Management of CML-blast crisis

https://doi.org/10.1016/j.beha.2016.10.005Get rights and content

Highlights

  • BCR-ABL is the driving force of progression.

  • Clonal evolution indicates progression.

  • BC patients who achieve a 2nd CP have the best prognosis.

  • Allo-SCT should be tried in 2nd CP whenever a donor is available and the patient can tolerate the procedure.

  • BC can be prevented by elimination (or reduction) of BCR-ABL.

  • Much is known on genetic instability and clonal evolution as causes of BC, but confirmation of our understanding by successful intervention as proof of principle is lacking.

Abstract

Tyrosine kinase inhibitors (TKI) have moderately improved survival in BC, but a median survival of less than 1 year is still unsatisfactory. This article reviews the various tests required for diagnosis of BC, features at diagnosis, treatment modalities (intensive chemotherapy, TKI, allo-SCT and a selection of investigational agents), options of prevention and predictors of progression. The best prognosis is observed in patients that achieve a 2nd CP. Allo-SCT probably further improves prognosis of patients in 2nd CP. The choice of TKI should be directed by the mutation profile of the patient. BC can be prevented. A careful analysis of risk factors for progression may help. Current treatment options are combined in a concluding strategy for the management of BC.

Introduction

CML-blast crisis (BC) remains a challenge in the management of CML, although it develops, as a rule, under the eyes of the treating physician. Indicators are clonal evolution reaching aneuploidy levels of up to 90% [1] and mutation levels including resistance mutations to TKI treatment in up to 80% [2]. The increase of blast cells with failure of normal hemopoiesis is only the end stage of this evolution. It therefore is not surprising that the exact definition of BC (e.g. 30% blasts in the blood or marrow) is controversial [3]. Inspite of these indicators, no effective therapy exists to date, except for the occasional return to chronic phase (CP) with chemotherapy and subsequent transplantation. Prevention of BC by careful monitoring of treatment response and intensification of treatment, if response milestones are not reached remains the mainstay of management.

For the advancement of prevention and treatment several open questions and possible answers need to be addressed:

  • 1.

    Can we prevent progression to BC better by early treatment intensification according to response milestones? Answer: Carefully designed clinical trials could provide the answer.

  • 2.

    Can we identify patients at risk for progression better to improve prevention? Answer: Optimizing risk prognosticators combined with “personal” molecular markers.

  • 3.

    Can we define a point in the course of the disease after which treatment cannot reverse clonal evolution (point of no return)? Possible answer: by systematic aligning genetic with hematologic and clinical findings.

  • 4.

    What indicators precede an increase of blasts? Can we arrive at a more pathophysiologic definition of BC in the interest of better and timelier intervention? Possible answer: further dissecting more molecular pathways underlying BC with targeted intervention as proof of principle.

  • 5.

    Is genetic instability by BCR-ABL the single causative factor for clonal evolution, or are there other predisposing factors? Answer: Molecular techniques such as comparative whole genome sequencing may help.

In this review we present a broad overview of the diagnosis, therapy, prediction of progression and prevention of BC and our opinion of the open questions.

Section snippets

Diagnosis

To diagnose BC, complete blood and differential counts and a bone marrow analysis with cytogenetics are required. Cytogenetic evolution is the most consistent predictor of blast transformation. Flow cytometry or cytochemistry is needed to determine the type of BC (myeloid, lymphoid or mixed). 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

Pathogenetic basis of therapy

Treatment of BC is guided by our understanding of BC pathogenesis. Good in-depth reviews on the biology of BC have been published [20], [21]. According to current evidence, BC is the direct consequence of continued BCR-ABL activity [20], [21], possibly via oxidative stress and reactive oxygen species [22], [23], causing DNA damage and impaired DNA repair [24] and, in a vicious circle, genomic instability by more mutations, gene doublings, translocations, and chromosomal breakages [25]. The

Intensive chemotherapy

Once BC has been diagnosed, management depends on prior therapy and type of leukemia (myeloid or lymphoid). 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) [27]. The cells of the responding BC frequently showed features of lymphoid morphology and were TdT+ [28]. These observations

