The BCR-ABL-negative myeloproliferative neoplasms (MPNs) have a variable risk of progressing to accelerated- or blast-phase MPN (MPN-AP/MPN-BP), defined by the presence of 10% to 19% and more than or equal to 20% myeloid blasts in the peripheral blood or bone marrow, respectively. The molecular processes underlying the progression to MPN-AP/MPN-BP are becoming increasingly understood with the acquisition of additional mutations in epigenetic modifiers (eg, ASXL1, EZH2, TET2), TP53, the Ras pathway, or splicing factors (eg, SRSF2, U2AF1), having been described as important steps in this evolutionary process. At least partially driven by the enrichment of these high-risk molecular features, the prognosis of patients with MPN-BP remains inferior to other patients with acute myeloid leukemia, with a median overall survival of 3 to 6 months. Allogeneic hematopoietic cell transplantation remains the only potentially curative therapeutic modality, but only a minority of patients are eligible. In the absence of curative intent, therapeutic strategies or palliative treatment with hypomethylating agents as monotherapy or in combination with ruxolitinib or venetoclax can be considered. Several novel agents are in various stages of clinical development but are not available for routine use at this point, highlighting the need for ongoing research and the prioritization of clinical trial enrollment when feasible.

Learning Objectives

  • Understand the definition, clinical characteristics, and prognosis of patients with MPN-AP or MPN-BP

  • Review the molecular evolution underlying the progression from chronic-phase MPN to MPN-AP or MPN-BP

  • Discuss current treatment options for MPN-AP and MPN-BP, including allogeneic hematopoietic cell transplantation, intensive chemotherapy, hypomethylating agents, emerging combinations, and molecularly targeted therapies

A 65-year-old male with a past medical history of atherosclerotic cardiovascular disease with myocardial infarction and coronary artery bypass grafting was referred to hematology for further workup of thrombocytosis, with a platelet count of 933 × 109/L found on routine blood work. The remainder of his laboratory test results included a normal white blood cell count of 8.3 × 109/L with normal differential; a hemoglobin level of 13.5  g/dL; normal electrolytes, kidney, and liver function; and an elevated lactate dehydrogenase level (352  U/L; normal range, 120-246  U/L). There was no evidence of leukoerythroblastic changes on peripheral blood smear review. A computed tomography scan of the abdomen and pelvis demonstrated a normal spleen size. Molecular testing from the peripheral blood was positive for a JAK2 V617F mutation, and the patient was diagnosed with essential thrombocythemia (ET). A bone marrow biopsy and aspirate was performed that was consistent with ET. Given his age and cardiovascular comorbidity, he received cytoreductive therapy with hydroxyurea as well as aspirin. Five years after the initial diagnosis, the patient developed worsening anemia requiring red blood cell transfusions. Bone marrow biopsy showed a markedly hypercellular marrow with profound myeloid hyperplasia, hypoplastic erythropoiesis, markedly hypoplastic megakaryopoiesis, marked fibrosis and osteosclerosis, and no increase in blasts, consistent with progression to post-ET myelofibrosis. Over the subsequent 5 months, the patient developed progressive leukocytosis, worsening red blood cell transfusion requirements, and thrombocytopenia. A bone marrow biopsy was repeated and was consistent with acute myeloid leukemia (AML) with 83% blasts.

The BCR-ABL1-negative myeloproliferative neoplasms (MPNs) compose a heterogeneous spectrum of diseases with a variable risk of progression to accelerated-phase MPN (MPN-AP) and blast-phase MPN (MPN-BP), which is defined as an increase in myeloid blasts in the peripheral blood or bone marrow to 10% to 19% and greater than or equal to 20%, respectively.1-3  While patients with chronic-phase MPN present with constitutional symptoms, thromboembolic and hemorrhagic complications, splenomegaly and variable degrees of abnormal peripheral blood counts, patients with MPN-AP or MPN-BP typically present with progressive anemia, thrombocytopenia, and leukocytosis and have a poor prognosis with a median overall survival (OS) of only 3 to 6 months in retrospective cohort studies.4-6  Although a better understanding of the molecular landscape of MPN has led to the development and approval of several agents for chronic-phase MPN, such as the Jak inhibitors ruxolitinib, fedratinib, and pacritinib for myelofibrosis as well as ropeg-interferon (IFN) for polycythemia vera (PV), no current standard of care for MPN-AP and MPN-BP exists.7-10  However, thanks to advances in molecular testing, the pathophysiology underlying the progression of chronic-phase MPN into MPN-AP and MPN-BP is becoming increasingly characterized, and several clinical trials of novel combination therapies have recently been conducted. Herein, we review the current risk assessment for patients with chronic- phase MPNs with regard to predicting disease progression, recent developments in understanding the clonal evolution driving MPN progression, and emerging treatment options.

While the majority of patients with chronic-phase MPNs do not experience disease progression, this cumulative lifetime risk is variable for patients with ET, PV, and primary myelofibrosis (PMF) and ranged from 3.8% for ET patients to 14.2% for PMF patients in a large cohort study of patients treated at the Mayo Clinic.11  As the only potentially curative therapeutic modality for MPNs remains allogeneic hematopoietic cell transplant (allo-HCT), risk-stratification tools allowing for the appropriate triaging and referral of patients for allo-HCT are of great clinical relevance.12,13  Various prognostic scoring systems for both PMF (eg, the dynamic prognostic scoring system) as well as post-ET and post-PV MF (myelofibrosis secondary to PV and ET Collaboration Prognostic Model) have been developed to predict OS but also correlate, albeit imperfectly, with the risk of progression to MPN-BP.14-16  More recently, the addition of selected high-risk mutations (ASXL1, EZH2, SRSF2, IDH1/2) and the absence of a type 1 CALR mutation to other prognostic disease and clinical characteristics has led to the development of the Molecularly Inspired Prognostic Scoring System (MIPSS).15  Conversely, the genetically inspired scoring system is based only on molecular features such as karyotype and selected high-risk mutations.17  Both scores have been shown to predict prognosis in PMF patients.15,17 

Table 1 provides an overview of prognostic factors associated with an increased risk of progression to MPN-BP. These can be classified into disease related (eg, increased peripheral blood blasts, higher-grade bone marrow fibrosis, depth of cytopenias), molecular (eg, the presence of certain high-risk somatic mutations), and treatment related.2,16,18  While the leukemogenic potential of hydroxyurea remains under evaluation, other agents used for cytoreduction in the past, such as busulfan and pipobroman, have been associated with an increased risk of progression to MPN-BP. Conversely, treatment with IFN may be associated with a reduced risk of progression to MPN-BP.16,19,20 

Table 1.

