Abstract
Imatinib mesylate has revolutionized the treatment landscape for patients with newly diagnosed chronic myeloid leukemia (CML). Imatinib at a dose of 400 mg/day is considered the standard treatment for all newly diagnosed chronic phase CML. Follow-up on the pivotal International Randomized Study of Interfreron versus STI571 (IRIS) study has shown excellent response rates, progression-free survival and overall survival after 8 years of follow-up. However, some patients will develop resistance to imatinib treatment due to a multitude of reasons. Numerous strategies to overcome resistance are available including dose escalation of imatinib, switching to a second generation tyrosine kinase inhibitor or to one of the newer non-tyrosine kinase inhibitors. This review guides the treating physician with a rational approach in the management of CML patients who fail initial treatment with imatinib or lose response while on therapy with imatinib.
Keywords: myeloproliferative disorders, tyrosine kinase inhibitors, mutation screening, homoharringtonine, multikinase inhibitors
INTRODUCTION
Chronic myeloid leukemia (CML) is a pluripotent hematopoietic stem cell disorder leading to myeloproliferation and its attendant consequences. In the United States, it is estimated that approximately 5050 cases of CML will be diagnosed in 2010 with an annual incidence of 1–2 cases per 100,000 adult individuals.1 The instigating factor in the pathogenesis of chronic myeloid leukemia (CML) is the formation of the Philadelphia chromosome resulting from the reciprocal translocation between chromosomes 9 and 22 (t(9;22)(q34;q11)), which is associated with the de novo creation of the BCR-ABL fusion oncogene.2,3 The gene product of the BCR-ABL gene constitutively activates numerous downstream targets including c-myc, Akt and Jun, all of which cause uncontrolled proliferation and survival of CML cells.
IMATINIB MESYLATE
Imatinib mesylate (Gleevec™, STI-571), a 2-phenylaminopyrimidine, is a selective and potent inhibitor of BCR-ABL and few other tyrosine kinases, including c-kit, PDGF-R alpha and beta, and ABL related gene (ARG).4 It is orally administered with 98% bioavailability and a half-life of 13–16 hours. Imatinib was first used in CML in patients who had developed resistance or intolerance to interferon-α (IFN-α). Among 532 such patients treated with imatinib, a complete cytogenetic response (CCyR) was achieved in 60%. The estimated 5-year survival rate was 76%.5,6
Based on these favorable results, a large, randomized trial was initiated among patients with CML in chronic phase (CML-CP) who had received no prior therapy. In this study, known as the International Randomized Study of Interfreron versus STI571 (IRIS) trial, patients were randomized to receive imatinib or IFN-α and ara-C which was the standard therapy at the time. Treatment with imatinib was significantly better in nearly all outcomes measured, including hematologic and cytogenetic response, toxicity and progression-free survival (PFS).7 After 8 years, the cumulative CCyR rate for first-line imatinib-treated patients was 82%.8 The event-free survival (EFS) was 81%, and the estimated rate of freedom from progression to accelerated phase (CML-AP) or blastic phase (CML-BP) was 92%. The estimated overall survival (OS) rate for patients treated with imatinib was 85%. At 8 years, 304 patients (55%) randomized to imatinib remained on treatment. The curves seem to plateau after the fourth year and yearly event rates have ranged from 0.3%–2%. With an annual mortality of 2%, the estimated survival of a newly diagnosed patient with CML may be in the range of 20–30 years.
MECHANISMS OF RESISTANCE
Despite the impressive results with imatinib, a subset of patients treated with imatinib will develop resistance. Failure to achieve a landmark response is considered primary resistance, and this is further subdivided into primary hematologic resistance, and primary cytogenetic resistance. Secondary resistance is defined by the achievement and then subsequent loss of a hematologic or cytogenetic response. Hematologic resistance occurs in 2–4% of cases, while cytogenetic resistance is more common, occurring in 15–25% of patients. Mutations in BCR-ABL are rarely responsible for primary resistance. Recent work suggests that primary resistance may be associated with increased transcript levels of the drug metabolism gene prostaglandin-endoperoxide synthase 1/cyclooxgenase 1 (PTGS1/COX1), and this may serve as a biomarker to distinguish patients with primary resistance to imatinib.9
Several mechanisms of resistance to imatinib have been described. These can be classified into two categories: BCR-ABL-dependent and BCR-ABL independent. The first group includes amplification or overexpression of BCR-ABL or its protein product,10 and point mutations of the ABL sequence.11 The second group includes multidrug-resistance (MDR) expression and overexpression of Src kinases.12 BCR-ABL-dependent mechanisms are more common, particularly point mutations, which have been identified in approximately 50% of patients who develop clinical resistance to imatinib.13,14 More than 90 different mutations have been described and occur in any of the different relevant domains of the kinase, including the ATP-binding domain (also known as P-loop), the catalytic domain, the activation loop, and amino acids that make direct contact with imatinib. The significance of these mutations varies. While some retain some sensitivity to imatinib at concentrations similar to those of the wild type sequence, others, particularly T315I, are nearly completely insensitive to imatinib.15 Most of the clinically relevant mutations develop in a few residues in the in the P-loop (G250E, Y253F/H, and E255K/V), the contact site (T315I), and the catalytic domain (M351T and F359V).16 The P-loop mutations have been suggested to carry an increased risk of rapid blastic transformation and short survival13 although the M.D. Anderson Cancer Center (MDACC) experience does not support this notion.17 In some patients, more than one mutation may be present at the same time. This phenomenon appears to increase in frequency after treatment with more than one tyrosine kinase mutation. Mutations are quantified by direct sequencing and the sensitivity of such assay varies between 10%–25%.18,19 Other methods, such as denatured high-performance liquid chromatography increase the sensitivity to 1% to 10%.19,20 However, it is unclear at this time if identification of small mutated clones with these highly sensitive methods is clinically relevant.
