Leukemic Transformation in Myeloproliferative Neoplasms

Leukemic Transformation in Myeloproliferative Neoplasms

SYMPOSIUM ON NEOPLASTIC HEMATOLOGY AND MEDICAL ONCOLOGY Leukemic Transformation in Myeloproliferative Neoplasms: A Literature Review on Risk, Charact...

245KB Sizes 0 Downloads 87 Views

SYMPOSIUM ON NEOPLASTIC HEMATOLOGY AND MEDICAL ONCOLOGY

Leukemic Transformation in Myeloproliferative Neoplasms: A Literature Review on Risk, Characteristics, and Outcome Meera Yogarajah, MD, and Ayalew Tefferi, MD From the Division of Hematology and Oncology, Brody School of Medicine, East Carolina University, Greenville, NC (M.Y.); and Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN (A.T.).

CME Activity Target Audience: The target audience for Mayo Clinic Proceedings is primarily internal medicine physicians and other clinicians who wish to advance their current knowledge of clinical medicine and who wish to stay abreast of advances in medical research. Statement of Need: General internists and primary care physicians must maintain an extensive knowledge base on a wide variety of topics covering all body systems as well as common and uncommon disorders. Mayo Clinic Proceedings aims to leverage the expertise of its authors to help physicians understand best practices in diagnosis and management of conditions encountered in the clinical setting. Accreditation: Mayo Clinic College of Medicine and Science is accredited by the Accreditation Council for Continuing Medical Education to provide continuing medical education for physicians. Credit Statement: Mayo Clinic College of Medicine and Science designates this journal-based CME activity for a maximum of 1.0 AMA PRA Category 1 Credit(s).TM Physicians should claim only the credit commensurate with the extent of their participation in the activity. Credit Statement: Successful completion of this CME activity, which includes participation in the evaluation component, enables the participant to earn up to 1 MOC point in the American Board of Internal Medicine’s (ABIM) Maintenance of Certification (MOC) program. Participants will earn MOC points equivalent to the amount of CME credits claimed for the activity. It is the CME activity provider’s responsibility to submit participant completion information to ACCME for the purpose of granting ABIM MOC credit. Learning Objectives: On completion of this article, you should be able to (1) describe risk factors for leukemic transformation in myeloproliferative neoplasms; (2) review current experience in the outcome of patients with blast-phase myeloproliferative neoplasms; and (3) discuss current challenges in the treatment of blast-phase myeloproliferative neoplasms. Disclosures: As a provider accredited by ACCME, Mayo Clinic College of Medicine and Science (Mayo School of Continuous Professional Development) must

ensure balance, independence, objectivity, and scientific rigor in its educational activities. Course Director(s), Planning Committee members, Faculty, and all others who are in a position to control the content of this educational activity are required to disclose all relevant financial relationships with any commercial interest related to the subject matter of the educational activity. Safeguards against commercial bias have been put in place. Faculty also will disclose any off-label and/or investigational use of pharmaceuticals or instruments discussed in their presentation. Disclosure of this information will be published in course materials so that those participants in the activity may formulate their own judgments regarding the presentation. In their editorial and administrative roles, Karl A. Nath, MBChB, Terry L. Jopke, Kimberly D. Sankey, and Nicki M. Smith, MPA, have control of the content of this program but have no relevant financial relationship(s) with industry. The authors report no competing interests. Method of Participation: In order to claim credit, participants must complete the following: 1. Read the activity. 2. Complete the online CME Test and Evaluation. Participants must achieve a score of 80% on the CME Test. One retake is allowed. Visit www.mayoclinicproceedings.org, select CME, and then select CME articles to locate this article online to access the online process. On successful completion of the online test and evaluation, you can instantly download and print your certificate of credit. Estimated Time: The estimated time to complete each article is approximately 1 hour. Hardware/Software: PC or MAC with Internet access. Date of Release: 7/1/2017 Expiration Date: 6/30/2019 (Credit can no longer be offered after it has passed the expiration date.) Privacy Policy: http://www.mayoclinic.org/global/privacy.html Questions? Contact [email protected].

Abstract Myeloproliferative neoplasms (MPNs) operationally include essential thrombocythemia, polycythemia vera, primary myelofibrosis (PMF), and prefibrotic PMF. All 4 MPN variants might progress into blast-phase disease (MPN-BP). For essential thrombocythemia, reported risk factors for leukemic transformation include advanced age, extreme thrombocytosis, anemia, leukocytosis, and sequence variants/mutations involving TP53 and EZH2 (for expansion of gene symbols, see www.genenames.org); for polycythemia vera, advanced age, leukocytosis, abnormal karyotype, mutations involving SRSF2 and IDH2, and treatment with pipobroman, chlorambucil, or P32; and for PMF, increased blast percentage, thrombocytopenia, abnormal karyotype, triple-negative driver mutational status, and sequence variants/mutations involving SRSF2, RUNX1, CEBPA, and SH2B3. The reported median survival figures for MPN-BP range from 1.5 to 2.5 months in patients treated with supportive care only, from 2.5 to 10 months in those receiving hypomethylating agents or low-dose chemotherapy, and from 3.9 to 9.4 months in those receiving induction chemotherapy. Three-year survival after allogeneic stem cell transplant was reported in 16% to 33% of patients. These observations validate the extremely poor prognosis associated with MPN-BP and the lack of effective drug therapy and highlight the need for urgent assessment of therapeutic values of investigational agents. In the meantime, affected patients might be best served with aggressive chemotherapy followed by allogeneic stem cell transplant after adequate blast clearance. ª 2017 Mayo Foundation for Medical Education and Research

1118

n

Mayo Clin Proc. 2017;92(7):1118-1128

Mayo Clin Proc. n July 2017;92(7):1118-1128 n http://dx.doi.org/10.1016/j.mayocp.2017.05.010 www.mayoclinicproceedings.org n ª 2017 Mayo Foundation for Medical Education and Research

