consecutive PCR tests) and under TKI treatment
in the United States, but only $12,000 to 20,000 in developing for at least 3 years were eligible. The duration of TKI treatment was
nations. Allo-SCT should not be offered as frontline therapy unless 8 years (range, 312.6 years), and median duration of MR4 before
the TKI prices are prohibitive to a nations health care budget or not TKI cessation was 5.4 years (range, 111.7 years). Overall, 123 of the
accessible to most. In India, the annual price of generic imatinib is 200 patients remained without relapse in the first 6 months. Recur-
$400. Considering the median age of patients with CML and the pro- rence of CML, defined as loss of MMR, was observed in 47/114
jected survival of 301 years ($400 X 30 years 5 $12,000), imatinib patients (47%) treated for <8 years, compared to 27/86 patients
therapy remains better than performing allo-SCT. In nations where (27%) treated for >8 years (P 5 0.003). The duration of MR4 >5
TKIs are not commonly accessible, it is better to perform allo-SCT years vs. <5 years was predictive factor for a lower relapse rate
on 100% and cure 60%, than having TKIs available to 10% (and func- (P 5 0.03).
tional cure 10%). In contrast to the frontline setting, in the salvage TKIs discontinuation studies in patients with durable CMR demon-
setting the value of allo-SCT versus second/third generation TKI is strate that stopping TKI therapy is feasible, and some patients may be
more pronounced. Allo-SCT in first salvage as cure costs $20,000 vs. cured. While the results thus far are encouraging, it is still recom-
$120,000-170,000/year for second-third generation TKIs. In these cir- mended that TKI therapy be stopped only under the auspices of a clini-
cumstances, allo-SCT could be offered as first salvage option in cal trial. Many laboratories are not able to produce precise and
nations with financial constraints [89,90]. reliable PCR results as those used in the above mentioned trials. Going

doi:10.1002/ajh.24275 American Journal of Hematology, Vol. 91, No. 2, February 2016 261
Jabbour and Kantarjian ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES

forward, it is important to continue to investigate the possibility of safe (i.e. achievement of a durable CMR and its persistence after discon-
treatment cessation. Measures of quality of life and adverse event tinuation of TKI therapy). This is not a trivial issue since, with effec-
avoidance should be studied in subsequent trials. The economic impact tive TKI therapy, and full treatment penetration worldwide (to 100%
of long-term discontinuation of imatinib is substantial. of all diagnosed patients and continuation of TKI therapy without
At MDACC, therapy discontinuation is offered only for patients in interruptions) the prevalence of CML would increase annually and
durable CMR and in the context of clinical trials. plateau at around 2,030 to 2,040 at a rate of 35 times the incidence.
The prevalence is estimated to be close to 1,60,000 patients with
Advanced Stage CML CML in the United States and about 3 million patients worldwide.
This may represent a considerable burden on patients and the health-
Patients with CML-AP or CML-BP may receive initial therapy care systems in relation to drug availability, compliance, potential
with TKIs (newer generation TKIs like dasatinib or ponatinib pre- development of long-term side effects, and costs. Therefore it is
ferred over imatinib) to reduce the CML burden, and be considered important to continue research into therapies that increase the rates
for early allo-SCT [97101]. Response rates with combinations of of durable CMRs. This may be achievable with the current more
TKIs and chemotherapy are 40% in nonlymphoid CML-BP and 70- potent new generation TKIs alone, or in combination with other
80% in lymphoid CML-BP [102104]. Median survival times are 6 available (peg-interferon alpha-2, omacetaxine, decitabine) or investi-
12 months, and 1224 months, respectively. The addition of TKIs to gational therapies (JAK2 inhibitors, hedgehog inhibitors, stem cell
chemotherapy has improved the response rates and prolonged the poisons, vaccines). Such strategies may improve the eradication of
median survival time in CML-BP. minimal residual disease, potentially obviating the need for indefinite
At present, allo-SCT is the only curative therapy for AP and BP therapy with TKIs. Further understanding of the pathophysiologic
CML: overall cure rates are in the range of 15 to 40% and 10 to 20%, events downstream of BCR-ABL1 may help in the development of
respectively [47,82]. Patients with cytogenetic clonal evolution as the new strategies to target them.
only AP criterion have a long-term event-free survival rate of about
60% [83]. Otherwise, TKIs provide hematologic responses in 80% of
patients and an estimated 4-year survival rates of 40 to 55% in CML- MDACC Approach
AP, but only a 40% response rate and a median survival of 9 to 12 ALL patients suspected to have CML undergo bone marrow exam-
months in CML-BP. Patients in the AP or BP should be encouraged ination which will confirm the diagnosis and provide information
to participate in investigational strategies to improve their prognosis. needed for staging. Baseline PCR is performed in order to identify
Patients with de novo CML-AP have a better outcome with frontline the specific type of rearrangement that can be appropriately followed
TKI therapy than patients who evolve from CP to AP. The estimated when assessing for response to TKI therapy.