Other mechanisms of resistance due to intrinsic factors include: BCR-ABL gene amplification, BCR-ABL overexpression, aberrations in other oncogenetic signaling pathways, and the persistence of leukemic stem cells.14,21,22 Extrinsic factors contributing to resistance include those that decrease the blood levels or bioavailability of imatinib, such as: patient compliance, drug–drug interactions, drug influx and efflux and multidrug resistance in sanctuary sites, as well as microenvironmental factors.21
MUTATION SCREENING DURING IMATINIB THERAPY
The European LeukemiaNet (ELN) and the National Comprehensive Cancer Network (NCCN) provide guidance for the monitoring of patients with CML.23,24 The criteria for defining optimal response, sub-optimal response and failure to respond are outlined in Table 1. The ELN recommends mutational analysis in instances of suboptimal response or failure to therapy, and always before changing therapy to a second-generation tyrosine kinase inhibitor (TKI). Patients failing TKI therapy should potentially be assessed for compliance to therapy before switch, as it has been shown that patient reported compliance and actual compliance reported can be discordant, and this may be a reason for treatment failure. The magnitude of increase in BCR-ABL transcript levels which should prompt mutation testing is a topic of debate. Five to 10-fold rises have been proposed as a reasonable trigger for mutation testing. A recent study demonstrated that increases in BCR-ABL mRNA levels of 5-fold or more were not sufficiently sensitive in detecting mutations, and that a 2.6-fold increase in BCR-ABL transcripts is a better threshold.25 In most clinics, however, it may be more reasonable to consider mutation testing when BCR-ABL levels increase at least 5-fold, confirmed in an independent test in the same laboratory to confirm that the observed increase is real, and not due to assay or laboratory variability.
Table 1.
Evaluation Time | Response | ||
---|---|---|---|
Optimal | Suboptimal | Failure | |
3 months | CHR and at least minor CyR | No CyR | No CHR |
6 months | At least partial CyR | Less than partial CyR | No CyR |
12 months | CCyR | Partial CyR | Less than partial CyR |
18 months | MMR | Less than MMR | Less than CCyR |
Any time | Stable or improving MMR | Loss of MMR, presence of mutations | Loss of CHR, loss of CCyR, clonal evolution |
CHR: complete hematologic response; CyR: cytogenetic response; CCyR: complete cytogenetic response; MMR: major molecular response
STRATEGIES TO OVERCOME IMATINIB RESISTANCE
Multiple strategies to overcome failure to standard dose (400 mg/day) imatinib are under investigation. These include dose escalation of imatinib, switch to a second-generation TKI, other novel TKIs in a clinical trial, non-TKI based therapy and allogeneic stem cell transplant (SCT) in eligible patients.
Imatinib Dose Escalation
Dose escalation can improve the response in a subset of patients with resistance to standard dose imatinib and was the main option for managing suboptimal responses and treatment failures before the introduction of second generation TKIs. In a retrospective analysis of patients enrolled in the IRIS trial, Kantarjian et al reported that among 106 patients who required dose escalation due to resistance to standard dose therapy, freedom-from-progression and OS rates were 89% and 84%, respectively, at 3 years from dose escalation.26 In another study from MDACC, 84 patients with CML-CP were dose escalated to imatinib 600–800 mg/day after developing hematologic failure (n = 21), or cytogenetic failure (n = 63) to standard dose imatinib.27 Among patients that met the criteria for cytogenetic failure, 75% (47/63) responded to imatinib dose escalation. In contrast, in patients where imatinib was dose escalated because of hematologic failure, 48% achieved a complete hematologic response and only 14% (3/21) achieved a cytogenetic response. Patients more likely to respond to imatinib dose increase are those that have previously achieved a cytogenetic response and then lost it and who have not developed any mutations unresponsive to imatinib. Even in these cases, a switch to a 2nd generation TKI is preferable unless the patient has no access to these agents.
Several Phase II studies examined the role of a higher dose of imatinib (800mg) upfront in the treatment of patients with CML. The Tyrosine Kinase Inhibitor Optimization and Selectivity (TOPS) study was a phase 3 trial comparing the efficacy and safety of high-dose (800 mg/day) with standard-dose imatinib (400 mg/day) in patients with newly diagnosed CML-CP.28 The primary endpoint of the study was rate of major molecular response (MMR) at 12 months of therapy. A 24-month update on the TOPS data was recently reported.29 It appears that there was no significant difference between the 800 mg/day and 400 mg/day arms for either the CCyR (76% vs 76%, respectively; P = 1.00) or MMR rate (51% vs 54%, respectively; P = .626) Most importantly, thus far at 24 months there were no differences between arms with respect to EFS (95% vs 95%, respectively; P = .71), PFS (98% vs 97%; P = .64), and OS (98% vs 97%, respectively; P = .70), although it is still relatively early. Adverse events tended to be more common among patients in the 800-mg/day arm vs the 400-mg/day arm, as was the rate of discontinuation due to adverse events (12% vs 5%, respectively). The results from TOPS study were confirmed in a randomized trial Gruppo Italiano Malattie Ematologiche dell’Adulto (GIMEMA) 021/ELN (021/ELN) assessing the efficacy of imatinib 800 mg/day vs 400 mg/day as front line therapy in high-risk Sokal patients.30 The primary study endpoint of CCyR at one year was not significantly different between patients treated with imatinib 400 mg/day (61%) vs 800 mg/day (64%). There was a trend toward higher rates of MMR with 800 mg/day compared with 400 mg/day, but the differences were not statistically significant. Adverse events were not significantly different between treatment arms, but compliance was lower in the 800-mg arm (62% received doses >600 mg) compared with the 400-mg arm (87% received doses >350 mg).
Although the aforementioned studies have shown improved CCyR and MMR with a higher dose of imatinib, the follow-up of these studies is short to evaluate for EFS and OS. Hence, at the writing of this chapter, imatinib at a dose of 400mg daily is still the preferred regimen of choice in newly diagnosed patients with CML-CP.
Dasatinib
Dasatinib (Sprycel®, Bristol-Myers Squib, Princeton, NJ) is an orally bioavailable, multi-kinase inhibitor that is be 325 fold more potent than imatinib against unmutated BCR-ABL.31 It is currently approved for the treatment of imatinib-resistant or imatininb-intolerant CML in all phases and Ph-positive acute lymphoblastic leukemia (Ph+ ALL). The response to dasatinib among patients in chronic, accelerated and blast phase (myeloid and lymphoid) after imatinib failure are summarized in Table 2.32–34 Dasatinib is overall well tolerated. Myelosuppression occurs frequently, with grade 3 or 4 neutropenia or thrombocytopenia occurring in nearly 50% of patients treated at a dose of 70mg twice daily. The most common non-hematologic grade 3–4 toxicities at a dose of 70 mg twice daily were pleural effusion (9%), dyspnea (6%), bleeding (4%), diarrhea (3%), and fatigue (3%). In an open-label phase III trial, 670 patients with imatinib-resistant/intolerant CML-CP were randomly assigned between four dasatinib treatment schedules: 100 mg once daily, 50 mg twice daily, 140 mg once daily, or 70 mg twice daily.35 Results of this trial showed that 100 mg once daily retained its activity and was associated with less toxicity, particularly pleural effusion and myelosuppression, with grade 3–4 neutropenia or thrombocytopenia occurring in approximately 30% each.