LEUKEMIC TRANSFORMATION IN MYELOPROLIFERATIVE NEOPLASMS

M

yeloproliferative neoplasms (MPNs) are clonal hematopoietic stem cell disorders characterized by proliferative bone marrow with varying degrees of reticulin/ collagen fibrosis, extramedullary hematopoiesis, abnormal peripheral blood count, and constitutional symptoms that are presumed to be secondary to abnormally expressed inflammatory cytokines.1,2 The classical categories of MPN consists of chronic myeloid leukemia, primary myelofibrosis (PMF), essential thrombocythemia (ET), and polycythemia vera (PV). Chronic myeloid leukemia is invariably associated with a specific genetic abnormality (BCR-ABL1) (for expansion of gene symbols, see www. genenames.org)3,4; accordingly, the other MPNs are operationally labeled as “BCR-ABL1enegative MPN,” and the World Health Organization (WHO) classification system currently recognizes 4 variants of BCR-ABL1enegative MPN: ET, PV, PMF, and prefibrotic PMF.5 There is considerable overlap among the 4 MPN variants in terms of symptomatology, laboratory findings, bone marrow morphology, and mutations, however, with marked differences in natural history and survival outcomes.5 Accordingly, diagnostic accuracy is prognostically relevant, especially in terms of differentiating WHO-defined ET from prefibrotic PMF and masked PV, which displays a higher risk of leukemic transformation.6,7 In general, the natural history of MPN might be interrupted by thrombohemorrhagic complications8 or disease transformation into post-ET or post-PV myelofibrosis (MF)9 or post-MPN acute leukemia, also known as blast-phase myeloproliferative neoplasms (MPN-BP).10,11 The typical definition of MPN-BP includes the documentation of 20% or more blasts in the peripheral blood or bone marrow.12 However, discordance in the content of peripheral blood vs bone marrow blasts is often seen.13 The propensity for leukemic transformation in MPN varies according to the MPN variant and is highest in PMF, with an incidence of 10% to 20% in the first 10 years of the disease14-16; lower in PV, with an estimated risk of 2.3% at 10 years and 7.9% at 20 years17; and lowest in ET, with less than 1% in the first decade of the disease.6,18 This varying incidence of MPN-BP was recently highlighted in a large study by Tefferi et al.10 Mayo Clin Proc. n July 2017;92(7):1118-1128 www.mayoclinicproceedings.org

n

CHARACTERISTICS OF MPN-BP Blast-phase myeloproliferative neoplasms commonly involve the myeloid lineage with rare involvement of the lymphoid lineage. The characteristics of MPN-BP have been reported to be different, both morphologically and cytogenetically, from primary (de novo) acute myeloid leukemia (AML). According to the French-American-British (FAB) classification of AML, erythroleukemia (FAB-M6) and megakaryoblastic leukemia (FAB-M7) were the most common subtypes reported in MPN-BP.14,19 A Mayo Clinic study of patients with PMF found M7 (25.4%) to be the most common subtype followed by M0 (22.4%) and M2 (17.9%).19 The incidence of AML in postET or post-PV MF is similar to that of postPMF AML.19 Patients with PV and ET can also directly develop AML without going through the fibrotic phase of the disease.16,20 A karyotype in MPN-BP is often complex, and a favorable karyotype is infrequent. Common mutations described in MPN-BP include JAK2, IDH1/2, TP53, ASXL1, and TET2, whereas their counterparts in primary AML include N/KRAS, DNMT3a, NPM1, and FLT3.21-24 A study that analyzed genetic information in paired samples of chronic MPN-BP reported acquisition of TET2 mutation during blast transformation and presence of ASXL1 in both phases of the disease.22 JAK2 mutations are important in MPN pathogenesis, and clonal changes in JAK2 clones, during leukemic transformation, have been observed with acquisition of new mutations and, in certain instances, loss of the prevailing mutations.25,26 RISK FACTORS FOR LEUKEMIC TRANSFORMATION IN MPN Various clinical and genetic risk factors have been identified as being useful in predicting leukemic transformation in MPN (Table 1). RISK FACTORS FOR LEUKEMIC TRANSFORMATION IN PMF Conventional drug therapies have not resulted in prolongation of survival in PMF, and allogeneic stem cell transplant (ASCT) remains the definitive treatment for purposes of cure or prolongation of survival. Accordingly, identification of risk factors for inferior overall

http://dx.doi.org/10.1016/j.mayocp.2017.05.010

1119

MAYO CLINIC PROCEEDINGS

TABLE 1. Risk Factors for Leukemic Transformation Risk factors

Essential thrombocytosis

Polycythemia vera

Clinical

Age 60 y Thrombosis6

Age >70 y Age >61 y17

Laboratory

Platelets (1000109/L)6,28 Anemia28,32 Leukocytosis (15109/L)33,34

Leukocytes (15109)17

Bone marrow

Prefibrotic primary myelofibrosis morphology, reticulin grading, and bone marrow cellularity6,34

27,28

Previous therapy

Myelofibrosis Age >65 y Red blood cell transfusion dependency31 White blood cells (>30109/L)35 Platelets (<50109/L)36 Peripheral blast (3%) and/or platelets (<100109/L)37 IL-8, IL-2R38 C-reactive protein (>7 mg/L) and peripheral blast (>1%)30 Bone marrow blasts (10%)36

29

30

Cytogenetic abnormalities

None

Cytoreductive agents, pipobroman, P32, chlorambucil39 Abnormal karyotype17

Mutations

TP53, EZH233

SRSF2, IDH233

Abnormal karyotype35 Chromosome 17 aberrations36 Monosomal karyotype (1q, 7q, 5q, 6p, 7p, 19q, 22q, and 3q, del17p, 5, 7) and/or complex40 Unfavorable karyotype (complex karyotype or sole or 2 abnormalities that include þ8, 7/7q-, i(17q), 5/ 5q-, 12p-, inv(3), or 11q23 rearrangement)41,42 Del17p, 5, 7, and/or complex43 Triple-negative mutational status10 ASXL1, SRSF2, IDH1, or IDH244

IL-2R ¼ interleukin 2 receptor; IL-8 ¼ interleukin 8.

and leukemia-free survival is a critical step in the management of patients with PMF or post-ET or post-PV MF. In a study of 311 patients with PMF, independent risk factors for leukemic transformation included circulating blast of more than 3% and platelet count of less than 100109/L.37 The same study also found no increased leukemogenicity from treatment with hydroxyurea, thalidomide, or many other drugs, although a potential detrimental effect from erythropoiesis stimulating agents, and danazol was suggested. Using the aforementioned 2 risk factors, the rate of leukemic transformation was only 6% in the absence of both risk factors and 18% in the presence of one or both risk factors. Leukocytosis (>30109/L) was also identified as a risk factor for leukemic transformation in another 1120

Mayo Clin Proc.

n

study.35 In a study by Passamonti et al,31 red blood cell (RBC) transfusion need was also identified as an independent risk factor for leukemic transformation, with an incidence of leukemic transformation at 7.4100 persons per year in RBC-transfused patients vs 1.5100 persons per year in nontransfused patients (P<.001). Other reported risk factors for leukemic transformation include increased serum interleukin 8 levels,38 C-reactive protein level of greater than 7 mg/L, age greater than 65 years, and peripheral blast count of more than 1%.30 The International Prognostic Scoring System45 and the Dynamic International Prognostic Scoring System (DIPSS)46 are currently used to risk stratify patients with PMF and predict survival. The DIPSS was further modified to DIPSS plus41 by including

