Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Apr 21.
Published in final edited form as: Acta Haematol. 2021 Apr 21;144(5):484–499. doi: 10.1159/000512929

Accelerated Phase of Myeloproliferative Neoplasms

Omar A Shahin 1,, Helen T Chifotides 1,, Prithviraj Bose 1, Lucia Masarova 1, Srdan Verstovsek 1,*
PMCID: PMC8458218  NIHMSID: NIHMS1657913  PMID: 33882481

Abstract

Background:

Myeloproliferative neoplasms (MPN) can transform to blast phase MPN (leukemic transformation; MPN-BP), typically via accelerated phase MPN (MPN-AP), in ~20–25% of the cases. MPN-AP and MPN-BP are characterized by 10–19% and ≥ 20% blasts, respectively. MPN-AP/BP portend a dismal prognosis with no established conventional treatment. Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is the sole modality associated with long-term survival.

Summary:

MPN-AP/BP has a markedly different mutational profile from de novo acute myeloid leukemia (AML). In MPN-AP/BP, TP53 and IDH1/2 are more frequent, whereas FLT3 and DNMT3A are rare. Higher incidence for leukemic transformation has been associated with the most aggressive MPN subtype, myelofibrosis MF); other risk factors for leukemic transformation include rising blast counts above 3–5%, advanced age, severe anemia, thrombocytopenia, leukocytosis, increasing bone marrow fibrosis, type 1 CALR-unmutated status, lack of driver mutations (negative for JAK2, CALR or MPL genes), adverse cytogenetics, and acquisition of ≥ 2 high molecular risk mutations (ASXL1, EZH2, IDH1/2, SRSF2, and U2AF1Q157). The aforementioned factors have been incorporated in several novel prognostic scoring systems for MF. Currently, elderly/unfit patients with MPN-AP/BP are treated with hypomethylating agents (HA) with/without ruxolitinib; these regimens appear to confer comparable benefit to intensive chemotherapy but with lower toxicity. Retrospective studies in patients who acquired actionable mutations during MPN-AP/BP showed positive outcomes with targeted AML regimens, such as IDH1/2 inhibitors, and require further evaluation in clinical trials.

Key Messages:

Therapy for MPN-AP patients represents an unmet medical need. MF patients, in particular, should be appropriately stratified regarding their prognosis and the risk for transformation. Higher-risk patients should be monitored regularly and treated prior to progression to MPN-BP. MPN-AP patients may be treated with hypomethylating agents alone or in combination with ruxolitinib or provided with the option to enroll in rationally designed clinical trials exploring combination regimens, including novel targeted drugs, with an ultimate goal to transition to transplant.

Keywords: accelerated phase MPN (MPN-AP), blast phase MPN (MPN-BP), myelofibrosis (MF), myeloproliferative neoplasms (MPN), post-MPN AML

Introduction

Aberrant proliferation of hematopoietic stem cells and pro-inflammatory cytokine release, megakaryocytic atypia, and bone marrow fibrosis are the main features of myelofibrosis (MF), the most aggressive among Philadelphia negative (Ph) myeloproliferative neoplasms (MPNs). Clinically, MF manifests with anemia and thrombocytopenia, hepatosplenomegaly, extramedullary hematopoiesis, and constitutional symptoms that compromise quality of life [1, 2]. MF can present as primary (PMF) or as a complication of preceding polycythemia vera (PV) or essential thrombocythemia (ET) (called post-PV MF and post-ET MF, respectively) [3]. Dysregulated JAK/STAT signaling is a hallmark of MPN, primarily arising from JAK2 mutations in exon 14 (primarily the valine to phenylalanine point mutation at codon 617 in the pseudokinase domain of JAK2) or other activating JAK2 mutations in exon 12, the myeloproliferative leukemia (MPL) gene, and indels in exon 9 of the calreticulin (CALR) gene [4, 5].

MF has the worst prognosis among MPNs whereas PV and ET are relatively indolent subtypes [6]. The most concerning complication of PMF and post-PV MF/post-ET MF is leukemic transformation, which occurs in ~20–25% of the cases [7]. At 10 years, the estimated incidence of leukemic transformation is highest for MF (10–20%) followed by PV (~2.3–14.4%), and is lowest in ET (0.7–3%) [7, 8]. Accordingly, in a retrospective analysis that assessed the outcomes of 795 MPN patients, the annual incidence of leukemic transformation was the highest in PMF patients when compared to PV (P = 0.002) or ET (1.09% and 0.37%, respectively, P = 0.02). [9].

Progression from MPN in the chronic phase (MPN-CP) to MPN in the blast phase (MPN-BP) usually evolves through MPN in the accelerated phase (MPN-AP) [10]. MPN-CP, MPN-AP and MPN-BP are defined on the basis of blast percentage in the bone marrow or peripheral blood, i.e., 0–9%, ≥10–19% and 20% or more, respectively (Table 1) [11, 12, 13]. In MPNs, leukemic transformation is associated with dismal prognosis, and great efforts have been made to identify the risk factors and molecular events associated with disease evolution and progression to MPN-BP. In general, advanced age, severe anemia, leukocytosis, circulating blasts >2%, thrombocytopenia, advanced bone marrow fibrosis, cytogenetic abnormalities, and acquisition of ≥ 2 high-risk mutations were defined as clear risk factors for leukemic transformation, in many studies [2, 7, 14, 15, 16]. Given that MPN-AP precedes MPN-BP in most patients, the aforementioned factors can be assessed as risk predictors of transformation from CP to MPN-AP [17]. Clinical data and treatments for MPN-AP are limited because the majority of investigations have been conducted in patients with MPN-CP, and not MPN-AP/BP [18]. Clinical practice, however, has clearly demonstrated that patients with MPN-AP/BP have a dismal outcome (worse than de novo acute myeloid leukemia [AML]), which is attributed to the inherent nature of the disease and the dearth of standard management or disease-modifying treatments [19, 20]. Currently, the sole modality that confers prolonged survival and potentially cures the disease is consolidative allogeneic hematopoietic stem cell transplantation (allo-HSCT) but a limited number of patients qualify for this option [19, 21, 22, 23, 24, 25, 26, 27, 28, 29]. In this review, we aim to highlight the key concepts and findings that have emerged for MPN-AP, primarily on the basis of retrospective studies, mounting evidence from clinical practice; and the limited number of clinical trials that have assessed therapeutic schemes in this population, facing adverse prognosis and a dearth of treatment modalities heretofore.

Table 1.

MPN-CP, MPN-AP, and MPN-BP and blast counts in the bone marrow or peripheral blood.

MPN Phase MPN Type BM blast count PB blast count
Chronic PMF, PV, ET >1–9% >1–9%
Accelerated PMF, PV, ET 10–19% 10–19%
Blast PMF, PV, ET ≥ 20% ≥ 20%

MPN-CP, myeloproliferative neoplasm in chronic phase; MPN-AP, myeloproliferative neoplasm in accelerated phase; MPN-BP, myeloproliferative neoplasm in blast phase; BM, bone marrow; PB, peripheral blood; PMF, primary myelofibrosis; PV, polycythemia vera; ET, essential thrombocythemia.

Retrospective studies, risk factors, and prognostication in MPN-AP

Our team retrospectively analyzed the clinical characteristics of 370 patients with PMF (79% of the patients) and secondary MF (post-PV MF and post-ET MF) that had a median overall survival (OS) of 12 months or less, to determine the characteristics of MPN-AP and identify a set of progression-associated criteria [10]. Multivariable analysis of the putative high-risk AP characteristics associated with median OS ≤ 12 months was performed; baseline bone marrow or circulating blasts ≥10%, aberrations of chromosome 17 and platelets below 50×109/L were associated with median OS of 10, 5, and 12 months, respectively. These high-risk features were also validated during the disease course; e.g., patients that were in the chronic phase and subsequently developed one of the aforementioned characteristics had similarly short survivals (median OS 12, 6 and 15 months, respectively). Moreover, the rate of leukemic transformation of patients who persistently were in MPN-CP was very low (1% at 3 years and 3 % at 10 years, P < 0.001) as compared to patients in MPN-AP. These findings clearly demonstrate that transformation to MPN-BP from an antecedent MPN transitions through the MPN-AP phase [10].

The role of increased blasts in the outcome of patients with MF was recently assessed by our group [30, 31]. On a cohort of 1316 MF patients with available peripheral and bone marrow blast percentages, we showed that patients with 4% peripheral blasts and ≥ 5% bone marrow blasts had comparable OS and clinical characteristics, and also resembled patients in AP (blasts ≥10–19%). The respective OS of patients with 0%, 1–3%, 4% peripheral blasts (and < 5% bone marrow blasts), 5–9% peripheral and/or bone marrow blasts and ≥10–19% peripheral and / or bone marrow blasts were 64, 48, 22, 24, and 13 months, respectively. Overall, 146 patients progressed to AML, with the highest rate among those with ≥10% blasts, as expected; however, patients with 5–9% blasts had an approximate twofold rate of leukemic transformation as compared to patients with lower blasts (AML incidence per 100 person-years was 7.7 and 24.7 cases for 5–9 % and ≥10% blasts, respectively, vs. 3.5 cases only for blasts ≤4%) [30, 31]. The estimated leukemia-free survival rate at 3 years was ~ 60% for those with < 5% blasts, and ~ 30% for those with ≥ 5% blasts [30]. Patients who had blasts <10% and were treated with ruxolitinib had superior overall survival compared to patients who did not receive ruxolitinib [31].

Geyer et al. conducted a retrospective study in which 92 patients with initial diagnosis of PMF, PV/ post-PV MF, and ET/ post-ET MF (median follow-up time was 11 years) and increased blasts were stratified in three categories according to blast counts: IB-1 (2–4% peripheral blood blasts and <5% bone marrow blasts), IB-2 (5–9% blasts in the bone marrow and/or peripheral blood), and AP (≥ 10–19% blasts in bone marrow or peripheral blood); the investigators also identified a control group comprising MPN patients that did not have increased blasts, leukocytosis or monocytosis [32]. The analysis demonstrated that patients in the IB-2 group (P = 0.00014) had a significantly inferior OS than both the IB-1 group and control patients; in contrast, the OS rates of group IB-1 and the controls were similar. As expected, patients in the combined IB-1/IB-2 groups (P = 0.0038) and the controls (P < 0.0001) had significantly longer OS than patients in AP [32]. The two aforementioned studies by Masarova at al. [30] and Geyer et al. [32] indicate that lowering the blast percentage threshold defining MPN-AP from 10% to 5% is justified [25, 33]. Notwithstanding designation of ≥ 20% bone marrow or peripheral blood blasts as indication of MPN-BP by the International Working Group-Myeloproliferative Neoplasms Research and Treatment (IWG-MRT) in 2013 [3], blasts in the range 10–19% have been associated with MPN-AP and clearly inferior outcomes [10, 30]; in clinical practice, the former blast range is currently applied as a criterion of advanced MPN disease and thus, could be included in the next revision of the criteria. In a retrospective study of 311 patients with PMF, aiming to assess the risk factors leading to leukemic transformation, peripheral blood blasts ≥ 3% (P < 0.0001) and a platelet count below 100 × 109/L (P = 0.004) were determined to be strong and independent risk factors in both univariate and multivariate analyses [34]. When the previous factors were taken into consideration, the frequency of leukemic transformation increased from 5.8% in the subgroup with neither one of the two risk factors to 18.2% in the subgroup that had both [34]. In a recent large study including 1306 patients with PMF, Vallapureddy et al. demonstrated that peripheral blasts ≥3% were associated with increased risk of leukemic transformation (HR 3.3) in the first five years of diagnosis [35]. Other retrospective studies led to the conclusion that red blood cell transfusion dependence and high white blood cell (WBC) counts were correlated with elevated risk of transformation [36, 37].