Patient, disease, and treatment characteristics associated with risk of progression to MPN-AP/MPN-BP

Patient factor 
 Advanced patient age (>60 vs >70 years)57,58  
Disease factors 
 Longer disease duration31  
 Higher risk for patients with PMF compared to ET and PV11  
 Platelet count <150 × 109/L, leukocytosis, transfusion dependence, peripheral blasts ≥3%16,58,59  
 Splenomegaly of ≥20 cm from left costal margin or requirement for splenectomy59  
 Bone marrow reticulin fibrosis57  
 Abnormal karyotype58,59  
 Lower risk with CALR mutations60  
 High-risk mutations (eg, TP53, ASXL1, SRSF2, IDH2, EZH2)4,60  
 Higher DIPSS, MYSEC-PM, or MIPSS70v2.0 score16  
Treatment factors 
 Prior treatment with alkylating agents and pipobroman20,58  
 Lower risk in patients treated with IFN16  
Patient factor 
 Advanced patient age (>60 vs >70 years)57,58  
Disease factors 
 Longer disease duration31  
 Higher risk for patients with PMF compared to ET and PV11  
 Platelet count <150 × 109/L, leukocytosis, transfusion dependence, peripheral blasts ≥3%16,58,59  
 Splenomegaly of ≥20 cm from left costal margin or requirement for splenectomy59  
 Bone marrow reticulin fibrosis57  
 Abnormal karyotype58,59  
 Lower risk with CALR mutations60  
 High-risk mutations (eg, TP53, ASXL1, SRSF2, IDH2, EZH2)4,60  
 Higher DIPSS, MYSEC-PM, or MIPSS70v2.0 score16  
Treatment factors 
 Prior treatment with alkylating agents and pipobroman20,58  
 Lower risk in patients treated with IFN16  

DIPSS, Dynamic International Prognostic Scoring System; MYSEC-PM, myelofibrosis secondary to PV and ET Collaboration Prognostic Model.

Significant advances have been made in elucidating the genetic landscape of MPN-BP. In addition to the mutations in JAK2, CALR, and MPL that are present in approximately 90% of patients with chronic-phase MPN, patients with MPN-BP have been shown to harbor additional somatic mutations, with ASXL1, TET2, TP53, EZH2, RUNX1, and splicing factor genes (eg, SRSF2, U2AF1) being the most frequently encountered genetic alterations.18,21-23  This mutational profile of MPN-BP is distinct from patients with de novo AML, and the enrichment of high-risk mutations might be contributing to the adverse prognosis of patients with MPN-BP.24  For example, mutations in FLT3 occur in up to 30% of AML cases but are rare in MPN-BP, which is characterized by a higher frequency of mutations in ASXL1, TET2, TP53, IDH1/2, and splicing factor genes (eg, SRSF2, U2AF1).18,21-23,25  Serial assessment of the mutational profile of patients progressing from chronic to MPN-AP and MPN-BP have demonstrated that the serial acquisition of additional mutations—for example, in IDH1/2 or TET2—as well as the expansion of a previously present subclone harboring a TP53 mutation can potentially drive disease progression.22,26-30  However, mutations in other genes such as ASXL1 or EZH2 can be acquired either before or after the canonical driver mutations in JAK2, MPL, or CALR.26  Additionally, recent studies have highlighted the contribution of a pro-inflammatory microenvironment in the bone marrow as a potential driver of clonal evolution and disease progression.30-32  Despite these advances, additional research is needed to understand the clonal evolution among patients undergoing various forms of treatment and whether serial sequencing can be used to predict transformation and inform treatment recommendations (eg, referral for allo-HCT).

A proposed treatment approach to MPN-BP is outlined in Figure 1. As the prognosis of patients with MPN-BP is limited and has not substantially improved over the last decade, clinical trial enrollment is encouraged wherever feasible.4  The only potentially curative therapeutic modality remains allo-HCT, which should be strongly considered for eligible patients with MPN-BP. Outcomes of allo-HCT in patients with MPN-BP are variable but appear to be inferior compared to patients with other subtypes of AML in large registry studies such as a recent analysis from the Center for International Blood and Marrow Transplant Research, which reported a hazard ratio (HR) of death of 1.4 (95% CI, 1.12-1.76) even among patients in remission at the time of transplant.33  This appears to be primarily driven by a higher rate of relapse and not by differences in nonrelapse mortality for MPN-BP compared to de novo or post-MDS AML.34  Interestingly, there was no statistically significant difference among patients with active disease at the time of allo-HCT and those who received myeloablative or reduced-intensity conditioning.33  Other studies have reported a 3-year OS of 18% to 33% for MPN-BP patients undergoing allo-HCT.35,36  Emerging data suggest that patients with TP53 mutations have inferior outcomes after allo-HCT, with TP53-mutant patients having a higher risk of death (HR, 1.99; 95% CI, 1.14-3.49) and relapse (HR, 2.59; 95% CI, 1.41-4.74).34  Given that disease relapse appears to be a main driver of mortality after allo-HCT among MPN-BP patients, efforts to reduce the risk of relapse are of great interest. Other areas of ongoing research include the influence of conditioning regimen intensity, graft source, and the influence of molecular characteristics on posttransplant outcomes.4,33,34 

Figure 1.

