You are on page 1of 18

Received: 13 December 2017 | Accepted: 17 December 2017

DOI: 10.1002/ajh.25011

ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL A JH


MALIGNANCIES

Chronic myeloid leukemia: 2018 update on diagnosis, therapy


and monitoring

Elias Jabbour | Hagop Kantarjian

Department of Leukemia, The University of


Texas M. D. Anderson Cancer Center,
Abstract
Houston, Texas
Disease overview: Chronic myeloid leukemia (CML) is a myeloproliferative neoplasm with an inci-
Correspondence dence of 1-2 cases per 100 000 adults. It accounts for approximately 15% of newly diagnosed
Elias Jabbour, MD Anderson Cancer cases of leukemia in adults.
Center, Box 428, 1515 Holcombe Blvd,
Houston, Texas 77030. Diagnosis: CML is characterized by a balanced genetic translocation, t(9;22)(q34;q11.2), involving
Email: ejabbour@mdanderson.org 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 chromo-
some. The molecular consequence of this translocation is the generation of a BCR-ABL1 fusion
oncogene, which in turn translates into a BCR-ABL1 oncoprotein.
Frontline therapy: Four tyrosine kinase inhibitors (TKIs), imatinib, nilotinib, dasatinib, and bosuti-
nib 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 second genera-
tion TKIs reported significantly deeper and faster responses; this has not translated into improved
long-term survival, because of the availability of effective salvage therapies.
Salvage therapy: For patients who fail frontline therapy, second-line options include second and
third generation TKIs. Second and third generation TKIs, although potent and selective, exhibit
unique pharmacological profiles and response patterns relative to different patient and disease char-
acteristics, 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 2 TKIs, and for all patients in CML advanced phases.

1 | DISEASE OVERVIEW cluster region (BCR) gene on chromosome 22. This results in expression
of an oncoprotein termed BCR-ABL1.3 BCR-ABL1 is a constitutively
Chronic myeloid leukemia (CML) is a myeloproliferative neoplasm with active tyrosine kinase that promotes growth and replication through
an incidence of 1–2 cases per 100 000 adults. It accounts for approxi- downstream signaling pathways such as RAS, RAF, JUN kinase, MYC,
mately 15% of newly diagnosed cases of leukemia in adults. In 2017, 1
and STAT.4–10 This influences leukemogenesis by creating a cytokine-
it is estimated about 9000 new CML cases will be diagnosed in the independent cell cycle with aberrant apoptotic signals in response to
United States, and about 1000 patients will die of CML. Since the cytokine withdrawal.
introduction of imatinib in 2000, the annual mortality in CML has Until 2000, drug therapy for CML was limited to nonspecific agents
1
decreased from 10%-20% down to 1%-2%. Consequently, the preva- such as busulfan, hydroxyurea, and interferon-alfa (IFN-a).11 IFN-a led to
lence of CML in the United States, estimated at about 25–30 000 in disease regression and improved survival but was hindered by its modest
2000, has increased to an estimated 80–100 0001 in 2017, and will efficacy and associated significant toxicities. Allogeneic stem cell trans-
reach a plateau of about 180 000 cases by 2030.2 plantation (allo-SCT) is curative, but carries risks of morbidity and mortal-
Central to the pathogenesis of CML is the fusion of the Abelson ity. Further, allo-SCT is an option only for patients with good performance
murine leukemia (ABL1) gene on chromosome 9 with the breakpoint status and organ functions, and who have an appropriate donor.

442 | V
C 2018 Wiley Periodicals, Inc. wileyonlinelibrary.com/journal/ajh Am J Hematol. 2018;93:442–459.
A JH |
JABBOUR AND KANTARJIAN 443

The CML therapeutic landscape changed dramatically with the related molecular BCR-ABL1 abnormalities by fluorescence in situ
development of the small molecule tyrosine kinase inhibitors (TKIs) hybridization (FISH) or by molecular studies.13–15
that potently interfered with the interaction between the BCR-ABL1 A FISH analysis relies on the colocalization of large genomic
oncoprotein and adenosine triphosphate (ATP), blocking cellular prolif- probes specific to the BCR and ABL genes. Comparison of simultaneous
eration of the malignant clone. This “targeted” approach altered the marrow and blood samples by FISH analysis shows high concordance.
natural history of CML, improving the 10-year survival rate from FISH studies may have a false positive range of 1%-5% depending on
approximately 20% to 80%-90%. 1,2,12 the probes used.
In this review, we will highlight the evidence supporting the use of Reverse transcriptase-polymerase chain reaction (RT-PCR) ampli-
each of the available TKIs, including how to select an agent under dif- fies the region around the splice junction between BCR and ABL1. It is
ferent circumstances and particular phases of CML. Allo-SCT is an highly sensitive in detecting minimal residual disease. PCR testing can
important treatment option in CML chronic phase (CP) post TKIs fail- either be qualitative (QPCR), providing information about the presence

ure, and in advanced CML phases, and its role will be reviewed. Cyto- of the BCR-ABL1 transcript, or quantitative, assessing the amount of

genetic and molecular benchmarks for patients on therapy will be BCR-ABL1 transcripts. Qualitative PCR is useful for diagnosing CML;

discussed. Finally, appropriate monitoring strategies for patients on quantitative PCR is ideal for monitoring residual disease. Simultaneous

TKIs will be addressed. peripheral blood and marrow QPCR studies show a high level of con-
cordance. False-positive and false-negative results can happen with
PCR. False-negative results may be from poor-quality RNA or failure of
1.1 | Manifestations and staging the reaction; false-positive results can be due to contamination. A 0.5-
1 log difference in some samples can occur depending on testing pro-
About 50% of patients diagnosed with CML in the United States are
cedures, sample handling, and laboratory experience.13–15 For correla-
asymptomatic, and are often diagnosed during a routine physical exami-
tive purpose and monitoring without necessarily performing repeat
nation or blood tests. CML can be classified into three phases: CP, accel-
marrow studies, a complete cytogenetic response (CcyR; 0% Ph-
erated phase (AP), and blast phase (BP). Most (90%-95%) patients
positive metaphases by cytogenetic) is equivalent to a negative FISH
present in CML-CP. Common signs and symptoms of CML-CP, when
test (6 2%) and BCR-ABL1 transcripts by International Standard [IS]
present, result from anemia and splenomegaly. These include fatigue,
<1%. A partial cytogenetic response (Ph-positive metaphases 35%) is
weight loss, malaise, easy satiety, and left upper quadrant fullness or
equivalent to BCR-ABL1 transcripts 10% [IS].
pain. Rare manifestations include bleeding (associated with a low platelet
The Ph chromosome is usually present in 100% of metaphases,
count and/or platelet dysfunction), thrombosis (associated with throm-
often as the sole abnormality. Ten to 15% of patients have additional
bocytosis and/or marked leukocytosis), gouty arthritis (from elevated
chromosomal changes (clonal evolution) involving trisomy 8, isochromo-
uric acid levels), priapism (usually with marked leukocytosis or thrombo-
some 17, additional loss of material from 22q or double Ph, or others.
cytosis), retinal hemorrhages, and upper gastrointestinal ulceration and
Ninety percent of patients have a typical t(9;22); 5% have variant
bleeding (from elevated histamine levels due to basophilia). Leukostatic
translocations which can be simple (involving chromosome 22 and a
symptoms (dyspnea, drowsiness, loss of coordination, confusion) due to
chromosome other than chromosome 9), or complex (involving one or
leukemic cells sludging in the pulmonary or cerebral vessels, are uncom-
more chromosomes in addition to chromosomes 9 and 22). Patients
mon in CP despite white blood cell (WBC) counts exceeding 100 3 109/ with Ph-variants have response to therapy and prognosis similar to Ph-
L. Splenomegaly is the most consistent physical sign detected in 40%- positive CML. About 2%-5% of patients present with a morphologic
50% of cases. Hepatomegaly is less common (less than 10%). Lymphade- picture of CML without the Ph-positivity by cytogenetic studies. FISH
nopathy and infiltration of skin or other tissues are rare. When present, and PCR document Ph-negative BCR-ABL1 rearranged CML. Such
they favor Ph-negative CML or AP or BP of CML. Headaches, bone patients have similar response and outcome on TKI therapy as patients
pain, arthralgias, pain from splenic infarction, and fever are more fre- with Ph-positive CML.
quent with CML transformation. Most patients evolve into AP prior to Bone marrow aspiration is mandatory for all patients in whom
BP, but 20% transition into BP without AP warning signals. CML-AP CML is suspected, as it will confirm the diagnosis (eg, cytogenetic anal-
might be insidious or present with worsening anemia, splenomegaly and ysis), and provide information needed for staging in terms of the blast
organ infiltration; CML-BP presents as an acute leukemia (myeloid in and basophil percentages. Baseline cytogenetic analysis allows the
60%, lymphoid in 30%, megakaryocytic or undifferentiated in 10%) with detection of clonal evolution, particularly i(17)(q10)-7/del7q, and
worsening constitutional symptoms, bleeding, fever and infections. 3q26.2 rearrangements, associated with a relatively poor prognosis.16
Baseline reverse transcriptase-polymerase chain reaction is imperative
to identify the specific type of rearrangement that can be appropriately
1.2 | Diagnosis
followed when assessing for response to TKI therapy. About 2%-5% of
The diagnosis of typical CML is simple and consists of documenting, in patients have e13a3 or e14a3 (not e13a2 or e14a2) variants of p210
the setting of persistent unexplained leukocytosis (or occasionally BCR-ABL1 or p230 transcripts that may yield a false negative PCR by
thrombocytosis), the presence of the Philadelphia (Ph) chromosome routine probes and (if not tested at diagnosis) would give the false
abnormality, the t(9;22)(q34;q11), by routine cytogenetics, or the Ph- impression that a patient is in “complete molecular response” on TKI.
|
A JH
444 JABBOUR AND KANTARJIAN

T AB LE 1 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 10 years At 10 years

IRIS Imatinib (n 5 304) 83 93 NR 80 83 11

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

At 12 months At 12 months

BEFORE Bosutinib 400 mg (n 5 268) 77 47 75 96 99.9 18 months

Imatinib (n 5 268) 66 37 57 94 99.7

NR, Not reported.

1.3 | Differential diagnosis 1.4 | Frontline treatment options


CML must be differentiated from leukemoid reactions, which usually The three commercially available TKIs for the frontline treatment of
produce white blood cell counts, lower than 50 3 109/L, toxic granulo- CML include imatinib, dasatinib, and nilotinib. Current guidelines
€ hle’s bodies in the granulocytes, absence of baso-
cytic vacuolation, Do endorse all three as options for the initial management of CML in the
philia, and normal or increased LAP levels. The clinical history and chronic phase (CML-CP) (Table 1).
physical examination generally suggest the origin of the leukemoid
1.4.1 | Imatinib
reaction. Corticosteroids can rarely cause extreme neutrophilia with a
left shift, but this abnormality is transient and short-lasting. Imatinib mesylate was the first TKI to receive the Food and Drug
CML may be more difficult to differentiate from other myeloproli- Administration (FDA) approval for the treatment of patients with CML-
ferative or myelodysplastic syndromes. Patients with agnogenic mye- CP. It acts via competitive inhibition at the ATP-binding site of the
loid metaplasia with or without myelofibrosis frequently have BCR-ABL1 oncoprotein, which results in the inhibition of phosphoryla-
splenomegaly, neutrophilia, and thrombocytosis. Polycythemia vera tion of proteins involved in cell signal transduction. It also blocks the
with associated iron deficiency, which causes normal hemoglobin and platelet-derived growth factor receptor and the C-KIT tyrosine
hematocrit values, can manifest with leukocytosis and thrombocytosis. kinase.17
Such patients usually have a normal or increased LAP score, a WBC The International Randomized Study of Interferon and STI571 (IRIS)
count less than 25 3 10 /L, and no Ph abnormality.
9 study is considered a landmark clinical trial for TKIs and CML.18 Investi-
The greatest diagnostic difficulty lies with patients who have spleno- gators randomized 1106 patients in CML-CP to receive imatinib
megaly and leukocytosis but who do not have the Ph chromosome. In 400 mg/day or IFN-a and low-dose cytarabine. After a median follow-up
some, the BCR-ABL1 hybrid gene can be demonstrated despite a normal of 19 months, the outcomes for patients receiving imatinib were signifi-
or atypical cytogenetic pattern. Patients who are Ph negative and BCR- cantly better than those treated with IFN-a and cytarabine, notably the
ABL1 negative are considered to have Ph-negative CML or chronic rates of complete cytogenetic response (CCyR) rate (74% versus. 9%,
myelomonocytic leukemia. Rarely, patients have myeloid hyperplasia, P < .001), and freedom-from-progression to AP or BP at 12 months
which involves almost exclusively the neutrophil, eosinophil, or basophil (99% versus 93%, P < .001). Further highlighting the challenge of using
cell lineage. These patients are described as having chronic neutrophilic, IFN-a was the high crossover rate to imatinib due to intolerance. With a
eosinophilic, or basophilic leukemia and do not have evidence of the Ph median follow-up of almost 11 years, the estimated overall survival rate
chromosome or the BCR-ABL1 gene. Isolated megakaryocytic hyperplasia for patients who had been assigned to receive imatinib was 83.3% with
can be seen in essential thrombocythemia, with marked thrombocytosis a cumulative CCyR rate of 83% and a 10-year major molecular response
and splenomegaly. Some patients who present with clinical characteris- (MMR) rate of 93%.19 Despite the high rate of early crossover among
tics of essential thrombocythemia (with marked thrombocytosis but patients assigned to receive IFN-a and cytarabine, the 10-year survival
without leukocytosis) have CML; cytogenetic and molecular studies rate favored the imatinib therapy arm (83.3% versus 78.8%).19
showing the Ph chromosome, the BCR-ABL1 rearrangement, or both While the results using imatinib are impressive, only 48% of
lead to the appropriate diagnosis and treatment. patients enrolled in the IRIS study remained on therapy at the 10-year
A JH |
JABBOUR AND KANTARJIAN 445