Several prognostic models were developed to assess the outcome of PMF patients [38]. The initial International Prognostic Scoring System (IPSS) [39] relies on clinical parameters at diagnosis and was modified to the Dynamic IPSS (DIPSS) to assess prognosis based on risk factors at diagnosis and during the natural history of the disease [40]. In DIPSS, age over 65 years (1 point), hemoglobin < 10 g/dL (2 points), WBC count > 25 × 109/L (1 point), peripheral blood blasts ≥1% (1 point), and constitutional symptoms (1 point) predicted poor survivals. In DIPSS, patients were stratified into 4 categories: low (0 points), intermediate-1 (1–2 points), intermediate-2 (3–4 points) and high-risk (5–6 points) [40]. The estimated hazard risk for progression to BP was 2 when the risk category progressed from low to intermediate-1, 3.8 from intermediate-1 to intermediate-2, and 3.2 from intermediate-2 to high. The risk to develop MPN-BP in the intermediate-2 and high categories was 7.8- and 24.9-fold higher than in the low-risk category, respectively [41]. On the basis of the evidence acquired from follow-up data of PMF patients, karyotype, transfusion-dependence and thrombocytopenia (platelet count <100 ×109/L) were integrated in the DIPSS, thereby developing DIPSS-plus [42], which turned out to be superior in assessing the LFS compared to both IPSS and DIPSS. Thrombocytopenia and anemia were also included in the more integrated Mutation-Enhanced International Prognostic Scoring System 70 (MIPSS70) for transplant-eligible patients with PMF aged ≤70 years [43] and in the Myelofibrosis SECondary to PV and ET-Prognostic Model (MYSEC-PM) for post-PV MF / post-ET MF [44].

Cytogenetic abnormalities constitute an important risk factor in progression of MF-CP to MF-AP/BP [7]; however, cytogenetics were not included as a factor in the IPSS [39] and DIPSS [40]. Hussein et al. showed the prognostic relevance of specific cytogenetic abnormalities (e.g., +8 or complex karyotype) [45] and the favorable prognostic impact of sole 20q-, 13q- or +9 [46, 47, 48]. In a more recent study on 1002 patients with MF, the same investigators showed that 45% of the patients had abnormal karyotype; 6% of the latter subgroup had complex karyotype (3 or more abnormalities), and of those, about one third had a monosomal karyotype [48]. In a cohort of patients with PMF, the 2-year leukemic transformation rate associated with monosomal karyotype was 29.4%, and the median survival was 6 months versus 8.3% and 24 months, respectively, for complex karyotype without monosomies, demonstrating the dismal prognosis associated with certain monosomies [49]. The unfavorable karyotype category of the two-tiered cytogenetic model for PMF includes complex karyotype and one or two abnormalities, for example, +8, −5/5q-, −7/7q-, 12p-, i(17q), inv(3), and 11q23 rearrangement [42, 46] (Table 2). Notably, Tefferi et al. recently proposed a more refined three-tiered cytogenetic stratification, comprising very high-risk (VHR), unfavorable, and favorable karyotype categories [48] (Table 2). The VHR karyotype showed inferiority in LFS regardless of other risk factors, such as high-risk mutations, thrombocytopenia, and DIPSS [48]. The three-tiered cytogenetic model has been integrated in the five-tiered Mutation and karyotype-enhanced MIPSS70-plus version 2.0 [50] and the four-tiered Genetically Inspired Prognostic Scoring System (GIPSS) [51] for PMF. On the other hand, the two-tiered cytogenetic model (comprising favorable and unfavorable categories) has been incorporated in DIPSS-plus [42] and MIPSS70-plus [43]. The recent refined three-tiered cytogenetic model was developed based on a cohort of 1002 PMF patients at Mayo Clinic [48] and was further evaluated in the studies that led to the development of MIPSS70-plus version 2.0 [50] and GIPSS [51]. Validation of the three-tiered model is necessary as preliminary results from our group did not confirm the same discrimination power of the three-tiered model [52].

Table 2.

Two-tiered versus the revised three-tiered cytogenetic risk models and risk categories in PMF.

Two-tiered cytogenetic risk categories according to Caramazza et al. [46] Cytogenetic risk category Revised three-tiered cytogenetic risk categories according to Tefferi et al. [48] Cytogenetic risk category
Complex karyotype
Sole or two abnormalities that include:
 • +8
 • −7/7q-
 • i(17q)
 • −5/5q-
 • 12p-
 • inv(3)
 • 11q23 rearrangement
Unfavorable Complex karyotype without a
VHR abnormality
Sole abnormalities that include:
 • +8
 • −7q-
Sole translocations not involving chromosome 1
Two abnormalities not including a VHR abnormality
Single/multiple 5q− abnormalities
Monosomal karyotype without a
VHR abnormality
Sole abnormalities not otherwise classified
Unfavorable
Single/multiple abnormalities of:
 • inv(3)/3q21
 • −7
 • 11q-/11q23
 • 12p-/12p11.2
 • i(17q)
Autosomal trisomies, not including
+8 or +9 (for example, +19, +21)
Very High Risk (VHR)
Normal karyotype
Sole abnormalities of:
 • +9
 • 13q-
 • 20q-
   chromosome 1 translocation/duplication
two abnormalities excluding unfavorable ones
Favorable Normal karyotype
Sole abnormalities of:
 • +9
 • 13q-
 • 20q-
   chromosome 1 translocation/duplication
sex chromosome abnormality, including –Y
Favorable

PMF: primary myelofibrosis; VHR: very high-risk

A recent study that Marcellino et al. conducted on the impact of chromosomal abnormalities in advanced MPN showed that 43% of the patients with MPN-AP/BP gained +1q versus 9% and 2% in the MF and PV/ET cohorts, respectively [53], and +1q was recurrently detected in MPN-AP/BP [53, 54]. Notably, +1q was detected in the majority of MPN patients (84%) that harbored JAK2V617F [53]. Furthermore, all the patients with MPN-AP/BP (33%), PMF (50%) and secondary MF (17%) exhibited loss of 17p13 (leading to TP53 deletion) versus none with PV/ET, findings clearly indicating the association of 17p deletions with advanced MPN [53].

A retrospective analysis of 649 patients with MF demonstrated that baseline bone marrow or peripheral blood blasts ≥ 5% (P = 0.02 and 0.01, respectively) and unfavorable karyotype (−5, −7, del17p, and/or complex karyotype, P = 0.04) were independent risk factors for leukemic transformation [55]. The subgroup that had one or both risk factors (80 patients, 12% of the cohort) had a median OS of 10 months and a considerably higher risk of leukemic transformation versus patients without either one of the two adverse factors; the one-year LFS was 82% compared to 98%, respectively (P = 0.001), at one year. During a 12-month follow-up, 13% (n = 10) of 80 patients who had either one of the two risk factors progressed to AML compared to 2% (n = 10) of 568 patients without the previous characteristics [55]. Acquisition of an abnormal karyotype has also been associated with higher incidence of disease progression in PV [16, 56]. In studies conducted at the MD Anderson Cancer Center, an abnormal karyotype was detected in about 14–20% of the patients with PV; in contrast, the frequency increased to 90% in the AP/BP patients, of whom 83% had complex karyotype [56]. In this study, another factor that was associated with higher risk of leukemia transformation, and later was confirmed by others, was the presence of moderate to severe bone marrow fibrosis and dysplasia [8, 16]. Expectedly, higher grades of bone marrow fibrosis correlated with advanced disease and adverse prognosis in PMF [57, 58] and have been incorporated in the MIPSS70-plus version 2.0 model [43].

Dobrowolski et al. demonstrated that correlation of leukemic transformation risk and persistent basophilia in PMF in the fibrotic stage was statistically significant [59]. Other groups showed that monocytosis was a strong adverse indicator of progression to the accelerated phase and inferior survival in patients with PMF [60, 61, 62]. A recent study conducted in 454 patients with PMF revealed that monocytosis was associated with leukemic transformation: an absolute monocyte count ≥ 1×109/L and > 3×109/L increased the hazard risk by two- and four-fold, respectively [62]. Furthermore, an association was found between monocytosis and older age, increased circulating blasts, higher DIPSS, and DIPSS-plus scores, and the presence of ASXL1 [62].

Radioactive phosphorus (32P), pipobroman and busulphan, used in PV treatment, have been associated with increased risk of leukemogenesis [7, 63]. In a retrospective analysis of 83 patients with MPN followed for nearly 8 years, 14%, 30% and 4% of the patients who were treated with hydroxyurea monotherapy, hydroxyurea and busulphan, or observed only developed AML/myelodysplastic syndrome (MDS).[64]. In a national study on a large Swedish cohort of patients with MPN (n = 11,039), transformation to AML/MDS (n = 162) significantly increased with high doses of 32P and alkylating agents (and the risk increased 2.9-fold with ≥ 2 treatments), whereas there was no association between the use of hydroxyurea and elevated risk of AML/MDS [65]. Researchers from Mayo Clinic also analyzed the effect of specific treatments on LFS and concluded that erythropoiesis-stimulating agents and danazol were both associated with significantly shorter LFS on univariate analysis [34]; however, these findings were not confirmed in subsequent studies by other researchers. Other proposed factors contributing to disease progression to MF-AP and MF-BP include high levels of circulating pro-inflammatory mediators, such as interleukin-8 and C-reactive protein (> 7 mg/L) [66, 67].

Molecular signatures of MPN-AP/BP

Approximately 90% of the patients with PMF harbor one of the three “driver” mutations. Janus kinase 2 (JAK2) V617F, calreticulin (CALR) exon 9 indels and myeloproliferative leukemia virus oncogene (MPL) W515L/K were detected in about 50–60%, 20–30% and 5–10% of the patients, respectively [68]. Nearly 10% of PMF patients have “triple negative” molecular status regarding “driver” mutations, which is a high-risk profile and has been associated with significantly worse LFS [69, 70]. In a retrospective analysis of 1581 patients with MPNs, including 428 with PMF, patients with “triple negative” status for the three driver mutations (JAK2, CALR, MPL) had significantly worse LFS compared to harboring JAK2 (HR 0.4; 95% CI, 0.2–0.7), CALR (HR 0.1; 95% CI, 0.05–0.35), and MPL (HR 0.3; 95%CI, 0.1–0.9) mutants [71]. Accordingly, OS was considerably longer in patients harboring CALR versus “triple-negative” (HR, 5.1; 95% CI) and JAK2 mutants (HR, 2.5; 95% CI) [71]. An international study of 570 patients with PMF showed that type 1 CALR mutations conferred the longest survival in the absence of ASXL1 (median 10.4 years); conversely, the shortest survival was detected in mutated-ASLX1 and unmutated-CALR cases [72]. A favorable correlation has been identified between CALR type 1 mutations and longer survival [70]. Unmutated- CALR (type 1) status is one of the factors incorporated in MIPSS70 [43] (1 point), MIPSS70-plus version 2.0 (2 points) [50], GIPSS (1 point) [51], and MYSEC-PM (2 points) [44].