July 2017;92(7):1118-1128

n

http://dx.doi.org/10.1016/j.mayocp.2017.05.010 www.mayoclinicproceedings.org

LEUKEMIC TRANSFORMATION IN MYELOPROLIFERATIVE NEOPLASMS

thrombocytopenia, unfavorable karyotype, and RBC transfusion need as additional risk factors. An analysis of risk factors for leukemic transformation, in the context of DIPSS plus, identified unfavorable karyotype and platelet count less than 100109/L as being the most important: patients with no risk factors had a risk of leukemic transformation at 5 years of 6% and at 10 years of 12%, whereas the risk was substantially higher in patients with 1 or more risk factors at 18% and 31% at 5 and 10 years, respectively.35 Subsequent studies have confirmed the adverse effect of specific cytogenetic abnormalities, including monosomal and complex karyotypes.40 In a study of 793 patients with PMF, the 2-year rate of leukemic transformation was 29.4% in patients with monosomal karyotype compared with 8.3% in patients with complex karyotype.40 Mutually exclusive driver mutations upregulating the JAK-STAT signaling pathway are frequent in MPN. JAK2 mutations are almost always present in patients with PV, whereas their incidence in PMF and ET is similar and estimated at 60%.47 In ET and PMF, the remaining 40% of patients harbor either CALR (25%-35%)48-54 or MPL (3%8%) mutations.47 Approximately 10% to 15% of patients with ET or PMF do not express any one of the aforementioned 3 driver mutations and are often referred to as being “triple negative.”10,55 The prognostic role of driver mutations, in terms of leukemic transformation, is best recognized for PMF where a higher risk has been associated with triple negative and a lower risk with CALR type 1 mutation status.10 The most elaborate studies assessing the prognostic role of mutations comes from 2 patient cohorts from Mayo Clinic and Florence, Italy.44 These studies identified ASXL1, SRSF2, IDH1, or IDH2 mutations as independent risk factors for leukemic transformation. The contribution of these mutations in conferring high risk for leukemic transformation was reported in other studies as well.56-59 A Genetics-Based Prognostic Scoring System for PMF that integrated karyotype information and mutation status was recently reported in an abstract form at the American Society of Hematology annual meeting.60 Cytogenetic risk categorization included very Mayo Clin Proc. n July 2017;92(7):1118-1128 www.mayoclinicproceedings.org

n

high (monosomal karyotype, inv(3), i(17q), 7/7q-, 11q, or 12p abnormalities), high (complex nonmonosomal, 2 abnormalities not included in the very high risk category, 5q-, þ8, other autosomal trisomies except þ9, and other sole abnormalities not included in other risk categories), intermediate (sole abnormalities of 20q-, 1qþ or any other sole translocation, and -Y or other sex chromosome abnormality), and low (normal or sole abnormalities of 13q- or þ9). Mutational status was considered favorable in the presence of CALR type 1/type 1elike variants and unfavorable in the presence of ASXL1, SRSF2, EZH2, and IDH1/2 mutations. The integration of karyotype and mutational status allowed risk stratification into high, intermediate 2, intermediate 1, and low risk groups, with the Genetics-Based Prognostic Scoring Systemeidentified high-risk group displaying the highest risk of leukemic transformation; leukemia-free survival was significantly different between the high-intermediate 2 group vs low-intermediate 1 group (median, 11.6 years vs not reached), and this was validated in another independent cohort.60 Most recently, a Mayo Clinic study of targeted sequencing in PMF identified ASXL1, SRSF2, CBL, and KIT mutations as interindependent risk factors for overall survival.57 Univariate analysis for leukemiafree survival identified SRSF2, RUNX1, CEBPA, SH2B3, and IDH2 mutations as risk factors, and the first 4 remained significant in that regard in multivariate analysis. RISK FACTORS FOR LEUKEMIC TRANSFORMATION IN PV It is now well established that treatment of patients with PV treated with P32, chlorambucil, or pipobroman results in a higher risk of leukemic transformation.39,61 Older age,17,29 leukocytosis,17,62 and abnormal karyotype17 have also been associated with a higher risk of leukemic transformation. A recent Mayo Clinic study evaluated 133 patients with PV, in which 5.4% had documented leukemic transformation.63 Sequence variants or mutations in PV other than JAK2, CALR, or MPL were seen in 52.6% of patients. ASXL1, SRSF2, and IDH2 were identified as adverse variants or mutations on the basis of their effect on overall, leukemia-free, or MF-free

http://dx.doi.org/10.1016/j.mayocp.2017.05.010

1121

MAYO CLINIC PROCEEDINGS

survival. In particular, SRSF2, IDH2, and RUNX1 mutations were associated with shortened leukemia-free survival in univariate analysis; SRSF2 and IDH2 mutations remained significant in multivariate analysis.

RISK FACTORS FOR LEUKEMIC TRANSFORMATION IN ET The reported incidence of leukemic transformation in ET has varied from less than 1% to almost 10% in some studies. In this regard, the 10-year rates from earlier studies have ranged from 2.6%27 to 8.3%-9.7%.33,64,65 More recent studies that have carefully excluded patients with prefibrotic PMF from their study population suggest a remarkably lower rate of less than 1% at 10 years and 2% in 15 years in WHO-defined ET.6,66 In regard to risk factors for leukemic transformation in ET, Gangat et al28 studied 605 consecutive patients and identified anemia with hemoglobin levels below sex-adjusted normal values, extreme thrombocytosis (1000109/L), and age as continuous variables for leukemic transformation; the first 2 risk factors were included in a prognostic model that predicted leukemic transformation, and the particular risk was low at 0.4% when both risk factors were absent and was significantly higher at 4.8% and 6.5% in the presence of one or both risk factors, respectively (P<.001). Other risk factors for leukemic transformation in ET identified by other studies include leukocytosis (15109/L),33,34 extreme thrombocytosis (1000109/L),6,28 anemia,28,32,34 older age,27,28 and increased reticulin content or bone marrow cellularity6,34; it is possible that the last 2 parameters segregated with incorrectly diagnosed cases with prefibrotic PMF. The effect of cytoreductive drugs in leukemic transformation of patients with ET was examined in several studies including a cohort of 338 patients67 in which single agent treatment with hydroxyurea or busulfan did not appear to increase the risk of leukemic transformation. Many other retrospective studies have also validated the absence of convincing evidence for drug leukemogenicity in ET, although reports to the contrary are also noted.33 Regardless, there is general agreement on the absence of controlled data 1122

Mayo Clin Proc.

