Skip to main content
Therapeutic Advances in Hematology logoLink to Therapeutic Advances in Hematology
. 2023 Jun 22;14:20406207231177282. doi: 10.1177/20406207231177282

Treating accelerated and blast phase myeloproliferative neoplasms: progress and challenges

Helen O Ajufo 1, Julian A Waksal 2, John O Mascarenhas 3,, Raajit K Rampal 4
PMCID: PMC10410182  PMID: 37564898

Abstract

Myeloproliferative neoplasms (MPNs) are a group of clonal hematologic malignancies that include polycythemia vera (PV), essential thrombocythemia (ET), and myelofibrosis (MF). MPNs are characterized by activating mutations in the JAK/STAT pathway and an increased risk of transformation to an aggressive form of acute leukemia, termed MPN-blast phase (MPN-BP). MPN-BP is characterized by the presence of ⩾20% blasts in the blood or bone marrow and is almost always preceded by an accelerated phase (MPN-AP) defined as ⩾10–19% blasts in the blood or bone marrow. These advanced forms of disease are associated with poor prognosis with a median overall survival (mOS) of 3–5 months in MPN-BP and 13 months in MPN-AP. MPN-AP/BP has a unique molecular landscape characterized by increased intratumoral complexity. Standard therapies used in de novo acute myeloid leukemia (AML) have not demonstrated improvement in OS. Allogeneic hematopoietic stem cell transplant (HSCT) remains the only curative therapy but is associated with significant morbidity and mortality and infrequently utilized in clinical practice. Therefore, an urgent unmet need persists for effective therapies in this advanced phase patient population. Here, we review the current management and future directions of therapy in MPN-AP/BP.

Keywords: accelerated phase, blast phase, myeloproliferative neoplasms, novel therapeutics

Background

Philadelphia chromosone negative (Ph-) myeloproliferative neoplasms (MPNs) are a heterogeneous group of clonal hematologic malignancies that include polycythemia vera (PV), essential thrombocythemia (ET), and myelofibrosis (MF). 1 Activating mutations in the JAK/STAT pathway are the biologic hallmark of the MPNs. 2 MPNs are associated with a risk of transformation to acute leukemia, termed MPN-blast phase (BP), which is characterized by the presence of ⩾20% blasts in the peripheral blood or bone marrow. 3 Transformation to MPN-BP occurs within 10 years from diagnosis in 1%, 4%, and 20% of patients with ET, PV, and MF, respectively.4,5 Clinically, MPN-BP resembles secondary acute myeloid leukemia (sAML) and is typically preceded by an accelerated phase (MPN-AP) that is defined as presence of ⩾10–19% blasts in the peripheral blood or bone marrow. 1 Rarely, MPNs can transform into acute lymphoblastic leukemia (ALL). In a literature review and report of 18 cases of lymphoid blast transformation of Ph-MPNs, the majority of cases had a B-cell phenotype, a median time to progression to ALL of approximately 10 years and poor prognosis, with a mortality of 80% in the published cases. 6 The mechanisms surrounding the progression to MPN-AP/BP are not entirely understood but are thought to involve the acquisition of somatic mutations and epigenetic alterations in hematopoietic progenitor and stem cells (HPSCs) leading to clonal expansion as well as a tumor supportive proinflammatory microenvironment.3,7,8 Both MPN-AP and MPN-BP have a poor prognosis with a median overall survival (mOS) of approximately 13 months in MPN-AP and 3–5 months in MPN-BP.3,9 Newer targeted therapies have improved progression-free survival (PFS) but have not significantly impacted overall survival (OS). The only curative therapy is allogeneic hematopoietic stem cell transplant (HSCT) that is utilized in a minority of patients due to the high rate of associated morbidity and mortality in unfit populations. The molecular landscape of MPN-AP/BP differs significantly from that of de novo acute myeloid leukemia (AML). MPNs are characterized by driver mutations in Janus Kinase 2 (JAK2), myeloproliferative leukemia virus oncogene (MPL), and Calreticulin (CALR). Over 50% of patients with MPN-CP have additional somatic mutations in epigenetic regulators, splicing modulators, RAS, and TP53. In addition to molecular mutations, cytogenetic abnormalities associated with MPNs often portend increased intratumoral heterogeneity and complexity. 10 Primary myelofibrosis (PMF) is associated with more molecular complexity and an increased frequency of transformation to MPN-AP/BP than both ET and PV. 10 Understanding the molecular landscape driving the progression to MPN-AP/BP is paramount in stratifying patients who are at higher risk of progression and appropriate for risk-adapted therapeutic interventions including HSCT while still in the chronic phase (CP). Here, we provide an overview of the current management of patients with MPN-AP/BP as well as the future direction of therapeutic interventions.

Risk of progression

For the majority of patients who progress from CP disease to MPN-BP, the development of MPN-AP is an obligatory step in disease evolution. 3 Several clinical characteristics associated with progression from MPN-CP to MPN-AP/BP include age ⩾60 years, red blood cell (RBC) transfusion dependence, leukocytosis, monosomal karyotype, platelet count of <100 × 109/l, and circulating blasts ⩾ 3%.1,1113 Three widely implemented prognostic scoring systems incorporate these clinical risk factors to risk stratify patients with MF. The Dynamic International Prognostic Scoring System (DIPSS) includes age, presence of systemic symptoms, as well as complete blood count abnormalities including leukocytosis, anemia, and presence of circulating blasts. 14 The DIPSS-plus includes cytogenetic abnormalities, thrombocytopenia, and RBC transfusion dependence. 15 The Mutation Enhanced International Prognostic Score System MIPSS70 incorporates the presence of known high-risk acquired somatic mutations. 16

Current understanding of molecular basis of advanced forms of disease

Activating driver mutations involving the JAK/STAT pathway are the hallmark of CP-MPNs with JAK2V617F occurring in approximately 98% of patients with PV and in 50–60% of patients with ET or MF. 11 In patients with MF, mutations involving CALR and MPL also contribute to hyperactivity of this signaling pathway and together with JAK2V617F account for 90% of driver mutations. 17 The presence of other genomic alterations in TET2, EZH2, and TP53 frequently further alter the biology of the disease and have prognostic implications. 11 The most common somatic mutations in MPN-BP outside of JAK2 occur in TET2, SRSF2, IDH1/2 and TP53. 11 Whereas, the most common somatic mutations in de novo AML occur in FLT3, NPM1, and DNMT3A. 11 At least two distinct routes to leukemic transformation have been proposed: one route includes the acquisition of mutations in the clone bearing a JAK/STAT driver mutation, most commonly TP53 that is seen in approximately 15% of patients with JAK2V617F, 25% of patients with CALR mutations, and 0% of patients with MPL mutations. 18 Co-operativity between JAK2 mutations and TP53 mutations/alterations has been credentialed in murine models, thus establishing the biologic basis of these clinical observations. 19 Alternatively, de novo emergence of a leukemic clone distinct from the clone bearing an activating driver mutation can emerge, ultimately leading to a decrease in variant allele frequency (VAF) or loss of pre-existing driver mutation, due to clonal expansion of the leukemic clones over the underlying MPN-clone (Figure 1).10,11,20 Even with a significant proportion of MPN-BP arising from a potential nondriver mutation MPN-CP clone, the molecular landscape and overall prognosis remains distinct from that in de novo AML.

Figure 1.

Figure 1.