Potential treatment algorithm for patients with blast-phase MPN.

In the absence of high-quality prospective studies specific to patients with blast-phase MPN, treatment recommendations are extrapolated from studies of patients with AML. At the time of diagnosis, molecular testing should be obtained. In patients who are potential candidates for allo-HCT, this constitutes the only potentially curative therapeutic modality and should therefore be considered. Induction chemotherapy prior to transplant is informed by molecular disease features, with the addition of midostaurin in patients with FLT3 mutations. Due to lower response rates with conventional cytotoxic chemotherapy in patients with TP53 mutations, combination therapy with HMAs plus venetoclax could be considered. Patients in remission after induction chemotherapy should proceed to allo-HCT if eligible. which may also be an option for a selected subset of patients who do not achieve a complete remission with induction therapy. For transplant-ineligible patients and those with R/R disease, clinical trial enrollment, HMAs alone or in combination, IDH1/2 inhibitors, or best supportive care are therapeutic options.

Figure 1.

Potential treatment algorithm for patients with blast-phase MPN.

In the absence of high-quality prospective studies specific to patients with blast-phase MPN, treatment recommendations are extrapolated from studies of patients with AML. At the time of diagnosis, molecular testing should be obtained. In patients who are potential candidates for allo-HCT, this constitutes the only potentially curative therapeutic modality and should therefore be considered. Induction chemotherapy prior to transplant is informed by molecular disease features, with the addition of midostaurin in patients with FLT3 mutations. Due to lower response rates with conventional cytotoxic chemotherapy in patients with TP53 mutations, combination therapy with HMAs plus venetoclax could be considered. Patients in remission after induction chemotherapy should proceed to allo-HCT if eligible. which may also be an option for a selected subset of patients who do not achieve a complete remission with induction therapy. For transplant-ineligible patients and those with R/R disease, clinical trial enrollment, HMAs alone or in combination, IDH1/2 inhibitors, or best supportive care are therapeutic options.

Close modal

Since outcomes of patients undergoing allo-HCT with active disease are potentially worse compared to patients in remission at the time of transplant,35,36  initial treatment at the time of disease progression is an important consideration and area of ongoing research. Selected clinical trials are summarized in Table 2. For patients eligible for intensive chemotherapy, such as an anthracycline-cytarabine–based regimen, response rates of 30% to 60% have been reported, with a median OS of only 3 to 9 months among patients who did not subsequently undergo an allo-HCT despite achieving a response to initial treatment.4,6,37  Patients who were able to proceed to allo-HCT had a significantly longer OS compared to patients who did not receive transplants.4,6,37  Furthermore, the survival of patients who receive intensive chemotherapy but do not receive consolidation with allo-HCT does not appear to differ from patients treated with nonintensive approaches.37  As such, the use of intensive chemotherapy should only be considered in patients planned or eligible to receive allo-HCT.

Table 2.

Selected studies of treatment outcomes in MPN-BP

Therapeutic regimenPatient populationStudy designKey outcomesReference
Intensive chemotherapy 
 Intensive chemotherapy (not specified) MPN-BP Retrospective cohort study Median OS 10.2 mo 61  
 “7 + 3” cytarabine plus an anthracycline MPN-BP Retrospective cohort study CR rate 67%; median OS 19.2 mo for allo-HCT vs 3.8 mo for nontransplant patients 5  
 AML-like induction chemotherapy MPN-BP Retrospective cohort study CR rate 35%; 3-y OS 32% for allo-HCT vs 19% for nontransplant patients 4  
HMA monotherapy 
 Azacitidine MPN-BP/AP Retrospective cohort study ORR 61.5%; median OS 13.5 mo 43  
MPN-BP Retrospective cohort study ORR 68%; median OS 9.9 mo 42  
MPN-BP Retrospective cohort study Median OS 9 mo 61  
 Azacitidine or decitabine MPN-BP Retrospective cohort study Median OS 6.7 mo 5  
 Decitabine MPN-BP, MPN-AP, high-risk PMF Retrospective cohort study MPN-BP: 29% ORR, 10.5 mo median OS
MPN-AP: 62% ORR, 11.8 mo median OS 
41  
Ruxolitinib monotherapy 
 Ruxolitinib MPN-BP Phase 2 clinical trial ORR 16.6% 48  
HMA + ruxolitinib 
 Ruxolitinib + decitabine MPN-AP, MPN-BP Phase 2 clinical trial ORR 44%; median OS 9.5 mo 49  
MPN-BP Phase 1/2 clinical trial ORR 45%; median OS 8.4 mo 50  
HMA + venetoclax 
 HMA + venetoclax MPN-BP Retrospective cohort study Newly diagnosed: ORR 50%; median OS 7 mo
R/R: no objective responses; median OS 3 mo 
45  
MPN-BP Retrospective cohort study ORR 44%; median OS 8 mo 44  
 MPN-AP, MPN-BP Retrospective multicenter cohort study ORR: 52%; median OS 6 mo 47  
IDH1/2 inhibitors 
 Enasidenib IDH2-mutant MPN-AP, MPN-BP Retrospective cohort study ORR: 37.5% per ELN 2017 and 75% per MPN-BP response criteria 54  
 Enasidenib or ivosidenib IDH1 or IDH2-mutant MPN-AP, MPN-BP Retrospective cohort study 43% CR rate among newly diagnosed patients; no objective responses in R/R cohort 53  
Therapeutic regimenPatient populationStudy designKey outcomesReference
Intensive chemotherapy 
 Intensive chemotherapy (not specified) MPN-BP Retrospective cohort study Median OS 10.2 mo 61  
 “7 + 3” cytarabine plus an anthracycline MPN-BP Retrospective cohort study CR rate 67%; median OS 19.2 mo for allo-HCT vs 3.8 mo for nontransplant patients 5  
 AML-like induction chemotherapy MPN-BP Retrospective cohort study CR rate 35%; 3-y OS 32% for allo-HCT vs 19% for nontransplant patients 4  
HMA monotherapy 
 Azacitidine MPN-BP/AP Retrospective cohort study ORR 61.5%; median OS 13.5 mo 43  
MPN-BP Retrospective cohort study ORR 68%; median OS 9.9 mo 42  
MPN-BP Retrospective cohort study Median OS 9 mo 61  
 Azacitidine or decitabine MPN-BP Retrospective cohort study Median OS 6.7 mo 5  
 Decitabine MPN-BP, MPN-AP, high-risk PMF Retrospective cohort study MPN-BP: 29% ORR, 10.5 mo median OS
MPN-AP: 62% ORR, 11.8 mo median OS 
41  
Ruxolitinib monotherapy 
 Ruxolitinib MPN-BP Phase 2 clinical trial ORR 16.6% 48  
HMA + ruxolitinib 
 Ruxolitinib + decitabine MPN-AP, MPN-BP Phase 2 clinical trial ORR 44%; median OS 9.5 mo 49  
MPN-BP Phase 1/2 clinical trial ORR 45%; median OS 8.4 mo 50  
HMA + venetoclax 
 HMA + venetoclax MPN-BP Retrospective cohort study Newly diagnosed: ORR 50%; median OS 7 mo
R/R: no objective responses; median OS 3 mo 
45  
MPN-BP Retrospective cohort study ORR 44%; median OS 8 mo 44  
 MPN-AP, MPN-BP Retrospective multicenter cohort study ORR: 52%; median OS 6 mo 47  
IDH1/2 inhibitors 
 Enasidenib IDH2-mutant MPN-AP, MPN-BP Retrospective cohort study ORR: 37.5% per ELN 2017 and 75% per MPN-BP response criteria 54  
 Enasidenib or ivosidenib IDH1 or IDH2-mutant MPN-AP, MPN-BP Retrospective cohort study 43% CR rate among newly diagnosed patients; no objective responses in R/R cohort 53  