n

that implicate any 1 drug or combination thereof as being leukemogenic in ET. More recent studies have examined the role of driver and other mutations in the development of leukemia in ET. Among the 3 MPN-associated driver mutations, MPL mutations have been associated with a higher risk of fibrotic progression but their effect on leukemic transformation was either not apparent68,69 or noted during longer-term follow-up.53 A Mayo Clinic study63 of targeted sequencing recently identified TP53, EZH2, SRSF2, and IDH2 variants or mutations as being associated with a higher risk of leukemic transformation in univariate analysis; TP53 and EZH2 remained significant in multivariate analysis. OUTCOME OF PATIENTS WITH MPN-BP The survival of patients with MPN-BP is generally considered to be extremely poor (Table 2). This is not unexpected, given its high-risk morphological and cytogenetic features compared with those of both chronic phase MPN and primary AML. Blast-phase myeloproliferative neoplasms are often, but not always, preceded by a fibrotic phase of the disease.36 In the largest retrospective review of 273 patients with MPN-BP treated with hypomethylating agents (n¼99), high-dose cytarabine (n¼71), or low-dose cytarabine (n¼52),81 progression-free or overall survival did not differ among the treatment groups; 46 patients subsequently received ASCT, with 27 in complete remission (CR), and median survival was longer in patients in CR vs patients not in CR (35.1 months vs 8.9 months). The authors also noted that treatment outcome had not changed between 1989 and 2016. In a similarly large study,19 we reported on 91 consecutive cases of MPN-BP; the overall outcome was poor, with a median survival of 2 to 3 months of the subgroup who received induction chemotherapy and responded faring a little better with a median survival of 6.2 months as opposed to 2.7 months of patients who did not respond to treatment (P¼NS). Only 1 patient had received ASCT and survived for more than 2 years.19 There are several other reports in the literature on MPN-BP. Tam et al16 reported on 74 patients and also documented

July 2017;92(7):1118-1128

n

http://dx.doi.org/10.1016/j.mayocp.2017.05.010 www.mayoclinicproceedings.org

Mayo Clin Proc. n July 2017;92(7):1118-1128 www.mayoclinicproceedings.org

n

99 pts, 7.8 mo, HMA, 52 pts, 8 mo, low dose

19 pts, 9.9 mo, azacytidine

21 pts, 6.9 mo, decitabine

18 pts, 0% response

18 pts, 17% response 15 pts, 6.6 mo

26 pts, 8 mo, azacitidine 6 pts, >9 mo, decitabine

8 pts, 2.5 mo, low dose 19 pts, 2.9 mo, low dose 12 pts, 7 mo, low dose

26 11 14 23 13 18 75 46 60 21 18 19 13 273 Thepot et al,70 2010 Mascarenhas et al,13 2010 Ciurea et al,71 2010 Noor et al,11 2011 Cherington et al,72 2012 Eghtedar et al,73 2012 Kennedy et al,74 2013 Alchalby et al,75 2014 Cahu et al,76 2014 Badar et al,77 2015 Pemmaraju et al,78 2015 Andriani et al,79 2015 Shanavas et al,80 2016 Chihara et al,81 2016

7 pts, 2.5 mo 48 pts, 2.1 mo 19 pts, 1.5 mo 23 91 74 Passamonti et al,20 2005 Mesa et al,19 2005 Tam et al,16 2008

http://dx.doi.org/10.1016/j.mayocp.2017.05.010

HMA ¼ hypomethylating agent; mOS ¼ median overall survival; OS ¼ overall survival; PFS ¼ progression-free survival; pts ¼ patients.

71 pts, 7.1 mo

13 pts, 32% 2-y OS 46 pts, 15.3 mo

17 pts, 47 mo 46 pts, 33% 3-y OS 60 pts, 16% 3-y OS

20 pts, 6 mo

13 pts, 9.4 mo

5 pts, >20 mo 14 pts, 31 mo 3 pts, 10.5 mo 8 pts, 75% 2-y PFS

1 pt, survived >70 d 1 pt, survived >57 mo 8 pts, 73% survival, median follow-up 31 mo 8 pts, 5.6 mo 24 pts, 3.9 mo 36 pts, 6 mo

Induction chemotherapy: n, mOS JAK2 inhibitor ruxolitinib HMA or low-dose chemotherapy: n, mOS Supportive care: n, mOS N Reference, study

TABLE 2. Survival Outcomes of Blast-Phase Myeloproliferative Neoplasms

responses to AML-like induction chemotherapy that were not, however, durable but with improved survival as compared with supportive care (median, 6 months vs 1.5 months); of note, patients who received ASCT at any point during their treatment had better survival than did those who did not receive ASCT, and the survival advantage was even more evident for early transplant with 73% survival at a median follow-up of 31 months; the authors identified older age, peripheral blasts of 20% or more, and previous splenectomy as independent risk factors for survival.16 Passamonti et al20 reported the outcome of MPN-BP after PV and found similar survival between patients who had received AML-like induction chemotherapy (median, 5.6 months) or palliation with low-dose chemotherapy (cytarabine or 6thioguanine) or supportive care (median, 2.5 months). Noor et al11 studied 23 patients with MPN-BP, among whom 20 received AML-like induction chemotherapy, with 12 achieving remission; median survival was higher in responding patients at 12.8 months compared with 2.9 months in patients who did not achieve remission; 3 patients underwent ASCT with a median survival of 10.5 months. The authors subsequently scanned the literature and gathered information on 112 patients with MPN-BP to identify favorable risk factors for survival: antecedent diagnosis of ET, absence of complex karyotype, age less than 60 years, and less than 3 previous therapies. In yet another study of 13 patients with MPN-BP, 11 were treated with AML-like induction chemotherapy, with 8 patients achieving complete response, reversal to chronic phase disease, or marrow blast clearance72; 8 patients underwent ASCT, including 5 patients with less than 5% blood and bone marrow blast, 2 patients with peripheral blasts, and 1 patient with more than 5% bone marrow blast; outcome in this small study was better in the absence of excess bone marrow blast, but patients with circulating blasts were still salvageable. Hypomethylating agents, such as azacitidine and decitabine, have also been shown to have activity in MPN-BP; in a study of 26 patients treated with azacitidine, a 38% response rate (median time to response,

Allogeneic stem cell transplant: n, mOS or survival

LEUKEMIC TRANSFORMATION IN MYELOPROLIFERATIVE NEOPLASMS

1123

MAYO CLINIC PROCEEDINGS

9 months) with an overall median survival of 8 months was reported.70 In that study, the authors observed better responses in post-ET, as opposed to post-PV, MPN-BP, with no significant difference in overall survival.70 Another study reviewed patients with MPN-BP treated with azacytidine (75 mg/m2) and found that the observed median survival of 9.9 months was better than that of their historical controls79; once again, the median survival of patients who achieved CR was even better at 19.6 months. In a retrospective comparison to 6 patients treated with decitabine, the 5 who received low-intensity ASCT survived longer, with a median survival of more than 20 months.13 Similar observations were noted in another study of 14 patients with MPN-BP who received ASCT after first being chemotherapy-induced and 2-year survival was reported at 49%.71 Badar et al77 studied the use of decitabine in 21 patients with MPN-BP; 6 patients responded (3 CR, 2 near CR, and 1 partial response), and survival was better in responders vs nonresponders (median, 10.5 months vs 4 months); the authors did not find survival difference between decitabine-treated patients and historical controls treated with AML-like induction chemotherapy without ASCT. Kennedy et al74 adopted an approach of using curative intent treatment with induction chemotherapy followed by ASCT in fit and transplant-eligible patients. Seventy-five patients were included in the study, with 39 patients receiving AML-like induction chemotherapy followed by ASCT in eligible patients (17 of 39). The 36 other patients were treated with noncurative intent using hypomethylating agents, novel agents, or supportive care. Two-year survival was superior at 25.6% vs 3.1% in patients treated with curative intent. Furthermore, median survival was impressive at 47 months in patients who received ASCT and was not affected by the type of conditioning regimen. Induction chemotherapy without ASCT was unable to induce long-term remissions and resulted in a median survival of 9.4 months. The transplant cohort and the induction chemotherapy arm with responses but without transplant were compared, and survival was significantly better in the transplant group (2-year survival of 47% vs 15%; P¼.03). The median survival for 1124