Pathophysiology of progression to MPN-AP/AP.

Current therapies

MPN-CP

The goal of therapy for patients with MPN-CP is to prevent thrombotic and hemorrhagic events, reduce the systemic symptom burden and splenomegaly, and minimize the risk of disease progression to MPN-AP/BP. The only curative therapy remains allogeneic HSCT, which is generally limited to patients with DIPSS intermediate-2 and high-risk MF due to associated morbidity and mortality. Current medical therapeutic agents for CP-MPNs include JAK inhibitors, pegylated interferon-alfa, and nonspecific cytoreductive agents. Ruxolitinib, fedratinib, and pacritinib are the three Food and Drug Administration (FDA) approved JAK inhibitors for select patients with MPN-CP. Fedratinib is typically reserved for patients who have failed ruxolitinib; however, it is approved for use in the front-line setting as well.21,22 Pacritinib is reserved for patients with MF with severe thrombocytopenia. 23 Momelotinib is another novel JAK inhibitor with positive phase III data seeking approval for MF patients with transfusion-dependent anemia. 24 While these therapeutic agents have demonstrated efficacy in terms of the ability to improve splenomegaly, and alleviate symptom burden, none has demonstrated the ability to effectively deplete the underlying malignant clone or reduce the risk of disease progression.22,23,2530 Despite the availability of these targeted agents, the majority of patients treated with selective JAK inhibitors discontinue treatment within 3 years. 31 Interferon derivatives have demonstrated the ability to deplete the underlying malignant clone in a minority of patients with PV and ET; however, whether or not this translates to a reduced risk of disease progression remains controversial and not yet confirmed in prospective studies.3234 Nonspecific cytoreductive agents include hydroxyurea and anagrelide, which are effective in preventing thrombotic and hemorrhagic events, but do not target the underlying malignant clone, and have not been shown to improve or reduce progression of disease to AP/BP.35,36 Rather than evaluating developmental agents based solely on thrombotic risk reduction, spleen size reduction, and symptom burden, there has been shift to develop novel anticlonal agents that can effectively target the underlying malignant clone and prevent progression of disease to AP/BP.

Potentially disease-modifying agents in development for patients with MF include inhibitors of MDM2, BET, BCL-2, PI3K, and telomerase, as well as recombinant PTX2 and an SMAC mimetic. 37 Trials have also evaluated histone deacetylase (HDAC) inhibitors in this patient population; however, toxicity profiles precluded further development.38,39 We are hopeful that as our understanding of disease biology continues to grow and more targeted treatment options become available, implementing these approaches will reduce the risk of disease progression and improve OS for CP-MPN. Even with optimal use of our current MPN-CP treatment armamentarium, however, a significant portion of patients with MPNs continue to progress to AP/BP. There remains no standardized approach for the management of MPNs that have progressed to AP or BP.

MPN-AP/BP

Cytotoxic chemotherapy

Cytotoxic chemotherapy with AML-inspired intensive induction regimens such as cytarabine and daunorubicin (7 + 3), fludarabine, cytarabine, and granulocyte colony-stimulating factor (G-CSF), idarubicin (FLAG-IDA), and mitoxantrone, etoposide, and cytarabine (MEC) without consolidative HSCT has been associated with poor outcomes.4,40,41 Complete response (CR) rates range from 0% to 30% often with incomplete count recovery (CRi), high treatment–related mortality, and short duration of leukemic-free interval.4,40,41 In a retrospective study of 91 patients with MF-BP, patients were split into 3 treatment groups; 19 received low-intensity therapy, 24 received an AML-like high-intensity induction regimen, and 48 received best supportive care. Low-intensity regimens included monotherapy with vincristine, etoposide, oral-alkylating agents, or low-dose cytarabine (LDAC). High-intensity regimens included 7 + 3 or monotherapy with high-dose cytarabine. No patients in the group receiving high-intensity therapy achieved a CR, and the mOS was 3.9 months. In a subset of patients who reverted to CP disease after high-intensity induction therapy, the mOS was 6.2 months compared with 2.7 months in those with persistently elevated blasts after induction therapy. The incidence of treatment-related mortality, however, was 33%. The mOS in the supportive care and low-intensity therapy arms was 2.1 months and 2.9 months, respectively. 41 Poor response to chemotherapy is likely due to the complex genetic profiles of MPN AP/BP that includes a high frequency of TP53 mutations.1,11 Owing to advanced age at diagnosis and often presence of comorbid conditions, patients with MPN BP/AP are frequently not candidates for intensive induction chemotherapy or HSCT. 1

Hematopoietic stem cell therapy

Consolidative HSCT is the only curative therapy in MPN-AP/BP. The significant morbidity and mortality associated with HSCT often precludes its use in older patients and those with significant comorbidities. In those who are transplant candidates, there are many transplant-related considerations including donor selection, optimal conditioning regimen, and post-transplant complications. In several retrospective studies of patients with MPN-BP who received HSCT, the only factor associated with prolonged OS was achievement of CR prior to transplant.40,42,43 In a retrospective review of 46 patients with MPN-BP, 42 of the 46 received induction therapy, followed by HSCT. Thirty eight of the 46 patients were assessable of which 24% achieved CR. At 3 years, the PFS and OS rates were 26% and 33%, respectively, and univariate analysis prior to HSCT demonstrated that only pretransplant CR impacted survival. Transplant-related factors such as donor source and conditioning regimen did not impact survival. 43 More recent data, however, suggest that CR is not required for improved outcomes in MPN-BP. A retrospective review of patients 46 patients with MPN-BP who received HSCT demonstrated that pre-HSCT blast count of <5% versus >5% or <10% versus >10% did not predict post-HSCT survival. 44 While there are no standard response criteria for patients with MPN AP/BP, the European LeukemiaNet (ELN) criteria are frequently employed to assess response. 45 Elevated circulating blasts and in the bone marrow >5%, however, can be seen in CP-MPNs due to aberrant MPN stem cell trafficking and resultant extramedullary hematopoiesis. A more comprehensive response assessment for MPN-BP has been proposed which incorporates five relevant components of MPN-CP and the leukemic clone: hematologic, clinical, pathologic, cytogenetic, and molecular changes. 46

Hypomethylating agents

Hypomethylating agents (HMAs) have demonstrated activity in MPN-AP/BP with less toxicity compared with intensive chemotherapy. In a retrospective analysis of 54 patients with Ph-negative MPNs who progressed to MPN-AP or BP and were treated with azacitidine, 24% achieved a CR. 47 The median duration of response was 9 months, and mOS was 11 months. 47 Another retrospective study that evaluated patients with high-risk MF and MPN-AP/BP, treated with single agent decitabine, demonstrated an overall response rate (ORR) of 29%, with an mOS of 10.5 months in those who responded and 4 months in nonresponders. 48 A retrospective review of a cohort of 410 patients with MPN-BP from the Mayo clinic and AIRC-Gruppo Italiano Malattie Mieloproliferative (AGIMM) treated with HMA monotherapy, the CR rate was only 4%. 12 While there is clearly a benefit for a certain subpopulation of patients with MPN-BP, when used as monotherapy, HMA therapy does not appear to provide a durable response in the majority of treated patients.

