Blast crisis (BC) remains the major challenge in the management of chronic myeloid leukemia (CML). It is now generally accepted that BC is the consequence of continued BCR-ABL activity leading to genetic instability, DNA damage, and impaired DNA repair. Most patients with BC carry multiple mutations, and up to 80% show additional chromosomal aberrations in a nonrandom pattern. Treatment with tyrosine kinase inhibitors has improved survival in BC modestly, but most long-term survivors are those who have been transplanted. Patients in BC should be treated with a tyrosine kinase inhibitor according to mutation profile, with or without chemotherapy, with the goal of achieving a second chronic phase and proceeding to allogeneic stem cell transplantation as quickly as possible. Although long-term remissions are rare, allogeneic stem cell transplantation provides the best chance of a cure in BC. Investigational agents are not likely to provide an alternative in the near future. In view of these limited options, prevention of BC by a rigorous and early elimination of BCR-ABL is recommended. Early response indicators should be used to select patients for alternative therapies and early transplantation. Every attempt should be made to reduce or eliminate BCR-ABL consistent with good patient care as far as possible.

Blast crisis (BC) is the major remaining challenge in the management of chronic myeloid leukemia (CML). The introduction of an inhibitor targeted at the BCR-ABL tyrosine kinase (imatinib) has fundamentally changed treatment of CML.1  BCR-ABL expression can be reduced by imatinib to very low or nondetectable levels in the majority of patients.2  Median survival in chronic phase (CP) is estimated at a median of 25 to 30 years. Progress to advanced phase CML or BC has been reduced to 1% to 1.5% per year1  compared with more than 20% per year in the pre-imatinib era.3  Prevalence of CML is estimated to increase by a factor of approximately 10 within the next 40 years.4  Once BC has appeared, however, the prognosis of imatinib-treated patients is not much better than that after conventional therapy.5  Median survival after diagnosis of BC currently ranges between 7 and 11 months compared with 3 to 4 months in the pre-imatinib era. Very few long-term survivors after diagnosis of BC have been reported. Most of these represent recipients of transplants during a second CP. The therapeutic dilemma of BC has recently been well summarized.6  More research is needed to fully understand the mechanisms underlying progression to BC. It is distressing that in CML BC a true malignancy evolves under our eyes. The 2 current burning questions in CML are: How can we best manage patients who progress to BC despite appropriate treatment? How can we best prevent BC?

First attempts at the definition of BC date back more than forty years.7  The generally used definition, which underlies virtually all current clinical CML trials and the European LeukemiaNet management recommendations, rests on at least 30% blasts in blood or marrow or the demonstration of extramedullary blastic infiltrates.8  The more recent World Health Organization definition proposes a blast count of 20% in analogy to the definition of acute myeloid leukemia (AML).9  Both definitions are not supported by biologic evidence. A new definition would regroup approximately 10% of patients.10  Patients with 20% to 29% blasts have significantly better prognoses than patients with more than 30% blasts. Because most clinicians and trialists would probably use the definition based on their own data and experience, I suggest awaiting the results of clinical and biologic research for a new evidence-based definition of BC.

To diagnose BC, I do complete blood and differential counts and a bone marrow analysis with cytogenetics (Table 1). Cytogenetic evolution is the most consistent predictor of blast transformation. Flow cytometry or cytochemistry is needed to determine the type of BC (myeloid or lymphoid). Molecular genetics with mutation analysis are needed to choose the appropriate tyrosine kinase inhibitor (TKI). Consensus recommendations when to perform mutation analyses have been published on behalf of the European LeukemiaNet.11  A donor search for allogeneic stem cell transplantation (allo-SCT) should be started immediately.

Table 1

BC diagnostics

Test rationale
Test at diagnosis of BC  
    CBC with differential and bone marrow Proportions of blasts, promyelocytes, and basophils? 
    Flow cytometry and/or cytochemistry Myeloid or lymphoid phenotype? 
    Cytogenetics Clonal evolution? 
    Molecular genetics Mutation profile? Choice of TKI 
    Donor search (if applicable) Allo-SCT 
Follow-up under therapy  
    Blood count and differential Return to CP? 
    Bone marrow and cytogenetics Ascertainment of second CP 
    Molecular genetics Monitoring of BCR-ABL transcript levels under TKI and after allo-SCT 
    In lymphoid BC: CSF cytology Intrathecal instillation for neuroprophylaxis 
Test rationale
Test at diagnosis of BC  
    CBC with differential and bone marrow Proportions of blasts, promyelocytes, and basophils? 
    Flow cytometry and/or cytochemistry Myeloid or lymphoid phenotype? 
    Cytogenetics Clonal evolution? 
    Molecular genetics Mutation profile? Choice of TKI 
    Donor search (if applicable) Allo-SCT 
Follow-up under therapy  
    Blood count and differential Return to CP? 
    Bone marrow and cytogenetics Ascertainment of second CP 
    Molecular genetics Monitoring of BCR-ABL transcript levels under TKI and after allo-SCT 
    In lymphoid BC: CSF cytology Intrathecal instillation for neuroprophylaxis 

BC indicates blast crisis; CP, chronic phase; CSF, cerebrospinal fluid; CBC, complete blood count; and TKI, tyrosine kinase inhibitor.

Clinically, BC may present with night sweats, weight loss, fever, bone pain, or symptoms of anemia. An increased risk of infections and of bleeding is also observed. The common laboratory features include high white blood cell and blast counts, decreased hemoglobin values, and platelet numbers and, in up to 80% of BC patients, additional cytogenetic aberrations (ACAs) in addition to the Philadelphia (Ph)–chromosome. Most frequent are the so called “major route” ACA (trisomy 8, additional Ph-chromosome, isochromosome (17q), trisomy 19), which are nonrandom and considered relevant for the pathogenesis of BC.12,14  Less frequent are the so-called “minor route” cytogenetic aberrations involving chromosome 3 aberrations, loss of the Y-chromosome, and other rarer aberrations. Minor route ACAs are less likely involved in BC pathogenesis and may mainly indicate genetic instability. The impact of major route ACA at diagnosis on progression and survival has been shown.15 

A variety of mutations has been associated with progression to BC. Mutations of the BCR-ABL tyrosine kinase domain have been observed in up to 80% of patients.11,16  ABL mutations in late CP with upfront imatinib resistance have been associated with a greater likelihood of progression to BC.17  Other mutations associated with BC include p53 mutations in approximately 24% of myeloid BC, p16 mutations in approximately 50% of lymphoid BC,18,19  and more recently characterized mutations, such as RUNX-1, IKZF1 (Ikaros), ASXL1, WT1, TET2, IDH1, NRAS, KRAS, and CBL in 3% to 33% of myeloid and/or lymphoid BC.20,22  In addition, a profoundly altered gene expression profile has been reported in CD34+ BC cells compared with CP cells.23,24  Genes overexpressed, down-regulated, or deregulated in BC include SOCS2, CD52, HLA antigens, PRAME, JunB, Fos, FosB, and Il8 and genes of the Wnt/β-catenin pathway.25 

Several features have been associated with an unfavorable prognosis, such as clonal evolution, more than 50% blast cells, high platelet counts, short duration of the CP, and extramedullary disease.26,28  Although nonrandom, chromosomal individuality of each clonal evolution is a characteristic feature of BC similar to other cancers, which has been compared with speciation in evolution.29,30  The most important predictor of a poor prognosis is an unsatisfactory response to initial therapy.

Treatment of BC is guided by our understanding of BC pathogenesis. Good in-depth reviews on the biology of BC have been published.31,33  According to current evidence, BC is the direct consequence of continued BCR-ABL activity,31  possibly via oxidative stress and reactive oxygen species,34,35  causing DNA damage and impaired DNA repair36  (Figure 1) and, in a vicious circle, genomic instability by more mutations, gene doublings, translocations, and chromosomal breakage.37  The latter effect of BCR-ABL would explain what is observed during clonal evolution and progression to BC. BCR-ABL has been shown to produce reactive oxygen species in hemopoietic cells.38 

Figure 1

Mechanisms of BCR-ABL activity in CML and blast crisis, leading to stimulation of proliferation and to induction of genetic instability, DNA damage, and impaired DNA repair. Reactive oxygen species induced by BCR-ABL are thought to mediate DNA damage and genetic instability. Data are from Skorski,34  Melo and Barnes,31  Radich,32  and Perrotti et al.33 

Figure 1

Mechanisms of BCR-ABL activity in CML and blast crisis, leading to stimulation of proliferation and to induction of genetic instability, DNA damage, and impaired DNA repair. Reactive oxygen species induced by BCR-ABL are thought to mediate DNA damage and genetic instability. Data are from Skorski,34  Melo and Barnes,31  Radich,32  and Perrotti et al.33 

Close modal

This consideration underlies the therapeutic principle in CML to hit “hard and early” to reduce the BCR-ABL–positive cell pool as early and as deep as possible and to thereby achieve the best possible outcome.39  The validity of this principle may be limited by quiescent CD34+ CML cells, which evade currently available pharmacotherapy40  or by a speculative preexisting genetic instability responsible for the generation of BCR-ABL.41  The clinical improvement by TKI treatment in parallel to BCR-ABL reduction and the postponement (or prevention) of BC in most patients with TK-inhibition (8-year incidence of BC in IRIS1  < 8% under standard imatinib) support the conclusion that BCR-ABL is the driving force behind disease progression. The transient nature of response to TK inhibition in BC demonstrates that most cells are still sensitive to BCR-ABL inhibition but that BCR-ABL independence has been achieved in some cells, which then have a growth advantage. It follows that the most effective management of BC would be its prevention by early reduction of tumor burden and elimination of BCR-ABL.