Table 2.
Response | Percent Response | ||||||||||
---|---|---|---|---|---|---|---|---|---|---|---|
Dasatinib | Nilotinib | Bosutinib | |||||||||
CP N=387 |
AP n=174 |
MyBP n=109 |
LyBP n=48 |
CP n=321 |
AP N=137 |
MyBP N=105 |
LyBP N=31 |
CP N=146 |
AP N=51 |
BP N=38 |
|
Median follow-up (mo) | 15 | 14 | 12+ | 12+ | 24 | 9 | 3 | 3 | 7 | 6 | 3 |
% Resistant to imatinib | 74 | 93 | 91 | 88 | 70 | 80 | 82 | 82 | 69 | NR* | NR* |
% Hematologic Response | - | 79 | 50 | 40 | 94 | 56 | 22 | 19 | 85 | 54 | 36 |
CHR | 91 | 45 | 27 | 29 | 76 | 31 | 11 | 13 | 81 | 54 | 36 |
NEL | - | 19 | 7 | 6 | - | 12 | 1 | 0 | - | 0 | 0 |
% Cytogenetic Response | NR | 44 | 36 | 52 | NR | NR | NR | NR | - | NR | NR |
Complete | 49 | 32 | 26 | 46 | 46 | 20 | 29 | 32 | 34 | 27 | 35 |
Partial | 11 | 7 | 7 | 6 | 15 | 12 | 10 | 16 | 13 | 20 | 18 |
% Survival (at 12 months) | 96 (15) | 82 (12) | 50 (12) | 50 (5) | 87 (24) | 67 (24) | 42 (12) | 42 (12) | 98 (12) | 60 (12) | 50 (10) |
CP: chronic phase; AP: accelerated phase; MyBP: myeloid blast phase; LyBP: lymphoid blast phase, mo: months; CHR: complete hematologic response; NEL: no evidence of leukemia
Based on these results, a Phase II trial from MDACC was recently reported in 50 patients with newly diagnosed chronic phase CML.36 Ninety-eight percent achieved CCyR, and 41 patients (82%) achieved a MMR. Responses occurred rapidly, with 94% of patients achieving CCyR by 6 months. The projected EFS rate at 24 months was 88%. A randomized phase 3 trial comparing the efficacy of dasatinib and imatinib in the first-line has completed accrual, and results are expected in late 2010.
Nilotinib
Nilotinib (Tasigna®; Novartis Pharmaceuticals, East Hanover, NJ) is a rationally-designed BCR-ABL inhibitor that is 30-fold more potent than imatinib in vitro, with greater specificity for BCR-ABL.37,38 It is currently approved for treatment of imatinib-resistant/intolerant patients with CML-CP and CML–AP (but not BP or Ph+ ALL) at a dose of 400 mg twice daily (BID). The response to nilotinib among patients in chronic, accelerated and blast phase (myeloid and lymphoid) after imatinib failure are summarized in Table 2.39,40 The most common grade 3 or 4 laboratory abnormalities were elevated lipase (17%), hypophosphatemia (16%), hyperglycemia (12%), and elevated total bilirubin (8%). Grade 3 or 4 non-hematologic adverse events were infrequent, with rash, headache, and diarrhea occurring in 2% of patients. The most common grade 3 or 4 hematological adverse events were neutropenia (31%), thrombocytopenia (31%), and anemia (10%). Pleural or pericardial effusions (all grades) occurred in 2% of patients, and grade 3 or 4 pleural or pericardial effusions were rare (<1%).
Nilotinib has also demonstrated promise as a front-line therapy in patients with CML-CP.41–43 In the first head-to-head comparison of a second-generation TKI (nilotinib at 300 mg BID or 400 mg BID) to imatinib (400 mg/day), nilotinib 300 mg BID and 400 mg BID showed higher rates of MMR (44% and 43% respectively) and CCyR (80% and 78% respectively) than imatinib at 400 mg/day (MMR: 22% [P < 0.0001 vs. both nilotinib doses), CCyR: 65% [P < 0.0001 vs. nilotinib 300 mg BID; P < .0005 vs. nilotinib 400 mg BID) at 12 months of follow-up.42 In a Phase II study from MDACC, 51 patients with newly diagnosed CML-CP were treated with nilotinib at 400mg BID. Ninety-eight percent patients achieved CCyR, while 76% (39/51) achieved MMR. Rapid responses were observed, with 96% and 98% of patients in CCyR by 3 and 6 months respectively.41 A randomized phase 3 trial comparing the efficacy of nilotinib and imatinib in the first-line has completed accrual, and results are expected in late 2010.
Bosutinib
Bosutinib (SKI606), an orally available dual SRC/ABL inhibitor, is 30 to 50 times more potent than imatinib, with minimal inhibitory activity against C-Kit and PDGFR, therefore expected to produce less myelosuppression and fluid retention.44 The phase I study identified a treatment dose of 500 mg daily and showed evidence of clinical efficacy. Phase II studies in patients with CML-CP who have failed imatinib and second generation TKIs therapy are ongoing.45,46 Preliminary data for response to nilotinib among patients in chronic, accelerated and blast phase (myeloid and lymphoid) after imatinib failure are summarized in Table 2. The most common adverse events with bosutinib were gastrointestinal (nausea, vomiting, diarrhea); these were usually grade 1–2, manageable and transient, diminishing in frequency and severity after the first 3–4 weeks of treatment. Bosutinib is currently being assessed in the frontline setting for treatment of patients with CML-CP.
OTHER MULTIKINASE INHIBITORS
One of the most promising agents for treatment of T315I mutation in clinical trials is AP24534, an orally available multi-TKI designed using a structure-based approach as a pan-BCR-ABL inhibitor.47 AP24534 potently inhibits the enzymatic activity of BCR-ABL-T315I, the native enzyme and all other tested mutants. It also prevents the emergence of resistant mutants at concentrations of 40 nM. In a Phase 1 clinical trial of AP24534 at doses from 2–60 mg in 27 patients with CML (19 with CP, 4 AP and 4 BP), complete hematologic response (CHR) was achieved or maintained in 83% of patients treated in CP; major hematologic responses were also achieved in 38% of patients treated in advanced stages of the disease.48 More importantly, 9 of 20 patients treated in CP achieved a MCyR (including 5 CCyR), including 3 of 7 with T315I (2 CCyR). The most common drug-related adverse events were elevations of lipase and amylase at a dose of 60mg daily. Grade 3 or 4 thrombocytopenia occurred in 9% of patients, with no grade 3–4 drug-related neutropenia. AP24534 will be tested in a large, multicenter study focusing on patients with imatinib-, nilotinib-, and dasatinib-resistant disease, including a subset with the T315I mutation.