Mayo Clin Proc.

n

low-intensity regimens was 6.6 months. This study further stressed the importance of ASCT as an adjunct to induction chemotherapy. The MPN Subcommittee of the Chronic Malignancies Working Party of the European Society for Blood and Marrow Transplantation studied 46 patients with MPN-BP who received ASCT.75 At a median follow-up of 37.4 months, progression-free survival and overall survival at 3 years were 26% and 33%, respectively. The major determinant of survival after ASCT was achievement of CR before transplant (69% vs 22%; P¼.008). A study in France analyzed the outcomes of ASCT in patients with MPN-BP or myelodysplastic syndrome (MDS)/MPN-BP76; 60 patients were studied and overall survival and leukemia-free survival at 3 years were 16% and 9%, respectively. As expected, outcome was better in patients receiving transplant in CR, while intensity of the conditioning regimen did not affect survival.76 A Canadian MPN group conducted a study to evaluate the survival effect of ASCT in patients with MF previously exposed to JAK inhibitors80; their patient cohort of 100 included 13 who progressed into MPN-BP while receiving JAK inhibitor; patients with MPN-BP received AML-like induction chemotherapy or hypomethylating agents before transplant; 2-year post-ASCT survival was worst in patients with MPN-BP (32%), and the poor outcome was attributed to a higher relapse rate. In regard to ruxolitinib activity in MPN-BP, a phase 2 study was conducted in refractory AML, including 18 patients with MPN-BP and exhibited limited activity.73 INVESTIGATIONAL AGENTS Blast-phase myeloproliferative neoplasms are relatively resistant to conventional chemotherapy with the only curative hope being ASCT, as discussed above, necessitating the need to develop novel therapies. Bromodomain and extra terminal domain inhibitors (BETis) target epigenetic proteins in cancer and were studied in patient-derived MPN blast progenitor cells and exhibited activity inducing apoptosis and inhibiting growth82; furthermore, synergism was observed when BETis were combined with ruxolitinib. In another study, combination of BETi with heat shock protein 90 inhibitor

July 2017;92(7):1118-1128

n

http://dx.doi.org/10.1016/j.mayocp.2017.05.010 www.mayoclinicproceedings.org

LEUKEMIC TRANSFORMATION IN MYELOPROLIFERATIVE NEOPLASMS

exerted effects on ruxolitinib-persisting or ruxolitinib-resistant MPN blast cells. This was based on a previous finding reported by this team that heat shock protein 90 inhibitor reduces mutant JAK2, AKT, and c-RAF as well as induces apoptosis in JAKi-resistant MPN blast cells.83 CPX-351 is a liposome formulation of cytarabine and daunorubicin and recently received breakthrough therapy designation in May 2016 for therapy-related AML or AML with myelodysplasia-related changes. Ex vivo sensitivity using CPX-351 in leukemia cells from different sources exhibited activity against myeloid blasts derived from patients with MPN/MDS overlap.84 Moreover, the improved survival (median, w10 months with CPX-351 vs w6 months with the standard “7þ3” AML therapy; P¼.005) reported in the relevant phase III trial of CPX-351 involved older patients and high-risk AML, including therapy related and with antecedent MDS or chronic myelomonocytic leukemia.85 Accordingly, it makes sense to investigate the therapeutic value of CPX-351 in MPN-BP, which happens to be a form of secondary AML and usually affects older patients. CONCLUSION The prognosis of MPN- BP remains dismal and appears to be worse than that of primary AML. Drug therapy alone, although capable of inducing CRs in a minority of patients, is ineffective in securing long-term remissions. Furthermore, despite increasing information on mutational repertoire in MPN-BP, the promise for targeted therapy in the near future is currently slim. Durable remissions have been noted in few patients who managed to receive ASCT, but the overall experience with the particular treatment modality has been less than impressive. It is possible that one can reduce the case numbers with MPN-BP using MPN treatment that modifies the natural history of disease, but the particular scenario has yet to be realized. Current prognostic models for MPN are useful in predicting leukemic transformation, and incorporation of molecular information might lead to further refinement of these models. For now, the only hope for cure or long-term remission in MPN-BP remains ASCT, and every effort should be made to identify Mayo Clin Proc. n July 2017;92(7):1118-1128 www.mayoclinicproceedings.org

n

patients with MPN who are at high risk for leukemic transformation so that they can be offered treatment with ASCT sooner rather than later. Abbreviations and Acronyms: AML = acute myeloid leukemia; ASCT = allogeneic stem cell transplant; BETi = bromodomain and extra terminal domain inhibitor; CR = complete remission; DIPSS = Dynamic International Prognostic Scoring System; ET = essential thrombocythemia; FAB = French-American-British; MDS = myelodysplastic syndrome; MF = myelofibrosis; MPN = myeloproliferative neoplasm; MPN-BP = blast-phase myeloproliferative neoplasm; PMF = primary myelofibrosis; PV = polycythemia vera; RBC = red blood cell; WHO = World Health Organization Correspondence: Address to Ayalew Tefferi, MD, Division of Hematology, Department of Medicine, Mayo Clinic, 200 First St SW, Rochester, MN 55905 (Tefferi.ayalew@mayo. edu). Individual reprints of this article and a bound reprint of the entire Symposium on Neoplastic Hematology and Medical Oncology will be available for purchase from our website www.mayoclinicproceedings.org. The Symposium on Neoplastic Hematology and Medical Oncology will continue in an upcoming issue.