follow-up time. This underscored the need for additional treatment 2012, 1551 newly diagnosed patients in CML-CP were randomized
options for patients who had resistance or intolerance to imatinib. This into a 5-arm study. The study was powered to detect a survival dif-
led to the rational development of second generation TKIs. ference of 5% at 5 years. After a median observation time of 9.5
years, the 10-year overall survival rate was 82%, the 10-year pro-
1.4.2 | Imatinib generics
gression-free survival rate was 80%, and the 10-year relative survival
Imatinib generics entered the market recently after the Novartis patent rate was 92%. In spite of a faster response with imatinib 800 mg,
for Gleevec expired. Several groups have reported on the efficacy and the survival difference between standard-dose and higher dose ima-
safety of generic imatinib compared to the branded one. The Polish tinib was only 3% at 5 years (less than 5%). In a multivariate analy-
Adult Leukemia Group (PALG) imatinib generics registry reported on a sis, standard-dose imatinib was equivalent to other “optimized “arms.
large series of patients who started imatinib generic (multiple manufac- Patients who reached the 6-months BCR-ABL1 transcripts [IS] <10%
turers) therapy after the diagnosis of CML (group A, n 5 99) or were milestone, had a significant survival advantage of about 6% after 10
switched to generic from branded imatinib (group B, n 5 627) and were years regardless of therapy.
observed for at least 12 months.20 Among patients treated in the front-
line setting (group A), the rates of 3-month partial cytogenetic response
1.4.4 | Dasatinib
(PCyR), 6-month CCyR, and 12-month MMR were 66%, 53%, and Dasatinib is an oral, second generation TKI that is 350 times more
50%, respectively, similar to the expected rates with branded imatinib. potent than imatinib in vitro.25–27 It also inhibits the Src family of
The rates of resistance and intolerance were also similar (26% and kinases, which may be important in blunting critical cell signaling path-
28%, respectively). Among patients who switched therapy, responses ways.28 Though initially evaluated in patients in the salvage setting, it
were maintained in the vast majority, with only 0.3% and 1% of was later compared to imatinib in frontline CML to test the possibility
patients losing a CCyR and MMR, respectively. Similar data were that frontline use of the more potent TKIs might improve outcomes.
reported from India. Among 174 patients treated with generics, The DASISION trial was a phase III randomized study comparing
response and survival rates were similar to those observed among imatinib 400 mg once daily to dasatinib 100 mg once daily in newly
1193 patients treated with the branded imatinib. Safety profiles were diagnosed patients with CML.29 The primary outcome was confirmed
similar as well.21 CCyR (cCCyR) at 12 months. A total of 519 patients were randomized
(1:1). Patients assigned to dasatinib achieved cCCyR at 12 months
1.4.3 | High-dose imatinib and imatinib-based combinations more often than those assigned to imatinib (77% versus 66%,
Other strategies for frontline therapy include using higher doses of P 5 .007). Many of the secondary endpoints of interest also favored
imatinib or combining a TKI with an additional agent, such as IFN-a. In the dasatinib arm. A five-year follow-up showed that dasatinib induced
the Tyrosine Kinase Inhibitor Optimization and Selectivity (TOPS) more rapid and deeper responses at early time points compared to ima-
study, patients were randomized to receive imatinib 400 mg once daily tinib.30 At 3 months, a higher proportion of patients treated with dasa-
22
or twice daily (800 mg). MMR rate at 12 months was the study pri- tinib achieved BCR-ABL1 transcripts [IS]  10% (84% versus 64%,
mary endpoint, with cytogenetic response and time to such responses P < .0001). Meeting this threshold in either arm predicted better
collected as secondary outcomes. Patients in the high-dose group progression-free survival and overall survival. Transformations to CML-
achieved faster CCyR and MMR, but the rates were not significantly AP or CML-BP were fewer in patients treated with dasatinib versus
different at 12 months. imatinib (4.6% versus 7.3%). However, the 5-year survival was similar
Interferon has re-emerged as an interesting therapeutic option in with dasatinib and imatinib (91%, and 90%)
CML with the advent of pegylated formulations requiring less frequent In another multicenter trial, several North American cooperative
administration and showing improved tolerability. In a phase III groups randomized patients with newly diagnosed CML-CP to either
randomized study, patients were assigned to one of four treatment dasatinib 100 mg once daily or imatinib 400 mg once daily.31 Similar to
arms: imatinib 400 mg once daily; imatinib 600 mg once daily; imatinib the results of the DASISION study, patients treated with dasatinib
400 mg once daily plus peginterferon alfa-2a; imatinib 400 mg once achieved higher rate of CCyR compared to patients receiving imatinib
daily plus subcutaneous cytarabine.23 Patients were initially assigned to (84% versus 69%, P 5 .04). There was more toxicity experienced in the
receive peginterferon alfa-2a at a dose of 90 mcg once weekly. dasatinib arm (Grades 3 or 4 adverse events 58% with dasatinib and
Because of a high rate of discontinuation due to toxicity, the dose was 35% imatinib), mostly hematologic toxicity.31
later reduced to 45 mcg once weekly. At 12 months, the rates of CCyR A third phase III randomized study, SPIRIT 2, compared imatinib
were similar among the four groups. The imatinib plus peginterferon 400 mg daily with dasatinib 100 mg daily.32 The primary endpoint
alfa-2a treated group obtained higher rates of MMR and deeper molec- of this trial is EFS at 5 years, with the rate of achievement of
ular responses, but follow-up was not sufficient to define the impact MMR, a key secondary endpoint. The interim results showed the
on long-term outcomes. 12-month MMR rates with dasatinib versus imatinib to be 58% ver-
The CML-study IV explored whether treatment with imatinib sus 43% (P < .001). The 12-month CCyR rates were 51% and 40%
400mg/day (n 5 400) could be optimized by doubling the dose respectively (P < .002). Progression rate to CML-AP was 0.7% with
(n 5 420), adding interferon (n 5 430) or cytarabine (n 5 158) or using imatinib and 0.5% with dasatinib. The progression rate to CML-BP
imatinib after interferon-failure (n 5 128).24 From July 2002 to March was 1.7% versus 1%.
|
A JH
446 JABBOUR AND KANTARJIAN

Pleural effusions occurred more frequently on dasatinib (19% ver- T AB LE 2 Frontline TKIs therapy: MDACC experience
sus <1%). Other side effects of dasatinib included myelosuppression
% Cumulative Imatinib 400 mg Imatinib 800 Nilotinib Dasatinib
(20%), and rare pulmonary hypertension (1–2%).
CCyR 85 90 99 97
1.4.5 | Lower dose dasatinib MR4.5 56 74 80 78
In early clinical trials, dasatinib was found active at lower doses with
better safety profile.33 In a later randomized trial, dasatinib 100 mg twice daily, or imatinib 400 mg orally once. The rates were 2%, 1%,
daily was found as effective as 140 mg daily, with a better safety pro- and 10% among patients with intermediate-Sokal risk and 9%, 5%, and
file.34 Furthermore investigators from the DASISION trial have 11% among patients with high-Sokal risk.
reported on the ability to maintain efficacy of dasatinib among patients In a second randomized trial with the same design that enrolled
who had their dose reduced while improving its safety profile.35 Based 267 Chinese patients, the 12-month MMR rate was 52% with nilotinib
on this rationale, we treated 75 patients with early CML-CP with dasa- compared with 28% with imatinib.40 However, the rates of both CCyR
tinib 50 mg daily.36 Ninety-five percent of patient treated achieved a (84% versus 87%) and progression-free survival (95% each) were simi-
3-month PCyR. At 12 months, the MMR and molecular response 4.5 lar at 24 months. In both arms, the estimated 2-year survival rate was
(MR4.5) rates were 83% and 74%. These rates compare favorably with 98%.
historical data with dasatinib 100 mg daily. In addition, the safety pro- While nilotinib therapy was overall well tolerated, there was an
file was favorable; only one patient discontinued therapy (subdural increased risk of accumulated vascular events on therapy. The 6-year
hematoma unlikely related) and one patient had his dose reduced to cumulative cardiovascular event rates were 9.9%, 15.9%, and 2.5%,
20 mg daily due to pleural effusion. Such strategy will have a significant among patients treated with nilotinib 300 mg twice daily, nilotinib
impact on our future practice, mainly due equivalent efficacy, better 400 mg twice daily, and imatinib 400 mg daily, respectively.39 Other
safety profile, and lower cost. notable side effects were headache and skin rashes (common 220–
30%- but mild to moderate; alleviated by dose reduction), self-limited
1.4.6 | Nilotinib
elevation of indirect bilirubin (10%), elevations of blood sugar (10%-
Nilotinib is a structural analog of imatinib. Its affinity for the ATP bind-
20%), and rare pancreatitis (1%-2%).
ing site on BCR-ABL1 is 30–50 times more in vitro.37 Like dasatinib,
nilotinib initially demonstrated the ability to induce hematologic and 1.4.7 | Bosutinib
cytogenetic responses in patients who had failed imatinib. Bosutinib is a potent dual SRC/ABL kinase inhibitor. The drug first
Similar to dasatinib, nilotinib was also compared to imatinib in a approved for adults with CML resistant and/or intolerant to prior ther-
large, international, randomized study (ENEST-nd). In ENEST-nd, two apy. Bosutinib was recently evaluated for front-line treatment CML-
doses of nilotinib (300 mg or 400 mg twice daily) were compared to CP.41 In a multinational, phase III study, 536 patients with newly diag-
38
imatinib 400 mg once daily. The primary endpoint was the rate of nosed chronic-phase CML were randomly assigned to receive 400 mg
MMR at 12 months. This endpoint was achieved at statistically signifi- of bosutinib once daily (n 5 268) or imatinib (n 5 268). The major MMR
cantly higher rates for both doses of nilotinib compared with imatinib rate at 12 months (primary end point) was significantly higher with
(44% and 43% versus 22%, P < .001). The cumulative incidence of bosutinib versus imatinib (47% versus 37%, respectively; P 5 .02), as
CCyR by 24 months was 87% with nilotinib 300 mg twice daily, 85% was the CCyR rate by 12 months (77% versus 66%, respectively;
with nilotinib 400 mg twice daily, and 77% with imatinib 400 mg daily P 5 .0075). Among treated patients, 22% of patients receiving bosuti-
(P < .001).38 nib and 27% of patients receiving imatinib discontinued treatment,
With a minimum follow-up of 5 years, the two arms of nilotinib mostly for drug-related toxicity (13% and 9%, respectively).41
demonstrated better early results compared with imatinib.39 The cumu- Grade  3 diarrhea (8% versus 0.8%) and increased ALT (19% versus
lative incidences of MMR by 60 months were 77%, 77%, and 60%, 1.5%) and AST (10% versus 2%) levels were more common with bosuti-
respectively (P < .0001%). The incidences of BCR-ABL1 transcripts nib. Cardiac and vascular toxicities were uncommon. Based on these
[IS] < 0.0032% (roughly equivalent to a 4.5 log reduction of disease) by results, bosutinib received an approval for frontline CML-CP therapy as
72 months were 54%, 52%, and 33%, respectively (P < .0001%). The of December 2017.
incidences of transformation to AP or BP were 3.9%, 2.1%, and 7.4%,
respectively (P 5 .06 and .003, respectively). The estimated 5-year EFS 1.4.8 | The MD Anderson Cancer Center (MDACC)
rates were not different, 95%, 97%, and 93%, respectively. The esti- experience (Table 2)
mated 5-year survival rates were 94%, 96%, and 92%, respectively. We published the long-term responses and outcomes of patients with
While nilotinib was superior to imatinib across all Sokal score catego- CML-CP and provided a comparison of four commonly used TKIs.32
ries in inducing higher rates of CCyR and MMR, the advantage in Unlike previous reports that compared only imatinib 400 mg with
reducing the rates of transformation was more pronounced in patients either imatinib 800 mg, dasatinib, or nilotinib in randomized trials, our
with intermediate- and high-Sokal risk CML. The rates of transforma- study provided a comparative analysis of the four TKIs after a long
tions were 1% and 1% and 0% among patients with low-Sokal risk follow-up. The analysis included 482 patients treated (July 2000 to
treated with nilotinib 300 mg orally twice daily, nilotinib 400 mg orally September 2013) in consecutive or parallel clinical trials, with imatinib
A JH |
JABBOUR AND KANTARJIAN 447