This is confirmed by experience of the German CML Study Group (Figure 2). The cumulative incidence of BC, as a consequence of more effective treatment early on, has decreased from close to 70% after 8 years 25 years ago to currently approximately 5% in CML Study IV under an optimized dose of imatinib.42 

Figure 2

Prevention of BC by more effective treatment in early CP as shown by the cumulative incidence of blast crisis (German CML Study Group experience 1983-2011). CML study I compared busulfan versus hydroxyurea (HU) versus interferon-α (IFN) monotherapy, CML study II IFN in combination with HU versus HU alone, CML study III and IIIA IFN in combination with intensive chemotherapy versus allo-SCT and CML study IV imatinib 400 mg versus imatinib in combination with low-dose cytosine arabinoside versus imatinib in combination with IFN versus imatinib after IFN failure versus imatinib at 800 mg.42 

Figure 2

Prevention of BC by more effective treatment in early CP as shown by the cumulative incidence of blast crisis (German CML Study Group experience 1983-2011). CML study I compared busulfan versus hydroxyurea (HU) versus interferon-α (IFN) monotherapy, CML study II IFN in combination with HU versus HU alone, CML study III and IIIA IFN in combination with intensive chemotherapy versus allo-SCT and CML study IV imatinib 400 mg versus imatinib in combination with low-dose cytosine arabinoside versus imatinib in combination with IFN versus imatinib after IFN failure versus imatinib at 800 mg.42 

Close modal

In the late 1960s/early 1970s, attempts were made to treat BC with treatment protocols designed for acute leukemia (AL). It was observed that 30% of the patients responded to a combination of vincristine and prednisone as used for acute lymphoblastic leukemia (ALL), whereas 70% did not.43,45  The cells of the responding BC frequently showed features of lymphoid morphology and were TdT+.46  These observations have led to the distinction of lymphoid and myeloid variants of BC. The response rates to vincristine and prednisone and other drugs used for ALL, such as 6-thioguanine, 6-mercaptopurine, cytosine arabinoside, and methotrexate, ranged between 15% and 50%. Response was only of short duration. Responders survived a median of 3 to 10 months compared with 1 to 5 months in nonresponders.

In the 1980s and 1990s, AML-type induction therapies were applied, including various combinations of anthracyclines, cytosine arabinoside, 5-azacytidine, etoposide, carboplatin, fludarabine, and decitabine.47  In a series of 162 patients with nonlymphoid BC, 31 patients treated with decitabine showed a trend for better survival at lower toxicity.48  In total, a return to CP was observed in approximately 10%, opening a window for transplantation. No cures in the absence of stem cell transplantations were observed.

Overall, treatment of BC turned out to be less successful than that of AL despite considerable intensity (and toxicity), but the chance offered by a second CP for allo-SCT was recognized.

Once BC has been diagnosed and without clear targets available for inhibition, management depends on previous therapy and type of leukemia (myeloid or lymphoid). Best results are achieved for the few patients who return to CP and are successfully transplanted.

1. If the patient has been pretreated with conventional therapy (IFN or hydroxyurea, meanwhile the exception), a TKI (imatinib 600-800 mg/d, dasatinib 140 mg once daily or nilotinib 2 × 400 mg/d according to mutation profile) should be given and allo-SCT planned. Outcomes of trials with imatinib and other TKIs in BC are summarized in Table 2. Imatinib and dasatinib have been approved for all phases of CML, including BC by the Food and Drug Administration and the European Medicine Agency.

Table 2

Treatment of BC by BCR-ABL TKI

DrugPatientsCR, %
Survival
MBC/LBC12 mo, %Median, mo
Imatinib     
    300-600 mg28  58 (20 LBC) 12 NA NA 
    400-600 mg49  229 (MBC only) 16 30 6.9 
    300-1000 mg50  75 (10 LBC) 16 22 6.5 
    600 mg51  30 13 36 10 
    600 mg52  92 (20 LBC) 17 29 
Dasatinib     
    50-100 mg bid54  33 (10 LBC) 52/90 ∼ 22* ∼ 6 
    70-100 mg bid55  157 (48 LBC) 35/56 49/30 11.8 (5.3) 
    70 bid vs 140 mg qd56  210 (61 LBC) 25-28/40-50 34-39/39-46 8 (10) 
Nilotinib     
    Up to 1200 mg58  33 (9 LBC) 18 NA NA 
    400-600 mg bid59  136 (31 LBC) 40 42 10 
DrugPatientsCR, %
Survival
MBC/LBC12 mo, %Median, mo
Imatinib     
    300-600 mg28  58 (20 LBC) 12 NA NA 
    400-600 mg49  229 (MBC only) 16 30 6.9 
    300-1000 mg50  75 (10 LBC) 16 22 6.5 
    600 mg51  30 13 36 10 
    600 mg52  92 (20 LBC) 17 29 
Dasatinib     
    50-100 mg bid54  33 (10 LBC) 52/90 ∼ 22* ∼ 6 
    70-100 mg bid55  157 (48 LBC) 35/56 49/30 11.8 (5.3) 
    70 bid vs 140 mg qd56  210 (61 LBC) 25-28/40-50 34-39/39-46 8 (10) 
Nilotinib     
    Up to 1200 mg58  33 (9 LBC) 18 NA NA 
    400-600 mg bid59  136 (31 LBC) 40 42 10 

CR indicates cytogenetic response (includes complete, partial, minimal, and minor response when available); LBC, lymphoid blast crisis; NA, not available; MBC, myeloid blast crisis; bid, twice a day; and qd, daily.

*

At 18 months.

Only complete and major cytogenetic response listed. Updated from Hehlmann and Saussele.5 

Imatinib

Five studies on 484 patients, 50 with lymphoid BC, showed hematologic remission rates of 50% to 70% (70% in patients with lymphoid BC), cytogenetic response rates of 12% to 17% (all responses), a 1-year survival of 22% to 36%, and a median survival of 6.5 to 10 months.28,49,,52 

2. If BC evolves under imatinib, treatment with a second-generation TKI (dasatinib 140 mg or nilotinib 2 × 400 mg according to mutation profile) combined with chemotherapy as necessary should be given and allo-SCT planned as quickly as possible. In case of V299L, T315A, or F317L/V/I/C mutations, nilotinib is probably more effective than dasatinib. In case of Y253H, E255K/V, or F359V/C/I mutations, dasatinib is probably more effective than nilotinib.11  In case of the T315I mutation, an investigational approach (eg, with ponatinib) should be tried.53  Cytopenias may necessitate TKI dose reduction or treatment interruption, substitution of erythrocytes and platelets, or, in case of neutropenia, treatment with G-CSF.

Dasatinib

Three studies on 400 BC patients pretreated with imatinib, including 119 with lymphoid BC, showed hematologic remission rates of 33% to 61% (lymphoid BC, 36%-80%), major cytogenetic remission (MCR) rates of 35% to 56%, a 1-year survival of 42% to 50%, a 2-year survival of 20% to 30%, and a median survival of 8 to 11 months.54,56 

The largest of the studies, a randomized open label phase 3 study on 214 patients with 61 in lymphoid BC, tried to optimize the dose-schedule of dasatinib, stratified for lymphoid and myeloid BC, and compared dasatinib at 140 mg once daily with 70 mg twice daily. The study yielded similar efficacy and improved tolerability for the once-daily regimen.56  Pleural effusion, which is observed in up to one-third of dasatinib-treated BC patients, may necessitate dose reduction, diuretics, and, in some cases, corticosteroids.

Dasatinib crosses the blood-brain barrier and shows long lasting responses in Ph+ CNS disease.57  It is speculated that these effects, which are different from imatinib, are the result of the dual specific SRC/BCR-ABL TK-inhibitory property of dasatinib. Dasatinib maintenance is recommended in responders not suitable for allo-SCT.