Ruxolitinib

Ruxolitinib, a selective JAK 1/2 inhibitor, has also been evaluated in MPN-BP in multiple clinical trials. A single-center phase II study evaluating high-dose ruxolitinib monotherapy was conducted in patients with R/R leukemia. Two of the 18 patients (11%) with MPN-BP treated with ruxolitinib at a dose of 25 mg twice daily attained a CR and one patient (5.5%) attained CRi. Notably, all three responding patients harbored JAK2V617F. 49 Overall, the regimen was well-tolerated, but led to limited and transient responses. 49 In a single-center phase I/II study in 27 patients testing three dose levels of ruxolitinib 50 mg twice daily (n = 4), 100 mg twice daily (n = 5), and 200 mg twice daily (n = 18) in patients with R/R AML and ALL, no CR was observed and the study was stopped due to lack of clinical benefit. 50 Similarly to HMA monotherapy, ruxolitinib alone has not demonstrated significant benefit in MPN-AP/BP. Several studies have evaluated ruxolitinib in combination with decitabine in this advanced phase setting. In a multicenter phase II study of 25 patients with MPN AP/BP treated with ruxolitinib at an induction dose of 25 mg twice daily and then 10 mg twice daily in subsequent 28-day cycles plus decitabine at 20 mg/m2 days 1–5 of a cycle, the ORR was 44% with an mOS of 9.5 months, and the combination was well-tolerated. 51 A single institution phase I study of ruxolitinib including 18 patients with R/R AML and phase II study including 29 patients with MPN-BP of ruxolitinib at a recommended phase II dose of 25 mg twice daily continuously and standard decitabine demonstrated an mOS of 6.9 months. 52 Because of the cross-trial mOS benefit of combination JAK2 inhibitor and HMA when compared with monotherapy data, and an improved toxicity profile when compared with intensive chemotherapy regimens, HMA and ruxolitinib are often administered in combination in this patient population when HSCT is not a therapeutic option for consolidation or as a bridge to HSCT in select patients (Table 1).

Table 1.

Therapeutic regimens and associated response rates and median OS.

Regimen Population Response rate Median OS References
Cytotoxic chemotherapy MPN-BP CR 0% 3.9 months Mesa et al. 41
Ruxolitinib MPN-BP CR 11% Eghtedar et al. 49
CR 0% Pemmaraju et al. 50
Ruxolitinib and decitabine MPN-AP/BP ORR (CR/CRi) 44% 9.5 months Mascarenhas et al. 51
IDH inhibitor MPN-AP/BP CR 25% Not reached at 9 months Patel et al. 60
IDH inhibitor monotherapy or in combination with ruxolitinib, venetoclax, HMA, or intensive chemotherapy MPN-BP CR 25% 10 months Chifotides et al. 59
HMA MPN-BP CR 24% 11 months Thepot et al. 47
MPN-AP/BP ORR 29% 10.5 months Badar et al. 48
MPN-BP CR 4% Tefferi et al. 12
Venetoclax and HMA MPN-BP ORR (CR/CRi) 44% 8 months Gangat et al. 54
MPN-BP CR/CRi 43% 7 months Masarova et al. 55

CR, complete response; Cri, incomplete count recovery; HMA, hypomethylating agent; IDH, isocitrate dehydrogenase; MPN-AP, myeloproliferative neoplasms–accelerated phase; MPN-BP, myeloproliferative neoplasms–blast phase; ORR, overall response rate; OS, overall survival.

Venetoclax

Venetoclax, a BCL-2 inhibitor, in combination with HMA is a well-described low-intensity regimen approved for the treatment of patients with AML. 53 These data have been extrapolated and employed in the anecdotal care of MPN-BP. This regimen has been evaluated in several retrospective studies in this patient population. A retrospective multicenter study of 32 patients with MPN-BP treated with a combination of venetoclax and azacitidine 75 mg/m2 (days 1–7) or decitabine 20 mg/m2 (days 1–5) demonstrated a 44% ORR (CR/CRi) and mOS of 8 months. 54 In a single-center study of 31 patients with MPN-BP treated with a combination venetoclax and HMA, 6 of 14 patients (43%) treated in the front-line setting achieved a CR/CRi and 1 patient (7%) achieved a partial response (PR). The mOS was 7 months. 55 In the R/R setting, no patients responded, and mOS was 3 months. 55 There was significant treatment-related toxicity with 83% of patients developing grade 3 or higher infections during the first cycle. 55 Another retrospective multicenter analysis that evaluated venetoclax and HMA or LDAC in 27 patients with MPN-AP or BP similarly demonstrated clinical activity with an ORR of 53% in MPN-BP and 50% in MPN-AP. There was limited survival benefit with an mOS of 6 months for both MPN-BP and MPN-AP. 56 While venetoclax plus HMA has significantly improved outcomes in patients with de novo AML, it has not demonstrated convincing beneficial impact in patients with MPN-BP. It is associated with significant toxicity, without survival benefit to merit its use in this patient population outside of a clinical trial.

IDH inhibitors

Mutations in isocitrate dehydrogenase 1/2 (IDH1/2) result in aberrant IDH1/2 activity and impaired myeloid differentiation. The use of small molecule inhibitors targeting these mutations, ivosidenib and enasidenib, which target IDH1 and IDH2, respectively, has led to improved outcomes in IDH-mutated AML in both the front-line and R/R setting.53,57 Importantly, IDH1 or IDH2 mutations occur in approximately 22% of MPN-AP/BP cases and thus represent a therapeutic opportunity in a significant portion of this patient population.58,59 In a retrospective analysis of MPN-BP patients with IDH1/2 mutations treated with an IDH inhibitor as monotherapy or in combination with ruxolitinib, venetoclax, HMA or intensive chemotherapy, 3 out of 12 (25%) patients achieved a CR with undetectable IDH1/2 mutations by next generation sequencing (NGS). 59 The mOS was 10 months for all patients and 19 months for responders. 59 Another group conducted a retrospective analysis of patients with IDH1 or IDH2-mutated MPN-AP/BP treated with IDH inhibitors. Of 8 patients treated, 2 (25%) achieved a CR, 4 (50%) achieved a PR, and 1 (12.5%) had stable disease. The median OS had not been reached at the median follow-up time of 9 months. 60 There is an ongoing multicenter Myeloproliferative Neoplasms Research Consortium (MPN-RC) phase II study assessing the safety and efficacy of the combination of ruxolitinib and enasidenib in patients with MPN AP/BP or CP MF and at least 5% circulating blasts with an IDH2 mutation (NCT04281498). Despite persistently low-response rates overall in published data, patients who responded have demonstrated favorable survival outcomes. Therefore, IDH inhibitors are typically incorporated into the treatment regimen for those MPN-AP/BPs with mutations in IDH1 or IDH2.