400 mg daily (n 5 68), imatinib 800 mg daily (n 5 200), dasatinib 50 mg with CML (e.g., age <50 years) versus older patients, may be
twice daily or 100 mg daily (n 5 106), or nilotinib 400 mg twice daily entertained.
(n 5 108). More patients receiving imatinib 800 mg or second-
generation TKIs (ie, dasatinib or nilotinib) achieved CcyR: 87% for ima-
2 | SELECTING A FRONTLINE THERAPY
tinib 400 mg; 90% for imatinib 800 mg; 96% for dasatinib; and 93% for
nilotinib. The MMR rates were 76%, 86%, 90%, and 91%, respectively.
2.1 | Patient’s age and comorbidities and TKI toxicity
The 45 log or higher reduction in BCR-ABL1 transcripts (MR4.5)
profile
response rates were 57%, 74%, 71%, and 71%, respectively. These
findings were consistent over time (3–60 months); where at any time While multiple TKIs are available for patients with newly diagnosed
point, imatinib induced lower rates of cytogenetic and molecular CML-CP, each has a distinct toxicity profile to consider when deciding
responses. These landmark assessments contrast with previous reports on a therapy. Most TKIs are reasonably well-tolerated with adequate
where results are reported cumulatively (“by” not “at”) and can be mis- monitoring and supportive care. For patients at risk of developing pleu-
leading. The 5-year event-free survival significantly differed between ral effusions (existing lung injuries), TKIs other than dasatinib should be
imatinib 400 mg and the other TKI groups (imatinib 800 mg P 5 .029, selected. This might be relevant for patients with a history of lung dis-
dasatinib P 5 .003, nilotinib P 5 .031). However, there was no signifi- ease (e.g., chronic obstructive pulmonary disease), cardiac disease (e.g.,
cant difference in the 5-year failure-free survival (P 5 .32, P 5 .075, congestive heart failure), or uncontrolled hypertension. Pulmonary arte-
P 5 .332), transformation-free survival (P 5 .053, P 5 .038, P 5 .493), or rial hypertension (PAH) is also a rare yet important complication of
overall survival (P 5 .563, P 5 .162, P 5 .981). We also compared event- dasatinib,44 and patients with preexisting PAH may be considered for
free and overall survival according to whether or not MMR or MR4.5 alternative TKIs in the frontline setting. Dasatinib also inhibits platelets
response was achieved in addition to CcyR. As expected, in patients function,45 and patients taking concomitant anticoagulants may be at
who achieved CcyR, no difference in outcomes was observed regard- an increased risk of hemorrhagic complications.46
less of whether MMR or MR4.5 response was achieved.42 Nilotinib has been associated with hyperglycemia. Caution should
Current guidelines recommend any of the three TKIs, imatinib, das- be exercised in patients with uncontrolled diabetes when initiating
tinib or nilotinib as good therapeutic options with a category 1 recom- therapy, and avoided or prescribed with caution in patients with diabe-
mendation for initial treatment of CML-CP.43 Second generation TKIs tes or history of pancreatitis. During preclinical development, nilotinib
produced higher rate of early optimal responses, but so far have no was shown to potentially prolong the QT interval, and parameters
impact on long-term survival (probably because of available effective were put in place to monitor for this complication after the drug was
salvage therapies). The main advantage of second generation TKI is approved; potassium and magnesium should be repleted to appropriate
obtained among patients with high-risk disease; a relevant decrease in serum levels before starting nilotinib or determining an individual
the rate of transformation to AP and a BP was achieved with nilotinib patient’s QT interval. Patients should always be counseled to take nilo-
and dasatinib. As such, second generation TKIs in the frontline setting tinib in a fasting state to avoid excess drug exposure. Nilotinib has also
could be reserved to patients with higher risk disease. Higher dose ima- been associated with vasospastic and vaso-occlusive vascular events,
tinib and combination approaches in the frontline setting are investiga- such as ischemic heart disease, ischemic cerebrovascular events, and
tional due to conflicting results of different studies to date. These peripheral artery occlusive disease (PAOD).47–50 In the 6-year follow-
strategies are also not benign interventions, as they add to the eco- up on the ENEST-nd trial,39 approximately 10% of patients experienced
nomic and toxicity burdens of the overall treatment plan. Allo-SCT or vascular events. Nilotinib use should be limited in patients with risk fac-
other chemotherapy agents are not recommended as upfront treat- tors such as diabetes mellitus or coronary or cerebrovascular artery dis-
ments for CML-CP, given the excellent outcomes and long term sur- ease. Avoiding nilotinib in patients with significant past vascular
vival achieved with the TKIs. An exception may be in emerging nations histories is warranted with the availability of other viable options. Nilo-
where allo-SCT is a one-time procedure costing $14–20 000, accessi- tinib is also given twice daily on empty stomach; this may result in com-
ble to most patients. Conversely, generic imatinib in such geographies pliance issues.
(e.g., India) costs only less than $400 per year of therapy. At MD Imatinib causes bothersome quality-of-life side-effects including
Anderson Cancer Center (MDACC), patients are currently offered ther- weight gain, fatigue, peripheral and periorbital edema, bone and muscle
apy with dasatinib 50 mg daily. Lower dose dasatinib showed equiva- aches, nausea and others. However, most are mild to moderate. Less
lent efficacy compared with standard dose in addition to a better than 5%-10% experience elevations in creatinine with long-term
safety profile. This cost-effective strategy may allow achievement of an therapy.
optimal response at a lower cost than standard-dose dasatinib, with a Finally, the patient’s age plays an important role in the treatment
potential molecular cure of CML (higher rate of complete molecular decision. Patients younger than 50 years are expected to live 301
responses; see below). Because of the higher rates of durable complete more years. Therefore, inducing a durable CMR may potentially lead to
molecular responses with second generation TKIs (which could lead to therapy discontinuation. Second generation TKIs induce a significantly
discontinuation of TKI therapy and potential molecular cures; discussed higher rate of CMR compared with imatinib. The issue of durable CMR
later), considerations of second generation TKIs in younger patients and potential therapy discontinuation plays a less important role in
|
A JH
448 JABBOUR AND KANTARJIAN

elderly patients where the expected survival is shorter and discontinu- have a lower likelihood of achieving the early milestones of CCyR and
ing therapy is less relevant. MMR and, particularly, higher chances of disease transformation to
AP-CML or BP-CML.
2.2 | Cost Any of the the TKIs currently approved for frontline CML therapy
may be selected. These include imatinib, dasatinib, or nilotinib. While
The cost of cancer drugs has risen exponentially over the past dec-
dasatinib and nilotinib have demonstrated superiority over imatinib
ade.51 All anticancer agents approved in the recent years were priced
when early surrogate markers are used, imatinib is still highly effective
at over $120 000 annually.51 When an international group of CML
in a large number of patients with CML. At MDACC, when choosing an
experts called attention to the high prices of TKIs, the cost history of
agent, we consider issues such as comorbidities, patient’s age, adverse
imatinib was used to illustrate the problem with the high cancer drug
event profile, risk stratification score, and cost. Kinase domain mutation
prices.52 When first approved in the United States, the annual price for
profile plays no role in selecting an initial TKI, but becomes relevant in
imatinib was less than $30 000, the price set to make the development
the relapse setting.
and commercialization of imatinib profitable based on cost of research,
population at risk, competitive market, estimated profits, and estimated
3 | MONITORING TREATMENT RESPONSE:
shorter-than-realized patient survival. Patients compliant with their
SURROGATE ENDPOINTS AND MILESTONES
treatment regimen now live a “normal” lifespan and remain on imatinib
indefinitely.53,54 Paradoxically, with a higher number of patients and a Because patients with CML on TKI therapy are expected to live for a
longer duration of therapy, the annual price of imatinib had quadrupled long period of time, surrogate markers of outcome are important. In
to $132 000. This price is in the same range as dasatinib and nilotinib, general, achieving a deeper response faster has been associated with
both priced above $130 000/year of therapy. improved outcome, though the result of molecular testing is dependent
In 2016, generic formulations of imatinib became available, on the laboratory and their techniques. Due to advances in technology,
although at a higher price than expected. This price should eventually there are less invasive tests available for treatment monitoring than tra-
fall dramatically below $5–8000/year (as in Canada; the annual price of ditional bone marrow examinations (except when changing TKI; or in
generics in India is only $400). Physicians will then have to assess the unusual situations like unexpected myelosuppression to exclude trans-
“treatment value” of second generation TKI (dasatinib or nilotinib) in formation or the development of myelodysplastic syndrome or other
the frontline setting against the generic imatinib in relation to benefits marrow conditions).
versus cost. Second generation TKIs may be offered for patients with
high-risk disease, while imatinib and/or its generic formulations will be 3.1 | Important points for monitoring and determining
offered for patients with low-risk disease. Experts recently assessed,
treatment failure
following a Markov model, the most cost-effective strategy for treating
newly diagnosed CML-CP after imatinib loses patent exclusivity.55 At baseline, all patients should undergo a bone marrow examination to

They found that initiating imatinib as frontline treatment and treating establish the diagnosis, assess percentage of blasts and basophils, and

in a stepwise approach, compared to physician choice, costs less and perform cytogenetic analysis to confirm the presence of the Philadel-

offer clinically-equivalent utility. In this analysis, the stepwise therapy phia chromosome and to exclude clonal evolution, particularly i(17)

was found to have an incremental cost-effectiveness ratio (ICER) of (q10) 27/del7q, and 3q26.2 rearrangements, associated with a rela-

$227 136/Quality-adjusted life years. The ICER was favorable for step- tively poor prognosis.16 The current recommendation that patients

wise therapy for each Sokal risk group. Furthermore, the efficacy and have a follow up bone marrow study at 3, 6, and 12 months after start-

safety of generic imatinib was compared to the patented drug (previ- ing therapy may no longer be necessary.58 An alternative method to
determine cytogenetic response is with the use of FISH and PCR on
ously discussed). Finally, lower dose dasatinib may offer the ultimate
peripheral blood. If a patient is responding optimally, and the FISH
solution with at least equivalent efficacy compared to second genera-
study is negative at 6 or 12 months and or BCR-ABL1 transcripts [IS]
tion TKIs but at a significant lower cost (50% of branded dasatinb
<1%, it may be reasonable to omit marrow exams, as the patient is
100 mg daily), a price comparable to generic formulation of imatinib.
likely to be in CCyR.59,60
For patients in durable CCyR receiving TKI therapy, periodic
2.3 | Disease characteristics
molecular monitoring using RT-Q-PCR is acceptable and useful, but
For patients with CML-CP it is common to use one of the available risk may lead to erroneous changes in a well-tolerated and effective treat-
stratification scores, such as Sokal56 or Hasford,57 to help predict out- ment due to discordant results between laboratories or even within the
comes. Patients with low-risk disease are expected to have optimal same laboratory. One strategy to minimize this is to use interphase
responses whith imatinib, dasatinib, or nilotinib. However, selecting a FISH as a complementary diagnostic test along with the molecular test
second generation TKI as frontline therapy (i.e., dasatinib or nilotinib) to detect possible false positive or negative results generated by either
based upon the Sokal or Hasford scores was proven more beneficial in assay.60 For patients in CCyR, the achievement and maintenance of a
patients with intermediate or high risk disease, as shown in the DASI- MMR is of debatable significance. Several studies evaluating patients
SION and ENEST-nd trials.29,30,38,39 Patients with higher risk disease receiving imatinib or second generation TKIs have found that patients
A JH |
JABBOUR AND KANTARJIAN 449