Nilotinib

Two studies have been published on 169 patients, including 40 with lymphoid BC58,59  reporting hematologic response rates of 60% (lymphoid BC 59%), major cytogenetic response rates of 38% (myeloid BC), and 52% (lymphoid BC), a 1-year survival of 42%, a 2-year survival of 27%, and a median survival of 10 months (7.9 months for lymphoid BC). Hyperglycemia, which is observed in up to 40% of nilotinib-treated patients, requires monitoring and may necessitate dose adaptation. Nilotinib has been approved for treating CP and accelerated phase (AP) CML, but not yet BC.

The outcomes with dasatinib and nilotinib are similar to those with imatinib.

Bosutinib, a third second-generation TKI, shows in preliminary analyses similar activity in advanced phase CML as dasatinib and nilotinib.60  Bosutinib has not yet been approved for CML.

3. If TKIs fail, conventional approaches remain an option, such as AML induction protocols with anthracyclines and cytosine arabinoside in myeloid BC or a trial with vincristine and prednisone (combined with dasatinib) in lymphoid BC, or third-generation TKI within a clinical trial.

In summary, survival after BC is better after treatment with TKI than after conventional therapies, but with a median survival of less than 1 year, outcome is still unsatisfactory.

The modest survival progress that is achieved by TKI after BC is illustrated by the experience of the German CML Study Group in Figure 3. Median survival has increased from 4 months in the pre-imatinib era (n = 699) to 9 months under imatinib (n = 65).

Figure 3

Survival with BC in the preimatinib and imatinib eras. Most long-term survivors (72%) are transplant recipients. German CML Study Group experience (1983-2011). Data are from the German CML-studies I to IV.42 

Figure 3

Survival with BC in the preimatinib and imatinib eras. Most long-term survivors (72%) are transplant recipients. German CML Study Group experience (1983-2011). Data are from the German CML-studies I to IV.42 

Close modal

If a return to CP or a complete remission has been achieved, I proceed to allo-SCT as quickly as possible, given that the patient can tolerate the procedure and has a donor. The search for a donor should be instituted as early as possible. The best outcome continues to be observed in patients after transplantation, although allo-SCT is successful in only a minority of BC patients mostly after prior return to a second CP. In an overview of the European Group for Blood and Marrow Transplantation from 1980 to 2003, 2-year survival rates are 16% to 22%.61  Most patients were transplanted in the pre-imatinib era. In a recent report from the German CML Study Group, the 3-year survival of 28 imatinib-pretreated patients transplanted in advanced phases (25 in BC) was 59%.62  The data show convincingly that allo-SCT represents the best chance of long-term remission or cure in BC. Current experience recommends allo-SCT in primary BC after an attempt has been made with a suitable TKI selected according to mutation profile in combination with chemotherapy as needed to achieve a second CP. In lymphoid BC, dasatinib should be combined with vincristine and prednisone.

In BC after imatinib failure, a second-generation TKI (according to mutation profile) has to be weighed against other options, such as AL-type therapy (also in combination with TKI) to give the best chance of a return to CP or cytoreduction. If patients carry the T315I mutation, this has to be considered in choosing the appropriate regimen (investigational agents; eg, ponatinib, AL-type therapy) followed by allo-SCT.63  Transplantation should be performed with an HLA-identical related or matched unrelated donor and an EBMT score 0 to 4.64  Standard conditioning with busulfan and cyclophosphamide or total body irradiation should be used. Reduced intensity conditioning is not recommended in this situation outside studies. Sudden-onset BC under imatinib is a rare event, but full disease eradication by allo-SCT may be successful65  and is warranted. Posttransplantation maintenance with TKI appears reasonable. Maintenance with dasatinib is recommended in lymphoid BC for neuroprophylaxis as it is known to cross the blood-brain barrier.57  Monitoring of BCR-ABL transcript levels should be done at regular intervals (3 months initially, 6 months later on, if transcripts are not detectable or stable).

As a consequence of these recommendations, more CML patients are now transplanted in second chronic or advanced phases than in first CP.66  Most long-term survivors shown in Figure 3 represent transplant recipients (72%).

A number of investigational approaches are under exploration. A selection is shown in Table 3. Some agents are in clinical trial and can be tried after conventional treatments (TKI and AL-type therapy) have failed. Some approaches may be suitable for BC prevention.

Table 3

Investigational approaches (selection)

Mode of actionAgent(s)PhaseTarget(s)
Third-generation TKI Ponatinib53  II Pan-BCR-ABL including T315I 
 DCC-203672  Abl-switch pocket 
PP2A activation Fingolimod (FTY720)75  Preclinical PP2A 
 SET antagonist OP44976  Preclinical SET 
 CIP2A inhibitor74  Preclinical CIP2A 
Survival of LSCs BCL6 + TK inhibitors78  Preclinical BCL6 + BCR-ABL 
 HIF1α inhibitor80  Preclinical HIF1α 
 IL1 RAP antibodies86  Preclinical IL1 RAP 
 Smoothened inhibitors in combination with TKI83  (dasatinib, nilotinib) Preclinical Smoothened (hedgehog pathway) + BCR-ABL 
 Jak2 inhibitor + dasatinib85  Preclinical Jak2 + BCR-ABL, LSC 
Activation of apoptosis BCL2-inhibitor ABT-73788  Preclinical Antiapoptotic proteins 
 Triptolide87,88  Preclinical Antiapoptotic proteins 
 Dual-kinase inhibitor ON04458091  Preclinical BC, T315I 
 MEK inhibitor PD184352 + farnesyltransferase inhibitor BMS-21466289  Preclinical MEK1, MEK2, RAS 
Others Omacetaxine92  II / III BCR-ABL, T315I, BC 
Mode of actionAgent(s)PhaseTarget(s)
Third-generation TKI Ponatinib53  II Pan-BCR-ABL including T315I 
 DCC-203672  Abl-switch pocket 
PP2A activation Fingolimod (FTY720)75  Preclinical PP2A 
 SET antagonist OP44976  Preclinical SET 
 CIP2A inhibitor74  Preclinical CIP2A 
Survival of LSCs BCL6 + TK inhibitors78  Preclinical BCL6 + BCR-ABL 
 HIF1α inhibitor80  Preclinical HIF1α 
 IL1 RAP antibodies86  Preclinical IL1 RAP 
 Smoothened inhibitors in combination with TKI83  (dasatinib, nilotinib) Preclinical Smoothened (hedgehog pathway) + BCR-ABL 
 Jak2 inhibitor + dasatinib85  Preclinical Jak2 + BCR-ABL, LSC 
Activation of apoptosis BCL2-inhibitor ABT-73788  Preclinical Antiapoptotic proteins 
 Triptolide87,88  Preclinical Antiapoptotic proteins 
 Dual-kinase inhibitor ON04458091  Preclinical BC, T315I 
 MEK inhibitor PD184352 + farnesyltransferase inhibitor BMS-21466289  Preclinical MEK1, MEK2, RAS 
Others Omacetaxine92  II / III BCR-ABL, T315I, BC 

LSC indicates leukemia stem cell; and MEK, mitogen-activated protein kinase kinase.

Imatinib in combination

Several small studies have focused on the combination of imatinib at 600 mg to 800 mg with chemotherapy or other agents. In a phase 1/2 trial on 16 BC patients, imatinib 600 mg daily was combined with mitoxantrone/etoposide.67  Hematologic response rate was 81% with a 1-year survival of approximately 50%, including 6 patients after allo-SCT. Another study combined imatinib 600 mg with decitabine in 10 patients and reported a median survival of 15 weeks.68  The combination of imatinib 600 mg with low-dose cytosine arabinoside and idarubicin in 19 patients with myeloid BC showed hematologic remissions in 47%. Median survival was 5 months.69  In a phase 1 study with the combination of the farnesyltransferase inhibitor lonafarnib with imatinib, 2 of 3 BC patients showed hematologic improvement.70  A study on 12 patients combining imatinib and homoharringtonine after priming with G-CSF reported hematologic or cytogenetic response in all patients.71  None of these studies has provided convincing evidence that the combinations are superior to imatinib alone.

Third-generation TKIs

New third-generation TKIs, such as the pan-BCR-ABL inhibitor ponatinib,53  show promise because, in addition to recognizing the T315I mutation, ponatinib also shows efficacy in BC and Ph+ ALL. A phase 2 study on 449 ponatinib-treated patients, 94 in BC or Ph+ ALL, showed after a median follow-up of approximately 5 months, complete cytogenetic remission (CCR) and major molecular remission (MMR) rates in BC of 27% and 22%, respectively.53  No data on survival were reported yet. Similarly, the ABL switch pocket inhibitor DCC-2036 showed efficacy against T315I and in BC in a phase 1 study.72  These TKIs may be the best choice of investigational agents in clinical trials.