XL228 (Exelixis Inc, San Francisco, USA) is a potent, multitargeted kinase inhibitor with potent activity against wild-type and T315I isoforms of BCR-ABL.49 In a prelim Phase 1 clinical study, XL228 was administered to 27 patients in six cohorts with a once-weekly dosing schedule (dose range from 0.45 mg/kg to 10.8 mg/kg). All patients were resistant or intolerant to at least two prior standard therapies (including imatinib, dasatinib, and nilotinib) or had a known BCR-ABL T315I mutation. Preliminary evidence of clinical activity was observed in patients treated at doses of 3.6 mg/kg and higher, including stable or decreasing white blood cell and/or platelet count within 2 months (in 14 patients, 5 with T315I), and/or >1-log reduction in BCR-ABL transcript levels by reverse transcriptase-polymerase chain reaction (RT-PCR) within 3 months (in 3 patients, 2 with T315I). XL 228 has been generally well tolerated. Dose limiting toxicities observed with once weekly dosing included grade 3 syncope and hyperglycemia in two patients dosed at 10.8 mg/kg. The most commonly reported grade 2 adverse effects were hyperglycemia, fatigue, nausea, vomiting, and bradycardia.
NON-TYROSINE KINASE INHIBITORS
Homoharringtonine is a plant alkaloid that has been used in China for many years in the treatment of patients with acute myeloid leukemia. Before the introduction of imatinib, it was the best treatment option for patients who failed IFN-α and were not transplant candidates, with cytogenetic responses in approximately 30% of patients.50,51 Omacetaxine mepesuccinate, a cephalotaxine ester and a derivative of homoharringtonine that has excellent bioavailability through the subcutaneous route, is a multitargeted protein synthase inhibitor that has been in clinical development for several years. Omacetaxine shows clinical activity against CML with a mechanism of action independent of tyrosine kinase inhibition and is thus not affected by the presence of mutations.52,53 In a recently reported Phase 2/3 clinical study of omacetaxine administered at a dose of 1.25 mg/m2 sc twice daily for 7 days (every 28 days) to 89 patients with CML (44 CP, 25 AP and 20 BP) who are either intolerant or resistant to at least 2 TKI’s (imatinib, dasatinib or nilotinib), the rates of CHR and MCyR were 82% and 23% in CP, respectively.54 In a similar trial enrolling 81 patients (49 CP, 17 AP and 15 BP) with T315I mutation who did not respond to imatinib; omacetaxine led to a CHR in 86% and MCyR in 27% among patients treated in CCyR. These responses were durable.55 The most commonly reported events were thrombocytopenia (58%), anemia (36%) and neutropenia (33%). Non-hematologic toxicities were primarily grade ½ with the most frequently reported events of diarrhea (44%), fatigue (35%), pyrexia (32%), nausea (26%), and asthenia (21%).
MUTATION STATUS AND CHOICE OF THERAPY
Although more than 100 BCR-ABL mutations have been identified in clinical samples,18 the presence of a mutation does not typically lead to resistance. Baseline mutation screening for newly diagnosed patients with CML has shown no benefit for predicting response, 56 and should not be routinely employed. In a study using highly sensitive DNA sequencing techniques, patients treated with imatinib showed no correlation between baseline mutation status and response, PFS or OS. Other studies have confirmed that the identification of mutations pre-therapy does not predict insensitivity to imatinib.57–59
The utility of using in vitro mutation data to select a second generation TKI remains a matter of controversy. In their seminal paper, Redaelli et al. report on the in vitro activity of nilotinib, dasatinib and bosutinib against 18 BCR-ABL mutations (Table 3).44 The 8 most common mutations (T315I, Y253F/H, E255D/K/R/V, M351T, G250A/E, F359C/L/V, H396P/R, M244V); found in 85% of patients with mutations were included in the analysis. The mutations were stratified using half maximal inhibitory concentration (IC50) values into sensitive, moderately resistant, resistant, or highly resistant. The authors conclude that this data offers physicians a tool for selecting a patient tailored TKI therapy. One of the major criticisms for using in vitro data in selecting the next line of therapy is that it does not fully predict the in vivo response.60 In a recent publication, Laneuville et al. report that adequate drug exposure to inhibit the BCR-ABL kinase located in the cytoplasm of leukemic cells requires satisfactory pharmacokinetics, which are affected by independent variables that might be related to molecular structure of the drug itself. They also note that the table as constructed in the Redaelli article does not allow a side-by-side comparison of data, as columns for each inhibitor are normalized to the data within that column. Indeed, none of these studies take into account factors such as protein binding and cell influx/efflux or a variety of other in vivo factors that could affect results. Therefore, until more definitive results are published, treating physicians must not solely rely on in vitro data to select the next TKI for their patients who are imatinib-resistant/intolerant.
Table 3.
IC50-fold increase (WT=1)
|
||||
---|---|---|---|---|
Imatinib | Bosutinib | Dasatinib | Nilotinib | |
WT | 1 | 1 | 1 | 1 |
L248V | 3.54 | 2.97 | 5.11 | 2.80 |
G250E | 6.86 | 4.31 | 4.45 | 4.56 |
Q252H | 1.39 | 0.31 | 3.05 | 2.64 |
Y253F | 3.58 | 0.96 | 1.58 | 3.23 |
E255K | 6.02 | 9.47 | 5.61 | 6.69 |
E255V | 16.99 | 5.53 | 3.44 | 10.31 |
D276G | 2.18 | 0.60 | 1.44 | 2.00 |
E279K | 3.55 | 0.95 | 1.64 | 2.05 |
V299L | 1.54 | 26.10 | 8.65 | 1.34 |
T315I | 17.50 | 45.42 | 75.03 | 39.41 |
F317L | 2.60 | 2.42 | 4.46 | 2.22 |
M351T | 1.76 | 0.70 | 0.88 | 0.44 |
F359V | 2.86 | 0.93 | 1.49 | 5.16 |
L384M | 1.28 | 0.47 | 2.21 | 2.33 |
H396P | 2.43 | 0.43 | 1.07 | 2.41 |
H396R | 3.91 | 0.81 | 1.63 | 3.10 |
G398R | 0.35 | 1.16 | 0.69 | 0.49 |
F486S | 8.10 | 2.31 | 3.04 | 1.85 |
Mutations can be classified as sensitive (IC50 fold increase ≤2), resistant (between 2.01 and 10) or highly resistant (>10; T315I mutation)
Prospective clinical studies evaluating the choice of second generation TKI’s based on in vitro sensitivity data in imatinib intolerant/resistant patients are limited. In a retrospective analysis of 169 imatinib-resistant patients treated with a second generation TKI at MDACC, 86 were found to have a mutation.61 Forty-one patients were treated with dasatinib and 45 with nilotinib. Mutations were stratified on the basis of IC50 values into high (n=42), intermediate (n=25), low (T315I, n=9), and unknown (n=10). Although response rates tended to be higher in patients without baseline mutations, there were no significant differences in CHR, MCyR, or CCyR between patients with and without baseline mutations. Response rates were higher in patients with CML-CP with low IC50 mutations, compared with intermediate IC50 mutations. The existence of a mutation at baseline was not shown to impact overall survival, but the presence of intermediate IC50 mutations was significantly associated with poorer EFS (p = 0.0006) and OS (p = 0.03).