Future directions

Tumor microenvironment

MPNs are characterized by dysregulated JAK/STAT signaling resulting in an increase in proinflammatory cytokines such as transforming growth factor-beta (TGF-β), tumor necrosis factor alpha (TNFα) and interleukins (ILs) 2, 6, and 8 that have been implicated in supporting malignant clonal hematopoiesis and promoting bone marrow fibrosis. 61 Specifically, IL-2 and IL-8 have demonstrated particular importance in the prognosis and phenotype of MF. Increased levels of both cytokines have been associated with constitutional symptoms and increased RBC transfusion requirements. 62 In treatment naïve patients, increased levels of circulating IL-8 were found to be associated with inferior survival, increased circulating blasts, and shorter leukemia-free survival. 62 A murine model of MF demonstrated high levels of TGF-β, mCXCL1 (the murine equivalent of hCXCL8, the gene coding IL-8) and its receptors CXCR1 and CXCR2. 63 After treatment with reparixin, a CXCR1/2 inhibitor, megakaryocytes demonstrated lower levels of TGF-β compared with control suggesting that IL-8 may be a viable therapeutic target. 63 There is evidence that constitutive JAK/STAT signaling in MF leads to an immunosuppressive tumor microenvironment by upregulation of PD-L1 expression, checkpoint receptor. 64 Oncogenic JAK2 activity via increased STAT3 and STAT5 activation has been shown to enhance PD-L1 promoter activity and, therefore, PD-L1 expression in murine JAK2V617F knock-in model. PD-L1 expression was also shown to be upregulated in human T cells of MF patients when compared with healthy controls. 64 In addition to PD-L1 overexpression, peripheral blood T cells have also demonstrated overexpression of other checkpoint receptors including CTLA4 and TIM-3. 65 Given these data, it was hypothesized that PD-1 inhibition with pembrolizumab may potentiate the immune system to target MPN HPSCs. 64 In a multicenter single-arm phase II study that included 10 patients with advanced MF and single patient with MPN-BP who received pembrolizumab monotherapy, there was no significant clinical or pathologic response. There, however, was evidence of T-cell activation and changes in T-cell clonality with shared epitope demonstrating changes in the immune phenotype suggesting an MPN cell–directed immune repsonse. 66 Despite a lack of clinical or pathologic response to anti-PD1 monotherapy, combination therapy targeting LAG3, CTLA4, or TGF-β may provide additional benefit. Further investigation into the role of targeted combination immunotherapy in MPN AP/BP is warranted.

TP53 pathway dysregulation

Dysregulation of the TP53 pathway via deletion, mutation, or inactivation of p53 plays an important role in the pathogenesis across a diverse array of both solid and hematologic malignancies. 67 TP53 loss is among the most common genomic alterations observed at the time of leukemic transformation in MPNs occurring in 30% of patients. 11 Paired patient samples in MPN-CP and post-MPN AML demonstrated that mutations in JAK2 and TP53 were almost always present at high VAFs (>50%) in patients with MPN-BP at the time of transformation. This suggests their presence in the dominant AML clone and consistent with loss of wild-type TP53. 11 Inactivation of p53 occurs commonly as a result of overexpression of mouse double-minute homolog 2 (MDM2) and mouse double-minute homolog 4 (MDM4) – two important negative regulators of TP53 transcription and activation. 68 MDM2 is an E3 ubiquitin ligase that regulates the stability of p53 by mediating its degradation thus acting as a negative regulator of the pathway. 69 MDM4 can increase or decrease the E3 ubiquitin ligase activity. 70 Several chromosomal analyses of patients with MPNs have demonstrated an association between abnormalities in chromosomes 1q, which encodes MDM4, with increased risk of disease progression to myelofibrosis and MPN-AP/BP.67,71,72 In addition, increased expression of MDM2 has been demonstrated in CD34+ HSPCs in MPNs. 73 JAK2V617F has been demonstrated to be a negative regulator of TP53 via increased MDM2 translation. 74 Several therapeutic candidates are being developed that target reactivation of the p53 pathway.

Expression of MDM2 is upregulated in the HPSCs of patients with JAK2V617F MPNs due to upregulation of La antigen, an enhancer of MDM2 translation. 74 A class of drugs called Nutlins, named after the town in which they were discovered (Nutley, NJ), was the first identified selective MDM2 inhibitors. 75 RG7112 is a small molecule inhibitor of p53-MDM2 binding and in preclinical leukemia models demonstrated sensitivity to RG7112 through induction of p53-dependent apoptosis. 76 In a phase I study of patients with R/R leukemias including AML and ALL, RG7112 demonstrated stabilization in p53 levels and transcriptional activation of p53 target genes. 77 The high doses required for efficacy, however, led to significant GI intolerance. 77 A phase II trial evaluated idasanutlin (RG7388, Roche Pharmaceuticals, Basel, ‎Switzerland), an oral MDM2 inhibitor, in patients with JAK2V617F positive PV resistant/intolerant to hydroxyurea. Subjects enrolled demonstrated a 39% median reduction in the JAKV617F VAF at the end of three treatment cycles and a median reduction of 76% at 32 weeks suggesting robust anticlonal activity. 38 Therapy, however, was associated with significant gastrointestinal toxicity leading to treatment discontinuation in greater than 50% of patients. 38 Navtemadlin (KRT232, Kartos Therapeutics, Redwood City, CA, USA), a more potent MDM2 inhibitor demonstrated dose-dependent activity in the reduction of leukemic burden and OS in a patient-derived xenograft (PDX) MPN-BP murine model. 78 Navtemadlin demonstrated clinical activity in a phase Ib dose escalation study in TP53 WT patients with MPN BP 79 and in a phase II study of patients with R/R MF and DIPSS intermediate 2-risk R/R patients with MF. 80 It is currently being investigated in a multicenter phase Ib/II study in patients with R/R AML (including those with MPN-BP) as monotherapy and in combination with LDAC or decitabine. 81 Interferon-alfa (IFNα) has also demonstrated the ability to decrease the JAK2V617F VAF in patients with ET and PV and induce molecular remissions in 17–18% of patients.33,82,83 In preclinical studies RG7112 and Peg-IFNα 2a, each demonstrated the ability to significantly decrease JAK2V617F VAF in MF- and PV-HPSCs. When combined in low doses, RG7112 and Peg-IFNα 2a were able to eliminate JAK2V617F HPSCs, suggesting that they work synergistically to enhance p53 expression. 84 Early generation MDM2 inhibitors have demonstrated efficacy in restoring p53 activity. The higher doses, however, needed to be effective as monotherapy have been precluded by GI toxicity and bone marrow suppression, as MDM2 is needed for normal hematopoiesis. ALRN-6924 (Aileron Therapeutics, Cambridge, MA, USA) is a dual MDM4/MDM2 that mimics the inhibitor binding region of TP53. 85 In preclinical studies involving AML cell, ALRN-6924 induced apoptosis and cell cycle arrest as well as improved survival in AML xenograft models. 85 A 71 patient phase I study of ALRN-6924 in advanced solid tumors and lymphomas irrespective of TP53 status was found to be safe with limited myelosuppression, a dose-limiting toxicity well-described in other MDM2 inhibitors. 86 In the solid tumor cohort, the disease-control rate was 59% with a median duration of clinical benefit of 7.5 months. 86 Preliminary results of a phase I/Ib study of ALRN-6924 alone and in combination with cytarabine in 32 patients with AML and myelodysplastic syndrome (MDS) also demonstrated safety with no drug-limiting toxicities and no maximum tolerated dose (MTD) identified. Out of 27 patients, there were two CRs.87,88 WIP1 is another negative regulator of TP53. Inhibition of WIP1 with GSK2830371 in combination with the MDM2 inhibitor Nutlin-3A synergistically decreased viability and apoptosis in a TP53 WT AML cell line. 89 More potent and selective MDM2 inhibitors like navtemadlin in combination with JAK2 inhibitors, immunomodulatory agents, or other negative regulators of TP53 may mitigate myelosuppression seen in MDM2 inhibitors and lead to significant advances in treatment of MPN-AP/BP.