PP2A activation

A new target of interest is the tumor suppressor protein phosphatase 2A (PP2A), which shows decreased activity in BC73  through up-regulation of its inhibitors suppressor of variegation, enhancer of zeste and trithorax (SET),73  and cancerous inhibitor of PP2A (CIP2A).74  The PP2A activator fingolimod (FTY720) induces apoptosis in CML-BC and Ph+ ALL progenitors33,75  and may be a candidate for BC treatment and prevention. Likewise, a novel SET antagonist (OP449) is selectively cytotoxic to CML cells and restores PP2A's tumor suppressive function.76  In addition, CIP2A inhibition increases PP2A activity.74 

Self-renewal of leukemia stem cells

Another target potentially relevant for BC management or prevention is the self-renewal of leukemia stem cells (LSCs) in vivo or leukemia-initiating cells in vitro. BCL6 has been identified as a critical effector of the BCR-ABL downstream target FoxO in self-renewal signaling of CML initiating cells.77  Pharmacologic inhibition of BCL6 in combination with BCR-ABL inhibition is proposed for eradication of leukemia-initiating cells in CML.78  Dual inhibition of BCL6 and BCR-ABL is an interesting approach that merits exploration for application to BC, but BCL6 inhibitors are not yet available for clinical use.79 

A similar role for survival maintenance of CML stem cells has been reported for the hypoxia-inducible factor 1α, a master transcriptional regulator of the cellular and systemic hypoxia response.80  Inhibition of the hypoxia-inducible factor 1α pathway may provide another strategy for eradicating LSCs in CML.

Clinical studies are ongoing to explore antagonists of the transmembrane protein smoothened, which plays a role in the hedgehog pathway and is essential for the maintenance of LSCs,81,82  such as cyclopamine, GDC-0449 (Genentech), LDE225 (Novartis), BMS833923, or PF0444913 (Pfizer), in combination with second-generation TKI for activity against BC-LSC and self-renewal.83  GDC-0449 has shown activity in basal cell carcinoma (18 of 33 patients responded)84  and in medulloblastoma. Similarly, the Jak2-inhibitor SAR503 in combination with dasatinib significantly reduced LSC, suggesting abolishment of LSC self-renewal capacity.85 

A new cell surface biomarker, IL1 receptor accessory protein (IL1 RAP), has been specifically identified on CML stem cells and might offer a new therapeutic target in the future.86 

Induction of apoptosis

Preclinical studies are investigating the activation of apoptosis in BC cells by various drugs and combinations. The BCL2 inhibitor ABT-737 combined with imatinib or with the diterpenoid triptolide reduces antiapoptotic proteins, thereby inducing apoptosis and cell death in K562 cells and in cells from BC patients.87,88  The MEK inhibitor PD184352 combined with the farnesyltransferase inhibitor BMS-214662 similarly induces apoptosis in K562 cells and CD34+ CML stem cells.89  In addition, p53 stabilization with the novel compound MI-219, which inhibits human homolog double minute 2, induces apoptosis in cell line and primary BC cells.90  And recently, the dual Jak2/Abl kinase inhibitor ON044580 was shown to induce apoptosis in cells from BC patients and in imatinib-resistant cells, including T315I.91 

More drugs are in clinical and in preclinical evaluation. These drugs include omacetaxine (a semisynthetic derivative of homoharringtonine),92  arsenic trioxide, which showed synergy with imatinib, histone deacetylase (HDAC) inhibitors, aurora kinase inhibitors alone or in combination (eg, with TK or HDAC inhibitors), HSP90 inhibitors, mTOR inhibitors (rapamycin), and other substances.4,93,95 

None of these approaches is likely to provide a breakthrough in the near future; because of the numerous blastic genotypes and their instability, no single therapeutic approach can soon be expected to be successful in all patients.

The low progression rates of CML under TKIs indicate that BC can be prevented (Figure 2). In addition, it is well known that very low or undetectable BCR-ABL transcripts after allo-SCT correlate with low relapse rates.96,97  Imatinib-treated patients who have achieved MMR enjoy durable responses with virtually no progression to AP or BC up to now.42,98  Patients who have achieved stable complete molecular remission experience, in approximately 40% of cases, continued remissions even in the absence of treatment.99  The challenge is how to identify early those patients who are at risk to proceed to BC to be able to offer alternative treatment to this patient group.

At diagnosis, risk scores provide information on the likelihood of progression.100,101  The EUTOS score, which was developed from imatinib-treated patients, has a predictive value of not reaching a CCR by 18 months of 34% and recognizes a small group of high-risk patients (∼ 12%), with a significantly higher progression rate. [The EUTOS score uses 2 variables at diagnosis (spleen size in centimeters below costal margin and percentage basophils) and separates 2 risk groups. It is calculated by the formula: EUTOS score = (7 × basophils) + (4 × spleen size). A score of > 87 indicates high risk.]102  In addition, distinct markers such as major route ACA,15  p190BCR-ABL,103  and signs of acceleration may be suitable for early prediction of progression (Table 4). In addition, BMI1 and CIP2A levels at diagnosis have been reported predictive of BC.74,104 

Table 4

Early prediction of progression

StudynBaseline3 mo6 mo12 moEnd point
Historical       
    Mahon et al (IFN)121  116 NA CHR NA NA MCR 
    Baccarani et al (imatinib, review)8  NA NA CHR NA CCR OS 
Baseline       
    Hasford et al (EUTOS)102  2060 High risk NA NA NA CCR* 
    Fabarius et al15  1151 Major route ACA NA NA NA OS 
    Verma et al103  1292 P190BCR-ABL NA NA NA PFS 
Clonal evolution       
    Baccarani et al (review)8  NA NA NA Any time NA OS 
Response       
    Hanfstein et al122  692 NA MR 10%, MCR MR 1%, CCR NA OS 
    Hehlmann et al42  1014 NA NA NA MMR (MR 0.1%) OS 
    Marin et al123  282 NA MR 9.84% MR 1.67% MR 0.53% OS 
    Jabbour et al124  435 NA MCR CCR NA OS 
StudynBaseline3 mo6 mo12 moEnd point
Historical       
    Mahon et al (IFN)121  116 NA CHR NA NA MCR 
    Baccarani et al (imatinib, review)8  NA NA CHR NA CCR OS 
Baseline       
    Hasford et al (EUTOS)102  2060 High risk NA NA NA CCR* 
    Fabarius et al15  1151 Major route ACA NA NA NA OS 
    Verma et al103  1292 P190BCR-ABL NA NA NA PFS 
Clonal evolution       
    Baccarani et al (review)8  NA NA NA Any time NA OS 
Response       
    Hanfstein et al122  692 NA MR 10%, MCR MR 1%, CCR NA OS 
    Hehlmann et al42  1014 NA NA NA MMR (MR 0.1%) OS 
    Marin et al123  282 NA MR 9.84% MR 1.67% MR 0.53% OS 
    Jabbour et al124  435 NA MCR CCR NA OS 

Patients at increased risk of progression can be detected by baseline markers, clonal evolution, and early molecular or cytogenetic response indicators. Failure to reach the defined response landmarks at 3, 6, and 12 months identifies a group of high risk patients with higher progression risks (25%-33% of patients at 3 months122,123 ) who might benefit from an early change of therapy. Percentages are according to international scale.130 

CHR indicates complete hematologic remission; MCR, major cytogenetic remission; NA, not applicable; OS, overall survival; ACA, additional cytogenetic aberrations; PFS, progression-free survival; and MR, molecular response.

*

CCR at 18 months.

Another indicator of progression risk is clonal evolution (ie, the acquisition of ACA in the course of the disease).105,,108  The relevance of clonal evolution has not changed in the imatinib era.109,,112  Mutations may be associated with clonal evolution.113  The pattern of chromosome abnormalities is not altered by TKI treatment.114  The prognostic impact of ACA may depend on the type of ACA.112  Some ACA types (major route, complex karyotypes) appear to imply poorer prognosis than others that may only indicate genetic instability.115  Acquired ACAs are high-risk features by European LeukemiaNet definition and indicate treatment failure if they appear under therapy.8  The prognostic relevance of rare clonal evolution in Ph-negative cells (observed in < 5% of cases) remains uncertain.116,,119  The evolution of gene expression profiles may also allow to diagnose disease progression.120 

Early response indicators are probably the best predictors of progression.8,121  These include cytogenetic and molecular responses determined by monitoring all patients. Failure to achieve defined landmarks will detect high-risk patients as early as 3 months after diagnosis.122,124 Table 4 summarizes the response levels and time points for response categorization.42,122,124  Patients who do not respond satisfactorily and are classified as high risk need alternative approaches, such as early second-generation TKI, treatment intensification, or an early allo-SCT.8,125  If the patients have a donor and have no medical contraindications, the risk of progression to BC has to be weighed against the risk of early transplantation and of chronic GVHD. With the current progress in donor selection and posttransplantation management, the risk of transplantation seems acceptable if compared with the risk of BC. If the patients are too old or have other medical contraindications that preclude allo-SCT or have no donor, investigational agents can be tried (Table 3).