REFERENCES 1. Tefferi A. Myelofibrosis with myeloid metaplasia. N Engl J Med. 2000;342(17):1255-1265. 2. Hasselbalch HC. The role of cytokines in the initiation and progression of myelofibrosis. Cytokine Growth Factor Rev. 2013; 24(2):133-145. 3. Verfaillie CM. Biology of chronic myelogenous leukemia. Hematol Oncol Clin North Am. 1998;12(1):1-29. 4. Faderl S, Talpaz M, Estrov Z, O’Brien S, Kurzrock R, Kantarjian HM. The biology of chronic myeloid leukemia. N Engl J Med. 1999;341(3):164-172. 5. Arber DA, Orazi A, Hasserjian R, et al. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood. 2016;127(20): 2391-2405. 6. Barbui T, Thiele J, Passamonti F, et al. Survival and disease progression in essential thrombocythemia are significantly influenced by accurate morphologic diagnosis: an international study. J Clin Oncol. 2011;29(23):3179-3184. 7. Barbui T, Thiele J, Gisslinger H, et al. Masked polycythemia vera (mPV): results of an international study. Am J Hematol. 2014; 89(1):52-54. 8. Elliott MA, Tefferi A. Thrombosis and haemorrhage in polycythaemia vera and essential thrombocythaemia. Br J Haematol. 2005;128(3):275-290. 9. Barosi G, Mesa RA, Thiele J, et al; International Working Group for Myelofibrosis Research and Treatment (IWGMRT). Proposed criteria for the diagnosis of postpolycythemia vera and post-essential thrombocythemia myelofibrosis: a consensus statement from the International Working Group for Myelofibrosis Research and Treatment. Leukemia. 2008;22(2):437-438. 10. Tefferi A, Guglielmelli P, Larson DR, et al. Long-term survival and blast transformation in molecularly annotated essential thrombocythemia, polycythemia vera, and myelofibrosis. Blood. 2014;124(16):2507-2513; quiz 2615.

http://dx.doi.org/10.1016/j.mayocp.2017.05.010

1125

MAYO CLINIC PROCEEDINGS

11. Noor SJ, Tan W, Wilding GE, et al. Myeloid blastic transformation of myeloproliferative neoplasmsda review of 112 cases. Leuk Res. 2011;35(5):608-613. 12. Mesa RA, Verstovsek S, Cervantes F, et al; International Working Group for Myelofibrosis Research and Treatment (IWGMRT). Primary myelofibrosis (PMF), post polycythemia vera myelofibrosis (post-PV MF), post essential thrombocythemia myelofibrosis (post-ET MF), blast phase PMF (PMF-BP): consensus on terminology by the international working group for myelofibrosis research and treatment (IWG-MRT). Leuk Res. 2007;31(6):737-740. 13. Mascarenhas J, Navada S, Malone A, Rodriguez A, Najfeld V, Hoffman R. Therapeutic options for patients with myelofibrosis in blast phase. Leuk Res. 2010;34(9):1246-1249. 14. Abdulkarim K, Girodon F, Johansson P, et al. AML transformation in 56 patients with Ph- MPD in two well defined populations. Eur J Haematol. 2009;82(2):106-111. 15. Cervantes F, Tassies D, Salgado C, Rovira M, Pereira A, Rozman C. Acute transformation in nonleukemic chronic myeloproliferative disorders: actuarial probability and main characteristics in a series of 218 patients. Acta Haematol. 1991;85(3):124-127. 16. Tam CS, Nussenzveig RM, Popat U, et al. The natural history and treatment outcome of blast phase BCRABL myeloproliferative neoplasms. Blood. 2008;112(5): 1628-1637. 17. Tefferi A, Rumi E, Finazzi G, et al. Survival and prognosis among 1545 patients with contemporary polycythemia vera: an international study. Leukemia. 2013;27(9):1874-1881. 18. Vardiman JW, Thiele J, Arber DA, et al. The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: rationale and important changes. Blood. 2009;114(5):937-951. 19. Mesa RA, Li CY, Ketterling RP, Schroeder GS, Knudson RA, Tefferi A. Leukemic transformation in myelofibrosis with myeloid metaplasia: a single-institution experience with 91 cases. Blood. 2005;105(3):973-977. 20. Passamonti F, Rumi E, Arcaini L, et al. Leukemic transformation of polycythemia vera: a single center study of 23 patients. Cancer. 2005;104(5):1032-1036. 21. Green A, Beer P. Somatic mutations of IDH1 and IDH2 in the leukemic transformation of myeloproliferative neoplasms. N Engl J Med. 2010;362(4):369-370. 22. Abdel-Wahab O, Manshouri T, Patel J, et al. Genetic analysis of transforming events that convert chronic myeloproliferative neoplasms to leukemias. Cancer Res. 2010;70(2):447-452. 23. Zhang SJ, Rampal R, Manshouri T, et al. Genetic analysis of patients with leukemic transformation of myeloproliferative neoplasms shows recurrent SRSF2 mutations that are associated with adverse outcome. Blood. 2012;119(19):4480-4485. 24. Rampal R, Ahn J, Abdel-Wahab O, et al. Genomic and functional analysis of leukemic transformation of myeloproliferative neoplasms. Proc Natl Acad Sci U S A. 2014;111(50):E5401E5410. 25. Campbell PJ, Baxter EJ, Beer PA, et al. Mutation of JAK2 in the myeloproliferative disorders: timing, clonality studies, cytogenetic associations, and role in leukemic transformation. Blood. 2006;108(10):3548-3555. 26. Theocharides A, Boissinot M, Girodon F, et al. Leukemic blasts in transformed JAK2-V617F-positive myeloproliferative disorders are frequently negative for the JAK2-V617F mutation. Blood. 2007;110(1):375-379. 27. Passamonti F, Rumi E, Arcaini L, et al. Prognostic factors for thrombosis, myelofibrosis, and leukemia in essential thrombocythemia: a study of 605 patients. Haematologica. 2008; 93(11):1645-1651. 28. Gangat N, Wolanskyj AP, McClure RF, et al. Risk stratification for survival and leukemic transformation in essential thrombocythemia: a single institutional study of 605 patients. Leukemia. 2007;21(2):270-276.

1126

Mayo Clin Proc.