Wee1 inhibition

Genomic instability due to JAK2V617F and TP53 loss leads to increased dependence on DNA repair mechanisms. 19 Wee1 is an important regulator of G2/M checkpoint of the cell cycle. 19 In a study of a JAK2V617F and Trp53 mutant leukemic mouse model, the combination of adavosertib, a selective Wee1 inhibitor in combination with a PARP inhibitor olaparib, demonstrated significant reduction in the leukocyte count and leukemic burden in both the peripheral blood and bone marrow. In addition, treated mice demonstrated a decrease in spleen size and increase in OS compared with controls. Inhibition of Wee1 is thought to induce replicative stress while PARP inhibition inhibits DNA damage repair. Additional preclinical models evaluating adavosertib demonstrated DNA repair suppression, increase in caspase-mediated apoptosis in ALL cell lines, and a synergistic effect when combined with adriamycin. 90 This cell cycle pathway regulator is a promising target for evaluation in MPN-AP/BP in a clinical setting.

Ruxolitinib and BCL-2 inhibitors

Constitutively activated JAK/STAT signaling is associated with a proinflammatory cytokine profile and overexpression of antiapoptotic B-cell lymphoma proteins BCL-BCL-XL, BCL-2, and MCL-1 (a STAT target gene). 91 Expression of MCL-1 has represented a barrier to selective BCL-2 inhibition. 91 JAK inhibition leads to a decrease in MCL-1 expression, shifting the burden to BCL-XL and BCL-2 to maintain survival and sensitizing the leukemic cells to BCL-2/BCL-XL inhibitors.9193 Navitoclax is a novel small molecule BCL-2/BCL-XL antagonist. In a phase II multicenter trial of MF patients with prior ruxolitinib exposure and suboptimal response, the combination of ruxolitinib and navitoclax demonstrated an improvement in spleen volume and symptoms in 58% of patients harboring high-risk mutations (HMRs). 94 Zn-d5 is another viable BCL-2 inhibitor under investigation that is a more selective inhibitor of BCL-2 over BCL-XL and may mitigate some of the on-target myelosuppression seen in combination therapy with venetoclax and HMA. 95 Another novel therapeutic targeting antiapoptotic proteins is the BCL-XL/BCL-2 proteolysis-targeting chimera (PROTAC). PROTACs are small molecule compounds that cause degradation of target proteins through a ubiquitin proteasome system. 96 753B is a BCL-XL/BCL-2 PROTAC that causes BCL-XL/BCL-2 ubiquitination in cells expressing von-Hippel Lindau (VHL) that spares platelets as VHL is minimally expressed in platelets. It demonstrated efficacy when evaluated in a diverse group of leukemia cell lines and was able to reduce cell viability and increase apoptosis via degradation of BCL-XL in all 17 cell lines as well as BCL-2 in 16 of the 17 leukemia cell lines. In addition, 753B was able to eliminate chemotherapy induced senescent leukemia cells. 97 Inhibition of both JAK/STAT and BCL-2-related pathways may provide synergy and significantly add to the benefit noted in data discussed above that have evaluated venetoclax in this context.

Novel therapies in MDS and AML

Novel therapeutic agents used to that have demonstrated efficacy in MDS and AML and could be explored in MPN-AP/BP as part of a combination regimen include Magrolimab and Eprenetapopt (APR-246). Magrolimab is an anti-CD47 antibody and macrophage checkpoint inhibitor which in combination with both venetoclax and HMA has demonstrated safety and efficacy in MDS and AML. A phase I/II study evaluated venetoclax, azacitidine, and magrolimab in newly diagnosed older, unfit, or high-risk patients with AML. The CR/CRi rate was 94%, and 8-week mortality was 0. In seven patients with TP53 mutation, the CR/CRi was 100%, CR 86%, measurable residual disease (MRD) negative, and CR 57%. 98 Eprenetapopt (APR-246) is a pro-drug that binds to cysteine residues in mutant p53 leading to stabilization of the p53 protein and restoration of the wild-type conformation in addition to increasing oxidative stress and the promotion of tumor cell death. 99 A phase IB/II study of APR-246 in combination with azacitidine in TP53 patients with TP53-mutated MDS or AML demonstrated an ORR of 71% and CR 44%. In the MDS patients, the ORR was 73% and CR was 50%. In the AML patients, the ORR was 64% and CR 36%. 100

When used in combination with azacitidine, it has demonstrated efficacy in both TP53 mutant and TP53 wild-type AML patients and was been well-tolerated. The phase III trial, however, failed to meet the primary endpoint, and this agent is no longer being developed (Table 2).

Table 2.

Mutation frequency in post-MPN AML and de novo AML.18,101

De novo AML Post-MPN-AML
Mutation Frequency (%) Mutation Frequency (%)
ASXL1 30 JAK2V617F 55
FLT3 28 ASXL1 30
NPM1 27 TET2 25
DNMT3A 26 SRSF2 22
IDH1/IDH2 20 RUNX1 20
NRAS/KRAS 12 TP53 17
RUNX1 10 DNMT3A 16
TET 2 8 IDH1 13
TP53 8 IDH2 13
CEBPA 6 CALR 13
KIT 4 MPL 6

AML, acute myeloid leukemia; MPN, myeloproliferative neoplasm.

Conclusion

Despite the development of a number of novel-targeted therapeutic agents and approaches, MPN-AP/BP carries a dismal prognosis. The cytogenetic and mutational landscape in MPN-AP/BP is associated with increased rates of TP53 alterations compared with de novo AML, rendering patients less responsive to cytotoxic chemotherapy. Current practice for transplant eligible patients remains intensive induction chemotherapy followed by consolidative HSCT. For those ineligible for transplant, current therapies include HMA, BCL-2 inhibitors, and JAK inhibitors, which have demonstrated modest but often transient clinical responses. Novel targeted agents including IDH1/2 inhibitors have demonstrated modest improvement in patients with IDH mutations in retrospective series, and prospective evaluation in this molecularly defined subgroup is still ongoing. The combination of HMA and a BCL-2 inhibitor has demonstrated synergy in poor risk CP-MF but has not led to a significant clinical benefit in the absence of consolidative HSCT. There remains an urgent need for improved therapies in this patient population. Further investigation into combination therapies involving targeted agents that are able to deplete malignant HPSCs and alter the microenvironment that supports these cells may provide improved and more durable responses. Novel agents targeting CXCR1/2 and Wee1 appear promising in the preclinical setting but do not yet have clinical data. Immunotherapeutic agents such as PD-1 pathway inhibitors have not shown promising clinical activity as monotherapy to date. Given the pathophysiology of these diseases, however, it is plausible that combination regimens or novel agents may potentiate the immune system to more effectively target the underlying malignant cell population. Despite slow incremental progress, we are hopeful for better outcomes for patients with MPN AP/BP as new mechanism–based therapeutic agents continue to be developed and combined in innovative approaches.

Acknowledgments

None.

Footnotes

ORCID iD: John O. Mascarenhas Inline graphic https://orcid.org/0000-0001-6167-4322

Contributor Information

Helen O. Ajufo, Memorial Sloan Kettering Cancer Center, New York, NY, USA

Julian A. Waksal, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA

John O. Mascarenhas, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1079, New York, NY 10029, USA.

Raajit K. Rampal, Memorial Sloan Kettering Cancer Center, New York, NY, USA

Declarations

Ethics approval and consent to participate: Not applicable.