T AB LE 3 Percentage of survival by early molecular response T AB LE 5 MDACC criteria for response/failure and change of
therapy
Study Q-PCR < 10% Q-PCR > 10%
Time (months) Imatinib Second generation TKIs
UK (8-year) 93 54
3–6 MCyR; PCR10% [IS] CCyR; PCR  1% [IS]
MDACC (10-year) 98 94
12 CCyR; PCR  1% [IS] CCyR; PCR  1% [IS]
ENEST-nd 97 87
Later CCyR; PCR  1% [IS] CCyR; PCR  1% [IS]
DASISION 97 86

reasonable to change the TKI from imatinib to second generation TKI


in CCyR have similar survival regardless of whether or not they
for BCR-ABL1 transcripts [IS] >10% after 6 months.66 However, a simi-
achieved MMR.61,62
lar situation in a patient on second generation TKI (dasatinib, nilotinib)
Early molecular response has been shown to have strong prognos-
does not necessarily imply consideration of allo-SCT.
tic value (Table 3). This has been shown with each of the 3 TKIs used
in the frontline setting. A BCR-ABL1 transcripts [IS] < 10% at 3 months
3.2 | When to switch therapy
separates patients into high and low risk categories for long term out-
comes (i.e., progression, survival).30,39,63,64 The important question is: The aim of therapy in CML should be the achievement of CCyR within
how should we advise a patient who does not meet the 3-month 12 months and to continue to be in CCyR at any time beyond 12
benchmark? One option is to switch TKIs early, but there are no data months, especially with standard dose imatinib therapy. For second
indicating this will alter long-term outcome. Several experts suggested generation TKIs, CCyR may need to be achieved sooner for an optimal
that a follow-up measurement at 6 months will help define patients outcome, for example, within 6 months.68 Patients who do not achieve
clearly in need of a change in therapy.58 This strategy has been retro- a complete hematologic response by 3 months should be considered
65–67
spectively analyzed by several large groups with conflicting results. for a change in therapy (Tables 4 and 5).
The results of two independent study groups have suggested that all One question is whether to change therapy at 3 months based on
patients with BCR-ABL1 transcripts > 10% at 3 months do not neces- the level of BCR-ABL1 transcripts [IS]. For patients on imatinib therapy,
sarily have an inferior outcome.66,67 Patients who continued on therapy if the 3-month transcript level is greater than 10%, we suggest an
and achieved transcripts levels less than 10% by 6 months had the approach similar to the ELN guidelines:58 perform serial molecular
same long-term favorable outcome as those with optimal molecular monitoring between 3 and 6 months to determine the response pre-
responses at 3 months. Patients with BCR-ABL1 transcripts [IS] >10% cisely. If patients still have greater than 10% BCR-ABL1 transcripts [IS]
after 6 months have a worse outcome. Still the 4-year survival with 6- at 6 months, the chances of attaining CCyR are low, and a change of
month BCR-ABL1 transcripts [IS] >10% versus <10% are 100% versus therapy might be in indicated. Furthermore, a 3-month value of 10%
74%. The CML-study IV has established a 6-month BCR-ABL1 tran- [IS] may not be accurate: in a recent report in 1 sample collected at 3
scripts [IS] <10% response as an optimal milestone. Patients who months and tested 96 times, the mean value was 11% (range, 5%-
reached the 6-monts milestone had a significant survival advantage of 16%), with only 31% of tests being 10%.69 This approach also applies
about 6% after 10 years regardless of therapy.24 Thus, it may be to patients on second generation TKIs, because, as mentioned earlier,

T AB LE 4 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% Ph1 36 – 95% Ph1 > 95%


And/or And/or And/or
BCR-ABL  10% BCR-ABL > 10% No CHR

6 months Ph1 0% Ph1 1 – 35% Ph1 > 35%


And/or And/or And/or
BCR-ABL < 1% BCR-ABL 1 – 10% 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 (-7 or 7q-) Loss of CHR

Loss or CCyR

Confirmed loss of MMR

CCA in Ph1 cells


|
A JH
450 JABBOUR AND KANTARJIAN

T AB LE 6 Important response categories in CML 4 | MANAGEMENT OF TKI RESISTANCE


Response Translates into:
A remaining problem with the widespread use of all commercially avail-
BCR-ABL1  10% Significantly improved survival
at 6 months; CCyR later able TKIs is increased drug resistance. A common mechanism of resist-
ance involves point mutations in the kinase domain of BCR-ABL1,
MMR Modest improvement in EFS;
possible longer duration CCyR; no which impairs the activity of the available TKIs. Second generation TKIs
survival benefit overcome most of the mutations that confer resistance to imatinib,
CMR Possibility of therapy discontinua- though novel mutations rendering the leukemia resistant to dasatinib
tion
and/or nilotinib have emerged. One important mutation, T315I known
as the “gatekeeper” mutation, displays resistance to all currently avail-
early switching has not yet shown to influence long-term outcome.70 able TKIs except ponatinib.
Another study randomized patients in CCyR on imatinib for at least 2 Before defining a patient as having TKI resistance and modifying
years to continue imatinib or switch to nilotinib.71 Switching to nilotinib therapy, treatment compliance and drug-drug interactions should be
induced deeper molecular responses, but did not translate into an assessed. Rates of imatinib adherence have been estimated to range
improvement in progression-free survival or in other meaningful from 75% to 90%, and lower adherence rates correlated with worse
outcomes. outcome.72–74 In a study of 87 patients with CML-CP treated with ima-
Patients who meet all the relevant benchmarks in the first 12
tinib 400mg daily, an adherence rate of 90% or less resulted in MMR
months are monitored periodically using FISH and molecular testing
rate of 28% compared with 94% with greater than 90% adherence rate
(molecular testing only if BCR-ABL1 [IS] transcripts consistently below
(P < .001).72 CMR rates were 0% versus 44% (P 5 .002), and no molec-
0.1%). If there are clear signs of possible failure, patients should
ular responses were observed when adherence rates were 80% or
undergo a bone marrow examination with cytogenetic studies, and
lower. Lower adherence rates have been described in younger patients,
molecular testing including analysis for mutations. Any degree of cyto-
those with adverse effects of therapy, and those who have required
genetic relapse calls for a change in therapy. Fluctuating molecular lev-
dose escalations.72
els during continuous CCyR would only prompt closer monitoring and
a compliance assessment.
In several studies, the achievement of a CCyR (Ph-positive meta- 4.1 | Second and third generation TKIs
phases 0%; BCR-ABL1 transcripts [IS]  1%) at 12 months or later on
Before their approval to treat first-line CML-CP, both nilotinib and
TKI therapy was associated with significant survival benefit compared
dasatinib were approved for use in second-line CML-CP following prior
with achievement of lesser degrees of response. Achievement of CCyR
therapy including imatinib.75,76 Clinical studies of second-line and third
is the primary endpoint of TKI therapy. Achievement of BCR-ABL1 tran-
line TKIs are summarized in Table 7. Based on these studies, several
scripts [IS]  0.1% (MMR) was associated with modest improvements in
noteworthy ideas have emerged. First, second-line treatment with nilo-
event-free survival rates, possible longer durations of CCyR, but not
tinib, dasatinib, or bosutinib can yield high rates of response in patients
with a survival benefit. The achievement of CMR (nonmeasurable BCR-
who have inadequate response to imatinib, including high rates of
ABL1 transcripts) offers the possibility of treatment discontinuation in
MMR. Second, dose escalation of imatinib can improve response rates
clinical trials only (Table 6). Lack of achievement of MMR or of CMR
in patients with inadequate response to standard-dose imatinib,77 but
should not be interpreted as a need to change TKI therapy or to consider
switching to second-line TKI is more effective.78 Several studies that
allo-SCT. Response assessments at earlier times on frontline TKI therapy
evaluated second-line nilotinib79,80, dasatinib78,80, or bosutinib81, and
(3–6 months) have shown better outcomes with achievement of a major
cytogenetic response by 3 to 6 months on imatinib therapy (Ph-positive high-dose imatinib (400 mg BID) have demonstrated significantly

metaphases  35%; BCR-ABL1 transcripts [IS]  10%). While this is higher rates of CHR, CCyR, and MMR with the newer TKIs than with

interpreted to mean that a change to second TKI therapy may be consid- high-dose imatinib. Moreover, PFS in these studies was better with the

ered if such outcome is not obtained, no studies have shown that chang- newer TKIs than with high-dose imatinib. Earlier switch to second-line
ing therapy from imatinib to second TKIs has improved patients’ TKI may be more effective than later switch. In the TIDEL-II study,
outcomes. When nilotinib or dasatinib are used in front-line therapy, patients who had suboptimal response to imatinib and were switched
achievement of complete cytogenetic response by 3–6 months of TKIs to nilotinib had a higher rate of CMR at 12 months than patients who
therapy has been associated with improved outcomes. had dose escalation of imatinib prior to being switched to nilotinib.82 In
At MDACC, the major treatment milestones are at 6 and 12 a retrospective pooled analysis of three clinical studies of second-line
months. Patients with BCR-ABL1 transcripts [IS] > 10% at 6 months, or dasatinib for patients resistant to or intolerant of imatinib, patients
without CCyR (BCR-ABL1 transcripts [IS]  1%) at 12 months, or with who were switched to dasatinib after the loss of MCyR (early interven-
loss of response at any time are candidates for a switch of therapy. The tion group) had higher rates of CHR, CCyR, and MMR, as well as 24-
choice of TKIs is based on the mutation profiles and patients comorbid- month EFS, TFS, and OS, than patients who were switched after the
ities. We do not consider a change of TKI therapy in patients in CCyR loss of both MCyR and CHR (late intervention group).83 Although this
but without MMR. analysis included studies with distinct study designs and various dosing
A JH |
JABBOUR AND KANTARJIAN 451

T AB LE 7 Summary of important phase II trials of 2nd and 3rd generation TKIs after prior TKI failure

Percent response
Dasatinib Nilotinib Bosutinib Ponatinib
CP AP MyBP LyBP CP AP MyBP LyBP CP AP BP CP AP BP
N 5 167 N 5 174 N 5 109 N 5 48 N 5 321 N 5 137 N 5 105 N 5 31 N 5 200 N 5 51 N 5 38 N 5 270 N 5 79 N 5 94

Median 24 14 12 12 48 9 3 3 24 6 3 36 36 36
follow-up
(mo)

% Resistant 74 93 91 88 70 80 82 82 69 NR NR 75 96 -
to imatinib

% Hematologic - 79 50 40 94 56 22 19 85 54 36 NR NR NR
Response

CHR 92 45 27 29 76 31 11 13 81 54 36 NR Ma Ma
HR: 57 HR: 34

NEL – 19 7 6 – 12 1 0 – 0 NR - NR NR

% Cytogenetic - 44 36 52 NR NR NR NR - NR NR - NR NR
Response

Complete 50 32 26 46 45 20 29 32 46 27 35 53 55 36

Partial 13 7 7 6 14 12 10 16 12 20 18 30 NR NR

% Survival At 6 years At 4 years At 4 years At 5 years

91 82 50 50 87 67 42 42 98 60 50 73 59 9

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.

schedules of dasatinib, the essential finding was that earlier switch to 45/64 (70%) patients with a T315I mutation. Patients responded more
dasatinib was associated with better outcomes. favorably if they had received fewer TKIs. After a median follow up of
Bosutinib was initially studied in patients who had resistance to or 5 years, 60% of patients achieved MCyR (primary endpoint) at any
intolerance of imatinib.81 After a dose escalation period, 500 mg once time; 82% of those remained in MCyR at 5 years.87 Furthermore, 40%
daily was selected as the phase II dose, with the potential for dose of patients achieved a MMR or better. The 5-years survival rate was
escalation to 600 mg once daily for patients not meeting predefined 73%.87 Arterial occlusive events occurred in 31% of patients (26% seri-
benchmarks. A total of 288 patients were enrolled in the pivotal phase ous). The most common all-grade treatment-emergent adverse events
II trial; more than two thirds had imatinib-resistant disease. The primary occurring in  40% of patients with CML-CP were abdominal pain
endpoint of MCyR at 6 months was achieved in 31%; 41% achieved a (46%), rash (46%), thrombocytopenia (45%), headache (43%), constipa-
CCyR. Bosutinib appeared to retain activity across most known muta- tion (41%), and dry skin (41%).87 Other notable toxicities include severe
tions that confer imatinib resistance, except for T315I. Responses were skin rashes (4%-7%), pancreatitis (7%), and severe hypertension (20%).
independent of whether patients had resistance to or intolerance of As of early 2014, ponatinib labeling included a revised warning
imatinib. The most common toxicities were diarrhea, nausea, vomiting, regarding the risk of thrombotic events (13% per year), vascular occlu-
and rash. Diarrhea occurred in 84% of patients, with 9% experiencing sions, heart failure, and hepatotoxicity, revised dosing information and
grade 3 diarrhea (there were no grade 4 events documented). Other indications limited to adults with T315I mutation and those for whom
notable adverse events included myelosuppression and liver function no other TKI is indicated.88 Vascular occlusive adverse events were
test abnormalities. more frequent with increasing age and in patients with a prior history
Ponatinib is a third generation TKI, and the first TKI in class to of ischemia, hypertension, diabetes, or hyperlipidemia.87 Factors associ-
exhibit activity against CML with T315I mutation.84 It is 500 times as ated with an increased risk of vascular occlusion events include older
potent than imatinib at inhibiting BCR-ABL1.85 The approval of ponati- age, higher dose, history of myocardial infarction or prior vascular
nib was based on the phase II PACE trial, where 449 patients with events, and longer duration of CML.87 Studies assessing lower dose-
heavily pretreated CML or Ph-positive acute lymphoblastic leukemia schedules of ponatinib are ongoing. In the community practice, it may
86
(ALL) were treated. Patients were considered for this trial if they had be safer to use ponatinib 30 mg daily (and lower the dose to 15 mg
resistance to or intolerance of dasatinib or nilotinib, or if they had CML daily for toxicities) rather than the FDA approved dose of 45 mg daily.
with T315I mutation. The dose of ponatinib was 45 mg once daily, and ABL001 (asciminib) is a small molecule allosteric BCR-ABL1 kinase
patients were stratified by the disease phase, and the presence or inhibitor that occupies the myristoyl pocket, leading to a kinase autoin-
absence of a T315I mutation. Of the 267 patients who received ponati- hibition conformation.89 ABL001 was designed to inhibit BCR-ABL1 in
nib in CML-CP, 56% achieved a MCyR by 12 months, which included a non-ATP-competitive manner to maintain activity against BCR-ABL1
|
A JH
452 JABBOUR AND KANTARJIAN

mutations that confer resistance to TKIs. Preclinically, combined with patient’s preexisting medical conditions. Mutational analysis are
ATP-competitive TKIs, ABL001 eliminates early leukemic progenitors required in patients who are failing imatinib or second generation TKIs,
and diminishes emergence of resistant clones.89 One hundred and one or those who progress to AP/BP. Baseline mutational analysis on
patients with relapsed refractory CML and Philadelphia-positive acute patients with newly diagnosed CML-CP are not done, as this has not
90
lymphoblastic leukemia were treated in a phase I trial. There were 5 proven to predict treatment outcome.
dose limiting toxicities (DLTs): 2 Grade 3 lipase elevations (40 mg BID At MDACC, post imatinib failure, the choice of second or third
and 200 mg QD), 1 Grade 2 arthralgia (80 mg BID), 1 acute coronary generation TKI is based on the disease phase, mutation profile, and
syndrome (150 mg BID), and 1 Grade 3 bronchospasm (200 mg BID). A patient’s comorbidities. In advanced phases, we favor a combination of
dose of 40 mg BID was recommended for CML-CP patients. MMR was chemotherapy and TKI (mainly ponatinib or dasatinib). In patients with
achieved at 13% and 38% by 6 and 12 months of therapy, respectively. CML-CP with T315I mutation, ponatinib will be the first choice fol-
Eighty percent (8/10) of patients with more than 35% Philadelphia- lowed by allo-SCT if a donor is available and an optimal response is not
positive metaphases achieved CCyR by 6 months. Further development achieved. Outside the context of T315I mutations, the type of muta-
of the drug is ongoing. tion will dictate the choice of therapy. Of note, patients with com-
pound mutations may not respond well to second generation TKI. A