The algorithm in Figure 4 gives an overview on how I approach management of a patient with BC. The treatment goal is the return to CP or the induction of a remission. Mainstays are TKIs taking into account the type of mutation and allo-SCT as quickly as possible. If TKIs alone are not sufficient, AL-type induction therapy should be tried, cytosine arabinoside and anthracyclines for myeloid BC, vincristine and prednisone in lymphoid BC, or TKI in combination with AL-type induction therapy. Management of primary BC follows the same principle, except that imatinib should be tried first in myeloid BC. Treatment decisions have to be adapted to the individual patients' situations and needs as required. Hematologic, cytogenetic, and molecular monitoring are mandatory (Table 1). Cytopenias may necessitate dose adaptation, substitution therapy, and treatment with G-CSF. In lymphoid BC, intrathecal neuroprophylaxis may be indicated. Investigational approaches are recommended only after all other options have failed. Allo-SCT without prior return to CP or at least cytoreduction is a high-risk procedure and discouraged. An option is transplantation in aplasia without waiting for marrow recovery.

Figure 4

Management algorithm of CML-BC. Mainstays are TKI and rapid allo-SCT. *2nd generation TKI and AL-induction therapy may be combined.

Figure 4

Management algorithm of CML-BC. Mainstays are TKI and rapid allo-SCT. *2nd generation TKI and AL-induction therapy may be combined.

Close modal

In view of the limited therapeutic options once BC has been diagnosed, the best management of BC is probably its prevention by a rigorous and early reduction to low levels or elimination of BCR-ABL. Regular molecular monitoring is required (Table 4). The current understanding of pathogenesis of CML-BC as a consequence of continued BCR-ABL activity provides the rationale for this approach. Patients with high-risk features at diagnosis,100,102  unsatisfactory response to therapy (eg, no major cytogenetic response or < 90% BCR-ABL reduction by 3 months),122,124  or signs of progression under therapy, such as clonal evolution, should receive more intensive therapies to prevent progression and BC. With the availability of optimized imatinib protocols42,126,127  and second-generation BCR-ABL inhibitors first line,128,129  which induce deeper remissions faster, I recommend every attempt to eliminate BCR-ABL as early as possible. I expect that more efficacious therapies and early treatment intensification in patients with high-risk features or unsatisfactory responses will further reduce progression and transformation to BC.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

The author thanks colleagues A. Hochhaus, M. C. Müller, S. Sauβele, M. Baccarani, and R. S. Silver for reading the manuscript; A. Gratwohl, R. Schwerdtfeger, and H.-J. Kolb for advice on transplantation in blast crisis; G. Bartsch and U. Böhm for technical support; and all members of the German CML Study Group for their continued patient care and cooperation.

This work was supported by the German CML Study Group, Deutsche Krebshilfe (106642), Novartis Germany, Kompetenznetz für Akute und Chronische Leukämien (BMBF 01GI0270), José-Carreras Leukämiestiftung (DJCLS H09/01f, H06/04v, H03/01), the European Commission (LSHC-CT-2004-503216), and Roche and Essex (now MSD).

Contribution: R.H. wrote the manuscript.

Conflict-of-interest disclosure: The author declares no competing financial interests.

Correspondence: Rüdiger Hehlmann, Medizinische Klinik, Medizinische Fakultät Mannheim der Universität Heidelberg, Pettenkoferstr 22, 68169 Mannheim, Germany; e-mail: sekretariat.hehlmann@medma.uni-heidelberg.de.