Among 1043 patients treated with second-line dasatinib in phase 2/3 trials, 39% had a preexisting BCR-ABL mutation, including 48% of 805 patients with imatinib resistance or suboptimal response.62 Sixty-three different BCR-ABL mutations affecting 49 amino acids were detected at baseline, with G250, M351, M244, and F359 most frequently affected. After 2 years of follow-up, dasatinib treatment of imatinib-resistant patients with or without a mutation resulted in notable response rates (CCyR: 43% vs 47%) and durable PFS (70% vs 80%). Impaired responses were observed with some mutations with a dasatinib median IC50 greater than 3nM; among patients with mutations with lower or unknown IC50, efficacy was comparable with those with no mutation. In a subanalysis of a phase II study of nilotinib in patients with imatinib-resistant or imatinib-intolerant CML-CP, baseline mutation data were assessed in 281 (88%) of 321 patients.63 Among imatinib-resistant patients, the frequency of mutations at baseline was 55%. After 12 months of therapy, MCyR was achieved in 60%, CCyR in 40%, and MMR in 29% of patients without baseline mutations versus 49% (P = 0.145), 32% (P = 0.285), and 22% (P = 0.366), respectively, of patients with mutations. Patients with mutations that were less sensitive to nilotinib in vitro (IC50 > 150 nM; Y253H, E255V/K, F359V/C) had less favorable responses, as 13%, 43%, and 9% of patients with each of these mutations, respectively, achieved MCyR; none achieved CCyR.
CURRENT RECOMMENDATIONS FOR TREATMENT OF CML
A proposed approach to the management of patients with CML is depicted in Figure 1. Imatinib at a dose of 400 mg/day is considered the standard treatment. If patients do not achieve the landmarks as established by the ELN, modification to this therapy should be strongly considered. Dose escalation of imatinib can be considered, but is not likely to be effective in patients who never achieved a cytogenetic response on imatinib or those with known imatinib-resistant mutations. A change to a second-generation therapy may be a better option for most patients. In vitro and in vivo data have demonstrated that both dasatinib and nilotinib have a small and distinct set of mutants that confer decreased sensitivity: Y253H, E255K/V, and F359C/V for nilotinib and Q252H, E255K/V, V299L, and F317L for dasatinib. Therefore, if the mutation analysis reveals any of these mutations, that particular second generation TKI should be avoided.
For the vast majority of patients who do not harbor a mutation, choice for a second generation TKI is based on co-morbid conditions present. Dasatinib use is associated with the development of pleural and pericardial effusion,64 bleeding65 and infection66. Therefore, caution should be exercised before prescribing dasatinib in patients with hypertension, asthma, pneumonia, gastrointestinal bleeding, chronic obstructive pulmonary disease, chest wall injury, congestive heart failure, auto-immune disorders and concomitant aspirin use. Severe, uncontrolled diabetes and past pancreatitis are considered risk factors for nilotinib use due to the occurrence of grade 3/4 lipase elevation (18%), bilirubin elevation (7%) and hyperglycemia (12%). QT prolongation is a concern with both agents, and the simultaneous use of agents prolonging the QT interval should be avoided. Although both dasatinib and nilotinib are ineffective against T315I BCR-ABL, this mutation is more likely to affect patients in the advanced phases of CML. Patients with T315I may achieve favorable outcomes with other therapies, e.g AP24534, omacetaxine. SCT is generally reserved for patients who have not responded to a second or third generation TKI and for those patients with T315I mutation who have not responded to newer agents.
CONCLUSION
Imatinib has dramatically altered the landscape of treatment for patients with CML. For most patients, the long-term outcomes including the PFS and OS are excellent. For a few subset of patients who are intolerant to or are resistant to imatinib, newer second generation TKI’s are becoming excellent choices of therapy. The mechanisms of resistance, in vivo and in vitro sensitivities and choice of agents are rapidly evolving. It is hoped that in the near future preclinical and clinical data will become available that will guide the treating physician to select the best TKI, both in the frontline and relapsed setting, for an individual patient with CML.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ. Cancer statistics, 2009. CA Cancer J Clin. 2009;59:225–249. doi: 10.3322/caac.20006. [DOI] [PubMed] [Google Scholar]
- 2.Bartram CR, de Klein A, Hagemeijer A, et al. Translocation of c-ab1 oncogene correlates with the presence of a Philadelphia chromosome in chronic myelocytic leukaemia. Nature. 1983;306:277–280. doi: 10.1038/306277a0. [DOI] [PubMed] [Google Scholar]
- 3.Rowley JD. Letter: A new consistent chromosomal abnormality in chronic myelogenous leukaemia identified by quinacrine fluorescence and Giemsa staining. Nature. 1973;243:290–293. doi: 10.1038/243290a0. [DOI] [PubMed] [Google Scholar]
- 4.Druker BJ, Tamura S, Buchdunger E, et al. Effects of a selective inhibitor of the Abl tyrosine kinase on the growth of Bcr-Abl positive cells. Nat Med. 1996;2:561–566. doi: 10.1038/nm0596-561. [DOI] [PubMed] [Google Scholar]
- 5.Druker BJ, Talpaz M, Resta DJ, et al. Efficacy and safety of a specific inhibitor of the BCR-ABL tyrosine kinase in chronic myeloid leukemia. N Engl J Med. 2001;344:1031–1037. doi: 10.1056/NEJM200104053441401. [DOI] [PubMed] [Google Scholar]