4.2 | How to select a second or third line option close monitoring should be offered.96 If an optimal response is not
achieved, a switch of therapy to a third generation TKI and/or allo-SCT
At the time of treatment failure, patients should undergo bone marrow is warranted. In patients with no mutation or a mutation sensitive to all
examination to allow proper determination of the CML phase and doc- second generation TKIs, the choice will be based on comorbidities.
umentation of any clonal evolution. All patients should have CML cells Patients with vascular risk factors and metabolic dysfunctions are not
tested for BCR-ABL1 kinase domain mutations, as this will help guide candidates for nilotinib. Patients with lung injuries are not candidates
the selection of the TKI.91,92 When selecting between dasatinib, bosuti- for dasatinib. Bosutinib may be the best choice for patients with car-
nib, and nilotinib, in vitro and in vivo data have identified distinct muta- diac problems and arrhythmias.
tions that exhibit decreased sensitivity to each of the agents.93,94
Physician may favor dasatinib or bosutinib if the patient has the follow-
ing mutations: Y253H, E255K/V, or F359C/V. Alternatively, nilotinib 4.3 | Allogeneic stem cell transplantation (allo-SCT)
may be favored in the presence of the V299L and F317L mutations. The number of patients undergoing allo-SCT for CML-CP has decreased
For patients lacking these mutations, the choice should be on preexist- significantly since TKIs were introduced, but will start to increase again
ing conditions, toxicity profiles, and cost. as the prevalence of CML increases, as about 2% of patients become
Bosutinib can be used for patients with most known mutations that resistant to many TKIs every year and require allo-SCT. Allo-SCT has a
lead to imatinib failure.81 Like dasatinib and nilotinib, bosutinib is not more important role when patients evolve into AP/BP (see below). Allo-
activie against T315I. Bosutinib may be a reasonable choice for patients SCT remains an important therapeutic option for patients in CML-CP
who fail imatinib who are not good candidates for dasatinib or nilotinib. whose CML has progressed after at least 2 TKIs, and those who poten-
Bosutinib has a relatively distinct toxicity profile from the other TKIs, tially harbore the T315I mutation (after a trial of ponatinib therapy).97
with the predominant problem being diarrhea and other gastrointestinal Prior exposure to TKIs does not impact transplant outcome negatively.
complaints. Recently, an analysis was conducted to closely characterize Patients referred to transplant may have a better outcome if entering
the toxicity of bosutinib and the management strategy.95 Diarrhea was the transplant with a better response to TKIs (lower CML burden).98
documented in 82% of patients (n 5 570), most being grades 1/2 in Finally, allo-SCT cost versus TKIs cost and availability should be
severity. Myelosuppression and liver function test abnormalities were considered. Allo-SCT, a curative one-time procedure costs $500 000 in
also common. With appropriate supportive care, monitoring, and dose the United States, but only $20 000 in developing nations. Allo-SCT
reductions for persistent diarrhea, most patients are able to continue should not be offered as frontline therapy unless the TKI prices are
therapy with periodic dose interruptions or adjustment. prohibitive to a nation’s health care budget or not accessible to most.
Ponatinib should be considered the agent of choice in patient with In India, the annual price of generic imatinib is $400. Considering the
CML and T315I mutation, and in instances where other TKIs are not median age of patients with CML and the projected survival of 301
indicated (patients with resistance to at least one second generation years ($400 3 30 years5 $12 000), imatinib therapy remains the best
TKI). No other commercially available TKIs have activity against the frontline options. In nations where TKIs are not commonly accessible,
T315I mutation. The risk for serious toxicities (vaso-occlusive disease, it may be better to perform allo-SCT on 100% of patients and cure
pancreatitis, hypertension, severe skin rashes) and of thrombotic events 60%, rather than having TKIs available to 10% (and functional cure
with ponatinib is significant, but the benefits outweigh the risks for most 10%). In contrast to the frontline setting, in the salvage setting the
patients with a T315I mutation, as there are no other options for disease value of allo-SCT versus second/third generation TKI is more pro-
control. These side effects are lower with ponatinib 15–30 mg daily. nounced. Allo-SCT in first salvage as cure costs $20 000 versus $120
Second and third generation TKIs have not been compared head- 000–170 000/year for second-third generation TKIs. In these circum-
to-head. Selection of one or the other is based on the side-effect pro- stances, allo-SCT could be offered as first salvage option in nations
files, mutations profile, drug interactions, compliance issues and the with financial constraints.99,100
A JH |
JABBOUR AND KANTARJIAN 453

5 | TREATMENT DURATION AND TKIs. Although the use of second-generation TKIs certainly increases
DISCONTINUATION the likelihood of obtaining a sufficiently deep molecular response to
consider TKI discontinuation, the use of these second-generation
Several studies have evaluated whether TKIs can be safely discontinued agents comes at significant financial cost. It has been estimated that
in patients who have achieved long-term deep molecular responses. In the use of frontline second-generation TKIs results in an increased cost
the Stop Imatinib (STIM) study, 100 patients who had achieved complete of $800 000 per quality-adjusted life-year, compared to generic imati-
molecular response (i.e., >5-log reduction in BCR-ABL1 and ABL1 levels nib.107,108 The financial burden associated with frontline use of
and undetectable transcripts on quantitative RT-PCR) after >2 years of second-generation TKIs should therefore be carefully weighed against
imatinib treatment discontinued TKI therapy and were followed prospec- the marginal improvement in cure fraction compared to imatinib.
tively.101 Forty-two patients (61%) experienced molecular relapse, all but Although it is encouraging that some patients may be functionally
2 within 6 months of TKI discontinuation. After reintroduction of imati- cured with long-term TKI therapy, alternative strategies should be
nib, 26 patients again achieved CMR; the other 16 patients had explored to increase the cure fraction of patients with CML. These
decreases in BCR-ABL1 levels. Similar results were observed in the com- approaches should focus on both inducing deeper molecular responses
parably designed TWISTER study.102 Among the 40 patients enrolled, and targeting the CML stem cell. Despite the presence of a constitutively
the actuarial estimate of stable treatment-free remission (TFR) was 47% active BCR-ABL1 kinase, these leukemic stem cells may be quiescent,
at 2 years; all patients were sensitive to imatinib re-treatment. which renders them relatively resistant to TKI therapy.109 The presence
Higher TFR rates can be achieved with second-generation TKIs of resistant leukemic stem cells, in part, explains why a substantial pro-
due to the relatively deeper and more sustained molecular responses
portion of patients rapidly relapse when TKI therapy is stopped. Future
achieved with these agents compared to imaitnib.29,38 In the ENEST-
TKI-independent therapeutic approaches that target the leukemic stem
freedom study, TKI discontinuation was evaluated in patients with
cell will be imperative to increasing the number of patients with CML
chronic phase CML who had achieved at least MR4.5 after 2 years of
who can be cured. The addition of pegylated interferon-a2b to imatinib
frontline nilotinib therapy, followed by 1 year of nilotinib consolidation.
or dasatinib results in promising deep molecular responses that compare
Nilotinib was reinitiated in patients who lost MMR. Among 190
favorably to those observed with either TKI alone, suggesting this may
patients who entered the TFR phase, 98 patients (51.6%) remained in
be a useful approach to increase the potential for TFR.111,112 There are
MMR or better at 2 years after stopping nilotinib. Similar to the experi-
also emerging data to suggest that the combination of Bcl-2 inhibitors
ence with imatinib, the vast majority of patients were able to regain a
such as venetoclax in combination with TKIs can enhance cytotoxicity
deep molecular response after nilotinib reinitiation, although it is nota-
and deplete CML stem cells.113,114 A study combining dasatinib 50 mg
ble 11.6% of patients were unable to regain MR4.5 after resuming nilo-
daily with venetoclax in frontline CML-CP is currently ongoing. Studies
tinib. In the STOP 2G-TKI study, patients receiving first-line or
of several other agents in combination with TKIs are currently ongoing,
subsequent dasatinib or nilotinib for 3 years and who were in MR4.5
including hypomethylating agents,115 JAK2 inhibitors,116 and immune-
with undetectable BCR-ABL1 transcripts for the preceding 2 years were
based therapies, including anti-PD-1 monoclonal antibodies and dendritic
eligible for TKI discontinuation.103 The 2-year TFR rate was 53.6%, and
cell vaccines.117 The hope is that these novel strategies will lead to
among patients who experienced molecular relapse and had to reinitia-
deeper and more durable responses than TKI therapy alone and will
tion TKI therapy, all patients were able to achieve MR4.5 again.
therefore facilitate functional cure in a higher proportion of patients with
Similar results were observed in the EURO-SKI trial, the largest
CML than does single-agent TKI therapy.
study to date of TKI discontinuation in CML.104 In this study of 821
TKIs discontinuation studies in patients with durable CMR demon-
patients with CML treated with frontline imatinib, nilotinib or dasatinib,
strate that stopping TKI therapy is feasible, and some patients may be
who had achieved at least MR4, and who subsequently stopped TKI
cured. At MDACC, therapy discontinuation is offered only for patients
therapy, the molecular recurrence-free survival at 2 years was 52%.
Among 321 (86%) patients who lost their MMR and were restarted on in chronic phase with a quantifiable transcript, who have been on TKI

TKI, 81% regained a molecular response (MMR/MR4). In a multivariate for at least 6 years, have achieved a sustained CMR for at least 3–4

analysis, duration of MR4 for 31 years was the only significant factor years, and in whom a close follow-up can be performed (Table 8).

for persistent deep molecular response after stopping therapy. Addi- Going forward, it is important to continue to investigate the possibility

tional factors include duration of TKI therapy for 61 years. 105 of safe treatment cessation. Measures of quality of life and adverse
The relatively higher TFR rates observed with patients initially event avoidance should be studied in subsequent trials. The economic
treated with second-generation TKIs suggests that second-generation impact of long-term discontinuation of imatinib is substantial.
TKIs may be preferred as initial therapy for patients in whom eventual
TKI discontinuation may be particularly valued (e.g., younger patients 6 | ADVANCED STAGE CML
106
expected to live more year benefitting from TKI discontinuation).
However, when factoring in the number of patients who achieve deep Patients with CML-AP or CML-BP may receive initial therapy with TKIs
enough molecular responses to be candidates for stopping TKI therapy, (newer generation TKIs like dasatinib or ponatinib preferred over imati-
the incidence of molecular cure remains relatively low, ranging from nib) to reduce the CML burden, and be considered for early allo-
approximately 15% with imatinib to 25%-30% with second-generation SCT.118–122 Response rates with combinations of TKIs and
|
A JH
454 JABBOUR AND KANTARJIAN