1
Druker
BJ
Guilhot
F
O'Brien
SG
et al
Five-year follow-up of patients receiving imatinib for chronic myeloid leukemia.
N Engl J Med
2006
355
23
2408
2417
2
Hughes
TP
Kaeda
J
Branford
S
et al
Frequency of major molecular responses to imatinib or interferon alfa plus cytarabine in newly diagnosed chronic myeloid leukemia.
N Engl J Med
2003
349
15
1423
1432
3
Sokal
JE
Evaluation of survival data for chronic myelocytic leukemia.
Am J Hematol
1976
1
4
493
500
4
Hehlmann
R
Jung-Munkwitz
S
Saussele
S
Treatment of chronic myeloid leukemia when imatinib fails.
Expert Opin Pharmacother
2011
12
2
269
283
5
Hehlmann
R
Saussele
S
Treatment of chronic myeloid leukemia in blast crisis.
Haematologica
2008
93
12
1765
1769
6
Silver
RT
The blast phase of chronic myeloid leukaemia.
Best Pract Res Clin Haematol
2009
22
3
387
394
7
Karanas
A
Silver
RT
Characteristics of the terminal phase of chronic granulocytic leukemia.
Blood
1968
32
3
445
459
8
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
35
6041
6051
9
Vardiman
JW
Melo
JV
Baccarani
M
Thiele
J
Swerdlow
SH
Campo
E
Harris
NL
et al
Chronic myelogenous leukemia, BCR-ABL1 positive.
WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues
2008
4th Ed
Lyon, France
International Agency for Research on Cancer
10
Cortes
JE
Talpaz
M
O'Brien
S
et al
Staging of chronic myeloid leukemia in the imatinib era: an evaluation of the World Health Organization proposal.
Cancer
2006
106
6
1306
1315
11
Soverini
S
Hochhaus
A
Nicolini
FE
et al
BCR-ABL kinase domain mutation analysis in chronic myeloid leukemia patients treated with tyrosine kinase inhibitors: recommendations from an expert panel on behalf of European LeukemiaNet.
Blood
2011
118
5
1208
1215
12
Mitelman
F
Levan
G
Nilsson
PG
Brandt
L
Non-random karyotypic evolution in chronic myeloid leukemia.
Int J Cancer
1976
18
1
24
30
13
Alimena
G
De Cuia
MR
Diverio
D
Gastaldi
R
Nanni
M
The karyotype of blastic crisis.
Cancer Genet Cytogenet
1987
26
1
39
50
14
Johansson
B
Fioretos
T
Mitelman
F
Cytogenetic and molecular genetic evolution of chronic myeloid leukemia.
Acta Haematol
2002
107
2
76
94
15
Fabarius
A
Leitner
A
Hochhaus
A
et al
Impact of additional cytogenetic aberrations at diagnosis on prognosis of CML: long-term observation of 1151 patients from the randomized CML Study IV.
Blood
2011
118
26
6760
6768
16
Hochhaus
A
La Rosee
P
Imatinib therapy in chronic myelogenous leukemia: strategies to avoid and overcome resistance.
Leukemia
2004
18
8
1321
1331
17
Soverini
S
Martinelli
G
Rosti
G
et al
ABL mutations in late chronic phase chronic myeloid leukemia patients with up-front cytogenetic resistance to imatinib are associated with a greater likelihood of progression to blast crisis and shorter survival: a study by the GIMEMA Working Party on Chronic Myeloid Leukemia.
J Clin Oncol
2005
23
18
4100
4109
18
Prokocimer
M
Rotter
V
Structure and function of p53 in normal cells and their aberrations in cancer cells: projection on the hematologic cell lineages.
Blood
1994
84
8
2391
2411
19
Sill
H
Goldman
JM
Cross
NCP
Homozygous deletions of the p16 tumor-suppressor gene are associated with lymphoid transformation of chronic myeloid leukemia.
Blood
1995
85
8
2013
2016
20
Grossmann
V
Kohlmann
A
Zenger
M
et al
A deep-sequencing study of chronic myeloid leukemia patients in blast crisis (BC-CML) detects mutations in 76.9% of cases.
Leukemia
2011
25
3
557
560
21
Roche-Lestienne
C
Deluche
L
Corm
S
et al
RUNX1 DNA-binding mutations and RUNX1-PRDM16 cryptic fusions in BCR-ABL(+) leukemias are frequently associated with secondary trisomy 21 and may contribute to clonal evolution and imatinib resistance.
Blood
2008
111
7
3735
3741
22
Mullighan
CG
Miller
CB
Radtke
I
et al
BCR-ABL1 lymphoblastic leukaemia is characterized by the deletion of Ikaros.
Nature
2008
453
7191
110
115
23
Zheng
C
Li
L
Haak
M
et al
Gene expression profiling of CD34+ cells identifies a molecular signature of chronic myeloid leukemia blast crisis.
Leukemia
2006
20
6
1028
1034
24
Radich
JP
Dai
H
Mao
M
et al
Gene expression changes associated with progression and response in chronic myeloid leukemia.
Proc Natl Acad Sci U S A
2006
103
8
2794
2799
25
Jamieson
CH
Ailles
LE
Dylla
SJ
et al
Granulocyte-macrophage progenitors as candidate leukemic stem cells in blast-crisis CML.
N Engl J Med
2004
351
7
657
667
26
Cervantes
F
Rozman
M
Rosell
J
Urbano Ispizua
A
Montserrat
E
Rozman
C
A study of prognostic factors in blast crisis of Philadelphia chromosome-positive chronic myelogenous leukaemia.
Br J Haematol
1990
76
1
27
32
27
Wadhwa
J
Szydlo
RM
Apperley
JF
et al
Factors affecting duration of survival after onset of blastic transformation of chronic myeloid leukemia.
Blood
2002
99
7
2304
2309
28
Druker
BJ
Sawyers
CL
Kantarjian
H
et al
Activity of a specific inhibitor of the BCR-ABL tyrosine kinase in the blast crisis of chronic myeloid leukemia and acute lymphoblastic leukemia with the Philadelphia chromosome.
N Engl J Med
2001
344
14
1038
1042
29
Fabarius
A
Li
R
Yerganian
G
Hehlmann
R
Duesberg
P
Specific clones of spontaneously evolving karyotypes generate individuality of cancers.
Cancer Genet Cytogenet
2008
180
2
89
99
30
Duesberg
P
Mandrioli
D
McCormack
A
Nicholson
JM
Is carcinogenesis a form of speciation?
Cell Cycle
2011
10
13
2100
2114
31
Melo
JV
Barnes
DJ
Chronic myeloid leukaemia as a model of disease evolution in human cancer.
Nat Rev Cancer
2007
7
6
441
453
32
Radich
JP
The biology of CML blast crisis.
Hematology Am Soc Hematol Educ Program
2007
1
384
391
33
Perrotti
D
Jamieson
C
Goldman
J
Skorski
T
Chronic myeloid leukemia: mechanisms of blastic transformation.
J Clin Invest
2010
120
7
2254
2264
34
Skorski
T
Oncogenic tyrosine kinases and the DNA-damage response.
Nat Rev Cancer
2002
2
5
351
360
35
Koptyra
M
Falinski
R
Nowicki
MO
et al
BCR/ABL kinase induces self-mutagenesis via reactive oxygen species to encode imatinib resistance.
Blood
2006
108
1
319
327
36
Nowicki
MO
Falinski
R
Koptyra
M
et al
BCR/ABL oncogenic kinase promotes unfaithful repair of the reactive oxygen species-dependent DNA double-strand breaks.
Blood
2004
104
12
3746
3753
37
Soverini
S
Gnani
A
Colarossi
S
et al
Philadelphia-positive patients who already harbor imatinib-resistant Bcr-Abl kinase domain mutations have a higher likelihood of developing additional mutations associated with resistance to second- or third-line tyrosine kinase inhibitors.
Blood
2009
114
10
2168
2171
38
Sattler
M
Verma
S
Shrikhande
G
et al
The BCR/ABL tyrosine kinase induces production of reactive oxygen species in hematopoietic cells.
J Biol Chem
2000
275
32
24273
24278
39
Hehlmann
R
Heimpel
H
Current aspects of drug therapy in Philadelphia-positive CML: correlation of tumor burden with survival.
Leuk Lymphoma
1996
22
Suppl 1
161
167
40
Graham
SM
Jorgensen
HG
Allan
E
et al
Primitive, quiescent, Philadelphia-positive stem cells from patients with chronic myeloid leukemia are insensitive to STI571 in vitro.
Blood
2002
99
1
319
325
41
Cortes
J
Jabbour
E
Kantarjian
H
et al
Dynamics of BCR-ABL kinase domain mutations in chronic myeloid leukemia after sequential treatment with multiple tyrosine kinase inhibitors.
Blood
2007
110
12
4005
4011
42
Hehlmann
R
Lauseker
M
Jung-Munkwitz
S
et al
Tolerability-adapted imatinib 800 mg/d versus 400 mg/d versus 400 mg/d plus interferon-alpha in newly diagnosed chronic myeloid leukemia.
J Clin Oncol
2011
29
12
1634
1642
43
Canellos
GP
DeVita
VT
Whang Peng
J
Carbone
P
Hematologic and cytogenetic remission of blastic transformation in chronic granulocytic leukemia.
Blood
1971
38
6
671
679
44
Marmont
AM
Damasio
EE
The treatment of terminal metamorphosis of chronic granulocytic leukaemia with corticosteroids and vincristine.
Acta Haematol
1973
50
1
1
8
45
Rosenthal
S
Canellos
GP
Whang-Peng
J
Gralnick
HR
Blast crisis of chronic granulocytic leukemia: morphologic variants and therapeutic implications.
Am J Hematol
1977
63
4
542
547
46
Marks
SM
Baltimore
D
McCaffrey
R
Terminal transferase as a predictor of initial responsiveness to vincristine and prednisone in blastic chronic myelogenous leukemia.
N Engl J Med
1978
298
15
812
814
47
Iacoboni
SJ
Plunkett
W
Kantarjian
HM
et al
High-dose cytosine arabinoside: treatment and cellular pharmacology of chronic myelogenous leukemia blast crisis.
J Clin Oncol
1986
4
7
1079
1088
48
Sacchi
S
Kantarjian
HM
O'Brien
S
et al
Chronic myelogenous leukemia in nonlymphoid blastic phase: analysis of the results of first salvage therapy with three different treatment approaches for 162 patients.