- 6.Kantarjian H, Sawyers C, Hochhaus A, et al. Hematologic and cytogenetic responses to imatinib mesylate in chronic myelogenous leukemia. N Engl J Med. 2002;346:645–652. doi: 10.1056/NEJMoa011573. [DOI] [PubMed] [Google Scholar]
- 7.O’Brien SG, Guilhot F, Larson RA, et al. Imatinib compared with interferon and low-dose cytarabine for newly diagnosed chronic-phase chronic myeloid leukemia. N Engl J Med. 2003;348:994–1004. doi: 10.1056/NEJMoa022457. [DOI] [PubMed] [Google Scholar]
- 8.Deininger M, O’Brien SG, Guilhot F, et al. International Randomized Study of Interferon Vs STI571 (IRIS) 8-Year Follow up: Sustained Survival and Low Risk for Progression or Events in Patients with Newly Diagnosed Chronic Myeloid Leukemia in Chronic Phase (CML-CP) Treated with Imatinib. ASH Annual Meeting Abstracts. 2009;114:1126. [Google Scholar]
- 9.Zhang WW, Cortes JE, Yao H, et al. Predictors of primary imatinib resistance in chronic myelogenous leukemia are distinct from those in secondary imatinib resistance. J Clin Oncol. 2009;27:3642–3649. doi: 10.1200/JCO.2008.19.4076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.le Coutre P, Tassi E, Varella-Garcia M, et al. Induction of resistance to the Abelson inhibitor STI571 in human leukemic cells through gene amplification. Blood. 2000;95:1758–1766. [PubMed] [Google Scholar]
- 11.Gorre ME, Mohammed M, Ellwood K, et al. Clinical resistance to STI-571 cancer therapy caused by BCR-ABL gene mutation or amplification. Science. 2001;293:876–880. doi: 10.1126/science.1062538. [DOI] [PubMed] [Google Scholar]
- 12.Weisberg E, Griffin JD. Mechanism of resistance to the ABL tyrosine kinase inhibitor STI571 in BCR/ABL-transformed hematopoietic cell lines. Blood. 2000;95:3498–3505. [PubMed] [Google Scholar]
- 13.Branford S, Rudzki Z, Walsh S, et al. Detection of BCR-ABL mutations in patients with CML treated with imatinib is virtually always accompanied by clinical resistance, and mutations in the ATP phosphate-binding loop (P-loop) are associated with a poor prognosis. Blood. 2003;102:276–283. doi: 10.1182/blood-2002-09-2896. [DOI] [PubMed] [Google Scholar]
- 14.Hochhaus A, Kreil S, Corbin AS, et al. Molecular and chromosomal mechanisms of resistance to imatinib (STI571) therapy. Leukemia. 2002;16:2190–2196. doi: 10.1038/sj.leu.2402741. [DOI] [PubMed] [Google Scholar]
- 15.Corbin AS, La Rosee P, Stoffregen EP, Druker BJ, Deininger MW. Several Bcr-Abl kinase domain mutants associated with imatinib mesylate resistance remain sensitive to imatinib. Blood. 2003;101:4611–4614. doi: 10.1182/blood-2002-12-3659. [DOI] [PubMed] [Google Scholar]
- 16.Soverini S, Colarossi S, Gnani A, et al. Contribution of ABL kinase domain mutations to imatinib resistance in different subsets of Philadelphia-positive patients: by the GIMEMA Working Party on Chronic Myeloid Leukemia. Clin Cancer Res. 2006;12:7374–7379. doi: 10.1158/1078-0432.CCR-06-1516. [DOI] [PubMed] [Google Scholar]
- 17.Jabbour E, Kantarjian H, Jones D, et al. Long-Term Incidence and Outcome of BCR-ABL Mutations in Patients (pts) with Chronic Myeloid Leukemia (CML) Treated with Imatinib Mesylate - P-Loop Mutations Are Not Associated with Worse Outcome. Blood (ASH Annual Meeting Abstracts) 2004;104:1007. [Google Scholar]
- 18.Jabbour E, Kantarjian H, Jones D, et al. Frequency and clinical significance of BCR-ABL mutations in patients with chronic myeloid leukemia treated with imatinib mesylate. Leukemia. 2006;20:1767–1773. doi: 10.1038/sj.leu.2404318. [DOI] [PubMed] [Google Scholar]
- 19.Soverini S, Martinelli G, Amabile M, et al. Denaturing-HPLC-based assay for detection of ABL mutations in chronic myeloid leukemia patients resistant to Imatinib. Clin Chem. 2004;50:1205–1213. doi: 10.1373/clinchem.2004.031112. [DOI] [PubMed] [Google Scholar]
- 20.Deininger MW, McGreevey L, Willis S, Bainbridge TM, Druker BJ, Heinrich MC. Detection of ABL kinase domain mutations with denaturing high-performance liquid chromatography. Leukemia. 2004;18:864–871. doi: 10.1038/sj.leu.2403307. [DOI] [PubMed] [Google Scholar]
- 21.Apperley JF. Part I: mechanisms of resistance to imatinib in chronic myeloid leukaemia. Lancet Oncol. 2007;8:1018–1029. doi: 10.1016/S1470-2045(07)70342-X. [DOI] [PubMed] [Google Scholar]
- 22.Hochhaus A, La Rosee P. Imatinib therapy in chronic myelogenous leukemia: strategies to avoid and overcome resistance. Leukemia. 2004;18:1321–1331. doi: 10.1038/sj.leu.2403426. [DOI] [PubMed] [Google Scholar]
- 23.National Comprehensive Cancer Network. Clinical Practice Guidelines in Oncology. 2010. Version 2. [Google Scholar]
- 24.Baccarani M, Cortes J, Pane F, et al. Chronic myeloid leukemia: an update of concepts and management recommendations of European LeukemiaNet. J Clin Oncol. 2009;27:6041–6051. doi: 10.1200/JCO.2009.25.0779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Press RD, Willis SG, Laudadio J, Mauro MJ, Deininger MW. Determining the rise in BCR-ABL RNA that optimally predicts a kinase domain mutation in patients with chronic myeloid leukemia on imatinib. Blood. 2009;114:2598–2605. doi: 10.1182/blood-2008-08-173674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Kantarjian HM, Larson RA, Guilhot F, et al. Efficacy of imatinib dose escalation in patients with chronic myeloid leukemia in chronic phase. Cancer. 2009;115:551–560. doi: 10.1002/cncr.24066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Jabbour E, Kantarjian HM, Jones D, et al. Imatinib mesylate dose escalation is associated with durable responses in patients with chronic myeloid leukemia after cytogenetic failure on standard-dose imatinib therapy. Blood. 2009;113:2154–2160. doi: 10.1182/blood-2008-04-154344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Cortes JE, Kantarjian HM, Goldberg SL, et al. High-dose imatinib in newly diagnosed chronic-phase chronic myeloid leukemia: high rates of rapid cytogenetic and molecular responses. J Clin Oncol. 2009;27:4754–4759. doi: 10.1200/JCO.2008.20.3869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Baccarani M, Druker BJ, Cortes-Franco J, et al. 24 Months Update of the TOPS Study: a Phase III, Randomized, Open-Label Study of 400mg/d (SD-IM) Versus 800mg/d (HD-IM) of Imatinib Mesylate (IM) in Patients (Pts) with Newly Diagnosed, Previously Untreated Chronic Myeloid Leukemia in Chronic Phase (CML-CP) Blood (ASH Annual Meeting Abstracts) 2009;114:337. [Google Scholar]