Only a minority of patients with MPN-BP are eligible for intensive chemotherapy and allo-HCT. In patients ineligible for intensive chemotherapy or allo-HCT, lower-intensity therapies that frequently use a hypomethylating agent (HMA) as monotherapy or in combination with other agents continue to be the mainstay of therapy. While the HMAs azacitidine and decitabine have been widely used in the treatment of MDS as well as AML for over a decade, the quality of evidence supporting their use in MPN-BP is limited, with no prospective, randomized data available.38-40  In small retrospective studies, the overall response rate (ORR) ranges from 29% to 61% for HMA monotherapy with median OS of 6 to 13.5 months.41-43  However, cross-study comparisons are limited by differences in patient populations and response assessments.

Inspired by the success of HMAs in combination with venetoclax in the treatment of older, intensive chemotherapy- ineligible patients with AML, this combination has been used off-label in patients with MPN-BP as well. In a retrospective, multicenter study of 32 MPN-BP patients treated with HMAs plus venetoclax, the rate of complete remission (CR) with incomplete count recovery (CRi) was 44% for the combination compared to 4% with an HMA alone in a historic control group. However, the median OS was 8 months for patients treated with an HMA plus venetoclax and was not statistically significantly better than HMA alone.44  In a similar study from MD Anderson Cancer Center, the CR rate with an HMA plus venetoclax was 43% among 14 patients with newly diagnosed post-MPN AML, resulting in a median OS of 7 months.45  Similar to studies in relapsed/refractory (R/R) AML, an HMA plus venetoclax had only limited efficacy in patients with R/R MPN-BP, with no objective responses seen and a median OS of 3 months.45,46  In another multicenter, retrospective analysis of both treatment-naive and R/R MPN-BP patients (n = 27) treated with venetoclax-based therapies, the ORR was 53%.47  However, this high response rate did not lead to a durable survival benefit, with a median OS of 6 months and an inferior survival rate (HR, 3.42; 95% CI, 1–11.8; P = .03) in patients with TP53 mutations.47  While HMA plus venetoclax combinations can be an option for some patients with MPN-BP, especially as a bridge to allo-HCT, additional studies are needed to inform patient selection based on molecular disease characteristics as well as potential combination therapies (eg, NCT03862157).

While the Jak inhibitor ruxolitinib has led to significant improvements in the treatment of patients with chronic-phase myelofibrosis, the role of ruxolitinib in MPN-BP is less clear, with limited efficacy if used as a single agent in MPN-BP.7,48  In a recent phase 2 study of ruxolitinib in combination with decitabine (NCT02076191), 25 patients with MPN-BP/MPN-AP received ruxolitinib (25  mg twice a day during induction followed by 10  mg twice a day during subsequent cycles) in combination with decitabine administered at 20  mg/m2 for 5 consecutive days on a 28-day cycle. The ORR, defined as a composite of CR, CRi, and partial remission, was 44% (11 out of 25 patients), with a median OS of 9.5 months (95% CI, 4.3-12.0 months).49  Treatment was generally well tolerated, with febrile neutropenia (7 patients, 28%), pneumonia (6 patients, 24%), and anemia (4 patients, 16%) being the most common grade 3/4 adverse events.49  These results were comparable to another single-center phase 2 study reporting an ORR of 45% with a median OS of 8.4 months.50 

