CME Information: Chronic Myeloid Leukemia: 2014 Update On Diagnosis, Monitoring, and Management
CME Information: Chronic Myeloid Leukemia: 2014 Update On Diagnosis, Monitoring, and Management
CME Information: Chronic Myeloid Leukemia: 2014 Update On Diagnosis, Monitoring, and Management
If you wish to receive credit for this activity, please refer to the website: www.wileyhealthlearning.com
Educational Objectives
Upon completion of this educational activity, participants will be better able to:
1. Define and personalize frontline therapy options in CML-CP
2. Identify milestones of response and when to switch therapy
Activity Disclosures
No commercial support has been accepted related to the development or publication of this activity.
Authors: Elias J. Jabbour discloses consulting work for BAS, Novartis, ARIPD, and Pfizer; as well as consulting work for and research grants
from TEVA. Hagop Kanterjian discloses research grants from BAS, Pfizer, Novartis, ARIPD, and TEVA.
CME Editor: Ayalew Tefferi, M.D. has no conflicts of interest 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 Servicess Policy on Activity Disclosure and
Conflict of Interest. The primary resolution method used was peer review and review by a non-conflicted expert.
Log on to http://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.
doi:10.1002/ajh.34
547
A JH
Introduction
Disease overview
Chronic Myeloid Leukemia (CML) is a myeloproliferative neoplasm with an incidence of 12 cases per 100,000 adults, and accounts for 15%
of newly diagnosed cases of leukemia in adults [1]. Central to the pathogenesis of CML is the fusion of the Abelson murine leukemia (ABL) gene
on chromosome 9 with the breakpoint cluster region (BCR) gene on chromosome 22, which results in expression of an oncoprotein, termed BCRABL [2]. BCR-ABL is a constitutively active tyrosine kinase that promotes growth and replication through downstream pathways such as RAS,
RAF, JUN kinase, MYC, and STAT [39]. 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 interferonalfa (INF-a) [10]. INF-a led to regression of the disease and improved survival but was hindered by a multitude of toxicities. Allogeneic stem cell
transplantation (AlloSCT) was a curative intervention, but carried with it a high risk of morbidity and mortality. Further, alloSCT is only an
option for patients with excellent performance status and an appropriate stem cell donor.
The landscape changed dramatically with the development of small molecule tyrosine kinase inhibitors (TKIs) that was shown to potently interfere with the interaction between the BCR-ABL protein and adenosine triphosphate (ATP), blocking cellular proliferation of the malignant clone
[4]. This targeted approach was found to dramatically alter the natural history of the disease, improving 10-year overall survival (OS) from 20
to 8090% [1,11]. In this review, we will briefly highlight the evidence supporting the use of each of the available TKIs, including how to select an
agent in various circumstances and phases of the disease. AlloSCT is performed rarely in CML, and patients who may benefit from such an intervention will be discussed. Cytogenetic and molecular benchmarks for patients on therapy will be mentioned throughout the article. Finally, appropriate monitoring strategies for patients on the various TKIs will be covered.
548
doi:10.1002/ajh.23691
Diagnosis
The diagnosis of typical CML is simple and consists of documenting, in the setting of persistent unexplained leukocytosis (or occasionally thrombocytosis), the presence of the Ph chromosome
abnormality, the t(9;22)(q34;q11), by routine cytogenetics, or the Phrelated molecular BCR-ABL abnormalities by fluorescent in situ
hybridization (FISH) or by molecular studies [1214].
A FISH analysis relies on the colocalization of large genomic
probes specific to the BCR and ABL genes. Comparison of simultaneous marrow and blood samples by FISH analysis shows high concordance. FISH studies may have a false positive range of 110%
depending on the probes used.
Reverse transcriptase-polymerase chain reaction (RT-PCR) amplifies
the region around the splice junction between BCR and ABL. It is highly
sensitive for the detection of minimal residual disease. PCR testing can
either be qualitative (QPCR), providing information about the presence
of the BCR-ABL transcript, or quantitative, assessing the amount of
BCR-ABL message. Qualitative PCR is useful for diagnosing CML;
quantitative PCR is ideal for monitoring residual disease. Simultaneous
peripheral blood and marrow QPCR studies show a high level of concordance. False-positive and false-negative results can happen with
PCR. False-negative results may be from poor-quality RNA or failure of
the reaction; false-positive results can be due to contamination. A 0.51
log difference in some samples can occur depending on testing procedures, sample handling, and laboratory experience [1214].