- 30.Baccarani M, Rosti G, Castagnetti F, et al. Comparison of imatinib 400 mg and 800 mg daily in the front-line treatment of high-risk, Philadelphia-positive chronic myeloid leukemia: a European LeukemiaNet Study. Blood. 2009;113:4497–4504. doi: 10.1182/blood-2008-12-191254. [DOI] [PubMed] [Google Scholar]
- 31.Lombardo LJ, Lee FY, Chen P, et al. Discovery of N-(2-chloro-6-methyl- phenyl)-2-(6-(4-(2-hydroxyethyl)- piperazin-1-yl)-2-methylpyrimidin-4- ylamino)thiazole-5-carboxamide (BMS-354825), a dual Src/Abl kinase inhibitor with potent antitumor activity in preclinical assays. J Med Chem. 2004;47:6658–6661. doi: 10.1021/jm049486a. [DOI] [PubMed] [Google Scholar]
- 32.Cortes J, Rousselot P, Kim DW, et al. Dasatinib induces complete hematologic and cytogenetic responses in patients with imatinib-resistant or -intolerant chronic myeloid leukemia in blast crisis. Blood. 2007;109:3207–3213. doi: 10.1182/blood-2006-09-046888. [DOI] [PubMed] [Google Scholar]
- 33.Guilhot F, Apperley J, Kim DW, et al. Dasatinib induces significant hematologic and cytogenetic responses in patients with imatinib-resistant or -intolerant chronic myeloid leukemia in accelerated phase. Blood. 2007;109:4143–4150. doi: 10.1182/blood-2006-09-046839. [DOI] [PubMed] [Google Scholar]
- 34.Hochhaus A, Baccarani M, Deininger M, et al. Dasatinib induces durable cytogenetic responses in patients with chronic myelogenous leukemia in chronic phase with resistance or intolerance to imatinib. Leukemia. 2008;22:1200–1206. doi: 10.1038/leu.2008.84. [DOI] [PubMed] [Google Scholar]
- 35.Shah NP, Kantarjian HM, Kim DW, et al. Intermittent target inhibition with dasatinib 100 mg once daily preserves efficacy and improves tolerability in imatinib-resistant and -intolerant chronic-phase chronic myeloid leukemia. J Clin Oncol. 2008;26:3204–3212. doi: 10.1200/JCO.2007.14.9260. [DOI] [PubMed] [Google Scholar]
- 36.Cortes JE, Jones D, O’Brien S, et al. Results of Dasatinib Therapy in Patients With Early Chronic-Phase Chronic Myeloid Leukemia. J Clin Oncol. 28:398–404. doi: 10.1200/JCO.2009.25.4920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Golemovic M, Verstovsek S, Giles F, et al. AMN107, a novel aminopyrimidine inhibitor of Bcr-Abl, has in vitro activity against imatinib-resistant chronic myeloid leukemia. Clin Cancer Res. 2005;11:4941–4947. doi: 10.1158/1078-0432.CCR-04-2601. [DOI] [PubMed] [Google Scholar]
- 38.Weisberg E, Manley PW, Breitenstein W, et al. Characterization of AMN107, a selective inhibitor of native and mutant Bcr-Abl. Cancer Cell. 2005;7:129–141. doi: 10.1016/j.ccr.2005.01.007. [DOI] [PubMed] [Google Scholar]
- 39.Hocchaus AGF, Appereley J, et al. Nilotinib in chronic myeloid leukemia patients in accelerated phase (CML-AP) with imatinib resistance or intolerance: 24-month follow-up results of a phase 2 study. Haematologica. 2009;94(suppl 2):Abstract 0631. [Google Scholar]
- 40.Kantarjian HM, Giles FJ, Bhalla KN, et al. Update On Imatinib-Resistant Chronic Myeloid Leukemia Patients in Chronic Phase (CML-CP) On Nilotinib Therapy at 24 Months: Clinical Response, Safety, and Long-Term Outcomes. ASH Annual Meeting Abstracts. 2009;114:1129. [Google Scholar]
- 41.Cortes JE, Jones D, O’Brien S, et al. Nilotinib as front-line treatment for patients with chronic myeloid leukemia in early chronic phase. J Clin Oncol. 28:392–397. doi: 10.1200/JCO.2009.25.4896. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.O’Dwyer MC, Kent E, Parker M, et al. Nilotinib 300 Mg Twice Daily Is Effective and Well Tolerated as First Line Treatment of Ph-Positive Chronic Myeloid Leukemia in Chronic Phase: Preliminary Results of the ICORG 0802 Phase 2 Study. ASH Annual Meeting Abstracts. 2009;114:3294. [Google Scholar]
- 43.Rosti G, Palandri F, Castagnetti F, et al. Nilotinib for the frontline treatment of Ph(+) chronic myeloid leukemia. Blood. 2009;114:4933–4938. doi: 10.1182/blood-2009-07-232595. [DOI] [PubMed] [Google Scholar]
- 44.Redaelli S, Piazza R, Rostagno R, et al. Activity of bosutinib, dasatinib, and nilotinib against 18 imatinib-resistant BCR/ABL mutants. J Clin Oncol. 2009;27:469–471. doi: 10.1200/JCO.2008.19.8853. [DOI] [PubMed] [Google Scholar]
- 45.Cortes J, Kantarjian HM, Kim D-W, et al. Efficacy and Safety of Bosutinib (SKI-606) in Patients with Chronic Phase (CP) Ph+ Chronic Myelogenous Leukemia (CML) with Resistance or Intolerance to Imatinib. Blood (ASH Annual Meeting Abstracts) 2008;112:1098. [Google Scholar]
- 46.Gambacorti-Passerini C, Kantarjian H, Bruemmendorf T, et al. Bosutinib (SKI-606) Demonstrates Clinical Activity and Is Well Tolerated among Patients with AP and BP CML and Ph+ ALL. Blood (ASH Annual Meeting Abstracts) 2007;110:473. [Google Scholar]