T AB LE 8 Requirements for TKI discontinuation in clinical practice

Discontinuation of TKI
Parameter Yes No

Sokal risk Low-intermediate High

BCR-ABL1 transcripts Quantifiable (e13a2 or e14a2) Not quantifiable

CML stage Chronic Accelerated/blast phase

Response to first TKI Optimal Failure

Duration of all TKIs therapy >6–8 years <3 years

Depth of molecular response CMR (MR4.5) Less than MR4

Duration of molecular response >31 years < 3 years

Monitoring availability Ideal (every 2 months in years 1–2) Poor; noncompliant

chemotherapy are 40% in nonlymphoid CML-BP and 70%-80% in novo CML-AP are treated with frontline second generation TKI infin-
lymphoid CML-BP.123–125 Median survival times are 6 to 12 months, itely if an optimal response (CCyR; BCR-ABL1 transcripts [IS] <1%) is
and 12 to 24 months, respectively. The addition of TKIs to chemother- achieved within 6 months of therapy All other patients in CML-AP are
apy has improved the response rates and prolonged the median sur- treated with second/third generation TKI followed by allo-SCT.
vival time in CML-BP. We recently assessed outcome of 477 patients
with CML-BP who were treated with a TKI at some point during the 7 | CONCLUSIONS AND FUTURE
course of their CML.126 The median overall survival was 12 months DIRECTIONS
and the median failure-free survival was 5 months. The combination of
a TKI with intensive chemotherapy followed by stem cell transplanta- In 2018, CML experts and patients with CML have multiple treatment
126
tion appeared to confer the best outcome. options in the CML therapeutic armamentarium, including 5 TKIs (ima-
At present, allo-SCT is the only curative therapy for CML-AP and tinib, nilotinib, dasatinib, bosutinib, ponatinib), omacetaxine (protein
CML-BP: overall cure rates are in the range of 15%-40% and 10%- synthesis inhibitor), and several older agents (hydroxurea, interferon
20%, respectively. Patients with cytogenetic clonal evolution as the alpha, busulfan, 6-mercaptopurine, cytarabine, decitabine, others).
only accelerated phase criterion have a long-term event-free survival Most patients with CML would be expected to have a normal life span,
rate of about 60%.127 Otherwise, TKIs provide hematologic responses and be potentially functionally, though not molecularly, cured, as long
in 80% of patients and an estimated 4-year survival rates of 40%-55% as they continue therapy with TKI based regimens, are compliant with
in CML-AP, but only a 40% response rate and a median survival of 9 to the treatment, and are monitored closely for signs of resistance, to
12 months in CML-BP. Patients in the AP or BP should be encouraged change therapy in a timely manner and/or consider allo-SCT before
to participate in investigational strategies to improve their prognosis. CML progression. Future directions will focus on the potential molecu-
Patients with de novo CML-AP have a better outcome with frontline lar cure of CML (i.e., achievement of a durable CMR and its persistence
TKI therapy than patients who evolve from CP to AP. The estimated after discontinuation of TKI therapy). This is not a trivial issue since,
6–8 year survival rates with TKI therapy in de novo CML-AP are 60%- with effective TKI therapy, and full treatment penetration worldwide
127
80%. Such patients may continue on TKI therapy as their long-term (to 100% of all diagnosed patients and continuation of TKI therapy
treatment if they achieve a complete cytogenetic response on TKI without interruptions) the prevalence of CML would increase annually
therapy. and plateau at around 2030 to 2040 at a rate of 35 times the inci-
Allo-SCT should be considered early in patients in AP based on dence. The prevalence is estimated to be close to 160 000 patients
response to TKI therapy. The only curative option for patients in BP with CML in the United States and about 3 million patients worldwide.
disease is allo-SCT. TKIs monotherapy or in combination with chemo- This may represent a considerable burden on patients and the health-
therapy may serve as a good option for those who are not candidates care systems in relation to drug availability, compliance, potential devel-
for transplant, or as a bridge to allo-SCT. The role of TKIs before and opment of long-term side effects, and costs. Therefore it is important
after transplant is being evaluated. Accumulating data show that TKIs to continue research into therapies that increase the rates of durable
do not increase transplant-related complications and when used after CMRs. This may be achievable with the current more potent new gen-
low intensity conditioning regimens, may delay relapse rates and need eration TKIs alone, or in combination with other available (Bcl-2 inhibi-
for donor lymphocyte infusions. tors, peg-interferon alpha-2, omacetaxine, decitabine) or investigational
At MDACC, patients with CML-BP are treated with combination therapies (JAK2 inhibitors, hedgehog inhibitors, stem cell poisons, vac-
of chemotherapy (type depends on the immunophenotype) and third cines). Such strategies may improve the eradication of minimal residual
generation TKI followed by allo-SCT once a complete response is disease, potentially obviating the need for indefinite therapy with TKIs.
achieved and placed on maintenance TKI therapy. Patients with de- Further understanding of the pathophysiologic events downstream of
A JH |
JABBOUR AND KANTARJIAN 455

BCR-ABL1 may help in the development of new strategies to target arrhythmias. Allo-SCT remains an important therapeutic option for
them. patients in CML-CP in the following situations: patients who fail at
least 2 TKIs or potentially harbor the T315I mutation (after a trial of
8 | MD ANDERSON CANCER CENTER ponatinib therapy).
APPROACH Patients with CML-BP are treated with combination of chemother-
apy (type depends on the immunophenotype) and third generation TKI
ALL patients suspected to have CML undergo bone marrow examina- followed by allo-SCT once a complete response is achieved and placed
tion which will confirm the diagnosis and provide information needed on maintenance TKI therapy post allo-SCT. Patients with de-novo
for staging. Baseline PCR is performed to identify the specific type of CML-AP are treated with frontline second generation TKI infinitely if
rearrangement that can be appropriately followed when assessing for an optimal response (CCyR; BCR-ABL1 transcripts [IS] <1%) is achieved
response to TKI therapy. within 6 months of therapy. All other patients in CML-AP are treated
Waiting for confirmation, patients are placed on transient cytore- with second/third generation TKI followed by allo-SCT. Therapy dis-
duction therapy with hydroxyurea. Tumor lysis syndrome prophylaxis is continuation is offered only for patients with CML-CP with a quantifi-
implemented as well. able transcript, who have been on TKI for at least 6 years, have
Patients are stratified into low-risk and intermediate/high-risk dis- achieved a sustained CMR for at least 3–4 years, and in whom a close
ease. Outside clinical trials, patients with low-risk disease are treated follow-up can be performed.
with imatinib; patients with high-risk disease are treated with second
generation TKI. The choice of second generation TKI is based on
patients ’comorbidities. ORC ID
Patients are monitored regularly every 3 months in the first year,
Elias Jabbour http://orcid.org/0000-0003-4465-6119
and later every 6 months. Monitoring by the use of peripheral blood
Hagop Kantarjian http://orcid.org/0000-0002-1908-3307
FISH and PCR are alternative to marrow examinations to determine
cytogenetic response. If a patient is responding optimally, and the FISH
RE FE RE NC ES
study is negative at 6 or 12 months and or BCR-ABL1 transcripts [IS]
[1] American Cancer Society. Cancer Facts & Figures 2017. Atlanta:
<1%, it may be reasonable to omit further marrow exams, as the
American Cancer Society; 2017.
patient is likely to be in stable CCyR . The major treatment milestones
[2] Huang X, Cortes J, Kantarjian H. Estimations of the increasing
are at 6 and 12 months. Patients with lack of PCyR (BCR-ABL1 tran- prevalence and plateau prevalence of chronic myeloid leukemia in
scripts [IS]  10%) at 6 months, lack of CCyR (BCR-ABL1 transcripts the era of tyrosine kinase inhibitor therapy. Cancer. 2012;118(12):
[IS]  1%) at 12 months, and loss of response at any time are candi- 3123–3127.
dates for a change of therapy. The choice of TKI is based on mutation [3] Rowley JD. Letter: A new consistent chromosomal abnormality in
profile and patients’ comorbidities. We do not change therapy for chronic myelogenous leukaemia identified by quinacrine fluores-
cence and Giemsa staining. Nature. 1973;243(5405):290–293.
patients in CCyR but without MMR. Patients who meet all the relevant
[4] Mandanas RA, Leibowitz DS, Gharehbaghi K, et al. Role of p21
benchmarks in the first 12 months are monitored periodically using
RAS in p210 bcr-abl transformation of murine myeloid cells. Blood.
FISH and molecular testing (molecular testing only if BCR-ABL1 tran-
1993;82(6):1838–1847.
scripts consistently below 0.1% [IS]).
[5] Okuda K, Matulonis U, Salgia R, Kanakura Y, Druker B, Griffin JD.
Post imatinib failure, patients should undergo a bone marrow Factor independence of human myeloid leukemia cell lines is asso-
examination with cytogenetics and molecular testing, including analysis ciated with increased phosphorylation of the proto-oncogene Raf-
for mutation. The choice of second or third generation TKIs is based on 1. Exp Hematol. 1994;22(11):1111–1117.
disease phase, mutation profile, and patient’s comorbidities. In [6] Raitano AB, Halpern JR, Hambuch TM, Sawyers CL. The Bcr-Abl
advanced phases, we favor a combination of chemotherapy and TKI leukemia oncogene activates Jun kinase and requires Jun for trans-
formation. Proc Natl Acad Sci USA. 1995;92(25):11746–11750.
(mainly ponatinib or dasatinib). In patients with CML-CP with T315I
[7] Sawyers CL, Callahan W, Witte ON. Dominant negative MYC
mutation, ponatinib is the first choice treatment followed by allo-SCT if
blocks transformation by ABL oncogenes. Cell. 1992;70(6):901–
a donor is available and an optimal response is not achieved. Outside 910.
the context of a T315I mutation, the type of mutation dictates the [8] Shuai K, Halpern J, ten Hoeve J, Rao X, Sawyers CL. Constitutive
choice of therapy. Patients with compound mutations may not respond activation of STAT5 by the BCR-ABL oncogene in chronic myelog-
well to second generation TKI, and a close monitoring is indicated. If an enous leukemia. Oncogene. 1996;13(2):247–254.
optimal response is not achieved, a switch of therapy to a third genera- [9] Carlesso N, Frank DA, Griffin JD. Tyrosyl phosphorylation and
tion TKI and/or allo-SCT is warranted. In patients with no mutation or DNA binding activity of signal transducers and activators of tran-
scription (STAT) proteins in hematopoietic cell lines transformed by
with mutations sensitive to all second generation TKIs, the choice is
Bcr/Abl. J Exp Med. 1996;183(3):811–820.
based on comorbidities. Patients with vascular risk factors and meta-
[10] Ilaria RL, Jr, Van Etten RA. P210 and P190 (BCR/ABL) induce the
bolic dysfunctions are not candidates for nilotinib therapy Patients
tyrosine phosphorylation and DNA binding activity of multiple spe-
with lung injuries are not candidates for dasatinib therapy. Bosutinib cific STAT family members. J Biol Chem. 1996;271(49):31704–
may be the best choice for patients with cardiac problems and 31710.
|
A JH
456 JABBOUR AND KANTARJIAN