The Ph chromosome is usually present in 100% of metaphases, often
as the sole abnormality. Ten to fifteen percent of patients have additional
chromosomal changes (clonal evolution) involving trisomy 8, isochromosome 17, additional loss of material from 22q or double Ph, or others.
Eight-five percent of patients have a typical t(9;22); 5% have variant translocations, which can be simple (involving chromosome 22
and a chromosome other than chromosome 9), or complex (involving
one or more chromosomes in addition to chromosomes 9 and 22).
Patients with Ph-variants have response to therapy and prognosis
similar to Ph-positive CML.
Differential Diagnosis
CML must be differentiated from leukemoid reactions, which usually
produce white blood cell counts lower than 50 3 109/L, toxic granulo-
doi:10.1002/ajh.23691
cytic vacuolation, Dohles bodies in the granulocytes, absence of basophilia, and normal or increased LAP levels. The clinical history and
physical examination generally suggest the origin of the leukemoid reaction. Corticosteroids can rarely cause extreme neutrophilia with a left
shift, but this abnormality is self-limited and of short duration.
CML may be more difficult to differentiate from other myeloproliferative or myelodysplastic syndromes. Patients with agnogenic myeloid metaplasia with or without myelofibrosis frequently have
splenomegaly, neutrophilia, and thrombocytosis. Polycythemia vera
with associated iron deficiency, which causes normal hemoglobin and
hematocrit values, can manifest with leukocytosis and thrombocytosis.
Such patients usually have a normal or increased LAP score, a WBC
count <25 3 109/L, and no Ph chromosome.
The greatest diagnostic difficulty lies with patients who have
splenomegaly and leukocytosis but do not have the Ph chromosome.
In some, the BCR-ABL hybrid gene can be demonstrated despite a
normal or atypical cytogenetic pattern. Patients who are Ph negative
and BCR-ABL negative are considered to have Ph-negative CML or
chronic myelomonocytic leukemia. Rarely, patients have myeloid
hyperplasia, which involves almost exclusively the neutrophil, eosinophil, or basophil cell lineage. These patients are described as having
chronic neutrophilic, eosinophilic, or basophilic leukemia and do not
have evidence of the Ph chromosome or the BCR-ABL gene. Isolated
megakaryocytic hyperplasia can be seen in essential thrombocythemia,
with marked thrombocytosis and splenomegaly. Some patients who
present with clinical characteristics of essential thrombocythemia
(with marked thrombocytosis but without leukocytosis) have CML;
cytogenetic and molecular studies showing the Ph chromosome, the
BCR-ABL rearrangement, or both lead to the appropriate diagnosis
and treatment.
Imatinib
Imatinib mesylate (Gleevec, Novartis Pharmaceutical Corporation,
NJ), was the first TKI to receive approval by the Food and Drug
Administration for the treatment of patients with CML-CP. It acts
via competitive inhibition at the ATP-binding site of the BCR-ABL
protein, which results in the inhibition of phosphorylation of proteins
involved in cell signal transduction. It efficiently inhibits the BCRABL kinase, but also blocks the platelet-derived growth factor receptor, as well as the C-KIT tyrosine kinase [15].
The International Randomized Study of Interferon and STI571
(IRIS) study is considered a landmark clinical trial for TKIs and
CML [16]. Investigators randomized 1,106 patients to receive imatinib
400 mg/day or INF-a plus low-dose subcutaneous cytarabine. After a
median follow-up of 19 months, relevant outcomes for patients
receiving imatinib were significantly better than in those treated with
INF-a plus cytarabine, notably the rate of complete cytogenetic
response (CCyR) rate (74 vs. 9%, P < 0.001), and freedom from progression to AP or BP at 12 months (99 vs. 93%, P < 0.001). Further
highlighting the challenge of using IFN-a was the high crossover rate
to imatinib due to intolerance. The responses to imatinib were also
durable, as shown in an 8-year follow up of the IRIS study [11]. Estimated event free survival rate was 81%, and OS rate was 93% when
only CML-related deaths were considered.