Cancer
1999
86
12
2632
2641
49
Sawyers
CL
Hochhaus
A
Feldman
E
et al
Imatinib induces hematologic and cytogenetic responses in patients with chronic myelogenous leukemia in myeloid blast crisis: results of a phase II study.
Blood
2002
99
10
3530
3539
50
Kantarjian
HM
Cortes
J
O'Brien
S
et al
Imatinib mesylate (STI571) therapy for Philadelphia chromosome-positive chronic myelogenous leukemia in blast phase.
Blood
2002
99
10
3547
3553
51
Sureda
A
Carrasco
M
de Miguel
M
et al
Imatinib mesylate as treatment for blastic transformation of Philadelphia chromosome positive chronic myelogenous leukemia.
Haematologica
2003
88
11
1213
1220
52
Palandri
F
Castagnetti
F
Testoni
N
et al
Chronic myeloid leukemia in blast crisis treated with imatinib 600 mg: outcome of the patients alive after a 6-year follow-up.
Haematologica
2008
93
12
1792
1796
53
Cortes
JE
Kim
D-W
Pinilla-Ibarz
J
et al
Initial findings from the PACE trial: a pivotal phase 2 study of ponatinib in patients with CML and Ph+ ALL resistant or intolerant to dasatinib or nilotinib, or with the T315I mutation [abstract].
Blood (ASH Annual Meeting Abstracts)
2011
118
21
52
Abstract 109
54
Talpaz
M
Shah
NP
Kantarjian
H
et al
Dasatinib in imatinib-resistant Philadelphia chromosome-positive leukemias.
N Engl J Med
2006
354
24
2531
2541
55
Cortes
J
Kim
DW
Raffoux
E
et al
Efficacy and safety of dasatinib in imatinib-resistant or -intolerant patients with chronic myeloid leukemia in blast phase.
Leukemia
2008
22
12
2176
2183
56
Saglio
G
Hochhaus
A
Goh
YT
et al
Dasatinib in imatinib-resistant or imatinib-intolerant chronic myeloid leukemia in blast phase after 2 years of follow-up in a phase 3 study: efficacy and tolerability of 140 milligrams once daily and 70 milligrams twice daily.
Cancer
2010
116
16
3852
3861
57
Porkka
K
Koskenvesa
P
Lundan
T
et al
Dasatinib crosses the blood-brain barrier and is an efficient therapy for central nervous system Philadelphia chromosome-positive leukemia.
Blood
2008
112
4
1005
1012
58
Kantarjian
H
Giles
F
Wunderle
L
et al
Nilotinib in imatinib-resistant CML and Philadelphia chromosome-positive ALL.
N Engl J Med
2006
354
24
2542
2551
59
Giles
FJ
Kantarjian
HM
le Coutre
PD
et al
Nilotinib is effective in imatinib-resistant or -intolerant patients with chronic myeloid leukemia in blastic phase.
Leukemia
2012
26
5
959
962
60
Keller
G
Schafhausen
P
Brümmendorf
TH
Martens
UM
Bosutinib.
Small Molecules in Oncology
2010
Vol. 184
Heidelberg, Germany
Springer Verlag
119
127
61
Gratwohl
A
Brand
R
Apperley
J
et al
Allogeneic hematopoietic stem cell transplantation for chronic myeloid leukemia in Europe 2006: transplant activity, long-term data and current results. An analysis by the Chronic Leukemia Working Party of the European Group for Blood and Marrow Transplantation (EBMT).
Haematologica
2006
91
4
513
521
62
Saussele
S
Lauseker
M
Gratwohl
A
et al
Allogeneic hematopoietic stem cell transplantation (allo SCT) for chronic myeloid leukemia in the imatinib era: evaluation of its impact within a subgroup of the randomized German CML Study IV.
Blood
2010
115
10
1880
1885
63
Nicolini
FE
Basak
GW
Soverini
S
et al
Allogeneic stem cell transplantation for patients harboring T315I BCR-ABL mutated leukemias.
Blood
2011
118
20
5697
5700
64
Gratwohl
A
Heim
D
Current role of stem cell transplantation in chronic myeloid leukaemia.
Best Pract Res Clin Haematol
2009
22
3
431
443
65
Jabbour
E
Kantarjian
H
O'Brien
S
et al
Sudden blastic transformation in patients with chronic myeloid leukemia treated with imatinib mesylate.
Blood
2006
107
2
480
482
66
Gratwohl
A
Baldomero
H
Schwendener
A
et al
The EBMT activity survey 2008: impact of team size, team density and new trends.
Bone Marrow Transplant
2011
46
2
174
191
67
Fruehauf
S
Topaly
J
Buss
EC
et al
Imatinib combined with mitoxantrone/etoposide and cytarabine is an effective induction therapy for patients with chronic myeloid leukemia in myeloid blast crisis.
Cancer
2007
109
8
1543
1549
68
Oki
Y
Kantarjian
HM
Gharibyan
V
et al
Phase II study of low-dose decitabine in combination with imatinib mesylate in patients with accelerated or myeloid blastic phase of chronic myelogenous leukemia.
Cancer
2007
109
5
899
906
69
Quintás-Cardama
A
Kantarjian
H
Garcia-Manero
G
et al
A pilot study of imatinib, low-dose cytarabine and idarubicin for patients with chronic myeloid leukemia in myeloid blast phase.
Leuk Lymphoma
2007
48
2
283
289
70
Cortes
J
Jabbour
E
Daley
GQ
et al
Phase 1 study of lonafarnib (SCH 66336) and imatinib mesylate in patients with chronic myeloid leukemia who have failed prior single-agent therapy with imatinib.
Cancer
2007
110
6
1295
1302
71
Fang
B
Li
N
Song
Y
Han
Q
Zhao
R
Standard-dose imatinib plus low-dose homoharringtonine and granulocyte colony-stimulating factor is an effective induction therapy for patients with chronic myeloid leukemia in myeloid blast crisis who have failed prior single-agent therapy with imatinib.
Ann Hematol
2010
89
11
1099
1105
72
Cortes
JE
Talpaz
M
Kantarjian
HM
et al
A phase 1 study of DCC-2036, a novel oral inhibitor of BCR-ABL kinase, in patients with Philadelphia chromosome positive (Ph+) leukemias including patients with T315I mutation [abstract].
Blood (ASH Annual Meeting Abstracts)
2011
118
21
276
Abstract 601
73
Neviani
P
Santhanam
R
Trotta
R
et al
The tumor suppressor PP2A is functionally inactivated in blast crisis CML through the inhibitory activity of the BCR/ABL-regulated SET protein.
Cancer Cell
2005
8
5
355
368
74
Lucas
CM
Harris
RJ
Giannoudis
A
Copland
M
Slupsky
JR
Clark
RE
Cancerous inhibitor of PP2A (CIP2A) at diagnosis of chronic myeloid leukemia is a critical determinant of disease progression.
Blood
2011
117
24
6660
6668
75
Neviani
P
Santhanam
R
Oaks
JJ
et al
FTY720, a new alternative for treating blast crisis chronic myelogenous leukemia and Philadelphia chromosome-positive acute lymphocytic leukemia.
J Clin Invest
2007
117
9
2408
2421
76
Agarwal
A
MacKenzie
R
Oddo
J
Vitek
MP
Christensen
DJ
Druker
BJ
A novel SET antagonist (OP449) is cytotoxic to CML cells, including the highly-resistant BCR-ABLT315I mutant, and demonstrates enhanced efficacy in combination with ABL tyrosine kinase inhibitors [abstract].
Blood (ASH Annual Meeting Abstracts)
2011
118
21
1603
Abstract 3757
77
Duy
C
Hurtz
C
Shojaee
S
et al
BCL6 enables Ph+ acute lymphoblastic leukaemia cells to survive BCR-ABL1 kinase inhibition.
Nature
2011
473
7347
384
388
78
Hurtz
C
Hatzi
K
Cerchietti
L
et al
BCL6-mediated repression of p53 is critical for leukemia stem cell survival in chronic myeloid leukemia.
J Exp Med
2011
208
11
2163
2174
79
Cerchietti
LC
Yang
SN
Shaknovich
R
et al
A peptomimetic inhibitor of BCL6 with potent antilymphoma effects in vitro and in vivo.
Blood
2009
113
15
3397
3405
80
Zhang
H
Li
H
Xi
HS
Li
S
HIF1alpha is required for survival maintenance of chronic myeloid leukemia stem cells.
Blood
2012
119
11
2595
2607
81
Zhao
C
Chen
A
Jamieson
CH
et al
Hedgehog signalling is essential for maintenance of cancer stem cells in myeloid leukaemia.
Nature
2009
458
7239
776
779
82
Dierks
C
Beigi
R
Guo
G-R
et al
Expansion of Bcr-Abl-positive leukemic stem cells is dependent on hedgehog pathway activation.
Cancer Cell
2008
14
3
238
249
83
Shih
AY
Schairer
A
Barrett
CL
et al
Cycling toward leukemia stem cell elimination with a selective sonic hedgehog antagonist [abstract].
Blood (ASH Annual Meeting Abstracts)
2011
118
21
1613
Abstract 3776
84
Von Hoff
DD
LoRusso
PM
Rudin
CM
et al
Inhibition of the Hedgehog pathway in advanced basal-cell carcinoma.
N Engl J Med
2009
361
12
1164
1172
85
Court Recart
AC
Sadarangani
A
Goff
D
et al
Combination targeted therapy to impair self-renewal capacity of human blast crisis leukemia stem cells [abstract].
Blood (ASH Annual Meeting Abstracts)
2011
118
21
737
Abstract 1693
86
Järås
M
Johnels
P
Hansen
N
et al
Isolation and killing of candidate chronic myeloid leukemia stem cells by antibody targeting of IL-1 receptor accessory protein.
Proc Natl Acad Sci U S A
2010
107
37
16280
16285
87
Mak
DH
Schober
WD
Chen
W
et al
Triptolide induces cell death independent of cellular responses to imatinib in blast crisis chronic myelogenous leukemia cells including quiescent CD34+ primitive progenitor cells.
Mol Cancer Ther
2009
8
9
2509
2516
88
Mak
DH
Wang
RY
Schober
WD
et al
Activation of apoptosis signaling eliminates CD34+ progenitor cells in blast crisis CML independent of response to tyrosine kinase inhibitors.
Leukemia
2012
26
4
788
794
89
Pellicano
F
Simara
P
Sinclair