n

29. Marchioli R, Finazzi G, Landolfi R, et al. Vascular and neoplastic risk in a large cohort of patients with polycythemia vera. J Clin Oncol. 2005;23(10):2224-2232. 30. Barbui T, Carobbio A, Finazzi G, et al. Elevated C-reactive protein is associated with shortened leukemia-free survival in patients with myelofibrosis. Leukemia. 2013;27(10):2084-2086. 31. Passamonti F, Rumi E, Elena C, et al. Incidence of leukaemia in patients with primary myelofibrosis and RBC-transfusiondependence. Br J Haematol. 2010;150(6):719-721. 32. Tefferi A, Gangat N, Wolanskyj AP, et al. 20þ yr without leukemic or fibrotic transformation in essential thrombocythemia or polycythemia vera: predictors at diagnosis. Eur J Haematol. 2008;80(5):386-390. 33. Chim CS, Kwong YL, Lie AK, et al. Long-term outcome of 231 patients with essential thrombocythemia: prognostic factors for thrombosis, bleeding, myelofibrosis, and leukemia. Arch Intern Med. 2005;165(22):2651-2658. 34. Abdulkarim K, Ridell B, Johansson P, Kutti J, Safai-Kutti S, Andréasson B. The impact of peripheral blood values and bone marrow findings on prognosis for patients with essential thrombocythemia and polycythemia vera. Eur J Haematol. 2011;86(2):148-155. 35. Dupriez B, Morel P, Demory JL, et al. Prognostic factors in agnogenic myeloid metaplasia: a report on 195 cases with a new scoring system. Blood. 1996;88(3):1013-1018. 36. Tam CS, Kantarjian H, Cortes J, et al. Dynamic model for predicting death within 12 months in patients with primary or post-polycythemia vera/essential thrombocythemia myelofibrosis. J Clin Oncol. 2009;27(33):5587-5593. 37. Huang J, Li CY, Mesa RA, et al. Risk factors for leukemic transformation in patients with primary myelofibrosis. Cancer. 2008; 112(12):2726-2732. 38. Tefferi A, Vaidya R, Caramazza D, Finke C, Lasho T, Pardanani A. Circulating interleukin (IL)-8, IL-2R, IL-12, and IL-15 levels are independently prognostic in primary myelofibrosis: a comprehensive cytokine profiling study. J Clin Oncol. 2011;29(10):1356-1363. 39. Finazzi G, Caruso V, Marchioli R, et al; ECLAP Investigators. Acute leukemia in polycythemia vera: an analysis of 1638 patients enrolled in a prospective observational study. Blood. 2005;105(7):2664-2670. 40. Vaidya R, Caramazza D, Begna KH, et al. Monosomal karyotype in primary myelofibrosis is detrimental to both overall and leukemia-free survival. Blood. 2011;117(21):5612-5615. 41. Gangat N, Caramazza D, Vaidya R, et al. DIPSS plus: a refined Dynamic International Prognostic Scoring System for primary myelofibrosis that incorporates prognostic information from karyotype, platelet count, and transfusion status. J Clin Oncol. 2011;29(4):392-397. 42. Caramazza D, Begna KH, Gangat N, et al. Refined cytogeneticrisk categorization for overall and leukemia-free survival in primary myelofibrosis: a single center study of 433 patients. Leukemia. 2011;25(1):82-88. 43. Quintás-Cardama A, Kantarjian H, Pierce S, Cortes J, Verstovsek S. Prognostic model to identify patients with myelofibrosis at the highest risk of transformation to acute myeloid leukemia. Clin Lymphoma Myeloma Leuk. 2013;13(3):315-318. e312. 44. Vannucchi AM, Lasho TL, Guglielmelli P, et al. Mutations and prognosis in primary myelofibrosis. Leukemia. 2013;27(9): 1861-1869. 45. Cervantes F, Dupriez B, Pereira A, et al. New prognostic scoring system for primary myelofibrosis based on a study of the International Working Group for Myelofibrosis Research and Treatment. Blood. 2009;113(13):2895-2901. 46. Passamonti F, Cervantes F, Vannucchi AM, et al. A dynamic prognostic model to predict survival in primary myelofibrosis: a study by the IWG-MRT (International Working Group for Myeloproliferative Neoplasms Research and Treatment). Blood. 2010;115(9):1703-1708.

July 2017;92(7):1118-1128

n

http://dx.doi.org/10.1016/j.mayocp.2017.05.010 www.mayoclinicproceedings.org

LEUKEMIC TRANSFORMATION IN MYELOPROLIFERATIVE NEOPLASMS

47. Tefferi A, Vainchenker W. Myeloproliferative neoplasms: molecular pathophysiology, essential clinical understanding, and treatment strategies. J Clin Oncol. 2011;29(5):573-582. 48. Klampfl T, Gisslinger H, Harutyunyan AS, et al. Somatic mutations of calreticulin in myeloproliferative neoplasms. N Engl J Med. 2013;369(25):2379-2390. 49. Nangalia J, Massie CE, Baxter EJ, et al. Somatic CALR mutations in myeloproliferative neoplasms with nonmutated JAK2. N Engl J Med. 2013;369(25):2391-2405. 50. Rotunno G, Mannarelli C, Guglielmelli P, et al; Associazione Italiana per la Ricerca sul Cancro Gruppo Italiano Malattie Mieloproliferative Investigators. Impact of calreticulin mutations on clinical and hematological phenotype and outcome in essential thrombocythemia. Blood. 2014;123(10):1552-1555. 51. Rumi E, Pietra D, Ferretti V, et al; Associazione Italiana per la Ricerca sul Cancro Gruppo Italiano Malattie Mieloproliferative Investigators. JAK2 or CALR mutation status defines subtypes of essential thrombocythemia with substantially different clinical course and outcomes. Blood. 2014;123(10):1544-1551. 52. Tefferi A, Lasho TL, Finke CM, et al. CALR vs JAK2 vs MPLmutated or triple-negative myelofibrosis: clinical, cytogenetic and molecular comparisons. Leukemia. 2014;28(7):1472-1477. 53. Tefferi A, Wassie EA, Lasho TL, et al. Calreticulin mutations and long-term survival in essential thrombocythemia. Leukemia. 2014;28(12):2300-2303. 54. Fu R, Xuan M, Zhou Y, et al. Analysis of calreticulin mutations in Chinese patients with essential thrombocythemia: clinical implications in diagnosis, prognosis and treatment. Leukemia. 2014; 28(9):1912-1914. 55. Vannucchi AM, Guglielmelli P. Molecular pathophysiology of Philadelphia-negative myeloproliferative disorders: beyond JAK2 and MPL mutations. Haematologica. 2008;93(7):972-976. 56. Tefferi A, Guglielmelli P, Lasho TL, et al. CALR and ASXL1 mutations-based molecular prognostication in primary myelofibrosis: an international study of 570 patients. Leukemia. 2014; 28(7):1494-1500. 57. Tefferi A, Lasho T, Finke C, et al. Targeted deep sequencing in primary myelofibrosis. Blood Adv. 2016;1(2):105-111. 58. Tefferi A, Lasho TL, Abdel-Wahab O, et al. IDH1 and IDH2 mutation studies in 1473 patients with chronic-, fibrotic- or blast-phase essential thrombocythemia, polycythemia vera or myelofibrosis. Leukemia. 2010;24(7):1302-1309. 59. Pardanani A, Lasho TL, Finke CM, Mai M, McClure RF, Tefferi A. IDH1 and IDH2 mutation analysis in chronic- and blast-phase myeloproliferative neoplasms. Leukemia. 2010; 24(6):1146-1151. 60. Tefferi A, Guglielmelli P, Finke C, et al. Integration of mutations and karyotype towards a Genetics-Based Prognostic Scoring System (GPSS) for primary myelofibrosis. Blood. 2014; 124(21):406. 61. Berk PD, Wasserman LR, Fruchtman SM, Goldberg JD. Treatment of polycythemia vera: a summary of clinical trials conducted by the Polycythemia Vera Study Group. In: Wasserman LR, Berk PD, Berlin NI, eds. Polycythemia Vera and the Myeloproliferative Disorders. Philadelphia: WB Saunders; 1995:166-194. 62. Gangat N, Strand J, Li CY, Wu W, Pardanani A, Tefferi A. Leucocytosis in polycythaemia vera predicts both inferior survival and leukaemic transformation. Br J Haematol. 2007;138(3): 354-358. 63. Tefferi A, Lasho T, Guglielmelli P, et al. Targeted deep sequencing in polycythemia vera and essential thrombocythemia. Blood Adv. 2016;1(1):21-30. 64. Girodon F, Dutrillaux F, Broséus J, et al. Leukocytosis is associated with poor survival but not with increased risk of thrombosis in essential thrombocythemia: a population-based study of 311 patients. Leukemia. 2010;24(4):900-903. 65. Cervantes F, Alvarez-Larrán A, Talarn C, Gómez M, Montserrat E. Myelofibrosis with myeloid metaplasia following essential thrombocythaemia: actuarial probability, presenting