Advances in the understanding of the underlying pathophysiology and molecular landscape have enabled the identification of novel therapeutic targets that are currently under investigation in clinical trials. Table 3 summarizes ongoing clinical trials in MPN-BP. Among the novel targets, BET inhibitors and MDM2 inhibitors have shown preclinical activity in MPN-BP models, but no clinical trial data are available to date.31,51,52  Only a small subset of MPN-BP patients with targetable mutations in IDH1 or IDH2 may benefit from treatment with the IDH1 inhibitor ivosidenib or the IDH2 inhibitor enasidenib, either as monotherapy or in combination with other agents. In a series of 12 patients treated with IDH1/2 inhibitor–based combinations, 3 out of 7 patients with newly diagnosed MPN-BP achieved a CR, while none of the patients with R/R disease responded.53  Another retrospective study of 8 patients with IDH2-mutant MPN-BP treated with enasidenib reported an ORR of 37.5% per European LeukemiaNet (ELN) 2017 and 75% per MPN-BP response criteria.54  Besides the need for additional prospective studies to further define the safety and efficacy of IDH inhibitors in MPN-BP and to identify potentially synergistic combination therapies, this latter study highlights an important aspect of clinical trial design and interpretation in MPN-BP. Current ELN 2017 response criteria for AML mandate the absence of blasts from the peripheral blood for a CR.55  However, a small percentage of circulating blasts are frequently seen in patients with chronic-phase MPNs, and it might be unrealistic to expect patients with MPN-BP to achieve peripheral blood blast clearance because treatment may not reverse the underlying fibrosis in the bone marrow and the extramedullary hematopoiesis underlying this small peripheral blast population. Converting patients back to a chronic-phase MPN state with hematologic improvement may provide important benefits to patients independent of the more stringent AML response criteria, which has led to the development of dedicated response criteria for MPN-BP. However, uptake in both routine clinical practice and trials has been limited.56 

Table 3.

Overview of selected ongoing clinical trials in MPN-BP

AgentNCTPhasePatient population
Decitabine + ruxolitinib or fedratinib NCT04282187 MPN-AP and MPN-BP as bridge to allo-HCT 
Ruxolitinib + enasidenib NCT04281498 IDH2-mutant MPN-AP, MPN-BP, chronic-phase myelofibrosis 
Fedratinib + ivosidenib or enasidenib NCT04955938 IDH1- or IDH2-mutant MPN with ≥5% blasts 
Azacitidine + venetoclax NCT05074355 MPN-AP and MPN-BP 
KRT-232 (MDM2 inhibitor) NCT04113616 1/2 post-MPN AML 
ZN-d5 (BCL2 inhibitor) + ZN-c3 (WEE1 inhibitor) NA 1/2 AML including post-MPN AML 
AgentNCTPhasePatient population
Decitabine + ruxolitinib or fedratinib NCT04282187 MPN-AP and MPN-BP as bridge to allo-HCT 
Ruxolitinib + enasidenib NCT04281498 IDH2-mutant MPN-AP, MPN-BP, chronic-phase myelofibrosis 
Fedratinib + ivosidenib or enasidenib NCT04955938 IDH1- or IDH2-mutant MPN with ≥5% blasts 
Azacitidine + venetoclax NCT05074355 MPN-AP and MPN-BP 
KRT-232 (MDM2 inhibitor) NCT04113616 1/2 post-MPN AML 
ZN-d5 (BCL2 inhibitor) + ZN-c3 (WEE1 inhibitor) NA 1/2 AML including post-MPN AML 

NA, not available.

Repeat molecular testing at the time of progression to MPN-BP showed IDH2, NPM1, TET2, and SRSF2. Following initial cytoreduction with hydroxyurea, the patient started treatment with azacitidine and venetoclax. Treatment with azacitidine and venetoclax was complicated by neutropenic sepsis and prolonged neutropenia and thrombocytopenia requiring hospital admission and transfusion support. Following 2 cycles of azacitidine and venetoclax, the patient achieved a complete acute leukemia response per the definitions proposed by the Post-Myeloproliferative Neoplasm Acute Myeloid Leukemia Consortium.56  He continues on azacitidine and venetoclax and is undergoing evaluation for an allo-HCT.

Patients with chronic-phase MPNs have a variable risk of progression to MPN-AP or MPN-BP, defined as an increase in blast count of 10% to 19% and greater than or equal to 20%, respectively. Adverse cytogenetic and molecular features are enriched in patients with MPN-BP, contributing to poor outcomes. While allo-HCT remains the only potentially curative therapeutic modality for patients with MPN-BP, only a minority of patients are eligible for this intense treatment modality. HMAs alone or in combination with Jak inhibitors or venetoclax, as well as IDH1/2 inhibitors in IDH1/2-mutant patients, can yield objective responses in a subset of patients. Several novel agents are in various stages of clinical development but are not yet available for routine use at this point, highlighting the need for ongoing research and the prioritization of clinical trial enrollment when feasible.

Jan Philipp Bewersdorf: no competing financial interests to declare.

Raajit K. Rampal: research funding: Constellation, Incyte, Stemline, Zentalis; consultancy: Abbvie, Blueprint, Celgene/Bristol Myers Squibb, Constellation, CTI, Disc Medicines, Galecto, Incyte, Jazz, Novartis, Pharmaessentia, Promedior, Sierra Oncology, Stemline, Sumitomo Pharma, Zentalis.

Jan Philipp Bewersdorf: nothing to disclose.

Raajit K. Rampal: nothing to disclose.