6-8 year survival rates with TKI therapy in de novo CML-AP are 60 Waiting for confirmation, patients are placed on transient cytore-
to 80% [105]. Such patients may continue on TKI therapy as their duction therapy with hydroxyurea. Tumor lysis syndrome prophylaxis
long-term treatment if they achieve a CCyR on TKI therapy. is implemented as well.
Allo-SCT should be considered early in patients in AP based on Patients are stratified into low-risk and intermediate/high-risk dis-
response to TKI therapy. The only curative option for patients in BP ease. Patients with low-risk disease are treated with imatinib; patients
disease is allo-SCT. TKIs monotherapy or in combination with chemo- with high-risk disease are treated with second generation TKI. The
therapy may serve as a good option for those who are not candidates choice of second generation TKI is based on patients comorbidities.
for transplant, or as a bridge to allo-SCT. The role of TKIs before and Patients are monitored regularly every 3 months in the first year,
after transplant is being evaluated. Accumulating data show that TKIs and later every 6 months. Monitoring by the use of peripheral blood
do not increase transplant-related complications and when used after FISH and PCR are alternative to marrow examinations to determine
low intensity conditioning regimens, may delay relapse rates and need cytogenetic response. If a patient is responding optimally, and the
for donor lymphocyte infusions. FISH study is negative at 6 or 12 months and or BCR-ABL1 tran-
At MDACC, patients with CML-BP are treated with combination of scripts [IS] <1%, it may be reasonable to omit further marrow exams,
chemotherapy (type depends on the immunophenotype) and third gen- as the patient is likely to be in stable CCyR . The major treatment
eration TKI followed by allo-SCT once a complete response is achieved milestones are at 6 and 12 months. Patients with lack of PCyR (BCR-
and placed on maintenance TKI therapy. Patients with de-novo CML- ABL1 transcripts [IS]  10%) at 6 months, lack of CCyR (BCR-ABL1
AP are treated with frontline second generation TKI infinitely if an transcripts [IS]  1%) at 12 months, and loss of response at any time
optimal response (CCyR; BCR-ABL1 transcripts [IS] <1%) is achieved are candidates for a change of therapy. The choice of TKI is based on
within 6 months of therapy all other patients in CML-AP are treated mutation profile and patients comorbidities. We do not change ther-
with second/third generation TKI followed by allo-SCT. apy for patients in CCyR but without MMR. Patients who meet all
the relevant benchmarks in the first 12 months are monitored peri-
Conclusions and Future Directions odically using FISH and molecular testing (molecular testing only if
BCR-ABL1 transcripts consistently below 0.1% [IS]).
In 2016, CML experts and patients with CML have multiple treat- Post imatinib failure, patients should undergo a bone marrow
ment options in the CML therapeutic armamentarium, including 5 examination with cytogenetics and molecular testing, including analy-
TKIs (imatinib, nilotinib, dasatinib, bosutinib, ponatinib), omacetax- sis for mutation. The choice of second or third generation TKIs is
ine (protein synthesis inhibitor), and several older agents (hydrox- based on disease phase, mutation profile, and patients comorbidities.
urea, interferon alpha, busulfan, 6-mercaptopurine, cytarabine, In advanced phases, we favor a combination of chemotherapy and
decitabine, others). Most patients with CML would be expected to TKI (mainly ponatinib or dasatinib). In patients with CML-CP with
have a normal life span, and be potentially functionally, though not T315I mutation, ponatinib is the first choice treatment followed by
molecularly, cured, as long as they continue therapy with TKI based allo-SCT if a donor is available and an optimal response is not
regimens, are compliant with the treatment, and are monitored achieved. Outside the context of a T315I mutation, the type of muta-
closely for signs of resistance, in order to change therapy in a timely tion dictates the choice of therapy. Patients with poor response to
manner and/or consider allo-SCT before CML progression. Future imatinib and compound mutations may not respond well to second
directions will focus on the potential molecular cure of CML generation TKI, and a close monitoring is indicated. If an optimal

262 American Journal of Hematology, Vol. 91, No. 2, February 2016 doi:10.1002/ajh.24275
ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES Chronic myeloid leukemia

response is not achieved, a switch of therapy to a third generation Patients with CML-BP are treated with combination of chemother-
TKI and/or allo-SCT is warranted. In patients with no mutation or apy (type depends on the immunophenotype) and third generation
with mutations sensitive to all second generation TKIs, the choice is TKI followed by allo-SCT once a complete response is achieved and
based on comorbidities. Patients with vascular risk factors and meta- placed on maintenance TKI therapy post allo-SCT. Patients with de-
bolic dysfunctions are not candidates for nilotinib therapy, while novo CML-AP are treated with frontline second generation TKI
patients with lung injuries are not candidates for dasatinib therapy. infinitely if an optimal response (CCyR; BCR-ABL1 transcripts [IS]
Bosutinib may be the best choice for patients with cardiac and rhyth- <1%) is achieved within 6 months of therapy. All other patients in
mic problems. Allo-SCT remains an important therapeutic option for CML-AP are treated with second/third generation TKI followed by
patients in CML-CP in the following situations: patients who fail at allo-SCT. Therapy discontinuation is offered only for patients with
least 2 TKIs or are potentially harboring the T315I mutation (after a CML-CP in durable CMR (2 years) and in the context of clinical
trial of ponatinib therapy). trials.