Mayo Clin Proc. n July 2017;92(7):1118-1128 www.mayoclinicproceedings.org

n

66.

67.

68.

69.

70.

71.

72.

73.

74.

75.

76.

77.

78.

79.

80.

81.

characteristics and evolution in a series of 195 patients. Br J Haematol. 2002;118(3):786-790. Passamonti F, Rumi E, Pungolino E, et al. Life expectancy and prognostic factors for survival in patients with polycythemia vera and essential thrombocythemia. Am J Med. 2004; 117(10):755-761. Palandri F, Catani L, Testoni N, et al. Long-term follow-up of 386 consecutive patients with essential thrombocythemia: safety of cytoreductive therapy. Am J Hematol. 2009;84(4): 215-220. Beer PA, Campbell PJ, Scott LM, et al. MPL mutations in myeloproliferative disorders: analysis of the PT-1 cohort. Blood. 2008; 112(1):141-149. Vannucchi AM, Antonioli E, Guglielmelli P, et al. Characteristics and clinical correlates of MPL 515W>L/K mutation in essential thrombocythemia. Blood. 2008;112(3):844-847. Thepot S, Itzykson R, Seegers V, et al; Groupe Francophone des Myelodysplasies (GFM). Treatment of progression of Philadelphia-negative myeloproliferative neoplasms to myelodysplastic syndrome or acute myeloid leukemia by azacitidine: a report on 54 cases on the behalf of the Groupe Francophone des Myelodysplasies (GFM). Blood. 2010; 116(19):3735-3742. Ciurea SO, de Lima M, Giralt S, et al. Allogeneic stem cell transplantation for myelofibrosis with leukemic transformation. Biol Blood Marrow Transplant. 2010;16(4):555-559. Cherington C, Slack JL, Leis J, et al. Allogeneic stem cell transplantation for myeloproliferative neoplasm in blast phase. Leuk Res. 2012;36(9):1147-1151. Eghtedar A, Verstovsek S, Estrov Z, et al. Phase 2 study of the JAK kinase inhibitor ruxolitinib in patients with refractory leukemias, including postmyeloproliferative neoplasm acute myeloid leukemia. Blood. 2012;119(20):4614-4618. Kennedy JA, Atenafu EG, Messner HA, et al. Treatment outcomes following leukemic transformation in Philadelphianegative myeloproliferative neoplasms. Blood. 2013;121(14): 2725-2733. Alchalby H, Zabelina T, Stübig T, et al; Chronic Malignancies Working Party of the European Group for Blood and Marrow Transplantation. Allogeneic stem cell transplantation for myelofibrosis with leukemic transformation: a study from the Myeloproliferative Neoplasm Subcommittee of the CMWP of the European Group for Blood and Marrow Transplantation. Biol Blood Marrow Transplant. 2014; 20(2):279-281. Cahu X, Chevallier P, Clavert A, et al. Allo-SCT for Philadelphia-negative myeloproliferative neoplasms in blast phase: a study from the Societe Française de Greffe de Moelle et de Therapie Cellulaire (SFGM-TC). Bone Marrow Transplant. 2014;49(6):756-760. Badar T, Kantarjian HM, Ravandi F, et al. Therapeutic benefit of decitabine, a hypomethylating agent, in patients with high-risk primary myelofibrosis and myeloproliferative neoplasm in accelerated or blastic/acute myeloid leukemia phase. Leuk Res. 2015;39(9):950-956. Pemmaraju N, Kantarjian H, Kadia T, et al. A phase I/II study of the Janus kinase (JAK)1 and 2 inhibitor ruxolitinib in patients with relapsed or refractory acute myeloid leukemia. Clin Lymphoma Myeloma Leuk. 2015;15(3):171-176. Andriani A, Montanaro M, Voso MT, et al. Azacytidine for the treatment of retrospective analysis from the Gruppo Laziale for the study of Ph-negative MPN. Leuk Res. 2015;39(8):801-804. Shanavas M, Popat U, Michaelis LC, et al. Outcomes of allogeneic hematopoietic cell transplantation in patients with myelofibrosis with prior exposure to Janus kinase 1/2 inhibitors. Biol Blood Marrow Transplant. 2016;22(3):432-440. Chihara D, Kantarjian HM, Newberry KJ, et al. Survival outcome of patients with acute myeloid leukemia transformed from myeloproliferative neoplasms. Blood. 2016;128(22):1940.

http://dx.doi.org/10.1016/j.mayocp.2017.05.010

1127

MAYO CLINIC PROCEEDINGS

82. Saenz DT, Fiskus W, Manshouri T, et al. BET protein bromodomain inhibitor-based combinations are highly active against post-myeloproliferative neoplasm secondary AML cells. Leukemia. 2017;31(3):678-687. 83. Fiskus W, Verstovsek S, Manshouri T, et al. Heat shock protein 90 inhibitor is synergistic with JAK2 inhibitor and overcomes resistance to JAK2-TKI in human myeloproliferative neoplasm cells. Clin Cancer Res. 2011;17(23):7347-7358.

1128

Mayo Clin Proc.

n

84. Gordon MJ, Tardi P, Loriaux MM, et al. CPX-351 exhibits potent and direct ex vivo cytotoxicity against AML blasts with enhanced efficacy for cells harboring the FLT3-ITD mutation. Leuk Res. 2017;53:39-49. 85. Lancet JE, Uy GL, Cortes JE, et al. Final results of a phase III randomized trial of CPX-351 versus 7þ3 in older patients with newly diagnosed high risk (secondary) AML. J Clin Oncol. 2016;34.

July 2017;92(7):1118-1128

n

http://dx.doi.org/10.1016/j.mayocp.2017.05.010 www.mayoclinicproceedings.org