1.
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
.
doi:10.1182/blood-2016-03-643544
.
2.
Odenike
O
.
How I treat the blast phase of Philadelphia chromosome– negative myeloproliferative neoplasms
.
Blood
.
2018
;
132
(
22
):
2339
-
2350
.
doi:10.1182/blood-2018-03-785907
.
3.
Shahin
OA
,
Chifotides
HT
,
Bose
P
,
Masarova
L
,
Verstovsek
S
.
Accelerated phase of myeloproliferative neoplasms
.
Acta Haematol
.
2021
;
144
(
5
):
484
-
499
.
doi:10.1159/000512929
.
4.
Tefferi
A
,
Mudireddy
M
,
Mannelli
F
, et al.
Blast phase myeloproliferative neoplasm: Mayo-AGIMM study of 410 patients from two separate cohorts
.
Leukemia
.
2018
;
32
(
5
):
1200
-
1210
.
doi:10.1038/s41375-018-0019-y
.
5.
Lancman
G
,
Brunner
A
,
Hoffman
R
,
Mascarenhas
J
,
Hobbs
G
.
Outcomes and predictors of survival in blast phase myeloproliferative neoplasms
.
Leuk Res
.
2018
;
70
(
July
):
49
-
55
.
doi:10.1016/j.leukres.2018.05.004
.
6.
Tam
CS
,
Nussenzveig
RM
,
Popat
U
, et al.
The natural history and treatment outcome of blast phase BCR-ABL-myeloproliferative neoplasms
.
Blood
.
2008
;
112
(
5
):
1628
-
1637
.
doi:10.1182/blood-2008-02-138230
.
7.
Verstovsek
S
,
Mesa
RA
,
Gotlib
J
, et al.
A double-blind, placebo-controlled trial of ruxolitinib for myelofibrosis
.
N Engl J Med
.
2012
;
366
(
9
):
799
-
807
.
doi:10.1056/NEJMoa1110557
.
8.
Harrison
CN
,
Schaap
N
,
Vannucchi
AM
, et al.
Janus kinase-2 inhibitor fedratinib in patients with myelofibrosis previously treated with ruxolitinib (JAKARTA-2): a single-arm, open-label, non-randomised, phase 2, multicentre study
.
Lancet Haematol
.
2017
;
4
(
7
):
e317
-
e324
.
doi:10.1016/S2352-3026(17)30088-1
.
9.
Mesa
RA
,
Vannucchi
AM
,
Mead
A
, et al.
Pacritinib versus best available therapy for the treatment of myelofibrosis irrespective of baseline cytopenias (PERSIST-1): an international, randomised, phase 3 trial
.
Lancet Haematol
.
2017
;
4
(
5
):
e225
-
e236
.
doi:10.1016/S2352-3026(17)30027-3
.
10.
Gisslinger
H
,
Klade
C
,
Georgiev
P
, et al.
PROUD-PV Study Group
.
Ropeginterferon alfa-2b versus standard therapy for polycythaemia vera (PROUD-PV and CONTINUATION-PV): a randomised, non-inferiority, phase 3 trial and its extension study
.
Lancet Haematol
.
2020
;
7
(
3
):
e196
-
e208
.
doi:10.1016/S2352-3026(19)30236-4
.
11.
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.
doi:10.1182/blood-2014-05-579136
.
12.
Kröger
N
,
Giorgino
T
,
Scott
BL
, et al.
Impact of allogeneic stem cell transplantation on survival of patients less than 65 years of age with primary myelofibrosis
.
Blood
.
2015
;
125
(
21
):
3347
-
3350
, quiz 3364.
doi:10.1182/blood-2014-10-608315
.
13.
Bewersdorf
JP
,
Sheth
AH
,
Vetsa
S
, et al.
Outcomes of allogeneic hematopoietic cell transplantation in patients with myelofibrosis—a systematic review and meta-analysis
.
Transplant Cell Ther
.
2021
;
27
(
10
):
873.e1
-
873873.e13
.
doi:10.1016/j.jtct.2021.05.016
.
14.
Passamonti
F
,
Cervantes
F
,
Vannucchi
AM
, et al.
Dynamic International Prognostic Scoring System (DIPSS) predicts progression to acute myeloid leukemia in primary myelofibrosis
.
Blood
.
2010
;
116
(
15
):
2857
-
2858
.
doi:10.1182/blood-2010-06-293415
.
15.
Guglielmelli
P
,
Lasho
TL
,
Rotunno
G
, et al.
MIPSS70: Mutation-Enhanced International Prognostic Score System for transplantation-age patients with primary myelofibrosis
.
J Clin Oncol
.
2018
;
36
(
4
):
310
-
318
.
doi:10.1200/JCO.2017.76.4886
.
16.
Palandri
F
,
Breccia
M
,
Tiribelli
M
, et al.
Risk factors for progression to blast phase and outcome in 589 patients with myelofibrosis treated with ruxolitinib: real-world data
.
Hematol Oncol
.
2020
;
38
(
3
):
372
-
380
.
doi:10.1002/hon.2737
.
17.
Tefferi
A
,
Guglielmelli
P
,
Nicolosi
M
, et al.
GIPSS: Genetically Inspired Prognostic Scoring System for primary myelofibrosis
.
Leukemia
.
2018
;
32
(
7
):
1631
-
1642
.
doi:10.1038/s41375-018-0107-z
.
18.
Lasho
TL
,
Mudireddy
M
,
Finke
CM
, et al.
Targeted next-generation sequencing in blast phase myeloproliferative neoplasms
.
Blood Adv
.
2018
;
2
(
4
):
370
-
380
.
doi:10.1182/bloodadvances.2018015875
.
19.
Björkholm
M
,
Derolf
AR
,
Hultcrantz
M
, et al.
Treatment-related risk factors for transformation to acute myeloid leukemia and myelodysplastic syndromes in myeloproliferative neoplasms
.
J Clin Oncol
.
2011
;
29
(
17
):
2410
-
2415
.
doi:10.1200/JCO.2011.34.7542
.
20.
Kiladjian
JJ
,
Rain
JD
,
Bernard
JF
,
Briere
J
,
Chomienne
C
,
Fenaux
P
.