TKI therapy

The clinical improvement with TKI treatment in parallel to BCR-ABL reduction and the postponement (or prevention) of BC in most patients with TKI (8-year incidence of BC in IRIS < 8% under standard imatinib [30]) supports the conclusion that BCR-ABL is the driving force behind disease progression. This is confirmed by the experience of the German CML Study Group which reported a decrease of 8-year BC-incidence from 70% 25 years ago to approximately 5% in CML Study IV [3] [31]. The transient

Scenario 1

If the patient has been pretreated with conventional therapy (IFN or hydroxyurea, currently the exception), imatinib 600–800 mg/d (or 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 have been summarized recently [32]. Imatinib and dasatinib have been approved for all phases of CML, including BC by the Food and Drug Administration and the European Medicine Agency.

Five

Dasatinib

Three studies of 400 BC patients pretreated with imatinib, including 119 with lymphoid BC, showed hematologic remission rates of 33%–61% (lymphoid BC, 36%–80%), major cytogenetic remission (MCR) rates of 35%–56%, a 1-year survival of 42%–50%, a 2-year survival of 20%–30%, and a median survival of 8–11 months [39], [40], [41].

The largest of the studies, a randomized open label phase 3 study of 214 patients with 61 in lymphoid BC, attempted to optimize the dose-schedule of dasatinib, stratified

Nilotinib

Two studies of 169 patients have been published including 40 with lymphoid BC [43], [44], 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 adjustment. Nilotinib has been

Imatinib in combination

Several small studies have focused on the combination of imatinib at 600 mg–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 [45]. Hematologic response rate was 81% with a 1-year survival of approximately 50%, including 6 patients who had an allo-SCT. Another study combined imatinib 600 mg with decitabine in 10 patients and reported a median survival of 15 weeks [46]. The combination of imatinib

Dasatinib and nilotinib in combination

Milojkovic et al. [52] reported 4 patients who progressed to BC while on imatinib and were successfully treated with dasatinib 100 mg daily combined with fludarabine 30 mg/m2 iv days 1–5, cytosine arabinoside 2 g/m2 iv days 1–5, idarubicin 12 mg/m2 iv days 1–3 and G-CSF 300 mg/day sc days 0–6 (FLAG-IDA). All patients were alive, three after and one prior to SCT. Strati et al. treated 42 BC patients with hyperfractionated cyclophosphamide, vincristine, Adriamycin, dexamethasone (HCVAD) plus

Bosutinib and ponatinib

Since 2012 [3], two additional TKI have been approved for CML, bosutinib and ponatinib. Bosutinib, a third second-generation TKI, shows in preliminary analyses on 48 BC-patients similar activity (CCR 29%, MMR 28%, PFS 7.8 months) as dasatinib and nilotinib [55].

The pan BCR-ABL inhibitor ponatinib shows, in addition to recognizing the T315I mutation, efficacy in BC and Ph + ALL. A phase 2 study on 449 ponatinib-treated patients included 62 patients in BC. After a median follow-up of the BC

Allo-SCT

Although allo-SCT is successful in only a minority of BC patients it probably has the best outcome in BC, if the patient can tolerate the procedure and if a donor is available after a return to CP, assuming a complete remission has been achieved. The search for a donor should be instituted as early as possible. In an overview of the European Group for Blood and Marrow Transplantation from 1980 to 2003, 2-year survival rates are 16%–22% [59]. Most patients were transplanted in the pre-imatinib

Investigational agents

A number of investigational approaches are under exploration. A selection is presented in Table 2. The approaches include activation of the tumor suppressor protein phosphatase 2A (PP2A), which shows decreased activity in BC [68] through up-regulation of its inhibitors suppressor of variegation, enhancer of zeste and trithorax (SET), and cancerous inhibitor of PP2A (CIP2A) [69] which upregulates the antiapoptotic protein BCR-XL [70]; inhibition of self-renewal of leukemia stem cells (LSCs) by