While the results using imatinib are quite impressive, only 55% of
patients enrolled in the IRIS study remained on therapy at the 8-year
follow up point. This underscored the need for additional options for
549
TABLE I. Summary of Pivotal Phase III Trials of Approved Tyrosine Kinase Inhibitors for the Treatment of Frontline or Relapsed CML
Follow-up data
Trial
Treatment
No. of patients
Ima 400 mg qd
IFN 1 ara-c
553
553
282
281
283
Dasa 100 mg qd
Ima 400 mg qd
259
260
IRIS
ENESTnd
DASISION
% MMR
% PFS at 18 months
97
91 (P < 0.001)
% MMR at 12 months
44
43
22 (P < 0.001 for
both comparisons)
% CCyR at 12 months
77
Imatinib: 66 (P 5 0.007)
Secondary endpoint:
MMR at 12 months:
Dasa: 46 Ima: 28 (P < 0.0001)
77
77
60 (P < 0.0001 for
both comparisons)
74
46
% PFS
% OS
6/8 years
93/92
6/8 years
88/85
5 years
92
95.5 (P 0.03 vs ima)
93.6
96 (P 5 0.04 vs ima)
4 years
90
90.2
92.9
92.1
ara-c: cytarabine; dasa: dasatinib; IFN: interferon; Ima: imatinib; MMR: major molecular response; nilo: nilotinib; OS: overall survival; PFS: progression-free
survival.
patients who had failed or were intolerant to imatinib. This led to the
rational development of second generation TKIs with hopes they would
effectively treat patients unable to continue on imatinib therapy.
Dasatinib
Dasatinib (Sprycel, Bristol-Myers Squibb) is an oral, second generation TKI that is 350 times more potent than imatinib in vitro [17
19]. In addition, it also is known to inhibit the Src family of kinases,
which may also be important in blunting critical cell signaling pathways [20]. Though initially evaluated in patients in the salvage setting, clinicians and researchers were excited to test the possibility that
frontline use of the more potent inhibitors might further improve the
outcome compared to imatinib.
The DASISION trial was a phase III, randomized study comparing
imatinib 400 mg once daily to dasatinib 100 mg once daily in newly
diagnosed patients [21]. Dose escalations were allowed for both drugs
in the setting of suboptimal response as defined per protocol. The
primary outcome was confirmed cCCyR at 12 months. A total of 519
patients were randomized in a 1:1 manner. Patients assigned to dasatinib achieved cCCyR at 12 months more frequently than those on
imatinib (77 vs. 66%, P 5 0.007). Many of the secondary endpoints of
interest were also significantly different between groups favoring the
dasatinib arm. A three-year follow up of the trial was recently published, and illustrated that dasatinib induces more rapid, deeper
responses at early time points compared to imatinib [22]. For
instance, at 3 months, a higher proportion of patients treated with
dasatinib achieved a BCR-ABL transcript level of less than or equal
to 10% (84 vs. 64%, P < 0.0001). Meeting this threshold in either arm
predicted for both progression-free survival and OS. As might be
expected, pleural effusions occurred more frequently in the group
receiving dasatinib (19 vs. <1%).
Nilotinib
Nilotinib (Tasigna, Novartis Pharmaceutical Corporation, NJ) is a
structural analog of imatinib, though its affinity for the ATP binding
site on BCR-ABL is up to 50 times more potent in vitro [23]. Like
dasatinib, nilotinib initially demonstrated the ability to induce hematologic and cytogenetic responses in patients who had failed imatinib.
Similar to the data with dasatinib, nilotinib has also been directly
compared with imatinib in a large, international, randomized study.
In ENESTnd, two doses of nilotinib (300 or 400 mg twice daily) were
compared with imatinib 400 mg once daily [24]. The primary end550
point in this study was the rate of major molecular response (MMR)
at 12 months. This endpoint was achieved at statistically significantly
higher rates for both doses of nilotinib compared with the imatinib
group (44 and 43% vs. 22%, P < 0.001). There was also much less
progression to AP or BP on the nilotinib arm. Recently, a 36 month
follow-up was published, and differences in deep molecular responses
continued to favor nilotinib at either dose [25].