Consent for publication: All authors have read and agreed to publication of this manuscript.

Author contributions: Helen O. Ajufo: Writing – original draft; Writing – review & editing.

Julian A. Waksal: Writing – original draft; Writing – review & editing.

John O. Mascarenhas: Supervision; Writing – review & editing.

Raajit K. Rampal: Supervision; Writing – review & editing.

Funding: The authors received no financial support for the research, authorship, and/or publication of this article.

Competing interests: The authors declared the following potential conflicts of interest with respect to the research, authorship, and/or publication of this article: H.O.A. reports no financial disclosures. J.A.W. reports no financial disclosures. R.K.R. has received research funding from Zentalis, Ryvu, Incyte, Constellation, Stemline, and has received consulting fees from Constellation, Zentalis, Incyte, Celgene/BMS, Novartis, Promedior, CTI, Blueprint, Stemline, Galecto, Pharmaessentia, AbbVie, Sierra Oncology/GSK, Disc Medicines, Sunimoto Dainippon, Servier, and Karyopharm. J.O.M. receives research funding paid to his institution from Incyte, CTI Bio, Novartis, PharmaEssentia, Merck, Kartos, Geron, BMS, Roche, and consulting fees from Incyte, CTI Bio, Novartis, Roche, BMS, Celgene, Geron, PharmaEssentia, MorphoSys, Sierra Oncology, GSK, Galecto, Karyopharm, Kartos, and Imago.

Availability of data and materials: Not applicable.