References phase- and hypermetaphase-FISH, qualitative


and quantitative PCR for diagnosis and for
myeloid leukemia. N Engl J Med 2010;362:
22512259.
1. American Cancer Society. Cancer Facts & Fig- follow-up in chronic myeloid leukemia: A study 30. Larson RA, Kim DW, Jootar S, et al. ENESTnd
ures 2015. Atlanta: American Cancer Society; on 350 cases. Leukemia 2002;16:53 5-year (y) update: Long-term outcomes of
2015. 16. Druker BJ, Lydon NB. Lessons learned from the patients (pts) with chronic myeloid leukemia in
2. Huang X, Cortes J, Kantarjian H. Estimations of development of an abl tyrosine kinase inhibitor chronic phase (CML-CP) treated with frontline
the increasing prevalence and plateau prevalence for chronic myelogenous leukemia. J Clin Invest nilotinib (NIL) versus imatinib (IM). J Clin
of chronic myeloid leukemia in the era of tyro- 2000;105:37. Oncol (Meeting Abstracts) 2014;32:7073.
sine kinase inhibitor therapy. Cancer 2012;118: 17. OBrien SG, Guilhot F, Larson RA, et al. Imati- 31. Wang J, Shen ZX, Saglio G, et al. Phase 3 study
31233127. nib compared with interferon and low-dose of nilotinib vs imatinib in Chinese patients with
3. Rowley JD. Letter: A new consistent chromo- cytarabine for newly diagnosed chronic-phase newly diagnosed chronic myeloid leukemia in
somal abnormality in chronic myelogenous leu- chronic myeloid leukemia. N Engl J Med 2003; chronic phase: ENESTchina. Blood 2015;125:
kaemia identified by quinacrine fluorescence 348:9941004. 27712778.
and Giemsa staining. Nature 1973;243:290293. 18. Cortes JE, Baccarani M, Guilhot F, et al. Phase 32. Jain P, Kantarjian H, Alattar ML, et al. Long-
4. Mandanas RA, Leibowitz DS, Gharehbaghi K, III, randomized, open-label study of daily imati- term molecular and cytogenetic response and
et al. Role of p21 RAS in p210 bcr-abl transfor- nib mesylate 400 mg versus 800 mg in patients survival outcomes with imatinib 400 mg, imati-
mation of murine myeloid cells. Blood 1993;82: with newly diagnosed, previously untreated nib 800 mg, dasatinib, and nilotinib in patients
18381847. chronic myeloid leukemia in chronic phase with chronic-phase chronic myeloid leukaemia:
5. Okuda K, Matulonis U, Salgia R. Factor inde- using molecular end points: Tyrosine kinase Retrospective analysis of patient data from five
pendence of human myeloid leukemia cell lines inhibitor optimization and selectivity study. clinical trials. Lancet Haematol 2015;2:e118
is associated with increased phosphorylation of J Clin Oncol 2010;28:424430. e128.
the proto-oncogene Raf-1. Exp Hematol 1994; 19. Preudhomme C, Guilhot J, Nicolini FE, et al. 33. OBrien S, Radich JP, Abboud CN, et al.
22:11111117. Imatinib plus peginterferon alfa-2a in chronic Chronic myelogenous leukemia, Version 1.2014.
6. Raitano AB, Halpern JR, Hambuch TM. The myeloid leukemia. N Engl J Med 2010;363: J Natl Compr Canc Netw 2013;11:13271340.