A retrospective study assessing a gene panel including 27 MPN-related mutations in 182 patients with PMF showed that 81% (n = 147 patients) harbored other mutations/variants apart from JAK2/CALR/MPL [69]. The most frequent mutations were ASXL1 (36%), SRSF2 (18%), TET2 (18%), and U2AF1 (16%) [69]. SRSF2, RUNX1, CEBPA and SH2B3 were associated with higher risk of leukemic transformation (HR was 4.9, 8.7, 5.4, and 5.8, respectively) [69]. In a larger analysis of 879 MF patients comprising 2 cohorts (European study group, n = 483, and validation group from Mayo Clinic, n = 396), the most common “non-driver” mutations were ASXL1 (21.7%), TET2 (9.7%), SRSF2 (8.5%), DNMT3A (5.7%), EZH2 (5.1%), CBL (4.4%) and IDH1/2 (2.6%) [73]. The study led to the conclusion that ASXL1, IDH1/2, and SRSF2 were independently correlated with leukemogenesis; ASXL1, EZH2 and SRSF2 were independent predictors of shortened survival; and ASXL1, SRSF2, EZH2 and IDH1/2 were identified as high molecular risk (HMR) mutations [73]. The cumulative risk of transformation to MPN-BP was considerably higher, and LFS was significantly lower in the HMR group as compared to the low-risk group (no HMR mutations) [73]. The aforementioned HMR mutations were incorporated in MIPSS70 [43], and U2AF1Q157 mutations were added to the MIPSS70-plus version 2.0 [50] after the negative prognostic value of U2AF1Q157 mutations in PMF was demonstrated [74]. Another study illustrated the association between significantly shorter LFS and the presence of ≥2 HMR mutations (HR 6.2) [71], a criterion that was incorporated in the MIPSS, MIPSS70-plus, MIPSS70-plus version 2.0, and GIPSS models [38, 68, 75].

Moreover, an extended exome-sequencing analysis of 69 genes in more than 2,000 MPN patients demonstrated that mutations in the RAS-pathway, and spliceosome and epigenetic regulators had a strong association with MF-AP [14]; and patients harboring NRAS/KRAS mutations had a higher incidence of progression to AML [76]. A recent study on 1306 patients with PMF corroborated association of ASXL1 (HR = 2.0), SRSF2 (HR 3.0) and IDH1 (HR = 4.3) with the highest transformation risk [35]. A few studies showed that TP53 mutations were strongly associated with progression to BP for all classic MPN subtypes [16, 54, 75, 77]. Accordingly, Marcellino et al. demonstrated that advanced forms of MPNs are accompanied by chromosomal abnormalities that lead to dysregulation of TP53 [53]. TP53 mutations co-occurred with JAK2V617F in patients with MPN-BP and increased during leukemic transformation [78]. Lundberg et al. demonstrated the strong association of TP53 loss with leukemogenesis as opposed to the presence of heterozygous TP53 at low allelic burden during MPN-CP [79]. In another recent study, multivariate analysis showed that RUNX1 mutations correlated with shorter survival in a cohort of 248 patients with MPN-BP [80].

The mutational profiles of MPN-AP and MPN-BP are similar given that AP precedes BP in the majority of the patients [10]. However, the mutational landscape of MPN-AP/BP has marked differences from that of both de novo AML and MPN-CP [17, 78]. For example, IDH1/2 and TP53 mutations have a higher frequency in MPN-BP versus MPN-CP [16, 78, 85]; and mutations in FLT3 and DNMT3A predominate in do novo AML, whereas both MPN-AP and MPN-BP are more frequently associated with mutated ASXL1, IDH1, IDH2, TET2, SRSF2 and TP53 [16, 78, 80, 81, 82, 83, 84, 85, 86]. In another deep sequencing study, more than 50% of the PV patients harbored the adverse mutations ASXL1, IDH2, and SRSF2, whereas in ET patients, IDH2, TP53, SH2B3, SF3B1, U2AF1, and EZH2 occurred more frequently, besides JAK2/CALR/MPL [87].

Canadian researchers performed targeted gene sequencing of patients with MPN-AP/BP (N=122, 14 in AP and 108 in BP) and correlated the mutational profiles with clinical outcomes [88]. Of 122 specimens that were analyzed, 34 (28%) patients had PMF, 40 (33%) had PV/post-PV MF, and 38 (31%) had ET/post-ET MF. The most common mutations were: JAK2V617F (55%), ASXL1 (30%), TET2 (25%), SRSF2 (22%), RUNX1 (20%), TP53 (17%), DNMT3A (16%), IDH1 and IDH2 (13% each) (Fig. 1) [88]. Twenty six percent of the patients were “triple-negative” for “driver” mutations, and had the shortest latency period between MPN-CP and diagnosis of MPN-AP/BP (2.8 years), whereas patients harboring CALR had the longest time (10.9 years) [88]. Shorter OS was associated with mutated TP53 (HR = 1.89, P = 0.03) and four or more mutations (P = 0.02) [88]. OS was significantly reduced and the risk to leukemic transformation increased when two or more somatic mutations (e.g., epigenetic regulators) were detected in MPN patients [79]. Increased peripheral blasts, low serum albumin and more than three cytogenetic abnormalities were correlated with shorter survival [10, 88]. Patients in AP/BP that harbored ASXL1 were more likely to have threefold higher WBC counts (P = 0.04) [88], and patients with ASXL1-mutated MPN had significantly lower hemoglobin levels than the wild-type subgroup [79]. Furthermore, acquisition of ASXL1 along with JAK2V617F cooperated in accelerating progression to MF in PV patients [89]. In a cohort comprising 42 patients with high-risk MF and MPN-AP/BP, the most frequent co-occurring mutations were ASXL1 and JAK2V617F [90].

Figure 1.

Figure 1.

Mutational landscape (excluding “driver” mutations) in patients with MPN-AP/BP (N=122, 14 in AP and 108 in BP). Data shown in the plot were reported by McNamara et al. [88].

Recently, our group showed that concomitant presence of JAK2V617F and JAK2 “variants” was associated with increased risk of leukemic transformation in patients with MF [91]. Analysis of the twenty-eight gene panels of bone marrow specimens from 2154 patients with MPN (n = 608), AML/MPN (n = 59) and de novo AML (n = 1487) revealed JAK2 “variants” (non-V617F/non-exon 12 JAK2 variants), in 114 patients [91]. Among all tested patients, 23 (20.2%) had a co-occurring JAK2V617F mutation. The three most frequent variants were N1108S, L393V, and R1063H across the three subgroups. JAK2 “variants” were considerably more frequent in the MPN-BP subgroup (~15 %) versus the MPN (~5%) and de novo AML (5.2%) subgroups. Patients harboring both JAK2V617F and a JAK2 “variant” had an increased cumulative risk to progress from MPN to AML compared to those with JAK2 V617F only (P = 0.003) [91]. Moreover, further mutational analysis demonstrated that TP53 was found in ~51% of the patients harboring a JAK2 “variant” [91].

Therapeutic Modalities

As noted in the previous sections, MPN-AP usually precedes MPN-BP [10], and both phases are inherently associated with a poor prognosis. Historically, the median OS for MPN-BP has been in the approximate range of 3–9 months [7, 19, 24, 92]. Treatment options and management for MPN-AP/BP are largely similar, and at present, there is no standard approved therapy [25, 92]. At our institution, we usually treat MPN-AP/BP patients with hypomethylating agents (decitabine, azacitidine) in combination with ruxolitinib - ruxolitinib is typically added at the dose of 10 mg twice daily to control splenomegaly and MPN symptoms to the degree possible, and this dose/schedule is maintained despite lowering of the blood count as expected from hypomethylating agents-, which has become standard practice as it appears to provide at least equal benefit (but lower toxicity) to intensive chemotherapy. In the cases of MPN-BP patients with IDH1 and IDH2 mutations, we administer combinations with ivosidenib or enasidenib or enroll them in clinical trials. In rare instances of FLT3-mutated MPN-BP, gilteritinib is included in the regimen. Patients who can achieve complete or partial remission or return to a second chronic phase are referred for allo-HSCT. Our experience and that of other investigators have shown that early detection of targetable mutations (e.g., IDH1, IDH2) and timely initiation of the appropriate treatment play an important role in efficacious treatment of patients with advanced-phase MPN. In patients who are fit for transplantation and do not harbor targetable mutations, we recommend allo-HSCT after intensive chemotherapy and achievement of CR or return to MPN-CP [94].

Allogeneic Hematopoietic Stem Cell Transplant

The importance of accurate risk stratification of MF patients [68, 75, 93, 94] and timely referral of MF patients for allo-HSCT evaluation in all categories except for low-risk were demonstrated in a recent study [95]. Allo-HSCT is recommended in patients with intermediate-2 or high risk MF according to the IPSS, DIPSS, or DIPSS-plus criteria or intermediate-1 disease in the presence of unfavorable karyotype according to the DIPSS-plus criteria [42], high molecular risk mutations (e.g., triple-negative MPN driver mutations, mutated ASXL1, IDH1/2), transfusion-dependent anemia, and peripheral blasts > 2% [95]. The National Cancer Comprehensive Network (NCCN) recommends consideration of blasts in the range 10–19% in the peripheral or bone marrow for MPN-AP diagnosis [96]. However, a few recent studies showed the association of blasts ≥ 3% with leukemic transformation [35] and ≥ 5% with leukemic transformation and inferior survival [3133]. As detailed in the section on risk factors of advanced phase MPN, Masarova et al. demonstrated the prognostic value of bone marrow and peripheral blood blasts ≥5% in patients with MF; the findings of the former studies clearly underscore that blasts ≥5% should be taken into consideration along with unfavorable karyotypes and high molecular risk mutations in patients with MF [30, 31].