- 47.O’Hare T, Shakespeare WC, Zhu X, et al. AP24534, a pan-BCR-ABL inhibitor for chronic myeloid leukemia, potently inhibits the T315I mutant and overcomes mutation-based resistance. Cancer Cell. 2009;16:401–412. doi: 10.1016/j.ccr.2009.09.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Cortes J, Talpaz M, Deininger M, et al. A Phase 1 Trial of Oral AP24534 in Patients with Refractory Chronic Myeloid Leukemia and Other Hematologic Malignancies: First Results of Safety and Clinical Activity against T315I and Resistant Mutations. Blood (ASH Annual Meeting Abstracts) 2009;114:643. [Google Scholar]
- 49.Cortes J, Paquette R, Talpaz M, et al. Preliminary Clinical Activity in a Phase I Trial of the BCR-ABL/IGF- 1R/Aurora Kinase Inhibitor XL228 in Patients with Ph++ Leukemias with Either Failure to Multiple TKI Therapies or with T315I Mutation. Blood (ASH Annual Meeting Abstracts) 2008;112:3232. [Google Scholar]
- 50.O’Brien S, Kantarjian H, Koller C, et al. Sequential homoharringtonine and interferon-alpha in the treatment of early chronic phase chronic myelogenous leukemia. Blood. 1999;93:4149–4153. [PubMed] [Google Scholar]
- 51.O’Brien S, Talpaz M, Cortes J, et al. Simultaneous homoharringtonine and interferon-alpha in the treatment of patients with chronic-phase chronic myelogenous leukemia. Cancer. 2002;94:2024–2032. doi: 10.1002/cncr.10436. [DOI] [PubMed] [Google Scholar]
- 52.Kantarjian HM, Talpaz M, Santini V, Murgo A, Cheson B, O’Brien SM. Homoharringtonine: history, current research, and future direction. Cancer. 2001;92:1591–1605. doi: 10.1002/1097-0142(20010915)92:6<1591::aid-cncr1485>3.0.co;2-u. [DOI] [PubMed] [Google Scholar]
- 53.O’Brien S, Kantarjian H, Keating M, et al. Homoharringtonine therapy induces responses in patients with chronic myelogenous leukemia in late chronic phase. Blood. 1995;86:3322–3326. [PubMed] [Google Scholar]
- 54.Cortes-Franco J, Raghunadharao D, Parikh P, et al. Safety and Efficacy of Subcutaneous-Administered Omacetaxine Mepesuccinate in Chronic Myeloid Leukemia (CML) Patients Who Are Resistant or Intolerant to Two or More Tyrosine Kinase Inhibitors - Results of A Multicenter Phase 2/3 Study. Blood (ASH Annual Meeting Abstracts) 2009;114:861. [Google Scholar]
- 55.Cortes-Franco J, Khoury HJ, Nicolini FE, et al. Safety and Efficacy of Subcutaneous-Administered Omacetaxine Mepesuccinate in Imatinib-Resistant Chronic Myeloid Leukemia (CML) Patients Who Harbor the Bcr- Abl T315I Mutation-Results of An Ongoing Multicenter Phase 2/3 Study. Blood (ASH Annual Meeting Abstracts) 2009;114:644. [Google Scholar]
- 56.Willis SG, Lange T, Demehri S, et al. High-sensitivity detection of BCR-ABL kinase domain mutations in imatinib-naive patients: correlation with clonal cytogenetic evolution but not response to therapy. Blood. 2005;106:2128–2137. doi: 10.1182/blood-2005-03-1036. [DOI] [PubMed] [Google Scholar]
- 57.Khorashad JS, Anand M, Marin D, et al. The presence of a BCR-ABL mutant allele in CML does not always explain clinical resistance to imatinib. Leukemia. 2006;20:658–663. doi: 10.1038/sj.leu.2404137. [DOI] [PubMed] [Google Scholar]
- 58.Mauro MJ. Mutational analysis and overcoming imatinib resistance in chronic myeloid leukemia with novel tyrosine kinase inhibitors. Curr Treat Options Oncol. 2007;8:287–295. doi: 10.1007/s11864-007-0036-y. [DOI] [PubMed] [Google Scholar]
- 59.Sherbenou DW, Wong MJ, Humayun A, et al. Mutations of the BCR-ABL-kinase domain occur in a minority of patients with stable complete cytogenetic response to imatinib. Leukemia. 2007;21:489–493. doi: 10.1038/sj.leu.2404554. [DOI] [PubMed] [Google Scholar]
- 60.Laneuville P, Dilea C, Yin OQ, Woodman RC, Mestan J, Manley PW. Comparative In vitro cellular data alone are insufficient to predict clinical responses and guide the choice of BCR-ABL inhibitor for treating imatinib-resistant chronic myeloid leukemia. J Clin Oncol. 28:e169–171. doi: 10.1200/JCO.2009.26.4945. author reply e172. [DOI] [PubMed] [Google Scholar]
- 61.Jabbour E, Jones D, Kantarjian HM, et al. Long-term outcome of patients with chronic myeloid leukemia treated with second-generation tyrosine kinase inhibitors after imatinib failure is predicted by the in vitro sensitivity of BCR-ABL kinase domain mutations. Blood. 2009;114:2037–2043. doi: 10.1182/blood-2009-01-197715. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Muller MC, Cortes JE, Kim DW, et al. Dasatinib treatment of chronic-phase chronic myeloid leukemia: analysis of responses according to preexisting BCR-ABL mutations. Blood. 2009;114:4944–4953. doi: 10.1182/blood-2009-04-214221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Hughes T, Saglio G, Branford S, et al. Impact of baseline BCR-ABL mutations on response to nilotinib in patients with chronic myeloid leukemia in chronic phase. J Clin Oncol. 2009;27:4204–4210. doi: 10.1200/JCO.2009.21.8230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Quintas-Cardama A, Kantarjian H, O’Brien S, et al. Pleural effusion in patients with chronic myelogenous leukemia treated with dasatinib after imatinib failure. J Clin Oncol. 2007;25:3908–3914. doi: 10.1200/JCO.2007.12.0329. [DOI] [PubMed] [Google Scholar]
- 65.Quintas-Cardama A, Kantarjian H, Ravandi F, et al. Bleeding diathesis in patients with chronic myelogenous leukemia receiving dasatinib therapy. Cancer. 2009;115:2482–2490. doi: 10.1002/cncr.24257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Sillaber C, Herrmann H, Bennett K, et al. Immunosuppression and atypical infections in CML patients treated with dasatinib at 140 mg daily. Eur J Clin Invest. 2009;39:1098–1109. doi: 10.1111/j.1365-2362.2009.02206.x. [DOI] [PubMed] [Google Scholar]