Bcr-Abl leukemia oncogene activates Jun kinase 25112521. 34. Montani D, Bergot E, Gunther S, et al. Pulmo-
and requires Jun for transformation. Proc Natl 20. Lombardo LJ, Lee FY, Chen P, et al. Discovery nary arterial hypertension in patients treated by
Acad Sci USA 1995;92:1174611750. of N-(2-chloro-6-methyl- phenyl)22-(6-(4-(2- dasatinib. Circulation 2012;125:21282137.
7. Sawyers CL, Callahan W, Witte ON. Dominant hydroxyethyl)- piperazin-1-yl)22-methylpyrimi- 35. Quintas-Cardama A, Han X, Kantarjian H,
negative MYC blocks transformation by ABL din-4- ylamino)thiazole-5-carboxamide (BMS- Cortes J. Tyrosine kinase inhibitor-induced pla-
oncogenes. Cell 1992;70:901910. 354825), a dual Src/Abl kinase inhibitor with telet dysfunction in patients with chronic mye-
8. Shuai K, Halpern J, ten Hoeve J. Constitutive potent antitumor activity in preclinical assays. loid leukemia. Blood 2009;114:261263.
activation of STAT5 by the BCR-ABL oncogene J Med Chem 2004;47:66586661. 36. Quintas-Cardama A, Kantarjian H, Ravandi F,
in chronic myelogenous leukemia. Oncogene 21. OHare T, Walters DK, Stoffregen EP, et al. In et al. Bleeding diathesis in patients with chronic
1996;13:247254. vitro activity of Bcr-Abl inhibitors AMN107 and myeloid leukemia receiving dasatinib therapy.
9. Carlesso N, Frank DA, Griffin JD. Tyrosyl phos- BMS-354825 against clinically relevant imatinib- Cancer 2009;115:24822490.
phorylation and DNA binding activity of signal resistant Abl kinase domain mutants. Cancer 37. Aichberger KJ, Herndlhofer S, Schernthaner G,
transducers and activators of transcription Res 2005;65:45004505. et al. Progressive peripheral arterial occlusive
(STAT) proteins in hematopoietic cell lines 22. Tokarski JS, Newitt JA, Chang CY, et al. The disease and other vascular events during niloti-
transformed by Bcr/Abl. J Exp Med 1996;183: structure of Dasatinib (BMS-354825) bound to nib therapy in CML. Am J Hematol 2011;86:
811820. activated ABL kinase domain elucidates its 533539.
10. Ilaria RL Jr., Van Etten RA. P210 and P190 inhibitory activity against imatinib-resistant 38. Quintas-Cardama A, Kantarjian H, Cortes J.
(BCR/ABL) induce the tyrosine phosphorylation ABL mutants. Cancer Res 2006;66:57905797. Nilotinib-Associated Vascular Events. Clin Lym-
and DNA binding activity of multiple specific 23. Shah NP, Tran C, Lee FY, et al. Overriding ima- phoma Myeloma Leuk 2012;12:337340.
STAT family members. J Biol Chem 1996;271: tinib resistance with a novel ABL kinase inhibi- 39. Tefferi A, Letendre L. Nilotinib treatment-
3170431710. tor. Science 2004;305:399401. associated peripheral artery disease and sudden
11. Silver RT, Woolf SH, Hehlmann R, et al. An 24. Kantarjian H, Shah NP, Hochhaus A, et al. death: Yet another reason to stick to imatinib as
evidence-based analysis of the effect of busulfan, Dasatinib versus imatinib in newly diagnosed front-line therapy for chronic myelogenous leu-
hydroxyurea, interferon, and allogeneic bone chronic-phase chronic myeloid leukemia. N kemia. Am J Hematol 2011;86:610611.
marrow transplantation in treating the chronic Engl J Med 2010;362:22602270. 40. Giles FJ, Mauro MJ, Hong F, et al. Rates of
phase of chronic myeloid leukemia: Developed 25. Cortes J, Saglio G, Baccarani M, et al. Final peripheral arterial occlusive disease in patients
for the American Society of Hematology. Blood study results of the Phase 3 dasatinib versus with chronic myeloid leukemia in the chronic
1999;94:15171536. imatinib in newly diagnosed chronic myeloid phase treated with imatinib, nilotinib, or non-
12. Deininger M, OBrien SG, Guilhot F, et al. leukemia in chronic phase (CML-CP) trial tyrosine kinase therapy: A retrospective cohort
International randomized study of interferon vs. (DASISION, CA180-056). Blood (ASH Annual analysis. Leukemia 2013;27:13101315.