The myelofibrosis transplant scoring system (MTSS) was recently developed to predict the outcome of patients with primary and secondary MF after allo-HSCT on the basis of pre-transplant factors [93]. Patients who can sustain intensive chemotherapy and achieve complete or partial remission or return to a second chronic phase have the option of consolidative allo-HSCT [21, 22, 24, 28, 29]. Up to the present, standardized criteria for response assessment to treatment of MPN-AP/BP have not been well established. Several studies have shown that complete remission (CR) prior to transplant was the most critical factor affecting survival in patients with MF that had transformed to AML [21, 22, 28, 29]. For example, Alchalby et al. showed that approximately three times more patients who underwent allo-HSCT in CR (69%) were alive compared to patients with residual disease (22%) after 3 years [22]. In general, there is no consensus about the depth of the response but typically, less than 5% blasts in the bone marrow and absence of peripheral blasts is considered a morphologic leukemia-free state [97]. To assess the response of patients with MPN-AP/BP to treatment, clinical investigators have been applying the European LeukemiaNet (ELN) criteria for AML published in 2017 [97], the proposed criteria for MPN-BP in 2012 [98] or both. The traditional ELN criteria assess elevation of the blast counts in the bone marrow [97], which is a hallmark of the disease. However, in advanced-phase MPN, a discordance can be found in the blast count of the bone marrow versus the peripheral blood due to extramedullary hematopoiesis. The proposed criteria for MPN-AP/BP assess both the AML and MPN components of the disease (cytogenetic/molecular responses and bone marrow fibrosis of the underlying MPN and other symptoms, such as splenomegaly) [98].

A retrospective study of 39 patients who had MPN-AP/BP and underwent allo-HSCT manifested that approximately one third of the patients achieved long-term survival, and the OS rate was ~29% at 2 years [21]. In a recent multicenter retrospective study, the OS of 551 MF patients who underwent allo-HSCT versus 1,377 non-HSCT patients were compared; allo-HSCT conferred long OS benefit to the former cohort, primarily in patients who had intermediate-1 and higher DIPSS risk scores and had been treated for more than one year [21,95]. Conversely, for the first year, the OS was longer in the non-HSCT cohort that had low-risk DIPSS score and received other treatments due to mortality related to transplant (frequently caused by graft-versus-host disease) [95]. Treatment of six patients in MPN-BP with a combination of intensive chemotherapy ([7 + 3] type; cytarabine with idarubicin or daunorubicin) and continuous ruxolitinib resulted in four responders and three patients who were able to undergo allo-HSCT [99]. Induction therapy can result in remission but the responses are not durable unless the treatment is followed by allo-HSCT [19, 23, 28, 88, 100]. Many patients are precluded from allo-HSCT due to age, poor performance status, other comorbidities, as well as lack of a suitable donor or inability to achieve a reasonable response [21, 25, 92, 95].

Hypomethylating Agents as Monotherapies and in Combination with Ruxolitinib

Hypomethylating agents or inhibitors of DNA methyltransferases, such as azacitidine and decitabine, initially as monotherapies or in combination with ruxolitinib (JAK1/2 inhibitor) have shown synergistic preclinical [78] and clinical activity [7] with comparable response rates to intensive chemotherapy but lower toxicity in patients with MPN-AP/BP [101]. In a previous study, 54 patients with MPN who progressed to MPN-BP (n = 26) or myelodysplastic syndrome (n = 28) were treated with azacitidine; the entire cohort had a median OS of 11 months, and the CR rate for the MPN-BP subgroup was 12% [102]. A retrospective analysis of 19 MPN patients (developing MPN-BP) that were treated with azacitidine showed that five patients (26.3%) achieved CR, one had a partial response, four had stable disease, and three displayed hematological improvement; since the time of BP evolution, the median cumulative survival was 9.9 months [103]. Andriani et al. conducted another recent retrospective study in 39 patients who were in MPN-AP/BP and were treated with azacitidine in the frontline setting at 10 hematologic centers in Italy [104]. Hematologic responses were noted in 24 patients: 8 (20.5%) and 7 (17.9%) achieved complete and partial responses, respectively, whereas hematologic improvement was noted in 9 (23.1%); 4 responders underwent allo-HSCT. The overall response rate was 61.5%, the entire cohort had a median OS of 13.5 months; and the median OS for responders was about 4 months longer (17.6 months) [104]. Another phase 1b study evaluated the safety and maximum tolerated dose of ruxolitinib in combination with azacitidine in a small cohort of patients comprising seven with MPN-AP (including MDS) and seven with MPN-BP. Of the 6 evaluable patients, one achieved complete remission, and one reached partial remission (MPN-BP patient), whereas two had stable disease [105]. A retrospective, multicenter study of 122 MPN patients (from eight French hospitals) who progressed to MPN-AP/BP demonstrated that the MPN-AP subgroup treated with azacitidine exhibited the longest OS in the cohort (13.6 months) [24]. In a phase 2 investigation of ruxolitinib and azacitdiine at our institution (NCT01787487), among 46 enrolled MF patients, 3 had MF-AP with 10% blasts or more; and one patient achieved clinical improvement in the spleen and Total Symptom Score (per International Working Group-Myeloproliferative Neoplasms Research and Treatment criteria) with a blast reduction to 2%, lasting for 28 months [106].

In 2015, we analyzed the efficacy of decitabine in 13 patients with MPN-AP at our institution [107]. Sixty two percent (8/13) of the patients with MPN-AP benefited from the treatment; the median duration of response was 6.5 months with a median OS of 9.7 months [107]. In another phase 2 study at our institution, 18 patients with MPN-BP were treated with ruxolitinib monotherapy, and 3/18 patients obtained CR and complete remission with incomplete hematological recovery (CRi) [108]. In a phase 1/2 study (NCT02257138), we treated 12 patients who had MPN-BP with a combination of ruxolitinib and decitabine. We determined that 50 mg ruxolitinib twice daily with the standard regimen for decitabine (20 mg/m2 for 5 days on a 4–6 week schedule) were tolerable and safe [109, 110] (Table 3). In the phase 2 study, among 18 patients with MPN-BP, treated with the recommended doses of ruxolitinib and decitabine, 61% were responders (11% CR + 50% CRi). Median OS was 8.4 months (range, 0.4 – 42.6 months) for all the patients and 9.4 months (range 3.1–42.6) for responders [110]. A multicenter phase 1 study (NCT02076191) that was designed and monitored by the Myeloproliferative Neoplasms Research Consortium, also assessed the combination of ruxolitinib and decitabine, in patients with MPN-AP/BP (21 patients, n = 8 in AP and n = 13 in BP) [111] (Table 3). The response rate and median OS were 66.7% and 16 months in the MPN-AP subgroup; and 45.5% and 7.2 months in the MPN-BP subgroup [111]. The optimum determined regimen was 10 mg ruxolitinib twice daily (after one cycle at 25 mg twice a day), and 20 mg/m2 per day of decitabine for 5 days every 4 weeks [111]. Mascarenhas and colleagues [112] published the results of the multi-center phase 2 trial evaluating the aforementioned regimen during the production of this article.

Table 3.

Selected clinical Trials on treatments for MPN-AP/BP.

Treatment Regimen Disease Clinical Trial
Ruxolitinib + decitabine MPN-AP/BP NCT02076191 (phases 1 and 2) #
Ruxolitinib + decitabine MPN-BP NCT02257138 (phase 1/2)
Ruxolitinib or fedratinib + decitabine MPN-AP/BP NCT04282187 (phase 2)
Ruxolitinib + enasidenib IDH2-mutated MPN-BP§ NCT04281498 (phase 2)
Ivosidenib + venetoclax +/− azacitidine IDH1-mutated
MPN-BP
NCT03471260 (phase 1b/2)
Ivosidenib + azacitidine IDH1-mutated MPN-BP NCT03173248 (phase 3; AGILE trial)
Guatecitabine (SGI-110)* MPN-AP NCT03075826 (phase 2)
Ivosidenib or Enasidenib + induction and consolidation therapy IDH1- or IDH2-mutated MPN-BP NCT02632708 (phase 1)
Gilteritinib FLT3-mutated MPN-BP (rare) NCT03836209 (phase 2)
KRT-232 + decitabine or low dose cytarabine MPN-BP‡, ** NCT04113616 (phase 1b/2)

The trials were conducted exclusively for patients in MPN-AP and/or MPN-BP.

Enrollment of patients with MPN-AP and or MPN-BP was allowed in the trial.

#

The phase 1 and 2 studies were conducted by the MPN-Research Consortium.

§

The MPN-Research Consortium is conducting the study. Patients with IDH2-mutated MPN-AP/BP or MF-CP (4–9% circulating blasts) are enrolled.

*

Decitabine –P– guanosine

**

Patients harboring TP53 mutation are excluded from this study.

Recently, Zhou et al. reported a retrospective study of 42 patients, comprising 14 with MPN-AP, 16 with MPN-BP and 12 with HR MF, who were treated with decitabine monotherapy or in combination with ruxolitinib [90]. Among the patients in AP, seven received decitabine monotherapy and six were treated with ruxolitinib/decitabine combination. Over a median follow-up of 12.4 months (range, 2.1–48.8), the median OS of HR MF and MPN-AP patients was not reached; however, only two and one patients were alive at 60 months, respectively. After initiation of decitabine therapy, the probability of complete or partial (50%) reduction in peripheral blasts was 54.6% in the entire cohort [90]. The investigators suggested that early initiation of decitabine or ruxolitinib/decitabine treatment in the accelerated phase conferred benefit to the patients; and that the combination of ruxolitinib with decitabine was superior to decitabine monotherapy (the integrated cohort of HR MF, MPN-AP and MPN-BP patients had a median OS of 21.0 months with the combination versus 12.9 months with decitabine only) [90].

Another study of 180 patients with MPN-AP/BP showed no difference in the outcomes with intensive chemotherapy compared to non-intensive therapy (hypomethylating agents, low-dose cytarabine or treatment in a clinical study); the median OS of the entire group was 5.8 months. Intensive chemotherapy treatment was advantageous only in patients with less than 4 mutations and wild-type TP53 (median OS of 8.1 months for the latter group) [88].

Treatments with Targeted Inhibitors

In preclinical studies on MPN-BP cell lines, significant synergistic activity was noted between ruxolitinib in combination with bromodomain and extraterminal (BET) protein inhibitors, targeting epigenetic proteins [113]. The former preclinical studies provide grounds to conduct clinical studies assessing BET inhibitors/ruxolitinib in patients with MPN-AP/BP. In the ongoing global phase 2 MANIFEST trial (NCT02158858), patients with advanced MF-CP who had either suboptimal response to ruxolitinib treatment or were JAK-inhibitor naïve [114] had notable clinical responses (spleen volume reduction and improvement in anemia, BM fibrosis, total symptom score, and transfusion-dependence) after therapy with CPI-0610 (a selective BET inhibitor) alone or in combination with ruxolitinib [115].

Co-treatment with ruxolitinib and BET proteolysis-targeting chimeras (BET-PROTACs, such as ARV-825) has also exhibited strong synergism against MPN-BP cells, and ARV-825 induced apoptosis in ruxolitinib-resistant MPN-BP cells in vitro [116, 117]. Accordingly, in another recent preclinical study, multi-targeted inhibition of the β-catenin-TCF7L2-JMJD6-c-Myc axis, through co-treatment with the BET-PROTAC ARV-771 and the inhibitor BC2059, overcame resistance of patient-derived MPN-BP blasts to BET inhibitors and improved survival of mice engrafted with BET inhibitor-resistant MPN-BP cells [118]. Inhibitors of the heat shock protein 90 (HSP90) degrade JAK2, and concomitant BET and HSP90 inhibition had synergistic lethal activity against MPN-BP cells that were resistant to ruxolitinib [113, 119].