Prevention

The low progression rates of CML with TKI maintenance therapy indicate that BC can be prevented. Also, it is well known that very low or undetectable BCR-ABL transcripts after allo-SCT correlate with low relapse rates [80]. Imatinib-treated patients who have achieved MMR enjoy durable responses with virtually no current progression to AP or BC [81]. Patients who have achieved stable complete molecular remission may experience in approximately 40% of cases complete continued remission in the

Prediction of progression

At diagnosis, risk scores provide information on the likelihood of progression. The EUTOS score [83] 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 ELTS-score [84] identifies three risk groups with significantly different probabilities of death due to CML. Distinct markers such as major route ACA [6], p190BCR−ABL [85],

Conclusion

The management strategy in Table 3 gives an overview how to prevent BC or approach management after BC has evolved. Goal is the return to chronic phase or the induction of a true cytogenetic and/or molecular remission. The main form of treatment should be a TKI preferably based upon the type of remission followed quickly by allo-SCT if possible. If TKIs are not sufficient, AL-type induction therapy should be tried, cytosine arabinoside and anthracyclines for myeloid BC, vincristine and

Conflict of interest

The authors declare no conflict of interest.

Acknowledgments

This work was supported by the Deutsche Krebshilfe (Nr. 109588), Novartis, Nürnberg, Germany, Kompetenznetz für Akute und Chronische Leukämien (BMBF 01GI0270), Deutsche José-Carreras Leukämiestiftung (DJCLS H09/01f, H06/04v, H03/01, R05/23), European LeukemiaNet (LSHC-CT-2004-503216), Roche, Grenzach-Wyhlen, Germany and Essex Pharma, München, Germany and the Cancer Research and Treatment Fund, Inc., New York, New York.

The contributions of A. Elett, G. Bartsch, S. Dean, E. Matzat, R.

References (100)

  • M.O. Nowicki et al.

    BCR/ABL oncogenic kinase promotes unfaithful repair of the reactive oxygen species-dependent DNA double-strand breaks

    Blood

    (2004)
  • S. Soverini 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)
  • M. Sattler et al.

    The BCR/ABL tyrosine kinase induces production of reactive oxygen species in hematopoietic cells

    J Biol Chem

    (2000)
  • M. Deininger 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 Annu Meet Abstr

    (2009)
  • C.L. Sawyers 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)
  • H.M. Kantarjian et al.

    Imatinib mesylate (STI571) therapy for Philadelphia chromosome–positive chronic myelogenous leukemia in blast phase

    Blood

    (2002)
  • K. Porkka et al.

    Dasatinib crosses the blood-brain barrier and is an efficient therapy for central nervous system Philadelphia chromosome-positive leukemia

    Blood

    (2008)
  • B. Deau et al.

    The addition of daunorubicin to imatinib mesylate in combination with cytarabine improves the response rate and the survival of patients with myeloid blast crisis chronic myelogenous leukemia (AFR01 study)

    Leuk Res

    (2011)
  • P. Valent et al.

    Vascular safety issues in CML patients treated with BCR/ABL1 kinase inhibitors

    Blood

    (2015)
  • S. Saussele 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)
  • A. Gratwohl et al.

    Current role of stem cell transplantation in chronic myeloid leukaemia

    Best Pract Res Clin Haematol

    (2009)
  • E. Jabbour et al.

    Sudden blastic transformation in patients with chronic myeloid leukemia treated with imatinib mesylate

    Blood

    (2006)
  • P. Neviani 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)
  • C.M. Lucas et al.

    Cancerous inhibitor of PP2A (CIP2A) at diagnosis of chronic myeloid leukemia is a critical determinant of disease progression

    Blood

    (2011)
  • H. Zhang et al.

    HIF1α is required for survival maintenance of chronic myeloid leukemia stem cells

    Blood

    (2012)
  • J. Xie et al.

    Combination of rapamycin and imatinib in treating refractory chronic myeloid leukemia myeloid blast crisis:a case report(025B3;)

    Chin Med Sci J

    (2013)
  • J. Kaeda 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)
  • J. Hasford et al.