STI571 (IRIS) 8-year follow up: Sustained sur- Meeting Abstracts) 2014;124:152. 41. Smith TJ, Hillner BE. Bending the cost curve in
vival and low risk for progression of events in 26. Radich JP, Kopecky KJ, Appelbaum FR, et al. A cancer care. N Engl J Med 2011;364:20602065.
patients with newly diagnosed chronic myeloid randomized trial of dasatinib 100 mg versus 42. Kantarjian HM, Fojo T, Mathisen M, Zwelling
leukemia in chronic phase (CML-CP) treated imatinib 400 mg in newly diagnosed chronic- LA. Cancer drugs in the United States: Justum
with imatinib. Blood 2009;114. Abstract #1126. phase chronic myeloid leukemia. Blood 2012; pretiumthe just price. J Clin Oncol 2013;31:
13. Jabbour E, Cortes JE, Kantarjian HM. Molecular 120:38983905. 36003604.
monitoring in chronic myeloid leukemia: 27. OBrien SG, Hedgley C, Adams S, et al. Spirit 2: 43. Experts in Chronic Myeloid Leukemia., et al.
Response to tyrosine kinase inhibitors and prog- An NCRI randomized study comparing dasati- The price of drugs for chronic myeloid leukemia
nostic implications. Cancer 2008;112:21122118. nib with imatinib in patients with newly diag- is a reflection of the unsustainable prices of can-
14. Kantarjian H, Schiffer C, Jones D, Cortes J. nosed CML. Blood (ASH Annual Meeting cer drugs: From the perspective of a large group
Monitoring the response and course of chronic Abstracts) 2014;124:517. of CML experts. Blood 2013; 121:44394442.
myeloid leukemia in the modern era of BCR- 28. Weisberg E, Manley PW, Breitenstein W, et al. 44. Gambacorti-Passerini C, Antolini L, Mahon X.
ABL tyrosine kinase inhibitors: Practical advice Characterization of AMN107, a selective inhibi- Multicenter independent assessment of out-
on the use and interpretation of monitoring tor of native and mutant Bcr-Abl. Cancer Cell comes in chronic myeloid leukemia patients
methods. Blood 2008;111:17741780. 2005;7:129141. treated with imatinib. J Natl Cancer Inst 2011;
15. Schoch C, Schnittger S, Bursch S, et al. Compar- 29. Saglio G, Kim DW, Issaragrisil S, et al. Nilotinib 103:553561.
ison of chromosome banding analysis, inter- versus imatinib for newly diagnosed chronic

doi:10.1002/ajh.24275 American Journal of Hematology, Vol. 91, No. 2, February 2016 263
Jabbour and Kantarjian ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES

45. Larson RA, Conti R, Padula WV, et al. What is ABL1 after 3 months of imatinib depends on ponatinib (AP24534): Lessons for overcoming
the most cost-effective strategy for treating the rate of BCR-ABL1 decline. Blood 2014;124: kinase inhibitor resistance. Chem Biol Drug Des
newly diagnosed chronic myeloid leukemia 511518. 2011;77:111.
(cml) after imatinib loses patent exclusivity? 62. Cortes JE, De Souza CA, Lopez JL, et al. Switch- 78. Cortes JE, Kim DW, Pinilla-Ibarz J, et al. A
Blood (ASH Annual Meeting Abstracts) 2014; ing to nilotinib in patients with chronic myeloid phase 2 trial of ponatinib in Philadelphia
124:738. leukemia in chronic phase with suboptimal chromosome-positive leukemias. N Engl J Med
46. Parikh P. Report of chronic myeloid leukemia cytogenetic response on imatinib: First results of 2013;369:17831796.
in chronic phase from Tata Memorial Hospital, the LASOR trial. Blood (ASH Annual Meeting 79. Cortes JE, Kim DW, Pinilla-Ibarz J, et al. Long-
Mumbai, 2002-2008. Indian J Med Paediatr Abstracts) 2013;122:95 term follow-up of ponatinib efficacy and safety
Oncol 2013;34:164167. 63. Leber B, Cervantes F, Spector N, et al. Achieve- in the Phase 2 PACE trial. Blood 2014;124:3135.
47. Sokal JE, Cox EB, Baccarani M, et al. Prognostic ment and maintenance of deeper molecular 80. Iclusig (ponatinib) [prescribing information].
Discrimination in Good-Risk Chronic Granu- response by switching to nilotinib in patients Cambridge, MA: ARIAD Pharmaceuticals, Inc;
locytic Leukemia. Blood 1984;63:789799. with chronic myeloid leukemia in chronic phase 2014.