Notwithstanding limited advancements, the overall clinical experience of treating patients with MPN-AP/BP clearly shows that a major unmet need still remains. As new targeted therapies are transforming the field of AML [18], molecularly targeted monotherapies or combinations with other agents engender promise in MPN-AP/BP [120]. Subgroups of patients harboring targetable mutations that are more frequent in MPN-AP/BP compared to the chronic phase [121] (e.g., IDH1/2 occur at ~ 20% frequency in MPN-BP versus ~ 4% in PMF [85]) may benefit from treatments with the corresponding inhibitors. For example, IDH1- and IDH2-mutated MPN-AP/BP can be treated with the oral IDH1 and IDH2 inhibitors ivosidenib and enasidenib, respectively, and their combinations with other drugs. Preclinical studies demonstrated synergism between enasidenib and ruxolitinib in double-mutant IDH2/JAK2V617F MPN and MPN-BP patient-derived cells [122]. Notably, Patel et al. and Chifotides et al. recently conducted two retrospective studies in small cohorts of patients with IDH2-mutated MPN-AP/BP [123] and IDH1/2-mutated MPN-BP [124], respectively. In the aforementioned studies, the patients were treated with enasidenib monotherapy (and azacitidine in one case) [123] and IDH1/2-inhibitor monotherapies or IDH1/2-inhibitor-based combinations with other drugs (hypomethylating agents, ruxolitinib, venetoclax, or intensive chemotherapy) [124], respectively; the patients exhibited durable clinical responses with acceptable tolerability and survivals that compared favorably with historical reports. A phase 2 clinical trial evaluating the combination of enasidenib and ruxolitinib in patients with IDH2-mutant MPN-AP/BP and MF-CP (4–9% circulating blasts) is currently underway (NCT04281498; Table 3). Moreover, in another ongoing phase 1 clinical trial (NCT02074839), 6/11 patients with IDH1-mutated MPN-BP and co-occurring JAK2V617F at baseline, were treated with ivosidenib monotherapy in the relapsed or refractory setting, and achieved CR [125].

Venetoclax monotherapy has exhibited limited efficacy in myeloid malignancies but combination strategies with other therapeutics are very promising in AML and are becoming the standard of care in patients aged > 75 years [126, 127, 128]. However, recent publications in a small number of patients indicate that venetoclax may not be an effective treatment in MPN-AP/BP patients (because Bcl-xl is elevated in MPN cells [129] and sensitivity to venetoclax correlates negatively with Bcl-xl levels) except for patients harboring IDH1/2 mutations due to the production of R-2-hydroxyglutarate. Tremblay et al. [130] and Gangat et al. [131] conducted retrospective studies on small cohorts of MPN-BP patients treated with the combination of venetoclax and hypomethylating agents (decitabine or azacitidine): the median overall survivals were not extended compared to the historically poor survivals reported for MPN-BP. Notwithstanding the short overall survivals, in both studies, a few patients achieved CR and CRi, and underwent allo-HSCT. Notably, in a few recent preclinical and clinical studies, the investigators reported an increased sensitivity of IDH1/2-mutated human AML cells and prolonged survival in IDH1/2-mutated AML patients with venetoclax [132, 133, 134, 135]. In the report by Gangat et al., the responses of 2 patients with IDH2-mutated BP-MPN revealed an analogous sensitivity to venetoclax-based combination treatment [131]. In another recent report, a patient with IDH2-mutated MPN-BP responded to venetoclax monotherapy [136]. A phase 1/2 trial assessing ivosidenib/venetoclax with or without azacitidine in IDH1-mutated patients with advanced malignancies, including MPN-AP/BP, is underway (NCT03471260; Table 3) [137]. Analysis of interim data from this study evaluating IDH1-mutated patients who were treated with ivosedinib/venetoclax showed efficacy of the combination and promising response rates [138]. Considering that MPN-AP/BP patients have been largely excluded from clinical trials heretofore, in this era, this population will have the option to enroll in rationally designed clinical studies evaluating novel promising regimens.

A phase 1/2 clinical study of KRT-232, an HDM2 inhibitor, in combination with either decitabine or low dose cytarabine is underway for patients with AML, including MPN-BP (NCT04113616; Table 3). In this study, patients with TP53 mutations are excluded due to lack of activity of HDM2 inhibitors on mutated TP53. Approximately 20% of the patients with MPN-AP/BP harbor TP53 [79]. Mutant TP53 AML [139] and TP53 MPN-AP/BP [88, 140] patients have a poor prognosis and do not respond well to chemotherapy treatment. Novel agents are being developed for patients with mutated TP53. Notably, preliminary results with anti-programmed death ligand 1 (anti-PD-L1) agents have not shown clinically relevant results in patients with MPN-AP/BP [141].

Conclusions

In summary, MPN-AP is an aggressive disease that is considered a continuous transitional phase between chronic phase MPN and MPN-BP, and portends poor prognosis. Clinical practice and the limited number of studies that have been conducted heretofore demonstrated that advanced clinical features, such as rising blast counts (≥ 3–5%), acquisition of HMR mutations (ASXL1, IDH1/2, SRSF2, EZH2, U2AF1Q157, and TP53) and complex karyotype or high-risk cytogenetics (e.g., 17p deletion and gain of 1q), type 1 CALR-unmutated status, severe anemia, and high-grade bone marrow fibrosis likely herald impending leukemic transformation. Regular clinical follow-up and evaluation of factors indicating disease progression in high-risk patients is recommended, aiming to implement early therapeutic interventions and direct the patients to allo-HSCT. In this respect, the prognostic models that were developed to assess the risk score of MPN patients can also predict the risk of transformation, and have evolved significantly in the last 10+ years. Hindrance of MPN progression to the AP/BP and timely referral of eligible patients for allo-HSCT are desirable goals whenever possible. Treatment with hypomethylating agents in combination with ruxolitinib provides an option for MPN-AP patients, instead of intensive chemotherapy, followed by an allo-HSCT. As more targeted therapies emerge for de novo AML, significant advancements are made in the molecular landscape of MPN-AP/BP; and patients with MPN-AP and actionable mutations will have the opportunity to be treated with new modalities and personalized schemes, e.g., IDH1/2 inhibitors alone or in combination with other drugs. In this respect, wide implementation of next generation sequencing is important. Additional novel inhibitors that are in clinical development, e.g., BET inhibitors, provide promising venues that merit further assessment.

Acknowledgements

This work was supported, in part, by the MD Anderson Cancer Center Support Grant P30 CA016672 from the National Cancer Institute (National Institutes of Health).

Disclosure Statement. P.B. has received research support from Incyte Corporation, Celgene (BMS), CTI Biopharma, Kartos Therapeutics, Blueprint Medicines, Constellation Pharmaceuticals, NS Pharma, Promedior, Astellas, and Pfizer. P.B. has received honoraria from Incyte Corporation, Celgene, CTI Biopharma, Kartos Therapeutics, and Blueprint Medicines. S.V. has received research support from Incyte Corporation, Roche, Celgene (BMS), Gilead, Promedior, CTI Biopharma Corporation, Genetech, Blueprint Medicines Corporation, NS Pharma, Novartis, Sierra Oncology, Pharma Essentia, Astra Zeneca, Italfarmaco, Kartos Therapeutics, Prelude Therapeutics, Protagonist Therapeutics, AbbVie, Constellation Pharmaceuticals, and Telios Pharmaceuticals. S.V. has received consultancy fees from Constellation Pharmaceuticals, Sierra Oncology, Incyte Corporation, Novartis, and Celgene. The remaining authors have no conflicts of interest to disclose.