A
et al
The MEK inhibitor PD184352 enhances BMS-214662-induced apoptosis in CD34+ CML stem/progenitor cells.
Leukemia
2011
25
7
1159
1167
90
Peterson
LF
Mitrikeska
E
Giannola
D
et al
p53 stabilization induces apoptosis in chronic myeloid leukemia blast crisis cells.
Leukemia
2011
25
5
761
769
91
Samanta
AK
Chakraborty
SN
Wang
Y
Schlette
E
Reddy
EP
Arlinghaus
RB
Destabilization of Bcr-Abl/Jak2 network by a Jak2/Abl kinase inhibitor ON044580 overcomes drug resistance in blast crisis chronic myelogenous leukemia (CML).
Genes Cancer
2010
1
4
346
359
92
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 [abstract].
Blood (ASH Annual Meeting Abstracts)
2009
114
22
267
Abstract 644
93
Giles
FJ
DeAngelo
DJ
Baccarani
M
et al
Optimizing outcomes for patients with advanced disease in chronic myelogenous leukemia.
Semin Oncol
2008
35
1 Suppl 1
S1
S17
94
Quintás-Cardama
A
Experimental non-ATP-competitive therapies for chronic myelogenous leukemia.
Leukemia
2008
22
5
932
940
95
Quintás-Cardama
A
Cortes
JE
The next generation of therapies for chronic myeloid leukemia.
Clin Lymphoma Myeloma
2009
9
Suppl 4
S395
S403
96
Cross
NCP
Feng
L
Chase
A
Bungey
J
Hughes
TP
Goldman
JM
Competitive polymerase chain reaction to estimate the number of BCR-ABL transcripts in chronic myeloid leukemia patients after bone marrow transplantation.
Blood
1993
82
6
1929
1936
97
Kaeda
J
O'Shea
D
Szydlo
RM
et al
Serial measurement of BCR-ABL transcripts in the peripheral blood after allogeneic stem cell transplantation for chronic myeloid leukemia: an attempt to define patients who may not require further therapy.
Blood
2006
107
10
4171
4176
98
Hughes
TP
Hochhaus
A
Branford
S
et al
Long-term prognostic significance of early molecular response to imatinib in newly diagnosed chronic myeloid leukemia: an analysis from the International Randomized Study of Interferon and STI571 (IRIS).
Blood
2010
116
19
3758
3765
99
Mahon
FX
Rea
D
Guilhot
J
et al
Discontinuation of imatinib in patients with chronic myeloid leukaemia who have maintained complete molecular remission for at least 2 years: the prospective, multicentre Stop Imatinib (STIM) trial.
Lancet Oncol
2010
11
11
1029
1035
100
Sokal
JE
Cox
EB
Baccarani
M
et al
Prognostic discrimination in “good-risk” chronic granulocytic leukemia.
Blood
1984
63
4
789
799
101
Hasford
J
Pfirrmann
M
Hehlmann
R
et al
A new prognostic score for survival of patients with chronic myeloid leukemia treated with interferon alfa: Writing Committee for the Collaborative CML Prognostic Factors Project Group.
J Natl Cancer Inst
1998
90
11
850
858
102
Hasford
J
Baccarani
M
Hoffmann
V
et al
Predicting complete cytogenetic response and subsequent progression-free survival in 2060 patients with CML on imatinib treatment: the EUTOS score.
Blood
2011
118
3
686
692
103
Verma
D
Kantarjian
HM
Jones
D
et al
Chronic myeloid leukemia (CML) with P190BCR-ABL: analysis of characteristics, outcomes, and prognostic significance.
Blood
2009
114
11
2232
2235
104
Mohty
M
Yong
ASM
Szydlo
RM
Apperley
JF
Melo
JV
The polycomb group BMI1 gene is a molecular marker for predicting prognosis of chronic myeloid leukemia.
Blood
2007
110
1
380
383
105
Krulik
M
Smadja
N
Degramont
A
et al
Sequential karyotype study on Ph-positive chronic myelocytic leukemia: significance of additional chromosomal abnormalities during disease evolution.
Cancer
1987
60
5
974
979
106
Geraci
L
Palka
G
Fioritoni
G
et al
Prognostic value of atypical chromosomal changes during chronic myeloid leukemia.
Haematologica
1987
72
6
515
521
107
Anastasi
J
Feng
J
Lebeau
MM
Larson
RA
Rowley
JD
Vardiman
JW
The relationship between secondary chromosomal abnormalities and blast transformation in chronic myelogenous leukemia.
Leukemia
1995
9
4
628
633
108
Majlis
A
Smith
TL
Talpaz
M
O'Brien
S
Rios
MB
Kantarjian
HM
Significance of cytogenetic clonal evolution in chronic myelogenous leukemia.
J Clin Oncol
1996
14
1
196
203
109
Schoch
C
Haferlach
T
Kern
W
et al
Occurrence of additional chromosome aberrations in chronic myeloid leukemia patients treated with imatinib mesylate.
Leukemia
2003
17
2
461
463
110
Marktel
S
Marin
D
Foot
N
et al
Chronic myeloid leukemia in chronic phase responding to imatinib: the occurrence of additional cytogenetic abnormalities predicts disease progression.
Haematologica
2003
88
3
260
267
111
Cortes
JE
Talpaz
M
Giles
F
et al
Prognostic significance of cytogenetic clonal evolution in patients with chronic myelogenous leukemia on imatinib mesylate therapy.
Blood
2003
101
10
3794
3800
112
O'Dwyer
ME
Mauro
MJ
Blasdel
C
et al
Clonal evolution and lack of cytogenetic response are adverse prognostic factors for hematologic relapse of chronic phase CML patients treated with imatinib mesylate.
Blood
2004
103
2
451
455
113
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
6
2128
2137
114
Haferlach
C
Bacher
U
Schnittger
S
Weiss
T
Kern
W
Haferlach
T
Similar patterns of chromosome abnormalities in CML occur in addition to the Philadelphia chromosome with or without tyrosine kinase inhibitor treatment.
Leukemia
2010
24
3
638
640
115
Verma
D
Kantarjian
H
Shan
J
et al
Survival outcomes for clonal evolution in chronic myeloid leukemia patients on second generation tyrosine kinase inhibitor therapy.
Cancer
2010
116
11
2673
2681
116
Terre
C
Eclache
V
Rousselot
P
et al
Report of 34 patients with clonal chromosomal abnormalities in Philadelphia-negative cells during imatinib treatment of Philadelphia-positive chronic myeloid leukemia.
Leukemia
2004
18
8
1340
1346
117
Bumm
T
Muller
C
Al Ali
HK
et al
Emergence of clonal cytogenetic abnormalities in Ph− cells in some CML patients in cytogenetic remission to imatinib but restoration of polyclonal hematopoiesis in the majority.
Blood
2003
101
5
1941
1949
118
Bacher
U
Hochhaus
A
Berger
U
et al
Clonal aberrations in Philadelphia chromosome negative hematopoiesis in patients with chronic myeloid leukemia treated with imatinib or interferon alpha.
Leukemia
2005
19
3
460
463
119
Jabbour
E
Kantarjian
HM
Abruzzo
LV
et al
Chromosomal abnormalities in Philadelphia chromosome-negative metaphases appearing during imatinib mesylate therapy in patients with newly diagnosed chronic myeloid leukemia in chronic phase.
Blood
2007
110
8
2991
2995
120
Oehler
VG
Yeung
KY
Choi
YE
Bumgarner
RE
Raftery
AE
Radich
JP
The derivation of diagnostic markers of chronic myeloid leukemia progression from microarray data.
Blood
2009
114
15
3292
3298
121
Mahon
FX
Faberes
C
Pueyo
S
et al
Response at three months is a good predictive factor for newly diagnosed chronic myeloid leukemia patients treated by recombinant interferon-alpha.
Blood
1998
92
11
4059
4065
122
Hanfstein
B
Müller
MC
Hehlmann
R
et al
Early molecular and cytogenetic response is predictive for long-term progression-free and overall survival in chronic myeloid leukemia (CML) [published online ahead of print March 26, 2012].
Leukemia
123
Marin
D
Ibrahim
AR
Lucas
C
et al
Assessment of BCR-ABL1 transcript levels at 3 months is the only requirement for predicting outcome for patients with chronic myeloid leukemia treated with tyrosine kinase inhibitors.
J Clin Oncol
2012
30
3
232
238
124
Jabbour
E
Kantarjian
H
O'Brien
S
et al
The achievement of an early complete cytogenetic response is a major determinant for outcome in patients with early chronic phase chronic myeloid leukemia treated with tyrosine kinase inhibitors.
Blood
2011
118
17
4541
4546
125
Jabbour
E
Kantarjian
H
O'Brien
S
et al
Predictive factors for outcome and response in patients treated with second-generation tyrosine kinase inhibitors for chronic myeloid leukemia in chronic phase after imatinib failure.
Blood
2011
117
6
1822
1827
126
Preudhomme
C
Guilhot
J
Nicolini
FE
et al
Imatinib plus peginterferon alfa-2a in chronic myeloid leukemia.
N Engl J Med
2010
363
26
2511
2521
127
Simonsson
B
Gedde-Dahl
T
Markevärn
B
et al
Combination of pegylated IFN-alpha2b with imatinib increases molecular response rates in patients with low- or intermediate-risk chronic myeloid leukemia.
Blood
2011
118
12
3228
3235
128
Saglio
G
Kim
DW
Issaragrisil
S
et al
Nilotinib versus imatinib for newly diagnosed chronic myeloid leukemia.
N Engl J Med
2010
362
24
2251
2259
129
Kantarjian
H
Shah
NP
Hochhaus
A
et al
Dasatinib versus imatinib in newly diagnosed chronic-phase chronic myeloid leukemia.
N Engl J Med
2010
362
24
2260
2270
130
Müller
MC
Cross
NC
Erben
P
et al
Harmonization of molecular monitoring of CML therapy in Europe.
Leukemia
2009
23
11
1957
1963
Sign in via your Institution