48. Hasford J, Pfirrmann M, Hehlmann R, et al. A with residual disease on long-term imatinib: 81. Cortes J, Jabbour E, Kantarjian H, et al. Dynam-
new prognostic score for survival of patients with Enestcmr 36-month follow up. Blood (ASH ics of BCR-ABL kinase domain mutations in
chronic myeloid leukemia treated with interferon Annual Meeting Abstracts) 2013;122:94 chronic myeloid leukemia after sequential treat-
alfa. J Natl Cancer Inst 1998;90:850858. 64. Marin D, Bazeos A, Mahon FX, et al. Adherence ment with multiple tyrosine kinase inhibitors.
49. Jain P, Kantarjian H, Patel KP, et al. Impact of is the critical factor for achieving molecular Blood 2007;110:40054011.
BCR-ABL transcript type on response and sur- responses in patients with chronic myeloid leu- 82. Jabbour E, Branford S, Saglio G, Jones D. Prac-
vival in patients with chronic phase chronic kemia who achieve complete cytogenetic tical advice for determining the role of BCR-
myeloid leukemia (CML-CP) treated with tyro- responses on imatinib. J Clin Oncol 2010;28: ABL mutations in guiding tyrosine kinase inhib-
sine kinase inhibitors (TKI). Blood (ASH 23812388. itor therapy in patients with chronic myeloid
Annual Meeting Abstracts) 2015;126. Abstract 65. Darkow T, Henk HJ, Thomas SK, et al. Treat- leukemia. Cancer 2011;117:18001811.
#598. ment interruptions and non-adherence with 83. Muller MC, Cortes JE, Kim DW, et al. Dasatinib
50. Baccarani M, Deininger MW, Rosti G, et al. imatinib and associated healthcare costs: A ret- treatment of chronic-phase chronic myeloid leu-
European LeukemiaNet recommendations for rospective analysis among managed care kemia: Analysis of responses according to preex-
the management of chronic myeloid leukemia: patients with chronic myelogenous leukaemia. isting BCR-ABL mutations. Blood 2009;114:
2013. Blood 2013;122:872884. PharmacoEconomics 2007;25:481496. 49444953.
51. Testoni N, Marzocchi G, Luatti S, et al. Chronic 66. Noens L, van Lierde MA, De Bock R, et al. 84. Hughes T, Saglio G, Branford S, et al. Impact of
myeloid leukemia: A prospective comparison of Prevalence, determinants, and outcomes of non- baseline BCR-ABL mutations on response to
interphase fluorescence in situ hybridization and adherence to imatinib therapy in patients with nilotinib in patients with chronic myeloid leuke-
chromosome banding analysis for the definition chronic myeloid leukemia: The ADAGIO study. mia in chronic phase. J Clin Oncol 2009;27:
of complete cytogenetic response: A study of the Blood 2009;113:54015411. 42044210.
R
GIMEMA CML WP. Blood 2009;114:49394943. 67. Bristol-Myers Squibb Company. SprycelV (dasa- 85. Kantarjian HM, Cortes JE, Kim DW, et al.
52. Kantarjian H, Cortes J. Considerations in the tinib) prescribing information. Princeton, NJ, Bosutinib safety and management of toxicity in
management of patients with Philadelphia 2011. leukemia patients with resistance or intolerance
chromosome-positive chronic myeloid leukemia 68. Novartis Pharmaceuticals Corporation. TasignaV R to imatinib and other tyrosine kinase inhibitors.
receiving tyrosine kinase inhibitors. J Clin Oncol (nilotinib) prescribing information. East Hano- Blood 2014;123:13091318.
2011;29:15121516. ver, NJ, 2011. 86. Jabbour E, Kantarjian H, Ghanem H, et al. The
53. Hehlmann R, Lauseker M, Jung-Munkwitz S, 69. Jabbour E, Kantarjian HM, Jones D, et al. Imati- achievement of a 3-month complete cytogenetic
et al. Tolerability-adapted imatinib 800 mg/d nib mesylate dose escalation is associated with response to second-generation tyrosine kinase
versus 400 mg/d versus 400 mg/d plus durable responses in patients with chronic mye- inhibitors predicts survival in patients with
interferon-alpha in newly diagnosed chronic loid leukemia after cytogenetic failure on chronic phase chronic myeloid leukemia after
myeloid leukemia. J Clin Oncol 2011;29:1634 standard-dose imatinib therapy. Blood 2009;113: imatinib failure. Clin Lymphoma Myeloma Leuk
1642. 21542160. 2013;13:302306.