References

  • 1.Bose P, Masarova L, Amin HM, Verstovsek S. Philadelphia chromosome-negative myeloproliferative neoplasms (Chapter 6) In: Kantarjian HM, Wolff RA, Rieber AG, editors. The MD Anderson Manual of Medical Oncology. 4th edition, China: McGraw-Hill Education; 2021, in press. [Google Scholar]
  • 2.Marcellino BK, Verstovsek S, Mascarenhas J. The myelodepletive phenotype in myelofibrosis: Clinical relevance and therapeutic implications. Clin Lymph Myeloma & Leukemia. 2020;20(7):415–421. [DOI] [PubMed] [Google Scholar]
  • 3.Tefferi A, Cervantes F, Mesa R, Passamonti F, Verstovsek S, Vannucchi AM, et al. Revised response criteria for myelofibrosis: International Working Group-Myeloproliferative Neoplasms Research and Treatment (IWG-MRT) and European LeukemiaNet (ELN) consensus report. Blood. 2013;122(8):1395–1398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Quintás-Cardama A, Verstovsek S. Molecular pathways: JAK/STAT pathway: mutations, inhibitors, and resistance. Clin Cancer Res. 2013;19(8):1933–1940. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Schieber M, Crispino JD, Stein B. Myelofibrosis in 2019: Moving beyond JAK2 inhibition. Blood Cancer J. 2019;9:74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Szuber N, Mudireddy M, Nicolosi M, Penna D, Vallapureddy RR, Lasho TL, et al. 3023 Mayo Clinic patients with myeloproliferative neoplasms: risk-stratified comparison of survival and outcomes data among disease subtypes. Mayo Clin Proc. 2019;94(4):599–610. [DOI] [PubMed] [Google Scholar]
  • 7.Yogarajah M, and Tefferi A. Leukemic transformation in myeloproliferative neoplasms: A literature review on risk, characteristics, and outcome. Mayo Clinic Proceedings. 2017;92(7):1118–1128. [DOI] [PubMed] [Google Scholar]
  • 8.Cerquozzi S and 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(11):e366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Abdulkarim K, Girodon F, Johansson P, Maynadié M, Kutti J, Carli P-M, et al. AML transformation in 56 patients with Ph MPD in two well defined populations. Eur J Haematol. 2009;82(2):106–111. [DOI] [PubMed] [Google Scholar]
  • 10.Tam CS, Kantarjian H, Cortes J, Lynn A, Pierce S, Zhou L, et al. Dynamic model for predicting death within 12 months in patients with primary or post-polycythemia vera/essential thrombocythemia myelofibrosis. J Clinical Oncol. 2009;27(33):5587–5593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Mudireddy M, Gangat N, Hanson CA, Ketterling RP, Pardanani A, Tefferi A. Validation of the WHO-defined 20% circulating blasts threshold for diagnosis of leukemic transformation in primary myelofibrosis. Blood Cancer J. 2018;8(6):57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Mesa RA, Verstovsek S, Cervantes F, Barosi G, Reilly JT, Dupriez B, et al. Primary myelofibrosis (PMF), post polycythemia vera myelofibrosis (post-PV MF), post essential thrombocythemia myelofibrosis (post-ET MF), blast phase PMF (PMF-BP): Consensus on terminology by the international working group for myelofibrosis research and treatment (IWG-MRT). Leuk Res. 2007;31(6):737–740. [DOI] [PubMed] [Google Scholar]
  • 13.Arber DA, Orazi A, Hasserjian R, Thiele J, Borowitz MJ, Le Beau MM, et al. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood. 2016; 127(20):2391–2405. [DOI] [PubMed] [Google Scholar]
  • 14.Grinfeld J, Nangalia J, Baxter EJ, Wedge DC, Angelopoulos N, Cantrill R, et al. Classification and Personalized Prognosis in Myeloproliferative Neoplasms. N Engl J Med. 2018;379:1416–1430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Masarova L and Verstovsek S. The evolving understanding of prognosis in post-essential thrombocythemia myelofibrosis and post-polycythemia vera myelofibrosis vs primary myelofibrosis. Clin Adv Hematol Oncol. 2019;17(5):299–307. [PubMed] [Google Scholar]
  • 16.Hidalgo López J, Carballo-Zarate A, Verstovsek S, Wang SA, Hu S, Li S, et al. Bone marrow findings in blast phase polycythemia vera. Ann Hematol. 2018;97:425–434. [DOI] [PubMed] [Google Scholar]
  • 17.Dunbar AJ, Rampal RK, Levine R. Leukemia secondary to myeloproliferative neoplasms. Blood. 2020;136(1):61–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Short N, Konopleva M, Kadia TM, Borthakur G, Ravandi F, DiNardo CD, et al. Advances in the treatment of acute myeloid leukemia: new drugs and new challenges. Cancer Disc. 2020;10(4):506–525. [DOI] [PubMed] [Google Scholar]
  • 19.Palandri F, Breccia M, Tiribelli M, Bonifacio M, Benevolo G, Iurlo A, 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] [PubMed] [Google Scholar]
  • 20.Mascarenhas J, Heaney ML, Najfeld V, Hexner E, Abdel-Wahab O, Rampal R, 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(12):1500–1504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Takagi S, Masuoka K, Uchida N, Kurokawa M, Nakamae H, Imada K, et al. Allogeneic hematopoietic cell transplantation for leukemic transformation preceded by Philadelphia chromosome-negative myeloproliferative neoplasms: A nationwide survey by the Adult Acute Myeloid Leukemia Working Group of the Japan Society for hematopoietic cell transplantation. Biol Blood Marrow Transplant. 2016;22:2208–2213. [DOI] [PubMed] [Google Scholar]
  • 22.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;20:279–287. [DOI] [PubMed] [Google Scholar]
  • 23.Odenike O How I treat the blast phase of Philadelphia chromosome-negative myeloproliferative neoplasms. Blood. 2018;132(22):2339–2350. [DOI] [PubMed] [Google Scholar]
  • 24.Mollard L-M, Chauveau A, Boyer-Perrard F, Douet-Guilbert N, Houot R, Quintin-Roué I, et al. Outcome of Ph-negative myeloproliferative neoplasms transforming to accelerated or leukemic phase. Leukemia & Lymph. 2018;59(12):2812–2820. [DOI] [PubMed] [Google Scholar]
  • 25.Marcellino B, Mascarenhas J. Management of advanced phase myeloproliferative neoplasms. Clin Adv Hematol & Oncol. 2019;17(7):405–411. [PubMed] [Google Scholar]
  • 26.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] [PubMed] [Google Scholar]
  • 27.Mannelli L, Guglielmelli P, Vannucchi AM. Stem cell transplant for the treatment of myelofibrosis. Expert Rev Hematol. 2020;13(4):363–374. [DOI] [PubMed] [Google Scholar]
  • 28.Lancman G, Brunner A, Hoffman R, Mascarenhas J, Hobbs G. Outcomes and predictors of survival in blast phase myeloproliferative neoplasms. Leuk Res. 2018;70:49–55. [DOI] [PubMed] [Google Scholar]
  • 29.Chihara D, Kantarjian HM, Newberry KJ, et al. Survival Outcome of patients with acute myeloid leukemia transformed from myeloproliferative neoplasms. Blood. 2016;128(22):1940.27503501 [Google Scholar]
  • 30.Masarova L, Bose P, Pemmaraju N, Daver NG, Zhou L, Pierce S, et al. Prognostic value of blasts in peripheral blood in myelofibrosis in the ruxolitinib era. Cancer. 2020;126(19):4322–4331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Masarova L, Bose P, Pemmaraju N, Daver N, Zhou L, Pierce S, et al. Clinical significance of bone marrow blast percentage in patients with myelofibrosis and the effect of Ruxolitinib therapy. Clin Lymph Myeloma Leuk. 2021;21(5):318–327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Geyer JT, Margolskee E, Krichevsky SA, Cattaneo D, Boiocchi L, Ronchi P, et al. Disease progression in myeloproliferative neoplasms: comparing patients in accelerated phase with those in chronic phase with increased blasts (<10%) or with other types of disease progression. Haematologica. 2020;105:e221–e224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Masarova L, Bose P, Zahr AA, Cortes J, Kantarjian H, Verstovsek S. Do we need to re-define accelerated phase of myelofibrosis? Correlation between blast percentage in myelofibrosis and outcomes. Clin Lymph Myeloma & Leukemia. 2017;17(Suppl. 2), S352. [Google Scholar]
  • 34.Huang J, Li C-Y, Mesa RA, Wu W, Hanson CA, Pardanani A, et al. Risk factors for leukemic transformation in patients with primary myelofibrosis. Cancer. 2008;112(12):2726–2732. [DOI] [PubMed] [Google Scholar]
  • 35.Vallapureddy RR, Mudireddy M, Penna D, Lasho TL, Finke CM, Hanson CA, et al. Leukemic transformation among 1306 patients with primary myelofibrosis: risk factors and development of a predictive model. Blood Cancer J. 2019;9(2):12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Dupriez B, Morel P, Demory JL, Lai JL, Simon M, Plantier I, et al. Prognostic factors in agnogenic myeloid metaplasia: a report on 195 cases with a new scoring system. Blood. 1996;88(3):1013–1018. [PubMed] [Google Scholar]
  • 37.Passamonti F, Rumi E, Elena C, Arcaini L, Merli M, Pascutto C, et al. Incidence of leukaemia in patients with primary myelofibrosis and RBC–transfusion-dependence. Br J Haematol. 2010;150(6):719–721. [DOI] [PubMed] [Google Scholar]
  • 38.Bose P, and Verstovsek S. The evolution and clinical relevance of prognostic classification systems in myelofibrosis. Cancer. 2016;122(5):681–692. [DOI] [PubMed] [Google Scholar]
  • 39.Cervantes F, Dupriez B, Pereira A, Passamonti F, Reilly JT. Morra E, et al. New prognostic scoring system for primary myelofibrosis based on a study of the International Working Group for Myelofibrosis Research and Treatment. Blood. 2009;113(13):2895–2901. [DOI] [PubMed] [Google Scholar]
  • 40.Passamonti F, Cervantes F, Vannucchi AM, Morra E, Rumi E, Pereira A, et al. A dynamic prognostic model to predict survival in primary myelofibrosis: a study by the IWG-MRT (International Working Group for Myeloproliferative Neoplasms Research and Treatment). Blood. 2010;115(9):1703–1708. [DOI] [PubMed] [Google Scholar]
  • 41.Passamonti F, Cervantes F, Vannucchi AM, Morra E, Rumi E, Cazzola M, et al. Dynamic International Prognostic Scoring System (DIPSS) predicts progression to acute myeloid leukemia in primary myelofibrosis. Blood. 2010;116(15):2857–2858. [DOI] [PubMed] [Google Scholar]
  • 42.Gangat N, Caramazza D, Vaidya R, George G, Begna K, Schwager S, 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 Clinical Oncol. 2011;29(4):392–397. [DOI] [PubMed] [Google Scholar]
  • 43.Guglielmelli P, Lasho TL, Rotunno G, Mudireddy M, Mannarelli C, Nicolosi M, et al. MIPSS70: Mutation-Enhanced International Prognostic Score System for transplantation-age patients with primary myelofibrosis. J Clinical Oncol. 2018;36(4):310–318. [DOI] [PubMed] [Google Scholar]
  • 44.Passamonti F, Giorgino T, Mora B, Guglielmelli P, Rumi E, Maffioli M, et al. A clinical-molecular prognostic model to predict survival in patients with post-polycythemia vera and post-essential thrombocythemia myelofibrosis. Leukemia. 2017;31:2726–2371. [DOI] [PubMed] [Google Scholar]
  • 45.Hussein K, Huang J, Lasho T, Pardanani A, Mesa RA, Williamson CM, et al. Karyotype complements the International Prognostic Scoring System for primary myelofibrosis. Eur J Haematol. 2009;82(4):255–259. [DOI] [PubMed] [Google Scholar]
  • 46.Caramazza D, Begna KH, Gangat N, Vaidya R, Siragusa S, Van Dyke DL, et al. Refined cytogenetic-risk categorization for overall and leukemia-free survival in primary myelofibrosis: a single center study of 433 patients. Leukemia. 2011;25(1):82–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Hussein K, Pardanani AD, Van Dyke DL, Hanson CA, Tefferi A. International Prognostic Scoring System–independent cytogenetic risk categorization in primary myelofibrosis. Blood. 2010;115(3):496–499. [DOI] [PubMed] [Google Scholar]