    Predicting complete cytogenetic response and subsequent progression-free survival in 2060 patients with CML on imatinib treatment: the EUTOS score

    Blood

    (2011)
  • D. Verma et al.

    Chronic myeloid leukemia (CML) with P190BCR-ABL: analysis of characteristics, outcomes, and prognostic significance

    Blood

    (2009)
  • J.E. Cortes et al.

    Prognostic significance of cytogenetic clonal evolution in patients with chronic myelogenous leukemia on imatinib mesylate therapy

    Blood

    (2003)
  • M. Baccarani et al.

    European LeukemiaNet recommendations for the management of chronic myeloid leukemia

    Blood

    (2013)
  • S. Branford et al.

    Prognosis for patients with CML and >10% BCR-ABL1 after 3 months of imatinib depends on the rate of BCR-ABL1 decline

    Blood

    (2014)
  • N.P. Shah et al.

    Dasatinib plus smoothened (SMO) inhibitor BMS-833923 in chronic myeloid leukemia (CML) with resistance or suboptimal response to a prior tyrosine kinase inhibitor (TKI): phase I study CA180323

    Blood ASH Annu Meet Abstr

    (2014)
  • P. Gallipoli et al.

    JAK2/STAT5 inhibition by nilotinib with ruxolitinib contributes to the elimination of CML CD34+ cells in vitro and in vivo

    Blood

    (2014)
  • W. Wang et al.

    The effect of nilotinib plus arsenic trioxide on the proliferation and differentiation of primary leukemic cells from patients with chronic myoloid leukemia in blast crisis

    Cancer Cell Int

    (2015)
  • B. Johansson et al.

    Cytogenetic and molecular genetic evolution of chronic myeloid leukemia

    Acta Haematol

    (2002)
  • F. Mitelman et al.

    Non-random karyotypic evolution in chronic myeloid leukemia

    Int J Cancer

    (1976)
  • Z. Chen et al.

    Differential impact of additional chromosomal abnormalities in myeloid vs lymphoid blast phase of chronic myelogenous leukemia in the era of tyrosine kinase inhibitor therapy

    Leukemia

    (2016)
  • W. Haaß et al.

    Clonal evolution and blast crisis correlate with enhanced proteolytic activity of separase in BCR-ABL b3a2 fusion type CML under imatinib therapy

    PLoS One

    (2015)
  • S. Soverini 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)
  • V. Grossmann 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)
  • C.G. Mullighan et al.

    BCR-ABL1 lymphoblastic leukaemia is characterized by the deletion of Ikaros

    Nature

    (2008)
  • C. Zheng et al.

    Gene expression profiling of CD34+ cells identifies a molecular signature of chronic myeloid leukemia blast crisis

    Leukemia

    (2006)
  • J.P. Radich et al.

    Gene expression changes associated with progression and response in chronic myeloid leukemia

    Proc Natl Acad Sci U S A

    (2006)
  • C.H. Jamieson et al.

    Granulocyte-macrophage progenitors as candidate leukemic stem cells in blast-crisis CML

    N Engl J Med

    (2004)
  • D. Perrotti et al.

    Chronic myeloid leukemia: mechanisms of blastic transformation

    J Clin Invest

    (2010)
  • B. Chereda et al.

    Natural course and biology of CML

    Ann Hematol

    (2015)
  • T. Skorski

    Oncogenic tyrosine kinases and the DNA-damage response

    Nat Rev Cancer

    (2002)
  • A.M. Marmont et al.

    The treatment of terminal metamorphosis of chronic granulocytic leukaemia with corticosteroids and vincristine

    Acta Haematol

    (1973)
  • S.M. Marks et al.

    Terminal transferase as a predictor of initial responsiveness to vincristine and prednisone in blastic chronic myelogenous leukemia

    N Engl J Med

    (1978)
  • Cited by (0)

    1

    Fax: +49 (0)621 383 6932.

    2

    Fax: +1 646 962 1606.

    View full text