54. Kantarjian HM, Shan J, Jones D, OBrien S, 70. Kantarjian H, Pasquini R, Levy V, et al. Dasati- 87. Jabbour E, Cortes J, Santos FP, et al. Results of
Rios MB, Jabbour E, et al. Significance of nib or high-dose imatinib for chronic-phase allogeneic hematopoietic stem cell transplanta-
increasing levels of minimal residual disease in chronic myeloid leukemia resistant to imatinib tion for chronic myelogenous leukemia patients
patients with Philadelphia chromosome-positive at a dose of 400 to 600 milligrams daily: Two- who failed tyrosine kinase inhibitors after devel-
chronic myelogenous leukemia in complete year follow-up of a randomized phase 2 study oping BCR-ABL1 kinase domain mutations.
cytogenetic response. J Clin Oncol 2009;27: (START-R). Cancer 2009;115:41364147. Blood 2011;117:36413647.
36593663. 71. Garcia-Gutierrez JV, Herrera P, Abalo LL, Rey 88. Lee SJ, Kukreja M, Wang T, et al. Impact of
55. Marin D, Ibrahim AR, Lucas C, et al. Assess- MD, Calbacho M. Impact of second-generation prior imatinib mesylate on the outcome of
ment of BCR-ABL1 transcript levels at 3 tyrosine kinase inhibitors as second line treat- hematopoietic cell transplantation for chronic
months is the only requirement for predicting ment for patients with chronic myeloid leuke- myeloid leukemia. Blood 2008;112:35003507.
outcome for patients with chronic myeloid leu- mia. Blood. 2011;118:632. 89. Ruiz-Arg uelles GJ, Tarin-Arzaga LC, Gonzalez-
kemia treated with tyrosine kinase inhibitors. 72. Goh HG, Jootar S, Kim HJ, Sohn SK, Park JS, Carrillo ML, et al. Therapeutic choices in
J Clin Oncol 2012;30:232238. Kim SH. Efficacy of nilotinib versus high-dose patients with Ph-positive CML living in Mexico
56. Jain P, Kantarjian H, Nazha A, et al. Early imatinib in early chronic phase CML patients in the tyrosine kinase inhibitor era: SCT or
responses predict better outcomes in patients who have suboptimal molecular responses to TKIs? Bone Marrow Transplant 2008;42:2328.
with newly diagnosed chronic myeloid leukemia: standard-dose imatinib (RE-NICE multicenter 90. Jaime-Perez JC, Heredia-Salazar AC, Cant u-
Results with four tyrosine kinase inhibitor study). Blood 2011;118:632. Rodrguez OG, et al. Cost structure and clinical
modalities. Blood 2013;121:48674874. 73. Cortes JE, Kantarjian HM, Brummendorf TH, outcome of a stem cell transplantation program
57. Neelakantan P, Gerrard G, Lucas C, et al. Com- et al. Safety and efficacy of bosutinib (SKI-606) in a developing country: The experience in
bining BCR-ABL1 transcript level at 3 and 6 in chronic phase Philadelphia chromosome- northeast Mexico. Oncologist 2015;20:386392.
months in chronic myeloid leukemia: Implica- positive chronic myeloid leukemia patients with 91. Mahon FX, Rea D, Guilhot J, et al. Discontinua-
tions for early intervention strategies. Blood resistance or intolerance to imatinib. Blood tion of imatinib in patients with chronic mye-
2013;121:37393742. 2011;118:45674576. loid leukaemia who have maintained complete
58. Nazha A, Kantarjian H, Jain P, et al. Assessment 74. Yeung DT, Osborn M, White DL, et al. Upfront molecular remission for at least 2 years: The
at 6 months may be warranted for patients with imatinib therapy in CML patients with rapid Prospective, multicenter Stop Imatinib (STIM)
chronic myeloid leukemia with no major cytoge- switching to nilotinib for failure to achieve trial. Lancet Oncol 2010;11:10291035.
netic response at 3 months. Haematologica molecular targets or intolerance achieves high 92. Mahon FX, Rea D, Guilhot J, et al. Long term
2013;98:16861688. overall rates of molecular response and a low follow-up after imatinib cessation for patients in
59. Branford S, Roberts N, Yeung DT, Altamura H, risk of progression - an update of the TIDEL-II deep molecular response: The updated results of
Parker WT, Hughes TP. Any BRC-ABL reduc- trial. Blood. 2011;118:632. the STIM1 study. Blood (ASH Annual Meeting
tion below 10% at 6 months of therapy signifi- 75. Quintas-Cardama A, Cortes JE, OBrien S, et al. Abstracts) 2013;122:255.