  • 48.Tefferi A, Nicolosi M, Mudireddy M, Lasho TL, Gangat N, Begna KH, et al. Revised cytogenetic risk stratification in primary myelofibrosis: analysis based on 1002 informative patients. Leukemia. 2018;32(5):1189–1199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Vaidya R, Caramazza D, Begna KH, Gangat N, Van Dyke DL, Hanson CA, Pardanani A, Tefferi A. Monosomal karyotype in primary myelofibrosis is detrimental to both overall and leukemia-free survival. Blood. 2011;117(21):5612–5615. [DOI] [PubMed] [Google Scholar]
  • 50.Tefferi A, Guglielmelli P, Lasho TL, Gangat N, Ketterling RP, Pardanani A, et al. MIPSS70+ Version 2.0: Mutation and Karyotype-Enhanced International Prognostic Scoring System for Primary Myelofibrosis. J Clin Oncol. 2018:36(17):1769–1770. [DOI] [PubMed] [Google Scholar]
  • 51.Tefferi A, Guglielmelli P, Nicolosi M, Mannelli F, Mudireddy M, Bartalucci N. et al. GIPSS: Genetically Inspired Prognostic Scoring System for primary myelofibrosis. Leukemia. 2018;32(7):1631–1642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Masarova L, Bose P, Pemmaraju N, Estrov ZE, Zhou L, Pierce SA, et al. Evaluation of cytogenetic stratifications in myelofibrosis. Blood. 2018;132(suppl.1):1763 (section #634). [Google Scholar]
  • 53.Marcellino BK, Hoffman R, Tripodi J, Lu M, Kosiorek H, Mascarenhas J, et al. Advanced forms of MPNs are accompanied by chromosomal abnormalities that lead to dysregulation of TP53. Blood Adv. 2018;2(24):3581–3589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Milosevic JD, and Kralovics R. Genetic and epigenetic alterations of myeloproliferative disorders. Int. J Hematol. 2013;97:183–197. [DOI] [PubMed] [Google Scholar]
  • 55.Quintás-Cardama A, Kantarjian H, Pierce S, Cortes J, Verstovsek S. Prognostic Model to Identify Patients With Myelofibrosis at the Highest Risk of Transformation to Acute Myeloid Leukemia. Clin Lymphoma Myeloma Leuk. 2013;13(3):315–8.e2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Tang G, Hidalgo López JE, Wang SA, Hu S, Ma J, Pierce S, et al. Characteristics and clinical significance of cytogenetic abnormalities in polycythemia vera. Haematologica. 2017;102(9):1511–1518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Guglielmelli P, Rotunno G, Pascilli A, Rumi E, Rosti V, Delaini F, et al. Prognostic impact of bone marrow fibrosis in primary myelofibrosis. A study of the AGIMM group on 490 patients. Am J Hematol. 2016;91(9):918–922. [DOI] [PubMed] [Google Scholar]
  • 58.Pozdnyakova O, Hasserjian RP, Verstovsek S, Orazi A. Impact of bone marrow pathology on the clinical management of Philadelphia chromosome-negative myeloproliferative neoplasms. Clin Lymph Myeloma & Leukemia. 2015;15(5):253–261. [DOI] [PubMed] [Google Scholar]
  • 59.Dobrowolski J, Pasca S, Teodorescu P, Selicean C, Rus I, Zdrenghea M, et al. Persistent basophilia may suggest an “accelerated phase” in the evolution of CALR-positive primary myelofibrosis toward acute myeloid leukemia. Front Oncol. 2019; 9: 872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Elliott MA, Verstovsek S, Dingli D, Schwager SM, Mesa RA, Li CY, Tefferi A. Monocytosis is an adverse prognostic factor for survival in younger patients with primary myelofibrosis. Leuk Res. 2007;31(11):1503–1509. [DOI] [PubMed] [Google Scholar]
  • 61.Boiocchi L, Espinal-Witter R, Geyer JT, Steinhilber J, Bonzheim I, Knowles DM, et al. Development of monocytosis in patients with primary myelofibrosis indicates an accelerated phase of the disease. Mod Pathol. 2013;26:204–212. [DOI] [PubMed] [Google Scholar]
  • 62.Tefferi A, Shah S, Mudireddy M, Lasho TL, Hanson CA, et al. Monocytosis is a powerful and independent predictor of inferior survival in primary myelofibrosis. Br J Haematol. 2018;183(5):835–838. [DOI] [PubMed] [Google Scholar]
  • 63.Cerquozzi S, and 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:e366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Nielsen I and Hasselbalch HC. Acute leukemia and myelodysplasia in patients with a Philadelphia chromosome negative chronic myeloproliferative disorder treated with hydroxyurea alone or with hydroxyurea after busulphan. Am J Hematol. 2003;74(1):26–31 [DOI] [PubMed] [Google Scholar]
  • 65.Björkholm M, Derolf AR, Hultcrantz M, Kristinsson SY, Ekstrand C, Goldin LR, et al. Treatment-related risk factors for transformation to acute myeloid leukemia and myelodysplastic syndromes in myeloproliferative neoplasms. J Clinical Oncol. 2011;29(17):2410–2415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Barbui T, Carobbio A, Finazzi G, Guglielmelli P, Salmoiraghi S, Rosti V, et al. Elevated C-reactive protein is associated with shortened leukemia-free survival in patients with myelofibrosis. Leukemia. 2013;27(10):2084–2086. [DOI] [PubMed] [Google Scholar]
  • 67.Tefferi A, Vaidya R, Caramazza D, Finke C, Lasho T, Pardanani A. Circulating Interleukin (IL)-8, IL-2R, IL-12, and IL-15 levels are independently prognostic in primary myelofibrosis: A comprehensive cytokine profiling study. J Clinical Oncol. 2011;29(10):1356–1363. [DOI] [PubMed] [Google Scholar]
  • 68.Bose P and Verstovsek S. Mutational profiling in myelofibrosis; implications for management. Inter J Hematol. 2020;111:192–199. [DOI] [PubMed] [Google Scholar]
  • 69.Tefferi A, Lasho TL, Finke CM, Elala Y, Hanson CA, Ketterling RP, et al. Targeted deep sequencing in primary myelofibrosis. Blood Adv. 2016;1(2):105–111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Rumi E, Pietra D, Pascutto C, Guglielmelli P, Martinez-Trillos A, Casetti I, et al. Clinical effect of driver mutations of JAK2, CALR, or MPL in primary myelofibrosis. Blood. 2014;124:1062–1069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Tefferi A, Guglielmelli P, Larson DR, Finke C, Wassie EA, Pieri L, et al. Long-term survival and blast transformation in molecularly annotated essential thrombocythemia, polycythemia vera, and myelofibrosis. Blood. 2014;124(16):2507–2513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Tefferi A, Guglielmelli P, Lasho TL, Rotunno G, Finke C, Mannarelli C, et al. CALR and ASXL1 mutations-based molecular prognostication in primary myelofibrosis: an international study of 570 patients. Leukemia. 2014;28:1494–1500. [DOI] [PubMed] [Google Scholar]
  • 73.Vannucchi AM, Lasho TL, Guglielmelli P, Biamonte F, Pardanani A, Pereira A, et al. Mutations and prognosis in primary myelofibrosis. Leukemia. 2013;27(9):1861–9. [DOI] [PubMed] [Google Scholar]
  • 74.Tefferi A, Finke CM, Lasho TL, Hanson CA, Rhett PK, Gangat N, Pardanani A. U2AF1 mutation types in primary myelofibrosis: phenotypic and prognostic distinctions. Leukemia. 2018;32:2274–2278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Tefferi A, Guglielmelli P, Pardanani A, Vannucchi AM. Myelofibrosis treatment algorithm. Blood Cancer J. 2018;8:72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Santos FPS, Getta B, Masarova L, Famulare C, Schulman J, Datoguia TS, et al. Prognostic impact of RAS-pathway mutations in patients with myelofibrosis. Leukemia. 2020;34(3):799–810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Harutyunyan A, Klampfl T, Cazzola M, Kralovics R. p53 lesions in leukemic transformation. N Engl J Med. 2011;364(5):488–490. [DOI] [PubMed] [Google Scholar]
  • 78.Rampal R, Ahn J, Abdel-Wahab O, Nahas M, Wang K, Lipson D, et al. Genomic and functional analysis of leukemic transformation of myeloproliferative neoplasms. Proc Natl Acad Sci., USA. 2014;111:E5401–E5410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Lundberg P, Karow A, Nienhold R, Looser R, Hao-Shen H, Nissen H, et al. Clonal evolution and clinical correlates of somatic mutations in myeloproliferative neoplasms. Blood. 2014;123(14):2220–2228. [DOI] [PubMed] [Google Scholar]
  • 80.Lasho TL, Mudireddy M, Finke CM, Hanson CA, Ketterling RP, Szuber N, et al. Targeted next-generation sequencing in blast phase myeloproliferative neoplasms. Blood Adv. 2018;2(4):370–380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Rampal R and Mascarenhas J Pathogenesis and management of acute myeloid leukemia that has evolved from a myeloproliferative neoplasm. Curr Opin Hematol. 2014;21:65–71. [DOI] [PubMed] [Google Scholar]
  • 82.Abdel-Wahab O, Manshouri T, Patel J, Harris K, Yao J, Hedvat C, et al. Genetic analysis of transforming events that convert chronic myeloproliferative neoplasms to leukemias. Cancer Research. 2010;70(2):447–452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Tallarico M, Odenike O. Secondary acute myeloid leukemias arising from Philadelphia chromosome negative myeloproliferative neoplasms: Pathogenesis, risk factors, and therapeutic strategies. Current Hematol Malign Rep. 2015;10(2):112–117. [DOI] [PubMed] [Google Scholar]
  • 84.Zhang S-J, Rampal R, Manshouri T, Patel J, Mensah N, Kayserian A, 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] [PMC free article] [PubMed] [Google Scholar]
  • 85.Tefferi A, Lasho TL, Abdel-Wahab O, Guglielmelli P, Patel J, Caramazza D, et al. IDH1 and IDH2 mutation studies in 1473 patients with chronic-, fibrotic- or blast-phase essential thrombocythemia, polycythemia vera or myelofibrosis. Leukemia. 2010;24:1302–1309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Pardanani A, Lasho TL, Finke CM, Mai M, McClure RF, Tefferi A. IDH1 and IDH2 mutation analysis in chronic- and blast-phase myeloproliferative neoplasms. Leukemia. 2010;24:1146–1151. [DOI] [PubMed] [Google Scholar]
  • 87.Tefferi A, Lasho TL, Guglielmelli P, Finke CM, Rotunno G, Elala Y, et al. Targeted deep sequencing in polycythemia vera and essential thrombocythemia. Blood. 2016;1(1):21–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.McNamara CJ, Panzarella T, Kennedy JA, Arruda A, Claudio JO, Daher-Reyes G, et al. The mutational landscape of accelerated- and blast-phase myeloproliferative neoplasms impacts patient outcomes. Blood Adv. 2018;2(20):2658–2671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Guo Y, Zhou Y, Yamatomo S, Yang H, Zhang P, Chen S, et al. ASXL1 alteration cooperates with JAK2V617F to accelerate myelofibrosis. Leukemia. 2019;33:1287–1291. [DOI] [PubMed] [Google Scholar]
  • 90.Zhou S, Tremblay D, Hoffman R, Kremyanskaya M, Najfeld V, Li L, et al. Clinical benefit derived from decitabine therapy for advanced phases of myeloproliferative neoplasms. Acta Haematol. 2021; 144(1): 48–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Benton CB, Boddu PC, DiNardo CD, Bose P, Wang F, Assi R, et al. Janus kinase 2 variants associated with the transformation of myeloproliferative neoplasms into acute myeloid leukemia. Cancer. 2019;125(11):1855–1866. [DOI] [PubMed] [Google Scholar]
  • 92.Mascarenhas J A concise update on risk factors, therapy, and outcome of leukemic transformation of myeloproliferative neoplasms. Clin Lymphoma Myeloma Leuk. 2016;16:Suppl:S124–9. [DOI] [PubMed] [Google Scholar]