[11] Silver RT, Woolf SH, Hehlmann R, et al. An evidence-based analy- imatinib-resistant Abl kinase domain mutants. Cancer Res. 2005;65
sis of the effect of busulfan, hydroxyurea, interferon, and alloge- (11):4500–4505.
neic bone marrow transplantation in treating the chronic phase of [27] Tokarski JS, Newitt JA, Chang CY, et al. The structure of Dasatinib
chronic myeloid leukemia: developed for the American Society of (BMS-354825) bound to activated ABL kinase domain elucidates
Hematology. Blood. 1999;94(5):1517–1536. its inhibitory activity against imatinib-resistant ABL mutants. Can-
[12] Deininger M, O’brien SG, Guilhot F. International randomized cer Res. 2006;66(11):5790–5797.
study of interferon vs. STI571 (IRIS) 8-year follow up: sustained [28] Shah NP, Tran C, Lee FY, et al. Overriding imatinib resistance with
survival and low risk for progression of events in patients with
a novel ABL kinase inhibitor. Science. 2004;305(5682):399–401.
newly diagnosed chronic myeloid leukemia in chronic phase (CML-
[29] Kantarjian H, Shah NP, Hochhaus A, et al. Dasatinib versus imati-
CP) treated with imatinib. Blood. 2009;114.
nib in newly diagnosed chronic-phase chronic myeloid leukemia. N
[13] Jabbour E, Cortes JE, Kantarjian HM. Molecular monitoring in
Engl J Med. 2010;362(24):2260–2270.
chronic myeloid leukemia: response to tyrosine kinase inhibitors
and prognostic implications. Cancer. 2008;112(10):2112–2118. [30] Cortes JE, Saglio G, Kantarjian HM, et al. Final 5-year study results
of DASISION: the dasatinib versus imatinib study in treatment-
[14] Kantarjian H, Schiffer C, Jones D, Cortes J. Monitoring the
naïve chronic myeloid leukemia patients trial. J Clin Oncol. 2016;34
response and course of chronic myeloid leukemia in the modern
(20):2333–2340.
era of BCR-ABL tyrosine kinase inhibitors: practical advice on the
use and interpretation of monitoring methods. Blood. 2008;111(4): [31] Radich JP, Kopecky KJ, Appelbaum FR, et al. A randomized trial of
1774–1780. dasatinib 100 mg versus imatinib 400 mg in newly diagnosed
chronic-phase chronic myeloid leukemia. Blood. 2012;120(19):
[15] Schoch C, Schnittger S, Bursch S, et al. Comparison of chromo-
3898–3905.
some banding analysis, interphase- and hypermetaphase-FISH,
qualitative and quantitative PCR for diagnosis and for follow-up in [32] O’Brien SG, Hedgley C, Adams S, et al. Spiri6 2: An NCRI random-
chronic myeloid leukemia: a study on 350 cases. Leukemia. 2002; ized study comparing dasatinib with imatinib in patients with
16(1):53. newly diagnosed CML. Blood (ASH Annual Meeting Abstracts).
2014;124:517.
[16] Wang W, Cortes JE, Tang G, et al. Risk stratification of chromo-
somal abnormalities in chronic myelogenous leukemia in the era of [33] Talpaz M, Shah NP, Kantarjian H, et al. Dasatinib in imatinib-
tyrosine kinase inhibitor therapy. Blood. 2016;127(22):2742–2750. resistant Philadelphia chromosome-positive leukemias. N Engl J
Med. 2006;354(24):2531–2541.
[17] Druker BJ, Lydon NB. Lessons learned from the development of
an abl tyrosine kinase inhibitor for chronic myelogenous leukemia. [34] Shah NP, Rousselot P, Schiffer C, et al. Dasatinib in imatinib-
J Clin Invest. 2000;105(1):3–7. resistant or -intolerant chronic-phase, chronic myeloid leukemia
patients: 7-year follow-up of study CA180-034. Am J Hematol.
[18] O’Brien SG, Guilhot F, Larson RA, et al. Imatinib compared with
2016;91(9):869–874.
interferon and low-dose cytarabine for newly diagnosed chronic-
phase chronic myeloid leukemia. N Engl J Med. 2003;348(11):994– [35] Cortes J, Hochhaus A, Kantarjian H, et al. Impact of dose reduc-
1004. tions on 5-year efficacy in newly diagnosed patients with chronic
[19] Hochhaus A, Larson RA, Guilhot F, et al. Long-term outcomes of myeloid leukemia in chronic phase (CML-CP) from DASISION. Am
imatinib treatment for chronic myeloid leukemia. N Engl J Med. Soc Clin Oncol. 2017;abstract 3036.
2017;376(10):917–927. [36] Naqvi K, Cortes J, Skinner J, et al. Evaluating the role of lower
[20] Sacha T, Go ra-Tybor J, Szarejko M, et al. A multicenter prospective dose dasatinib in newly diagnosed early chronic phase-chronic
study on efficacy and safety of imatinib generics: A report from myeloid leukemia (CML-CP). Blood. 2017;130:abstract 1611.
Polish Adult Leukemia Group imatinib generics registry. Am J Hem- [37] Weisberg E, Manley PW, Breitenstein W, et al. Characterization of
atol. 2017;92(7):E125–E128. AMN107, a selective inhibitor of native and mutant Bcr-Abl. Can-
[21] Madhav D. India Generic imatinib in chronic myeloid leukemia: sur- cer Cell. 2005;7(2):129–141.
vival of the cheapest. Blood. 2016;128:abstract 630. [38] Saglio G, Kim DW, Issaragrisil S, et al. Nilotinib versus imatinib for
[22] Cortes JE, Baccarani M, Guilhot F, et al. Phase III, randomized, newly diagnosed chronic myeloid leukemia. N Engl J Med. 2010;
open-label study of daily imatinib mesylate 400 mg versus 800 mg 362(24):2251–2259.
in patients with newly diagnosed, previously untreated chronic [39] Hochhaus A, Saglio G, Hughes TP, et al. Long-term benefits and
myeloid leukemia in chronic phase using molecular end points: risks of frontline nilotinib vs imatinib for chronic myeloid leukemia
tyrosine kinase inhibitor optimization and selectivity study. J Clin in chronic phase: 5-year update of the randomized ENESTnd trial.
Oncol. 2010;28:424–430. Leukemia. 2016;30(5):1044–1054.
[23] Preudhomme C, Guilhot J, Nicolini FE, et al. Imatinib plus peginter- [40] Wang J, Shen ZX, Saglio G, et al. Phase 3 study of nilotinib vs ima-
feron alfa-2a in chronic myeloid leukemia. N Engl J Med. 2010;363 tinib in Chinese patients with newly diagnosed chronic myeloid
(26):2511–2521. leukemia in chronic phase: ENESTchina. Blood. 2015;125(18):
[24] Hehlmann R, Lauseker M, Saussele S, et al. Final evaluation of 2771–2778.
randomized CML-study IV: 10-year survival and evolution of ter- [41] Cortes JE, Gambacorti-Passerini C, Deininger MW, et al. Bosutinib
minal phase. Blood. 2017;130:abstract 897. versus Iimatinib for newly diagnosed chronic myeloid leukemia:
[25] Lombardo LJ, Lee FY, Chen P, et al. Discovery of N-(2-chloro-6- results from the randomized BFORE Trial. J Clin Oncol. 2017.
methyl- phenyl)-2-(6-(4-(2-hydroxyethyl)- piperazin-1-yl)-2-methyl- [42] Jain P, Kantarjian H, Alattar ML, et al. Long-term molecular and
pyrimidin-4- ylamino)thiazole-5-carboxamide (BMS-354825), a dual cytogenetic response and survival outcomes with imatinib 400 mg,
Src/Abl kinase inhibitor with potent antitumor activity in preclinical imatinib 800 mg, dasatinib, and nilotinib in patients with chronic-
assays. J Med Chem. 2004;47(27):6658–6661. phase chronic myeloid leukaemia: retrospective analysis of patient
[26] O’Hare T, Walters DK, Stoffregen EP, et al. In vitro activity of Bcr- data from five clinical trials. Lancet Haematol. 2015;2(3):e118–
Abl inhibitors AMN107 and BMS-354825 against clinically relevant e128.
A JH |
JABBOUR AND KANTARJIAN 457

[43] O’Brien S, Radich JP, Abboud CN, et al. Chronic myelogenous leu- leukemia receiving tyrosine kinase inhibitors. J Clin Oncol. 2011;29:
kemia, Version 1.2014. J Natl Compr Canc Netw. 2013;11:1327– 1512–1516.
1340. [61] Hehlmann R, Lauseker M, Jung-Munkwitz S, et al. Tolerability-
[44] Montani D, Bergot E, Gunther S, et al. Pulmonary arterial hyper- adapted imatinib 800 mg/d versus 400 mg/d versus 400 mg/d
tension in patients treated by dasatinib. Circulation. 2012;125(17): plus interferon-alpha in newly diagnosed chronic myeloid leukemia.
2128–2137. J Clin Oncol. 2011;29:1634–1642.
[45] Quintas-Cardama A, Han X, Kantarjian H, Cortes J. Tyrosine kinase [62] Kantarjian HM, Shan J, Jones D, et al. Significance of increasing
inhibitor-induced platelet dysfunction in patients with chronic levels of minimal residual disease in patients with Philadelphia
myeloid leukemia. Blood. 2009;114(2):261–263. chromosome-positive chronic myelogenous leukemia in complete
[46] Quintas-Cardama A, Kantarjian H, Ravandi F, et al. Bleeding diath- cytogenetic response. J Clin Oncol. 2009;27:3659–3663.
esis in patients with chronic myeloid leukemia receiving dasatinib [63] Marin D, Ibrahim AR, Lucas C, et al. Assessment of BCR-ABL1
therapy. Cancer. 2009;115(11):2482–2490. transcript levels at 3 months is the only requirement for predicting
[47] Aichberger KJ, Herndlhofer S, Schernthaner G, et al. Progressive outcome for patients with chronic myeloid leukemia treated with
peripheral arterial occlusive disease and other vascular events dur- tyrosine kinase inhibitors. J Clin Oncol. 2012;30:232–238.
ing nilotinib therapy in CML. Am J Hematol. 2011;86(7):533–539. [64] Jain P, Kantarjian H, Nazha A, et al. Early responses predict better
[48] Quintas-Cardama A, Kantarjian H, Cortes J. Nilotinib-Associated outcomes in patients with newly diagnosed chronic myeloid leuke-
Vascular Events. Clin Lymphoma Myeloma Leuk. 2012;12(5):337– mia: results with four tyrosine kinase inhibitor modalities. Blood.
340. 2013;121(24):4867–4874.
[49] Tefferi A, Letendre L. Nilotinib treatment-associated peripheral [65] Neelakantan P, Gerrard G, Lucas C, et al. Combining BCR-ABL1
artery disease and sudden death: Yet another reason to stick to transcript level at 3 and 6 months in chronic myeloid leukemia:
imatinib as front-line therapy for chronic myelogenous leukemia. implications for early intervention strategies. Blood. 2013;121:
Am J Hematol. 2011;86(7):610–611. 3739–3742.
[50] Giles FJ, Mauro MJ, Hong F, et al. Rates of peripheral arterial [66] Nazha A, Kantarjian H, Jain P, et al. Assessment at 6 months may
occlusive disease in patients with chronic myeloid leukemia in the be warranted for patients with chronic myeloid leukemia with no
chronic phase treated with imatinib, nilotinib, or non-tyrosine major cytogenetic response at 3 months. Haematologica. 2013;98
kinase therapy: a retrospective cohort analysis. Leukemia. 2013;27 (11):1686–1688.
(6):1310–1315. [67] Branford S, Roberts N, Yeung DT, Altamura H, Parker WT, Hughes
[51] Smith TJ, Hillner BE. Bending the cost curve in cancer care. N Engl TP. Any BRC-ABL reduction below 10% at 6 months of therapy
J Med. 2011;364(21):2060–2065. significantly improves outcome for CML patients with a poor
[52] Kantarjian HM, Fojo T, Mathisen M, Zwelling LA. Cancer drugs in response at 3 months. Blood (ASH Annual Meeting Abstracts). 2013;
the United States: justum pretium—the just price. J Clin Oncol. 122:254.
2013;31(28):3600–3604. [68] Jabbour E, Kantarjian HM, O’brien S, et al. Front-line therapy with
[53] Experts in Chronic Myeloid Leukemia. The price of drugs for second-generation tyrosine kinase inhibitors in patients with early
chronic myeloid leukemia is a reflection of the unsustainable prices chronic phase chronic myeloid leukemia: what is the optimal
of cancer drugs: from the perspective of a large group of CML response? J Clin Oncol. 2011;29:4260–4265.
experts. Blood. 2013;121:4439–4442. [69] Branford S. Monitoring and defining early response: Where to
[54] Gambacorti-Passerini C, Antolini L, Mahon X, et al. Multicenter draw the line? Best Pract Res Heamtol. 2016;29:284–294.
independent assessment of outcomes in chronic myeloid leukemia [70] Cortes JE, De Souza CA, Ayala M, et al. Switching to nilotinib ver-
patients treated with imatinib. J Natl Cancer Inst. 2011;103:553– sus imatinib dose escalation in patients with chronic myeloid leu-
561. kaemia in chronic phase with suboptimal response to imatinib
[55] Larson RA, Conti R, Padula WV, et al. What is the most cost- (LASOR): a randomised, open-label trial. Lancet Haematol. 2016;3
effective strategy for treating newly diagnosed chronic myeloid (12):e581–e591.
leukemia (CML) after imatinib loses patent exclusivity? Blood (ASH [71] Hughes TP, Leber B, Cervantes F, et al. Sustained deep molecular
Annual Meeting Abstracts). 2014;124:738. responses in patients switched to nilotinib due to persistent BCR-
[56] Sokal JE, Cox EB, Baccarani M, et al. Prognostic discrimination in ABL1 on imatinib: final ENESTcmr randomized trial results. Leuke-
“good-risk” chronic granulocytic leukemia. Blood. 1984;63(4):789– mia. 2017;31(11):2529–2531.
799. [72] Marin D, Bazeos A, Mahon FX, et al. Adherence is the critical fac-
[57] Hasford J, Pfirrmann M, Hehlmann R, et al. A new prognostic tor for achieving molecular responses in patients with chronic
score for survival of patients with chronic myeloid leukemia myeloid leukemia who achieve complete cytogenetic responses on
treated with interferon Alfa. J Natl Cancer Inst. 1998;90:850–858. imatinib. J Clin Oncol. 2010;28(14):2381–2388.
[58] Baccarani M, Deininger MW, Rosti G, et al. European LeukemiaNet [73] Darkow T, Henk HJ, Thomas SK, et al. Treatment interruptions
recommendations for the management of chronic myeloid leuke- and non-adherence with imatinib and associated healthcare costs:
mia: 2013. Blood. 2013;122(6):872–884. a retrospective analysis among managed care patients with chronic
[59] Testoni N, Marzocchi G, Luatti S, et al. Chronic myeloid leukemia: myelogenous leukaemia. PharmacoEconomics. 2007;25(6):481–496.
a prospective comparison of interphase fluorescence in situ hybrid- [74] Noens L, van Lierde MA, De Bock R, et al. Prevalence, determi-
ization and chromosome banding analysis for the definition of nants, and outcomes of nonadherence to imatinib therapy in
complete cytogenetic response: a study of the GIMEMA CML WP. patients with chronic myeloid leukemia: the ADAGIO study. Blood.
Blood. 2009;114(24):4939–4943. 2009;113(22):5401–5411.
[60] Kantarjian H, Cortes J. Considerations in the management of [75] Bristol-Myers Squibb Company. Sprycel® (dasatinib) prescribing
patients with Philadelphia chromosome-positive chronic myeloid information. Princeton, NJ; 2011.
|
A JH
458 JABBOUR AND KANTARJIAN