References

  • 1. Jain T, Rampal RK. Accelerated and blast phase myeloproliferative neoplasms. Hematol Oncol Clin North Am 2021; 35: 325–331. [DOI] [PubMed] [Google Scholar]
  • 2. 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: 447–452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Marcellino B, Mascarenhas J. Management of advanced phase myeloproliferative neoplasms. Clin Adv Hematol Oncol 2019; 17: 405–411. [PubMed] [Google Scholar]
  • 4. Tam CS, Nussenzveig RM, Popat U, et al. The natural history and treatment outcome of blast phase BCR-ABL− myeloproliferative neoplasms. Blood 2008; 112: 1628–1637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Cervantes F, Tassies D, Salgado C, et al. Acute transformation in nonleukemic chronic myeloproliferative disorders: actuarial probability and main characteristics in a series of 218 patients. Acta Haematol 1991; 85: 124–127. [DOI] [PubMed] [Google Scholar]
  • 6. Alhuraiji A, Naqvi K, Huh YO, et al. Acute lymphoblastic leukemia secondary to myeloproliferative neoplasms or after lenalidomide exposure. Clin Case Rep 2018; 6: 155–161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Bowman RL, Busque L, Levine RL. Clonal hematopoiesis and evolution to hematopoietic malignancies. Cell Stem Cell 2018; 22: 157–170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Lasho TL, Mudireddy M, Finke CM, et al. Targeted next-generation sequencing in blast phase myeloproliferative neoplasms. Blood Adv 2018; 2: 370–380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Mudireddy M, Gangat N, Hanson CA, et al. Validation of the WHO-defined 20% circulating blasts threshold for diagnosis of leukemic transformation in primary myelofibrosis. Blood Cancer J 2018; 8: 57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Grinfeld J, Nangalia J, Baxter EJ, et al. Classification and personalized prognosis in myeloproliferative neoplasms. N Engl J Med 2018; 379: 1416–1430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Rampal R, Ahn J, Abdel-Wahab O, et al. Genomic and functional analysis of leukemic transformation of myeloproliferative neoplasms. Proc Natl Acad Sci 2014; 111: E5401–E5410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. 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: 1200–1210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Huang J, Li CY, Mesa RA, et al. Risk factors for leukemic transformation in patients with primary myelofibrosis. Cancer 2008; 112: 2726–2732. [DOI] [PubMed] [Google Scholar]
  • 14. 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: 1703–1708. [DOI] [PubMed] [Google Scholar]
  • 15. 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: 392–397. [DOI] [PubMed] [Google Scholar]
  • 16. 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: 310–318. [DOI] [PubMed] [Google Scholar]
  • 17. Tefferi A, Lasho TL, Finke CM, et al. CALR vs JAK2 vs MPL-mutated or triple-negative myelofibrosis: clinical, cytogenetic and molecular comparisons. Leukemia 2014; 28: 1472–1477. [DOI] [PubMed] [Google Scholar]
  • 18. McNamara CJ, Panzarella T, Kennedy JA, et al. The mutational landscape of accelerated- and blast-phase myeloproliferative neoplasms impacts patient outcomes. Blood Adv 2018; 2: 2658–2671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Li B, An W, Wang H, et al. BMP2/SMAD pathway activation in JAK2/p53-mutant megakaryocyte/erythroid progenitors promotes leukemic transformation. Blood 2022; 139: 3630–3646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Mannelli F. Acute myeloid leukemia evolving from myeloproliferative neoplasms: many sides of a challenging disease. J Clin Med 2021; 10: 436. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Harrison C, Kiladjian JJ, Verstovsek S, et al. MPN-164: overall survival (OS) and progression-free survival (PFS) in patients treated with fedratinib as first-line myelofibrosis (MF) therapy and after prior ruxolitinib (RUX): results from the JAKARTA and JAKARTA2 trials. Clin Lymphoma Myeloma Leuk 2021; 21: S356. [Google Scholar]
  • 22. 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: e317–e324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Mascarenhas J, Hoffman R, Talpaz M, et al. Pacritinib vs best available therapy, including ruxolitinib, in patients with myelofibrosis: a randomized clinical trial. JAMA Oncol 2018; 4: 652–659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Harrison CN, Vannucchi AM, Platzbecker U, et al. Momelotinib versus best available therapy in patients with myelofibrosis previously treated with ruxolitinib (SIMPLIFY 2): a randomised, open-label, phase 3 trial. Lancet Haematol 2018; 5: e73–e81. [DOI] [PubMed] [Google Scholar]
  • 25. Verstovsek S, Gotlib J, Mesa RA, et al. Long-term survival in patients treated with ruxolitinib for myelofibrosis: COMFORT-I and -II pooled analyses. J Hematol Oncol 2017; 10: 156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Harrison CN, Mead AJ, Panchal A, et al. Ruxolitinib vs best available therapy for ET intolerant or resistant to hydroxycarbamide. Blood 2017; 130: 1889–1897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. 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: e225–e236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Martí-Carvajal AJ, Anand V, Solà I. Janus kinase-1 and Janus kinase-2 inhibitors for treating myelofibrosis. Cochrane Database Syst Rev 2015: CD010298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Vannucchi AM, Kantarjian HM, Kiladjian JJ, et al. A pooled analysis of overall survival in COMFORT-I and COMFORT-II, 2 randomized phase III trials of ruxolitinib for the treatment of myelofibrosis. Haematologica 2015; 100: 1139–1145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Kremyanskaya M, Mascarenhas J, Rampal R, et al. Development of extramedullary sites of leukaemia during ruxolitinib therapy for myelofibrosis. Br J Haematol 2014; 167: 144–146. [DOI] [PubMed] [Google Scholar]
  • 31. Palandri F, Breccia M, Bonifacio M, et al. Life after ruxolitinib: reasons for discontinuation, impact of disease phase, and outcomes in 218 patients with myelofibrosis. Cancer 2020; 126: 1243–1252. [DOI] [PubMed] [Google Scholar]
  • 32. Barbui T, Vannucchi AM, De Stefano V, et al. Ropeginterferon alfa-2b versus phlebotomy in low-risk patients with polycythaemia vera (low-PV study): a multicentre, randomised phase 2 trial. Lancet Haematol 2021; 8: e175–e184. [DOI] [PubMed] [Google Scholar]
  • 33. Kiladjian JJ, Cassinat B, Chevret S, et al. Pegylated interferon-alfa-2a induces complete hematologic and molecular responses with low toxicity in polycythemia vera. Blood 2008; 112: 3065–3072. [DOI] [PubMed] [Google Scholar]
  • 34. Mascarenhas J, Kosiorek HE, Prchal JT, et al. A randomized phase 3 trial of interferon-α vs hydroxyurea in polycythemia vera and essential thrombocythemia. Blood 2022; 139: 2931–2941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Godfrey AL, Campbell PJ, MacLean C, et al. Hydroxycarbamide plus aspirin versus aspirin alone in patients with essential thrombocythemia age 40 to 59 years without high-risk features. J Clin Oncol 2018; 36: 3361–3369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Mazzucconi MG, Baldacci E, Latagliata R, et al. Anagrelide in essential thrombocythemia (ET): results from 150 patients over 25 years by the ‘Ph1-negative myeloproliferative neoplasms latium group’. Eur J Haematol 2020; 105: 335–343. [DOI] [PubMed] [Google Scholar]
  • 37. Waksal JA, Mascarenhas J. Novel therapies in myelofibrosis: beyond JAK inhibitors. Curr Hematol Malig Rep 2022; 17: 140–154. [DOI] [PubMed] [Google Scholar]
  • 38. Mascarenhas J, Passamonti F, Burbury K, et al. The MDM2 antagonist idasanutlin in patients with polycythemia vera: results from a single-arm phase 2 study. Blood Adv 2022; 6: 1162–1174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Guerini V, Barbui V, Spinelli O, et al. The histone deacetylase inhibitor ITF2357 selectively targets cells bearing mutated JAK2(V617F). Leukemia 2008; 22: 740–747. [DOI] [PubMed] [Google Scholar]
  • 40. Kennedy JA, Atenafu EG, Messner HA, et al. Treatment outcomes following leukemic transformation in Philadelphia-negative myeloproliferative neoplasms. Blood 2013; 121: 2725–2733. [DOI] [PubMed] [Google Scholar]
  • 41. Mesa RA, Li CY, Ketterling RP, et al. Leukemic transformation in myelofibrosis with myeloid metaplasia: a single-institution experience with 91 cases. Blood 2005; 105: 973–977. [DOI] [PubMed] [Google Scholar]
  • 42. Ciurea SO, De Lima M, Giralt S, et al. Allogeneic stem cell transplantation for myelofibrosis with leukemic transformation. Biol Blood Marrow Transplant 2010; 16: 555–559. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Alchalby H, Zabelina T, Stübig T, et al. 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: 279–281. [DOI] [PubMed] [Google Scholar]
  • 44. Shah MV, Saliba RM, Varma A, et al. Allogeneic stem cell transplant for patients with myeloproliferative neoplasms in blast phase: improving outcomes in the recent era. Br J Haematol 2021; 193: 1004–1008. [DOI] [PubMed] [Google Scholar]
  • 45. 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: 424–447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Mascarenhas J, Heaney ML, Najfeld V, et al. 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: 1500–1504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Thepot S, Itzykson R, Seegers V, et al. 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: 3735–3742. [DOI] [PubMed] [Google Scholar]
  • 48. 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: 950–956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. 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: 4614–4618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. 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: 171–176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. 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: 5246–5256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Bose P, Verstovsek S, Cortes JE, et al. A phase 1/2 study of ruxolitinib and decitabine in patients with post-myeloproliferative neoplasm acute myeloid leukemia. Leukemia 2020; 34: 2489–2492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. DiNardo CD, Jonas BA, Pullarkat V, et al. Azacitidine and venetoclax in previously untreated acute myeloid leukemia. N Engl J Med 2020; 383: 617–629. [DOI] [PubMed] [Google Scholar]