Current guidelines recommend any of the three TKIs: imatinib,
dastinib, or nilotinib as options with a category 1 recommendation
for initial treatment of CML-CP [26]. Second generation TKIs have
shown inducing higher rates of early optimal responses; their impact
on long-term over survival remains to be determined. AlloHSCT or
other chemotherapy agents are not any longer recommended as
upfront treatments for CML-CP given the excellent outcomes and
long-term survival achieved with the TKIs.
doi:10.1002/ajh.23691
Cost
The price of therapies to treat cancer has been on an exponential rise
over the past decade [32]. Of the 12 agents used in cancer care that
were approved in 2012, at least nine of them were priced at over
$100,000 annually [33]. Most of these therapies did not appear to provide a substantial improvement in survival or other measures to justify
such a cost. Recently, an international group of CML experts called
attention to the high prices of TKIs in particular [34]. The case of imatinib was used to illustrate an example of how things have spun out of
control. When first approved, the annual price for imatinib was
$30,000 in the United States. This was the price set to make the development and commercialization of imatinib profitable when it was
expected that most patients would take the drug for perhaps 510 years.
Patients can now remain on imatinib and live a normal lifespan as long
as they are compliant with the medication [35]. Imatinib has also gone
on to receive additional indications, such as the treatment of gastrointestinal stromal tumor [36]. Paradoxically though, with a higher number of patients and a longer duration of therapy, the annual price of
imatinib in 2013 was nearly triple at $90,000. This price is in the same
range as dasatinib and nilotinib, both of which are over $100,000.
At this point, the costs of TKIs are all fairly similar, as they are
priced against each other. However, in the coming years, imatinib is
set to be available as a generic formulation, and the expectation is that
the price will eventually fall dramatically. When this happens, clinicians
will have to weigh the early benefits some patients attain with dasatinib
or nilotinib in the frontline setting against the disparity in cost.
In general, any of the three TKIs currently approved for frontline
use may be selected. These include imatinib, dasatinib, or nilotinib.
While dasatinib and nilotinib have demonstrated superiority over imatinib when early surrogate markers are used, imatinib is still expected
to be highly effective in a large number of CML patients. When
choosing an agent, one can consider issues such as comorbidities,
adverse event profile, and cost to help make the final determination.
Kinase domain mutation profile plays very little, if any role in selecting an initial TKI, but becomes highly relevant in the relapsed setting.
doi:10.1002/ajh.23691
551
Baseline
3 months
6 months
12 months
Any time
Optimal
Warning
Failure
perform serial molecular monitoring between 3 and 6 months to sufficiently determine the response. If patients still have >10% BCRABL transcripts at 6 months, the chances of attaining CCyR are low,
and a change of therapy might be in order. We would also consider
this approach for patients on second generation TKIs, because, as
mentioned previously, very early switching has not yet been shown to
influence the long-term outcome [48]. Another study randomized
patients in CCyR on imatinib for at least 2 years to continue imatinib
or switch to nilotinib [49]. While switching to nilotinib induced
deeper molecular responses, it has yet to translate into an improvement in progression-free survival or other meaningful outcomes.
Patients who meet all of the relevant benchmarks in the first 12
months are monitored periodically using FISH and molecular testing.
If there are clear signs of possible failure, patients should undergo a
bone marrow biopsy with cytogenetics to assess for relapse. Any
degree of cytogenetic relapse calls for a change in therapy, while fluctuating molecular levels during continuous CCyR should only prompt
closer monitoring and a compliance assessment.
In several studies, the achievement of a CCyR (Ph-positive metaphases 0%; BCR-ABL transcripts [IS] 1%) at 12 months or later on
TKI therapy was associated with significant survival benefit compared
with achievement of lesser degrees of response. Therefore, achievement of CCyR is now the primary endpoint of TKI therapy. Achievement of BCR-ABL transcripts ( 0.1% [IS]) was associated with
modest improvements in event-free survival rates, possible longer
durations of CCyRs, but not with a survival benefit. The achievement
of complete molecular response (CMR; nonmeasurable BCR-ABL
transcripts) offers the possibility of treatment discontinuation in clinical trials only. Lack of achievement of MMR or of CMR should not
be interpreted as a need to change TKI therapy or to consider
alloHSCT. Response assessments at earlier times on front-line TKI
therapy (36 months) have shown better outcomes with achievement
of a major cytogenetic response by 36 months on imatinib therapy
(Ph-positive metaphases 35%, or BCR-ABL1 transcripts 10%).