  • 93.Gagelmann N, Ditschkowski M, Bogdanov R, Robin M, Cassinat B, Shahswar R, et al. Comprehensive clinical-molecular transplant scoring system for myelofibrosis undergoing stem cell transplantation. Blood. 2019;133(20): 2233–2242. [DOI] [PubMed] [Google Scholar]
  • 94.Mannelli L, Guglielmelli P, Vannucchi AM. Stem cell transplant for the treatment of myelofibrosis. Expert Review Hematol. 2020;13(4):363–374. [DOI] [PubMed] [Google Scholar]
  • 95.Gowin K, Ballen K, Ahn KW, Hu Z-H, Ali H, Arcasoy MO, et al. Survival following allogeneic transplant in patients with myelofibrosis. Blood Adv. 2020;4(9):1965–1973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines). Myeloproliferative Neoplasms (Version I.2020); https://www.nccn.org/professionals/physician_gls/default.aspx. Last accessed October 3, 2020.
  • 97.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] [PMC free article] [PubMed] [Google Scholar]
  • 98.Mascarenhas J, Heaney ML, Najfeld V, Hexner E, Abdel-Wahab O, Rampal R, 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]
  • 99.Devillier R, Raffoux E, Rey J, Lengline E, Ronchetti A-M, Serbet M, et al. Combination therapy with ruxolitinib plus intensive treatment strategy is feasible in patients with blast-phase myeloproliferative neoplasms. Br J Haematol. 2016;172:625–644. [DOI] [PubMed] [Google Scholar]
  • 100.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] [PMC free article] [PubMed] [Google Scholar]
  • 101.Dumas P-Y; Bertoli S, Bérard E, Médiavilla C, Yon E, Tavitian S, et al. Azacitidine or intensive chemotherapy for older patients with secondary or therapy-related acute myeloid leukemia. Oncotarget. 2017;8(45):79126–79136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Thepot S, Itzykson R, Seegers V, Raffoux E, Quesnel B, Chait Y, 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(19):3735–3742. [DOI] [PubMed] [Google Scholar]
  • 103.Andriani A, Montanaro M, Voso MT, Villiva N, Ciccone F, Andrizzi C, 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] [PubMed] [Google Scholar]
  • 104.Andriani A, Elli E, Trapè G, Villivà N, Fianchi L, Di Veroli A, 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] [PubMed] [Google Scholar]
  • 105.Drummond MW, Harrison CN, Vicente SM, Mead AJ, Yap C, Chen F, et al. A Phase Ib to assess the safety and tolerability of ruxolitinib in combination with azacitidine in patients with advanced phase myeloproliferative neoplasms (MPN), including myelodysplastic syndromes (MDS) or acute myeloid leukemia (AML) arising from MPN (The Bloodwise/TAP PHAZAR study on behalf of the UK MPN CGS). Blood. 2017;130 Suppl. 1):1649.28733324 [Google Scholar]
  • 106.Masarova L, Verstovsek S, Hidalgo-Lopez JE, Bose P, Estrov Z, Jabbour EJ, et al. A phase 2 study of ruxolitinib in combination with azacitidine with myelofibrosis. Blood. 2018;132(16):1664–1674. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Badar T, Kantarjian HM, Ravandi F, Jabbour E, Borthakur G, Cortes JE, 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. Leukemia Research. 2015;39(9):950–956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Eghtedar A, Verstovsek S, Estrov Z, Burger J, Cortes J, Bivins C, 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] [PMC free article] [PubMed] [Google Scholar]
  • 109.Bose P, Verstovsek S, Gasior Y, Jain N, Jabbour EJ, Estrov Z, et al. Phase I/II study of ruxolitinib (RUX) with decitabine (DAC) in patients with post-Myeloproliferative Neoplasm Acute Myeloid Leukemia (post-MPN AML): Phase I Results. Blood. 2016;128(22):4262. [Google Scholar]
  • 110.Bose P, Verstovsek S, Cortes JE, Tse S, Gasior Y, Jain N, et al. A phase 1/2 study of ruxolitinib and decitabine in patients with post-myeloproliferative neoplasm acute myeloid leukemia. Leukemia.2020;34(9):2489–2492. [DOI] [PubMed] [Google Scholar]
  • 111.Rampal RK, Mascarenhas JO, Kosiorek HE, Price L, Berenzon D, Hexner E, et al. Safety and efficacy of combined ruxolitinib and decitabine in accelerated and blast-phase myeloproliferative neoplasms. Blood Adv. 2018;2(24):3572–3580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Mascarenhas J, 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] [PMC free article] [PubMed] [Google Scholar]
  • 113.Saenz DT, Fiskus W, Manshouri T, Rajapakshe K, Krieger S, Sun B, 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] [PMC free article] [PubMed] [Google Scholar]
  • 114.Mascarenhas J, Harrison C, Patriarca A, Devos T, Palandri F, Rampal R, et al. CPI-0610, a Bromodomain and Extraterminal Domain Protein (BET) inhibitor, in combination with ruxolitinib, in JAK inhibitor treatment naïve myelofibrosis patients: Update from MANIFEST phase 2 study. HemaSphere. 2020;4(S1, abstract EP1084):499–500. [Google Scholar]
  • 115.Verstovsek S, Mascarenhas J, Kremyanskaya M, Hoffman R, Rampal R, Gupta V, et al. CPI-0610, Bromodomain and Extraterminal domain protein (BET) inhibitor, as ‘add-on’ to ruxolitinib (RUX), in advanced myelofibrosis patients with suboptimal response: Update of MANIFEST phase 2 study. HemaSphere. 2020;4(S1, abstract EP1083):499. [Google Scholar]
  • 116.Saenz DT, Fiskus W, Qian Y, Manshouri T, Rajapakshe K, Raina K, et al. Novel BET protein proteolysis-targeting chimera exerts superior lethal activity than bromodomain inhibitor (BETi) against post-myeloproliferative neoplasm secondary (s)AML cells. Leukemia. 2017;31(9):1951–1961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Verstovsek S, Fiskus W, Mashouri T, Bhalla KN. Targeting cistrome and dysregulated transcriptome of post-MPN sAML. Oncotarget. 2017;8(55):93301–93302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Saenz D Fiskus W, Mill CP, Perera D, Manshouri T, Lara BH, et al. Mechanistic basis and efficacy of targeting β-catenin-TCF7L2-JMJD6-c-Myc axis to overcome resistance to BET inhibitors. Blood. 2020;135(15):1255–1269. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Bhagwat N, Koppikar P, Keller M, Marubayashi S, Shank K, Rampal R, et al. Improved targeting of JAK2 leads to increased therapeutic efficacy in myeloproliferative neoplasms. Blood. 2014;123(13):2075–2083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Scherber RM, Mesa RA. Management of challenging myelofibrosis after JAK inhibitor failure and/or progression. Blood Rev. 2020May30;100716. doi: 10.1016/j.blre.2020.100716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Green A and Beer P. Somatic Mutations of IDH1 and IDH2 in the leukemic transformation of myeloproliferative neoplasms. N Engl J Med. 2010;362(4):369–370. [DOI] [PubMed] [Google Scholar]
  • 122.McKenney AS, Allison NL, Hanasoge Somasundra AV, Spitzer B, Intlekofer AM, et al. JAK2/IDH-mutant–driven myeloproliferative neoplasm is sensitive to combined targeted inhibition. J Clinical Invest. 2018;128(2):789–804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.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] [PubMed] [Google Scholar]
  • 124.Chifotides HT, Masarova L, Alfayez M, Daver N, Alvarado Y, Jabbour E, 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] [PMC free article] [PubMed] [Google Scholar]
  • 125.Choe S, Wang H, DiNardo CD, Stein EM, de Botton S, Roboz GJ, et al. Molecular mechanisms mediating relapse following ivosidenib monotherapy in IDH1-mutant relapsed or refractory AML. Blood Adv. 2020;4(9):1894–1905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.DiNardo CD, Jonas BA, Pullarkat V, et al. Azacitidine and venetoclax in previously untreated acute myeloid leukemia. N Engl J Med. 2020;383(7):617–629. [DOI] [PubMed] [Google Scholar]
  • 127.Lachowiez C, DiNardo CD, Konopleva M. Venetoclax in acute myeloid leukemia-current and future directions. Leukemia & Lymphoma. 2020;61(6):1313–1322. [DOI] [PubMed] [Google Scholar]
  • 128.DiNardo CD, Maiti A, Rausch CR, et al. 10-day decitabine with venetoclax for newly diagnosed intensive chemotherapy ineligible, and relapsed or refractory acute myeloid leukaemia: a single-centre, phase 2 trial. Lancet Haematol. 2020;7(10):e724–e736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Petiti J, Lo lacono M, Rosso V, Andreani G, Jovanovski A, Podesta M, et al. Bcl-xl represents a therapeutic target in Philadelphia negative myeloproliferative neoplasms. J. Cell Mol Med 2020; 24: 10978–10986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Tremblay D, Feld J, Dougherty M, et al. Venetoclax and hypomethylating agent combination therapy in acute myeloid leukemia secondary to a myeloproliferative neoplasm. Leuk Res. 2020;98:106456. [DOI] [PubMed] [Google Scholar]
  • 131.Gangat N, Morsia E, Foran JM, et al. Venetoclax plus hypomethylating agent in blast-phase myeloproliferative neoplasm: preliminary experience with 12 patients. Br. J Haematol. 2020;191(5):e120–e124. [DOI] [PubMed] [Google Scholar]
  • 132.Pollyea DA, Amaya M, Konopleva MY. Venetoclax for AML: changing the treatment paradigm. Blood Adv. 2019;3(24):4326–4335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Konopleva M, Pollyea DA, Potluri J, et al. Efficacy and biological correlates of response in a phase II study of venetoclax monotherapy in patients with acute myelogenous leukemia. Cancer Discov. 2016;6(10):1106–1117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Chyla B, Daver N, McKeegan E, et al. Genetic biomarkers of sensitivity and resistance to venetoclax monotherapy in patients with relapsed acute myeloid leukemia. Am J Hematol. 2018;93(8):E202–E205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.DiNardo CD et al. Molecular patterns of response and treatment failure after frontline venetoclax combinations in older patients with AML. Blood 2020;135(11):791–803. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Huemer F, Melchardt T, Jansko B, et al. Durable remissions with venetoclax monotherapy in secondary AML refractory to hypomethylating agents and high expression of BCL‐2 and/or BIM. Eur J Haematol. 2019;102:437–441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Lachowiez CA, Borthakur G, Loghavi S, et al. Phase Ib/II study of the IDH1-mutant inhibitor ivosidenib with the BCL2 inhibitor venetoclax +/− azacitidine in IDH1-mutated hematologic malignancies. J Clin Oncol. 2020;38(suppl 15):7500. [Google Scholar]
  • 138.DiNardo CD, Takahashi K, Kadia T. et al. A phase 1b/2 clinical study of targeted IDH1 inhibition with ivosidenib in combination with the Bcl-2 inhibitor venetoclax, for patients with IDH1-mutated myeloid malignancies. HemaSphere. 2019;3(S1):97 (# PF 291). [Google Scholar]
  • 139.Rücker FG, Schlenk RF, Bullinger L, et al. TP53 alterations in acute myeloid leukemia with complex karyotype correlate with specific copy number alterations, monosomal karyotype, and dismal outcome Blood. 2012;119(9):2114–2121. [DOI] [PubMed] [Google Scholar]
  • 140.Venton G, Courtier F, Charbonnier A, et al. Impact of gene mutations on treatment response and prognosis of acute myeloid leukemia secondary to myeloproliferative neoplasms. Am J Hematol. 2018;93(3):330–338. [DOI] [PubMed] [Google Scholar]
  • 141.Verstovsek S. Personal communication.

RESOURCES