Long-term incidence of hematological evolution in three French prospective studies of hydroxyurea and pipobroman in polycythemia vera and essential thrombocythemia
.
Semin Thromb Hemost
.
2006
;
32
(
4, pt 2
):
417
-
421
.
doi:10.1055/s-2006-942762
.
21.
Tefferi
A
,
Lasho
TL
,
Finke
CM
, et al.
Targeted deep sequencing in primary myelofibrosis
.
Blood Adv
.
2016
;
1
(
2
):
105
-
111
.
doi:10.1182/bloodadvances.2016000208
.
22.
Rampal
R
,
Ahn
J
,
Abdel-Wahab
O
, et al.
Genomic and functional analysis of leukemic transformation of myeloproliferative neoplasms
.
Proc Natl Acad Sci USA
.
2014
;
111
(
50
):
E5401
-
E5410
.
doi:10.1073/pnas.1407792111
.
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
.
doi:10.1182/blood-2011-11-390252
.
24.
Papaemmanuil
E
,
Gerstung
M
,
Bullinger
L
, et al.
genomic classification and prognosis in acute myeloid leukemia
.
N Engl J Med
.
2016
;
374
(
23
):
2209
-
2221
.
doi:10.1056/NEJMoa1516192
.
25.
Daver
N
,
Schlenk
RF
,
Russell
NH
,
Levis
MJ
.
Targeting FLT3 mutations in AML: review of current knowledge and evidence
.
Leukemia
.
2019
;
33
(
2
):
299
-
312
.
doi:10.1038/s41375-018-0357-9
.
26.
Lundberg
P
,
Karow
A
,
Nienhold
R
, et al.
Clonal evolution and clinical correlates of somatic mutations in myeloproliferative neoplasms
.
Blood
.
2014
;
123
(
14
):
2220
-
2228
.
doi:10.1182/blood-2013-11-537167
.
27.
Miles
LA
,
Bowman
RL
,
Merlinsky
TR
, et al.
Single-cell mutation analysis of clonal evolution in myeloid malignancies
.
Nature
.
2020
;
587
(
7834
):
477
-
482
.
doi:10.1038/s41586-020-2864-x
.
28.
Ortmann
CA
,
Kent
DG
,
Nangalia
J
, et al.
Effect of mutation order on myeloproliferative neoplasms
.
N Engl J Med
.
2015
;
372
(
7
):
601
-
612
.
doi:10.1056/NEJMoa1412098
.
29.
Maslah
N
,
Verger
E
,
Giraudier
S
, et al.
Single-cell analysis reveals selection of TP53-mutated clones after MDM2 inhibition
.
Blood Adv
.
2022
;
6
(
9
):
2813
-
2823
.
doi:10.1182/bloodadvances.2021005867
.
30.
Rodriguez-Meira
A
,
Norfo
R
,
Wen
WX
, et al.
Deciphering TP53 mutant cancer evolution with single-cell multi-omics. Preprint
. Posted online
29
March
2022
.
bioRxiv
.
2022
:2022.2003.2028.485984.
doi:10.1101/2022.03.28.485984
.
31.
Saha
C
,
Attwell
L
,
Harrison
CN
,
McLornan
DP
.
Addressing the challenges of accelerated and blast phase myeloproliferative neoplasms in 2022 and beyond
.
Blood Rev
.
2022
;
55
(
September
):
100947
.
doi:10.1016/j.blre.2022.100947
.
32.
Koschmieder
S
,
Chatain
N
.
Role of inflammation in the biology of myeloproliferative neoplasms
.
Blood Rev
.
2020
;
42
(
suppl 1
):
100711
.
doi:10.13039/501100001659
.
33.
Gupta
V
,
Kim
S
,
Hu
ZH
, et al.
Comparison of outcomes of HCT in blast phase of BCR-ABL1− MPN with de novo AML and with AML following MDS
.
Blood Adv
.
2020
;
4
(
19
):
4748
-
4757
.
doi:10.1182/bloodadvances.2020002621
.
34.
Gupta
V
,
Kennedy
JA
,
Capo-Chichi
JM
, et al.
Genetic factors rather than blast reduction determine outcomes of allogeneic HCT in BCR-ABL- negative MPN in blast phase
.
Blood Adv
.
2020
;
4
(
21
):
5562
-
5573
.
doi:10.1182/bloodadvances.2020002727
.
35.
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 Transpl
.
2014
;
49
(
6
):
756
-
760
.
doi:10.1038/bmt.2014.31
.
36.
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
.
doi:10.1016/j.bbmt.2013.10.027
.
37.
Kennedy
JA
,
Atenafu
EG
,
Messner
HA
, et al.
Treatment outcomes following leukemic transformation in Philadelphia-negative myeloproliferative neoplasms
.
Blood
.
2013
;
121
(
14
):
2725
-
2733
.
doi:10.1182/blood-2012-10-464248
.
38.
Fenaux
P
,
Mufti
GJ
,
Hellstrom-Lindberg
E
, et al.
International Vidaza High-Risk Myelodysplastic Syndrome Survival Study Group
.
Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomised, open-label, phase III study
.
Lancet Oncol
.
2009
;
10
(
3
):
223
-
232
.
doi:10.1016/S1470-2045(09)70003-8
.
39.
Fenaux
P
,
Mufti
GJ
,
Hellström-Lindberg
E
, et al.
Azacitidine prolongs overall survival compared with conventional care regimens in elderly patients with low bone marrow blast count acute myeloid leukemia
.
J Clin Oncol
.
2010
;
28
(
4
):
562
-
569
.
doi:10.1200/JCO.2009.23.8329
.
40.
Kantarjian
H
,
Issa
JP
,
Rosenfeld
CS
, et al.
Decitabine improves patient outcomes in myelodysplastic syndromes: results of a phase III randomized study
.
Cancer
.
2006
;
106
(
8
):
1794
-
1803
.
doi:10.1002/cncr.21792
.
41.
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
.
doi:10.1016/j.leukres.2015.06.001
.
42.