While this is interpreted to mean that a change to second TKI therapy may be considered if such outcome is not obtained, no studies
have shown that changing therapy from imatinib to second TKIs has
improved patients outcome. When nilotinib or dasatinib are used for
front-line therapy, achievement of CCyR by 36 months of TKIs
therapy has been associated with improved outcomes.
doi:10.1002/ajh.23691
Nilotinib
Bosutinib
Ponatinib
CP
AP
MyBP
LyBP
CP
AP
MyBP LyBP
CP
AP
BP
CP
N 5 387 N 5 174 N 5 109 N 5 48 N 5 321 N 5 137 N 5 105 N 5 31 N 5 146 N 5 51 N 5 38 N 5 271
15
74
91
NR
49
11
96
4
93
79
45
19
44
32
7
82
12a
91
50
27
7
36
26
7
50
12a
88
40
29
6
52
46
6
50
24
70
94
76
NR
46
15
87
9
80
56
31
12
NR
20
12
67
3
82
22
11
1
NR
29
10
42
3
82
19
13
0
NR
32
16
42
7
69
85
81
34
13
98
6
NRb
54
54
0
NR
27
20
60
3
NRb
36
36
NR
NR
35
18
50
11
NR
NR
NR
46
NR
91
AP
N 5 79
BP
N 5 94
13
6
96
NR
NR
Ma HR: 57 MaHR: 34
NR
NR
NR
NR
55
36
NR
NR
42
35
AP: accelerated phase; BP: blast phase; CHR: complete hematologic response; CP: chronic phase; LyBP: lymphoid blast phase; MaHR: major hematologic
response; MyBP: myeloid blast phase; NEL: no evidence of leukemia; NR: not reported.
nib retain activity against most of the known mutations that confer
resistance to imatinib [65,66]. When selecting between dasatinib and
nilotinib, in vitro and in vivo data have identified distinct mutations
that exhibit decreased sensitivity to each of the agents [67,68]. The
clinician may tend to favor dasatinib if the patient has the following
mutations at the time of disease progression: Y253H, E255K/V, or
F359C/V. Alternatively, nilotinib may be a better selection in the
presence of the V299L and F317L mutations. For patients lacking
these mutations, the choice once again calls into consideration toxicity profile and cost.
The available data also indicate that bosutinib can be used for
patients with most known mutations that lead to imatinib failure
[61]. Like dasatinib and nilotinib, bosutinib lacks activity against
T315I. Bosutinib may be a reasonable choice for patients who fail
imatinib who are not good candidates for dasatinib or nilotinib.
Bosutinib has a relatively distinct toxicity profile from the other TKIs,
with the predominant problem being diarrhea and other gastrointestinal complaints. Recently, an analysis was conducted to closely characterize the toxicity of bosutinib and the management strategy [69].
Diarrhea was documented in 82% of the overall cohort (n 5 570), the
majority of which was grades 1/2 in severity. Myelosuppression and
liver function test abnormalities were also common. The authors
found that with appropriate supportive care and monitoring, most
patients are able to continue on therapy with periodic dose interruptions or adjustment.
Ponatinib should be considered the agent of choice for any patient
that develops a T315I mutation and in instances where other TKIs
are not indicated. There are currently no other commercially available
TKIs that have activity against this mutation. The risk for thrombotic
events with ponatinib is serious, but the benefits outweigh the risks
for most patients with a T315I mutation, as there are very few viable
options for disease control.
Second and third generation TKIs have not been compared headto-head, so we currently select one or the other based on the side
effect profile, mutations profile, drug interactions, compliance issues,
and the patients preexisting medical conditions. We do a mutational
analysis in patients who are failing imatinib or second generation
TKIs, or those who progress to AP/BP. We do not test baseline mutational analysis on patients with newly diagnosed CML in CP, as it
has not proven to predict treatment outcome either.
553
doi:10.1002/ajh.23691
References
doi:10.1002/ajh.23691
19.
20.
21.
22.
23.
24.
25.
26.
27.
28.
29.
30.
31.
32.
33.
34.
35.
36.
37.
38.
39.
BMS-354825 against clinically relevant imatinibresistant Abl kinase domain mutants. Cancer
Res 2005;65:45004505.
Tokarski JS, Newitt JA, Chang CY, et al. The
structure of Dasatinib (BMS-354825) bound
to activated ABL kinase domain elucidates
its inhibitory activity against imatinibresistant ABL mutants. Cancer Res 2006;66:
57905797.
Shah NP, Tran C, Lee FY, et al. Overriding imatinib resistance with a novel ABL kinase inhibitor. Science 2004;305:399401.
Kantarjian H, Shah NP, Hochhaus A, et al.
Dasatinib versus imatinib in newly diagnosed
chronic-phase chronic myeloid leukemia. N
Engl J Med 2010;362:22602270.