[76] Novartis Pharmaceuticals Corporation. Tasigna® (nilotinib) pre- [92] Jabbour E, Branford S, Saglio G, Jones D, Cortes JE, Kantarjian
scribing information. East Hanover, NJ; 2011. HM. Practical advice for determining the role of BCR-ABL muta-
[77] Jabbour E, Kantarjian HM, Jones D, et al. Imatinib mesylate dose tions in guiding tyrosine kinase inhibitor therapy in patients with
escalation is associated with durable responses in patients with chronic myeloid leukemia. Cancer. 2011;117(9):1800–1811.
chronic myeloid leukemia after cytogenetic failure on standard- [93] Mu€ller MC, Cortes JE, Kim D-W, et al. Dasatinib treatment of chronic-
dose imatinib therapy. Blood. 2009;113(10):2154–2160. phase chronic myeloid leukemia: analysis of responses according to
[78] Kantarjian H, Pasquini R, Levy V, et al. Dasatinib or high-dose ima- preexisting BCR-ABL mutations. Blood. 2009;114(24):4944–4953.
tinib for chronic-phase chronic myeloid leukemia resistant to imati- [94] Hughes T, Saglio G, Branford S, et al. Impact of baseline BCR-ABL
nib at a dose of 400 to 600 milligrams daily: two-year follow-up of mutations on response to nilotinib in patients with chronic myeloid
a randomized phase 2 study (START-R). Cancer. 2009;115(18): leukemia in chronic phase. J Clin Oncol. 2009;27(25):4204–4210.
4136–4147. [95] Kantarjian HM, Cortes JE, Kim DW, et al. Bosutinib safety and
[79] Garcia-Gutierrez JV, Herrera P, Abalo LL, Rey MD, Calbacho M. management of toxicity in leukemia patients with resistance or
Impact of second-generation tyrosine kinase inhibitors as second intolerance to imatinib and other tyrosine kinase inhibitors. Blood.
line treatment for patients with chronic myeloid leukemia. Blood. 2014;123(9):1309–1318.
2011;118:632. [96] Jabbour E, Kantarjian H, Ghanem H, et al. The achievement of a
[80] Goh HG, Jootar S, Kim HJ, Sohn SK, Park JS, Kim SH. Efficacy of 3-month complete cytogenetic response to second-generation
nilotinib versus high-dose imatinib in early chronic phase CML tyrosine kinase inhibitors predicts survival in patients with chronic
patients who have suboptimal molecular responses to standard- phase chronic myeloid leukemia after imatinib failure. Clin Lym-
dose imatinib (RE-NICE multicenter study). Blood. 2011;118:632. phoma Myeloma Leuk. 2013;13(3):302–306.
[81] Cortes JE, Kantarjian HM, Brummendorf TH, et al. Safety and effi- [97] Jabbour E, Cortes J, Santos FP, et al. Results of allogeneic hemato-
cacy of bosutinib (SKI-606) in chronic phase Philadelphia poietic stem cell transplantation for chronic myelogenous leukemia
chromosome-positive chronic myeloid leukemia patients with patients who failed tyrosine kinase inhibitors after developing BCR-
resistance or intolerance to imatinib. Blood. 2011;118(17):4567– ABL1 kinase domain mutations. Blood. 2011;117(13):3641–3647.
4576. [98] Lee SJ, Kukreja M, Wang T, et al. Impact of prior imatinib mesylate
[82] Yeung DT, Osborn M, White DL, Branford S, Kornhauser M, Slader on the outcome of hematopoietic cell transplantation for chronic
C. Upfront imatinib therapy in CML patients with rapid switching myeloid leukemia. Blood. 2008;112(8):3500–3507.
to nilotinib for failure to achieve molecular targets or intolerance €elles GJ, Tarin-Arzaga LC, Gonzalez-Carrillo ML, et al.
[99] Ruiz-Argu
achieves high overall rates of molecular response and a low risk of Therapeutic choices in patients with Ph-positive CML living in
progression - an update of the TIDEL-II trial. Blood. 2011;118:632. Mexico in the tyrosine kinase inhibitor era: SCT or TKIs? Bone
[83] Quintas-Cardama A, Cortes JE, O’brien S, et al. Dasatinib early Marrow Transplant. 2008;42(1):23–28.
intervention after cytogenetic or hematologic resistance to imati- [100] Jaime-Perez JC, Heredia-Salazar AC, Cantu-Rodríguez OG, et al.
nib in patients with chronic myeloid leukemia. Cancer. 2009;115 Cost structure and clinical outcome of a stem cell transplantation
(13):2912–2921. program in a developing country: the experience in northeast
[84] O’Hare T, Shakespeare WC, Zhu X, et al. AP24534, a pan-BCR- Mexico. Oncologist. 2015;20(4):386–392.
ABL inhibitor for chronic myeloid leukemia, potently inhibits the [101] Mahon FX, Rea D, Guilhot J, et al. Discontinuation of imatinib in
T315I mutant and overcomes mutation-based resistance. Cancer patients with chronic myeloid leukaemia who have maintained
Cell. 2009;16:401–412. complete molecular remission for at least 2 years: the prospective,
[85] Zhou T, Commodore L, Huang WS, et al. Structural mechanism of multicentre Stop Imatinib (STIM) trial. Lancet Oncol. 2010;11(11):
the pan-BCR-ABL inhibitor ponatinib (AP24534): lessons for over- 1029–1035.
coming kinase inhibitor resistance. Chem Biol Drug Des. 2011;77:1– [102] Ross DM, Branford S, Seymour JF, et al. Safety and efficacy of
11. imatinib cessation for CML patients with stable undetectable mini-
[86] Cortes JE, Kim DW, Pinilla-Ibarz J, et al. A phase 2 trial of ponati- mal residual disease: results from the TWISTER study. Blood.
nib in Philadelphia chromosome-positive leukemias. N Engl J Med. 2013;122(4):515–522.
2013;369(19):1783–1796. [103] Rea D, Nicolini FE, Tulliez M, et al. Discontinuation of dasatinib or
[87] Kantarjian HM, Pinilla-Ibarz J, Le Coutre PD, et al. Five-year results nilotinib in chronic myeloid leukemia: interim analysis of the STOP
of the ponatinib phase II PACE trial in heavily pretreated CP-CML 2G-TKI study. Blood. 2017;129(7):846–854.
patients. Am Soc Clin Oncol. 2017;abstract 7012. [104] Mahon FX, Richter J, Guilhot J, et al. Cessation of tyrosine kinase
[88] Iclusig (Ponatinib) [Prescribing Information]. Cambridge, MA: ARIAD inhibitors treatment in chronic myeloid leukemia patients with
Pharmaceuticals, Inc; 2014. deep molecular response: results of the euro-ski trial. Blood. 2016;
[89] Wylie AA, Schoepfer J, Jahnke W, et al. The allosteric inhibitor 128(22):787–787.
ABL001 enables dual targeting of BCR-ABL1. Nature. 2017;543 [105] Duration of deep molecular response has most impact on the suc-
(7647):733–737. cess of cessation of tyrosine kinase inhibitor treatment in chronic
[90] Hughes TP, Goh YT, Ottmann OG, et al. Expanded phase 1 study myeloid leukemia - results from the EURO-SKI trial. Blood. 2017;
of ABL001, a potent, allosteric inhibitor of BCR-ABL, reveals sig- 130: abstract 313.
nificant and durable responses in patients with CML-chronic phase [106] Jabbour E. Chronic myeloid leukemia: First-line drug of choice. Am
with failure of prior TKI therapy. Blood. 2016;128:abstract 625. J Hematol. 2016;91(1):59–66.
[91] Cortes J, Jabbour E, Kantarjian H, et al. Dynamics of BCR-ABL [107] Padula WV, Larson RA, Dusetzina SB, et al. Cost-effectiveness of
kinase domain mutations in chronic myeloid leukemia after tyrosine kinase inhibitor treatment strategies for chronic myeloid
sequential treatment with multiple tyrosine kinase inhibitors. Blood. leukemia in chronic phase after generic entry of imatinib in the
2007;110(12):4005–4011. United States. J Natl Cancer Inst. 2016;108(7).
A JH |
JABBOUR AND KANTARJIAN 459

[108] Chhatwal J, Mathisen M, Kantarjian H. Are high drug prices for [120] Carpenter PA, Snyder DS, Flowers ME, et al. Prophylactic adminis-
hematologic malignancies justified? A critical analysis. Cancer. tration of imatinib after hematopoietic cell transplantation for
2015;121(19):3372–3379. high-risk Philadelphia chromosome-positive leukemia. Blood. 2007;
[109] Holyoake TL, Vetrie D. The chronic myeloid leukemia stem cell: 109(7):2791–2793.
stemming the tide of persistence. Blood. 2017;129(12):1595–1606. [121] Jabbour E, Cortes J, Kantarjian H, et al. Novel tyrosine kinase
[110] Holyoake TL, Vetrie D. The chronic myeloid leukemia stem cell: inhibitor therapy before allogeneic stem cell transplantation in
stemming the tide of persistence. Blood. 2017;129(12):1595–1606. patients with chronic myeloid leukemia: no evidence for increased
transplant-related toxicity. Cancer. 2007;110(2):340–344.
[111] Simonsson B, Gedde-Dahl T, Markevarn B, et al. Combination of
pegylated IFN-alpha2b with imatinib increases molecular response [122] Le Coutre PD, Giles FJ, Hochhaus A, et al. Nilotinib in patients
rates in patients with low- or intermediate-risk chronic myeloid with Ph1 chronic myeloid leukemia in accelerated phase following
leukemia. Blood. 2011;118(12):3228–3235. imatinib resistance or intolerance: 24-month follow-up results. Leu-
kemia. 2012;26(6):1189–1194.
[112] Hjorth-Hansen H, Stentoft J, Richter J, et al. Safety and efficacy of
the combination of pegylated interferon-alpha2b and dasatinib in [123] Thomas DA, Faderl S, Cortes J, et al. Treatment of Philadelphia
newly diagnosed chronic-phase chronic myeloid leukemia patients. chromosome-positive acute lymphocytic leukemia with hyper-
Leukemia. 2016;30(9):1853–1860. CVAD and imatinib mesylate. Blood. 2004;103(12):4396–4407.

[113] Carter BZ, Mak PY, Mu H, et al. Combined targeting of BCL-2 and [124] Ravandi F, O’brien S, Thomas D, et al. First report of phase 2
BCR-ABL tyrosine kinase eradicates chronic myeloid leukemia study of dasatinib with hyper-CVAD for the frontline treatment of
stem cells. Sci Transl Med. 2016;8(355):355ra117. patients with Philadelphia chromosome-positive (Ph1) acute lym-
phoblastic leukemia. Blood. 2010;116(12):2070–2077.
[114] Ko TK, Chuah CT, Huang JW, Ng KP, Ong ST. The BCL2 inhibitor
ABT-199 significantly enhances imatinib-induced cell death in [125] Quintas-Cardama A, Kantarjian H, Garcia-Manero G, et al. A pilot
chronic myeloid leukemia progenitors. Oncotarget. 2014;5(19): study of imatinib, low-dose cytarabine and idarubicin for patients
9033–9038. with chronic myeloid leukemia in myeloid blast phase. Leuk Lym-
phoma. 2007;48(2):283–289.
[115] Schnekenburger M, Grandjenette C, Ghelfi J, et al. Sustained expo-
sure to the DNA demethylating agent, 2’-deoxy-5-azacytidine, [126] Jain P, Kantarjian HM, Ghorab A, et al. Prognostic factors and sur-
leads to apoptotic cell death in chronic myeloid leukemia by pro- vival outcomes in patients with chronic myeloid leukemia in blast
moting differentiation, senescence, and autophagy. Biochem Phar- phase in the tyrosine kinase inhibitor era: Cohort study of 477
macol. 2011;81(3):364–378. patients. Cancer. 2017;123(22):4391–4402.

[116] Gallipoli P, Cook A, Rhodes S, et al. JAK2/STAT5 inhibition by [127] Ohanian M, Kantarjian HM, Quintas-Cardama A, et al. Tyrosine
nilotinib with ruxolitinib contributes to the elimination of CML kinase inhibitors as initial therapy for patients with chronic mye-
CD341 cells in vitro and in vivo. Blood. 2014;124(9):1492–1501. loid leukemia in accelerated phase. Clin Lymphoma Myeloma Leuk.
2014;14(2):155–162.
[117] Held SA, Heine A, Mayer KT, Kapelle M, Wolf DG, Brossart P.
Advances in immunotherapy of chronic myeloid leukemia CML.
Curr Cancer Drug Targets. 2013;13(7):768–774.
[118] Apperley JF, Cortes JE, Kim DW, et al. Dasatinib in the treatment How to cite this article: Jabbour E, Kantarjian H. Chronic mye-
of chronic myeloid leukemia in accelerated phase after imatinib
loid leukemia: 2018 update on diagnosis, therapy and monitor-
failure: the START-A trial. J Clin Oncol. 2009;27:3472–3479.
ing. Am J Hematol. 2018;93:442–459. https://doi.org/10.1002/
[119] Oehler VG, Gooley T, Snyder DS, et al. The effects of imatinib
ajh.25011
mesylate treatment before allogeneic transplantation for chronic
myeloid leukemia. Blood. 2007;109(4):1782–1789.

You might also like