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
.
doi:10.1016/j.leukres.2015.03.001
.
43.
Andriani
A
,
Elli
E
,
Trapè
G
, et al.
Treatment of Philadelphia-negative myeloproliferative neoplasms in accelerated/blastic phase with azacytidine: clinical results and identification of prognostic factors
.
Hematol Oncol
.
2019
;
37
(
3
):
291
-
295
.
doi:10.1002/hon.2635
.
44.
Gangat
N
,
Guglielmelli
P
,
Szuber
N
, et al.
Venetoclax with azacitidine or decitabine in blast-phase myeloproliferative neoplasm: a multicenter series of 32 consecutive cases
.
Am J Hematol
.
2021
;
96
(
7
):
781
-
789
.
doi:10.1002/ajh.26186
.
45.
Masarova
L
,
DiNardo
CD
,
Bose
P
, et al.
Single-center experience with venetoclax combinations in patients with newly diagnosed and relapsed AML evolving from MPNs
.
Blood Adv
.
2021
;
5
(
8
):
2156
-
2164
.
doi:10.1182/bloodadvances.2020003934
.
46.
Bewersdorf
JP
,
Giri
S
,
Wang
R
, et al.
Venetoclax as monotherapy and in combination with hypomethylating agents or low dose cytarabine in relapsed and treatment refractory acute myeloid leukemia: a systematic review and meta-analysis
.
Haematologica
.
2020
;
105
(
11
):
2659
-
2663
.
doi:10.3324/haematol.2019.242826
.
47.
King
AC
,
Weis
TM
,
Derkach
A
, et al.
Multicenter evaluation of efficacy and toxicity of venetoclax-based combinations in patients with accelerated and blast phase myeloproliferative neoplasms
.
Am J Hematol
.
2022
;
97
(
1
):
E7
-
E10
.
doi:10.1002/ajh.26381
.
48.
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
.
doi:10.1182/blood-2011-12-400051
.
49.
Mascarenhas
JO
,
Rampal
RK
,
Kosiorek
HE
, et al.
Phase 2 study of ruxolitinib and decitabine in patients with myeloproliferative neoplasm in accelerated and blast phase
.
Blood Adv
.
2020
;
4
(
20
):
5246
-
5256
.
doi:10.1182/bloodadvances.2020002119
.
50.
Bose
P
,
Verstovsek
S
,
Cortes
JE
, et al.
A phase ½ study of ruxolitinib and decitabine in patients with post-myeloproliferative neoplasm acute myeloid leukemia
.
Leukemia
.
2020
;
34
(
9
):
2489
-
2492
.
doi:10.1038/s41375-020-0778-0
.
51.
Wang
X
,
Hu
CS
,
Gillespie
V
,
Krejsa
CM
,
Hoffman
R
.
Navtemadlin (KRT-232), a small molecule MDM2 inhibitor, is more effective than decitabine against myeloproliferative neoplasm-blast phase in a patient-derived xenograft model
.
Blood
.
2021
;
138
(
suppl 1
):
3591
.
doi:10.1182/blood-2021-153071
.
52.
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
.
doi:10.1038/leu.2016.260
.
53.
Chifotides
HT
,
Masarova
L
,
Alfayez
M
, et al.
Outcome of patients with IDH1/2-mutated post–myeloproliferative neoplasm AML in the era of IDH inhibitors
.
Blood Adv
.
2020
;
4
(
21
):
5336
-
5342
.
doi:10.1182/bloodadvances.2020001528
.
54.
Patel
AA
,
Cahill
K
,
Charnot-Katsikas
A
, et al.
Clinical outcomes of IDH2- mutated advanced-phase Ph-negative myeloproliferative neoplasms treated with enasidenib
.
Br J Haematol
.
2020
;
190
(
1
):
e48
-
e51
.
doi:10.1111/bjh.16709
.
55.
Döhner
H
,
Estey
E
,
Grimwade
D
, et al.
Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel
.
Blood
.
2017
;
129
(
4
):
424
-
447
.
doi:10.1182/blood-2016-08-733196
.
56.
Mascarenhas
J
,
Heaney
ML
,
Najfeld
V
, et al.
Post-Myeloproliferative Neoplasm Acute Myeloid Leukemia Consortium
.
Proposed criteria for response assessment in patients treated in clinical trials for myeloproliferative neoplasms in blast phase (MPN-BP): formal recommendations from the post-myeloproliferative neoplasm acute myeloid leukemia consortium
.
Leuk Res
.
2012
;
36
(
12
):
1500
-
1504
.
doi:10.1016/j.leukres.2012.08.013
.
57.
Cerquozzi
S
,
Tefferi
A
.
Blast transformation and fibrotic progression in polycythemia vera and essential thrombocythemia: a literature review of incidence and risk factors
.
Blood Cancer J
.
2015
;
5
(
November
):
e366
.
doi:10.1038/bcj.2015.95
.
58.
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
.
doi:10.1038/leu.2013.163
.
59.
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
.
doi:10.1200/JCO.2009.22.8833
.
60.
Alvarez-Larrán
A
,
Senín
A
,
Fernández-Rodríguez
C
, et al.
Impact of genotype on leukaemic transformation in polycythaemia vera and essential thrombocythaemia
.
Br J Haematol
.
2017
;
178
(
5
):
764
-
771
.
doi:10.1111/bjh.14762
.
61.
Mollard
LM
,
Chauveau
A
,
Boyer-Perrard
F
, et al.
Outcome of Ph negative myeloproliferative neoplasms transforming to accelerated or leukemic phase
.
Leuk Lymphoma
.
2018
;
59
(
12
):
2812
-
2820
.
doi:10.1080/10428194.2018.1441408
.