Jabbour E, Kantarjian HM, Saglio G, et al. Early
response with dasatinib or imatinib in chronic
myeloid leukemia: 3-year follow-up from a
randomized phase 3 trial (DASISION). Blood
2014;123:494500.
Weisberg E, Manley PW, Breitenstein W, et al.
Characterization of AMN107, a selective inhibitor of native and mutant Bcr-Abl. Cancer Cell
2005;7:129141.
Saglio G, Kim DW, Issaragrisil S, et al. Nilotinib
versus imatinib for newly diagnosed chronic
myeloid leukemia. N Engl J Med 2010;362:
22512259.
Larson RA, Hochhaus A, Clark RE, et al. Nilotinib vs imatinib in patients with newly diagnosed
Philadelphia chromosome-positive chronic myeloid leukemia in chronic phase: ENESTnd 3year follow up. Leukemia 2012;26:21972203.
OBrien S, Radich JP, Abboud CN, et al.
Chronic myelogenous leukemia, Version 1.2014.
J Natl Compr Canc Netw 2013;11:13271340.
Montani D, Bergot E, Gunther S, et al. Pulmonary arterial hypertension in patients treated by
dasatinib. Circulation 2012;125:21282137.
Quintas-Cardama A, Han X, Kantarjian H,
Cortes J. Tyrosine kinase inhibitor-induced platelet dysfunction in patients with chronic myeloid leukemia. Blood 2009;114:261263.
Quintas-Cardama A, Kantarjian H, Ravandi F,
et al. Bleeding diathesis in patients with chronic
myeloid leukemia receiving dasatinib therapy.
Cancer 2009;115:24822490.
Kim TD, le Coutre P, Schwarz M, et al. Clinical
cardiac safety profile of nilotinib. Haematologica
2012;97:883889.
Quintas-Cardama A, Kantarjian H, Cortes J.
Nilotinib-associated vascular events. Clin Lymphoma Myeloma Leuk 2012;12:337340.
Smith TJ, Hillner BE. Bending the cost curve
in cancer care. N Engl J Med 2011;364:2060
2065.
Kantarjian HM, Fojo T, Mathisen M, Zwelling
LA. Cancer drugs in the United States: justum
pretiumthe just price. J Clin Oncol 2013;31:
36003604.
Experts in Chronic Myeloid Leukemia. The
price of drugs for chronic myeloid leukemia is a
reflection of the unsustainable prices of cancer
drugs: from the perspective of a large group of
CML experts. Blood 2013;121:44394442.
Gambacorti-Passerini C, Antolini L, Mahon X,
et al. Multicenter independent assessment of
outcomes in chronic myeloid leukemia patients
treated with imatinib. J Natl Cancer Inst 2011;
103:553561.
Gleevec (imatinib) Package Insert. East Hanover,
NJ. Novartis Pharmaceuticals Corporation; 2013.
Baccarani M, Deininger MW, Rosti G, et al.
European LeukemiaNet recommendations for
the management of chronic myeloid leukemia:
2013. Blood 2013;122:872884.
Testoni N, Marzocchi G, Luatti S, et al. Chronic
myeloid leukemia: a prospective comparison of
interphase fluorescence in situ hybridization and
chromosome banding analysis for the definition
of complete cytogenetic response: a study of the
GIMEMA CML WP. Blood 2009;114:4939
4943.
Kantarjian H, Cortes J. Considerations in the
management of patients with Philadelphia
chromosome-positive chronic myeloid leukemia
40.
41.
42.
43.
44.
45.
46.
47.
48.
49.
50.
51.
52.
53.
54.
55.
555
56.
57.
58.
59.
60.
61.
62.
63.
64.
65.
556
66.
67.
68.
69.
70.
71.
72.
73.
74.
75.
chronic myeloid leukemia after sequential treatment with multiple tyrosine kinase inhibitors.
Blood 2007;110:40054011.
Jabbour E, Branford S, Saglio G, Jones D. Practical advice for determining the role of BCRABL mutations in guiding tyrosine kinase inhibitor therapy in patients with chronic myeloid
leukemia. Cancer 2011;117:18001811.
Muller MC, Cortes JE, Kim DW, et al. Dasatinib
treatment of chronic-phase chronic myeloid leukemia: analysis of responses according to preexisting BCR-ABL mutations. Blood 2009;114:
49444953.