cantly improves outcome for CML patients with Dasatinib early intervention after cytogenetic or 93. Ross DM, Branford S, Seymour JF, et al. Safety
a poor response at 3 months. Blood (ASH hematologic resistance to imatinib in patients and efficacy of imatinib cessation for CML
Annual Meeting Abstracts) 2013;122:254. with chronic myeloid leukemia. Cancer 2009; patients with stable undetectable minimal resid-
60. Jabbour E, Kantarjian HM, OBrien S, et al. 115:29122921. ual disease: Results from the TWISTER study.
Front-line therapy with second-generation tyro- 76. OHare T, Shakespeare WC, Zhu X, et al. Blood 2013;122:515522.
sine kinase inhibitors in patients with early AP24534, a pan-BCR-ABL inhibitor for chronic 94. Mahon FX, Nicolini FE, Noel MP, et al. Prelimi-
chronic phase chronic myeloid leukemia: What myeloid leukemia, potently inhibits the T315I nary report of the STIM2 study: A multicenter
is the optimal response? J Clin Oncol 2011;29: mutant and overcomes mutation-based resist- stop imatinib trial for chronic phase chronic
42604265. ance. Cancer Cell 2009;16:401412. myeloid leukemia de novo patients on imatinib.
61. Branford S, Yeung DT, Parker WT, et al. Prog- 77. Zhou T, Commodore L, Huang WS, et al. Struc- Blood (ASH Annual Meeting Abstracts) 2013;
nosis for patients with CML and >10% BCR- tural mechanism of the pan-BCR-ABL inhibitor 122:654.

264 American Journal of Hematology, Vol. 91, No. 2, February 2016 doi:10.1002/ajh.24275
ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES Chronic myeloid leukemia

95. Rousselot P, Charbonnier A, Cony-Makhoul P, 99. Carpenter PA, Snyder DS, Flowers ME, et al. 103. Ravandi F, OBrien S, Thomas D, et al. First
et al. Loss of major molecular response as a trig- Prophylactic administration of imatinib after report of phase 2 study of dasatinib with
ger for restarting tyrosine kinase inhibitor ther- hematopoietic cell transplantation for high-risk hyper-CVAD for the frontline treatment of
apy in patients with chronic-phase chronic Philadelphia chromosome-positive leukemia. patients with Philadelphia chromosome-
myelogenous leukemia who have stopped imati- Blood 2007;109:27912793. positive (Ph1) acute lymphoblastic leukemia.
nib after durable undetectable disease. J Clin 100. Jabbour E, Cortes J, Kantarjian H, et al. Novel Blood 2010;116:20702077.
Oncol 2014;32:424430. tyrosine kinase inhibitor therapy before alloge- 104. Quintas-Cardama A, Kantarjian H, Garcia-
96. Mahon F, Richter J, Guilhot JG, et al. Interim neic stem cell transplantation in patients with Manero G, et al. A pilot study of imatinib,
analysis of a pan European Stop Tyrosine chronic myeloid leukemia: No evidence for low-dose cytarabine and idarubicin for patients
Kinase Inhibitor Trial in chronic myeloid leuke- increased transplant-related toxicity. Cancer with chronic myeloid leukemia in myeloid
mia: The EURO-SKI study. Blood (ASH Annual 2007;110:340344. blast phase. Leuk Lymph 2007;48:283289.
Meeting Abstracts) 2014;124:151. 101. le Coutre PD, Giles FJ, Hochhaus A, et al. Nilo- 105. Ohanian M, Kantarjian HM, Quintas-Cardama
97. Apperley JF, Cortes JE, Kim DW, et al. Dasati- tinib in patients with Ph1 chronic myeloid leu- A, Jabbour E, Abruzzo L, Verstovsek S, et al.
nib in the treatment of chronic myeloid leuke- kemia in accelerated phase following imatinib Tyrosine kinase inhibitors as initial therapy for
mia in accelerated phase after imatinib failure: resistance or intolerance: 24-month follow-up patients with chronic myeloid leukemia in
The START-A trial. J Clin Oncol 2009;27:3472 results. Leukemia 2012;26:11891194. accelerated phase. Clin Lymphoma Myeloma
3479. 102. Thomas DA, Faderl S, Cortes J, et al. Treat- Leuk 2014;14:155162.
98. Oehler VG, Gooley T, Snyder DS, et al. The ment of Philadelphia chromosome-positive
effects of imatinib mesylate treatment before acute lymphocytic leukemia with hyper-CVAD
allogeneic transplantation for chronic myeloid and imatinib mesylate. Blood 2004;103:4396
leukemia. Blood 2007;109:17821789. 4407.

doi:10.1002/ajh.24275 American Journal of Hematology, Vol. 91, No. 2, February 2016 265

You might also like