  • 54. 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: 781–789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. 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: 2156–2164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. 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: E7–E10. [DOI] [PubMed] [Google Scholar]
  • 57. Stein EM, DiNardo CD, Fathi AT, et al. Molecular remission and response patterns in patients with mutant-IDH2 acute myeloid leukemia treated with enasidenib. Blood 2019; 133: 676–687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Tefferi A, Jimma T, Sulai NH, et al. IDH mutations in primary myelofibrosis predict leukemic transformation and shortened survival: clinical evidence for leukemogenic collaboration with JAK2V617F. Leukemia 2012; 26: 475–480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. 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: 5336–5342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. 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: e48–e51. [DOI] [PubMed] [Google Scholar]
  • 61. Mondet J, Hussein K, Mossuz P. Circulating cytokine levels as markers of inflammation in Philadelphia negative myeloproliferative neoplasms: diagnostic and prognostic interest. Mediators Inflamm 2015; 2015: 670580–670510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Tefferi A, Vaidya R, Caramazza D, et al. 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: 1356–1363. [DOI] [PubMed] [Google Scholar]
  • 63. Verachi P, Martelli F, Zingariello M, et al. The CXCL1 inhibitor reparixin rescues myelofibrosis in the gata1 low model of the disease. Blood 2021; 138(Suppl. 1): 3579–3579. [Google Scholar]
  • 64. Prestipino A, Emhardt AJ, Aumann K, et al. Oncogenic JAK2 V617F causes PD-L1 expression, mediating immune escape in myeloproliferative neoplasms. Sci Transl Med 2018; 10: eaam7729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Cimen Bozkus C, Roudko V, Finnigan JP, et al. Immune checkpoint blockade enhances shared neoantigen-induced T-cell immunity directed against mutated calreticulin in myeloproliferative neoplasms. Cancer Discov 2019; 9: 1192–1207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Hobbs G, Cimen Bozkus C, Moshier E, et al. PD-1 inhibition in advanced myeloproliferative neoplasms. Blood Adv 2021; 5: 5086–5097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Marcellino BK, Hoffman R, Tripodi J, et al. Advanced forms of MPNs are accompanied by chromosomal abnormalities that lead to dysregulation of TP53. Blood Adv 2018; 2: 3581–3589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Wade M, Li YC, Wahl GM. MDM2, MDMX and p53 in oncogenesis and cancer therapy. Nat Rev Cancer 2013; 13: 83–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Konopleva M, Martinelli G, Daver N, et al. MDM2 inhibition: an important step forward in cancer therapy. Leukemia 2020; 34: 2858–2874. [DOI] [PubMed] [Google Scholar]
  • 70. Linares LK, Hengstermann A, Ciechanover A, et al. HdmX stimulates Hdm2-mediated ubiquitination and degradation of p53. Proc Natl Acad Sci 2003; 100: 12009–12014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Tang G, Hidalgo Lopez JE, Wang SA, et al. Characteristics and clinical significance of cytogenetic abnormalities in polycythemia vera. Haematologica 2017; 102: 1511–1518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Klampfl T, Harutyunyan A, Berg T, et al. Genome integrity of myeloproliferative neoplasms in chronic phase and during disease progression. Blood 2011; 118: 167–176. [DOI] [PubMed] [Google Scholar]
  • 73. Lu M, Wang X, Li Y, et al. Combination treatment in vitro with nutlin, a small-molecule antagonist of MDM2, and pegylated interferon-α 2a specifically targets JAK2V617F-positive polycythemia vera cells. Blood 2012; 120: 3098–3105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Nakatake M, Monte-Mor B, Debili N, et al. JAK2(V617F) negatively regulates p53 stabilization by enhancing MDM2 via La expression in myeloproliferative neoplasms. Oncogene 2012; 31: 1323–1333. [DOI] [PubMed] [Google Scholar]
  • 75. Vassilev LT. MDM2 inhibitors for cancer therapy. Trends Mol Med 2007; 13: 23–31. [DOI] [PubMed] [Google Scholar]
  • 76. Tovar C, Graves B, Packman K, et al. MDM2 small-molecule antagonist RG7112 activates p53 signaling and regresses human tumors in preclinical cancer models. Cancer Res 2013; 73: 2587–2597. [DOI] [PubMed] [Google Scholar]
  • 77. Andreeff M, Kelly KR, Yee K, et al. Results of the phase I trial of RG7112, a small-molecule MDM2 antagonist in leukemia. Clin Cancer Res 2016; 22: 868–876. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Wang X, Hu CS, Gillespie V, et al. 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. [Google Scholar]
  • 79. Erba HP, Becker PS, Shami PJ, et al. Phase 1b study of the MDM2 inhibitor AMG 232 with or without trametinib in relapsed/refractory acute myeloid leukemia. Blood Adv 2019; 3: 1939–1949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Vachani P, Lange A, Delgado RG, et al. Potential disease-modifying activity of navtemadlin (KRT-232), a first-in-class MDM2 inhibitor, correlates with clinical benefits in relapsed/refractory myelofibrosis (MF). Blood 2021; 138(Suppl. 1): 3581–3581. [Google Scholar]
  • 81. Rampal R, Ramanathan S, Papayannidis C, et al. An open-label, multicenter, phase 1b/2 study of navtemadlin (KRT-232) in patients with relapsed/refractory acute myeloid leukemia secondary to myeloproliferative neoplasms. J Clin Oncol 2022; 40(Suppl.): TPS7063–TPS7063. [Google Scholar]
  • 82. Quintás-Cardama A, Abdel-Wahab O, Manshouri T, et al. Molecular analysis of patients with polycythemia vera or essential thrombocythemia receiving pegylated interferon α-2a. Blood 2013; 122: 893–901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Hasselbalch HC. A new era for IFN-α in the treatment of Philadelphia-negative chronic myeloproliferative neoplasms. Expert Rev Hematol 2011; 4: 637–655. [DOI] [PubMed] [Google Scholar]
  • 84. Lu M, Xia L, Li Y, et al. The orally bioavailable MDM2 antagonist RG7112 and pegylated interferon α 2a target JAK2V617F-positive progenitor and stem cells. Blood 2014; 124: 771–779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Carvajal LA, Neriah DB, Senecal A, et al. Dual inhibition of MDMX and MDM2 as a therapeutic strategy in leukemia. Sci Transl Med 2018; 10: eaao3003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Saleh MN, Patel MR, Bauer TM, et al. Phase 1 trial of ALRN-6924, a dual inhibitor of MDMX and MDM2, in patients with solid tumors and lymphomas bearing wild-type TP53. Clin Cancer Res 2021; 27: 5236–5247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87. Sallman DA, Borate U, Cull EH, et al. Phase 1/1b study of the stapled peptide ALRN-6924, a dual inhibitor of MDMX and MDM2, as monotherapy or in combination with cytarabine for the treatment of relapsed/refractory AML and advanced MDS with TP53 wild-type. Blood 2018; 132(Suppl. 1): 4066–4066. [Google Scholar]
  • 88. Khurana A, Shafer DA. MDM2 antagonists as a novel treatment option for acute myeloid leukemia: perspectives on the therapeutic potential of idasanutlin (RG7388). Onco Targets Ther 2019; 12: 2903–2910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. Fontana MC, Nanni J, Ghelli Luserna di Rorà A, et al. Pharmacological inhibition of WIP1 sensitizes acute myeloid leukemia cells to the MDM2 inhibitor nutlin-3a. Biomedicines 2021; 9: 388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Duan Y, Dong X, Nie J, et al. Wee1 kinase inhibitor MK‑1775 induces apoptosis of acute lymphoblastic leukemia cells and enhances the efficacy of doxorubicin involving downregulation of Notch pathway. Oncol Lett 2018; 16: 5473–5481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Tognon R, Gasparotto EPL, Neves RP, et al. Deregulation of apoptosis-related genes is associated with PRV1 overexpression and JAK2 V617F allele burden in essential thrombocythemia and myelofibrosis. J Hematol Oncol 2012; 5: 2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Guo J, Roberts L, Chen Z, et al. JAK2V617F drives Mcl-1 expression and sensitizes hematologic cell lines to dual inhibition of JAK2 and Bcl-xL. PLoS ONE 2015; 10: e0114363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93. Waibel M, Solomon VS, Knight DA, et al. Combined targeting of JAK2 and Bcl-2/Bcl-xL to cure mutant JAK2-driven malignancies and overcome acquired resistance to JAK2 inhibitors. Cell Rep 2013; 5: 1047–1059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Harrison CN, Garcia JS, Somervaille TCP, et al. Addition of navitoclax to ongoing ruxolitinib therapy for patients with myelofibrosis with progression or suboptimal response: phase II safety and efficacy. J Clin Oncol 2022; 40: 1671–1680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Izadi H, Hoang T, Ibrahim N, et al. Abstract 2591: combination of the BCL-2 inhibitor ZN-d5 with the WEE1 inhibitor ZN-c3 shows additive or synergistic anti-tumor activity in acute myeloid leukemia (AML) models. Cancer Res 2022; 82(Suppl.): 2591–2591. [Google Scholar]
  • 96. Khan S, Zhang X, Lv D, et al. A selective BCL-XL PROTAC degrader achieves safe and potent antitumor activity. Nat Med 2019; 25: 1938–1947. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Jia Y, Zhang Q, Zhang W, et al. Targeting BCL-XL and BCL-2 By protac 753B effectively eliminates AML cells and enhances efficacy of chemotherapy by targeting senescent cells. Blood 2021; 138(Suppl. 1): 2230–2230. [Google Scholar]
  • 98. Daver N, Konopleva M, Maiti A, et al. Phase I/II study of azacitidine (AZA) with venetoclax (VEN) and magrolimab (Magro) in patients (pts) with newly diagnosed older/unfit or high-risk acute myeloid leukemia (AML) and relapsed/refractory (R/R) AML Blood 2021; 138(Suppl. 1): 371. [Google Scholar]
  • 99. Zhang Q, Bykov VJN, Wiman KG, et al. APR-246 reactivates mutant p53 by targeting cysteines 124 and 277. Cell Death Dis 2018; 9: 439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Sallman DA, DeZern AE, Garcia-Manero G, et al. Eprenetapopt (APR-246) and azacitidine in TP53-mutant myelodysplastic syndromes. J Clin Oncol 2021; 39: 1584–1594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. The Cancer Genome Atlas Research Network. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med 2013; 368: 2059–2074. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Therapeutic Advances in Hematology are provided here courtesy of SAGE Publications

RESOURCES