Hughes T, Saglio G, Branford S, et al. Impact of
baseline BCR-ABL mutations on response to nilotinib in patients with chronic myeloid leukemia in
chronic phase. J Clin Oncol 2009;27:42044210.
Kantarjian HM, Cortes JE, Kim DW, et al.
Bosutinib safety and management of toxicity in
leukemia patients with resistance or intolerance
to imatinib and other tyrosine kinase inhibitors.
Blood, in press.
Jabbour E, Cortes J, Santos FP, et al. Results of
allogeneic hematopoietic stem cell transplantation for chronic myelogenous leukemia patients
who failed tyrosine kinase inhibitors after developing BCR-ABL1 kinase domain mutations.
Blood 2011;117:36413647.
Mahon FX, Rea D, Guilhot J, et al. Discontinuation of imatinib in patients with chronic myeloid leukaemia who have maintained complete
molecular remission for at least 2 years: the prospective, multicenter Stop Imatinib (STIM) trial.
Lancet Oncol 2010;11:10291035.
Mahon FX, Rea D, Guilhot J, et al. Long term
follow-up after imatinib cessation for patients in
deep molecular response: the updated results of
the STIM1 study. Blood (ASH Annual Meeting
Abstracts) 2013;122:255.
Ross DM, Branford S, Seymour JF, et al. Safety
and efficacy of imatinib cessation for CML
patients with stable undetectable minimal residual disease: results from the TWISTER study.
Blood 2013;122:515522.
Mahon FX, Nicolini FE, Noel MP, et al. Preliminary report of the STIM2 study: a multicenter
stop imatinib trial for chronic phase chronic
myeloid leukemia de novo patients on imatinib.
Blood (ASH Annual Meeting Abstracts) 2013;
122:654.
Rousselot P, Charbonnier A, Cony-Makhoul P,
et al. Loss of major molecular response as a trig-
76.
77.
78.
79.
80.
81.
82.
83.
84.
ger for restarting tyrosine kinase inhibitor therapy in patients with chronic-phase chronic
myelogenous leukemia who have stopped imatinib after durable undetectable disease. J Clin
Oncol 2014;32:424430.
Apperley JF, Cortes JE, Kim DW, et al. Dasatinib
in the treatment of chronic myeloid leukemia in
accelerated phase after imatinib failure: the
START-A trial. J Clin Oncol 2009;27:34723479.
Oehler VG, Gooley T, Snyder DS, et al. The
effects of imatinib mesylate treatment before
allogeneic transplantation for chronic myeloid
leukemia. Blood 2007;109:17821789.
Carpenter PA, Snyder DS, Flowers ME, et al.
Prophylactic administration of imatinib after
hematopoietic cell transplantation for high-risk
Philadelphia chromosome-positive leukemia.
Blood 2007;109:27912793.
Jabbour E, Cortes J, Kantarjian H, et al. Novel
tyrosine kinase inhibitor therapy before allogeneic stem cell transplantation in patients with
chronic myeloid leukemia: no evidence for
increased transplant-related toxicity. Cancer
2007;110:340344.
le Coutre PD, Giles FJ, Hochhaus A, et al. Nilotinib in patients with Ph1 chronic myeloid leukemia in accelerated phase following imatinib
resistance or intolerance: 24-month follow-up
results. Leukemia 2012;26:11891194.
Thomas DA, Faderl S, Cortes J, et al. Treatment
of Philadelphia chromosome-positive acute lymphocytic leukemia with hyper-CVAD and imatinib mesylate. Blood 2004;103:43964407.
Ravandi F, OBrien S, Thomas D, et al. First report
of phase 2 study of dasatinib with hyper-CVAD
for the frontline treatment of patients with Philadelphia chromosome-positive (Ph1) acute lymphoblastic leukemia. Blood 2010;116:20702077.
Quintas-Cardama A, Kantarjian H, GarciaManero G, et al. A pilot study of imatinib, lowdose cytarabine and idarubicin for patients with
chronic myeloid leukemia in myeloid blast
phase. Leuk Lymphoma 2007;48:283289.
Ohanian M, Kantarjian HM, Quintas-Cardama
A, et al. Tyrosine kinase inhibitors as initial
therapy for patients with chronic myeloid leukemia in accelerated phase. Clin Lymphoma
Myeloma Leuk, in press.
doi:10.1002/ajh.23691