Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Jan 1.
Published in final edited form as: Clin Lymphoma Myeloma Leuk. 2018 Jan;18(1):1–12. doi: 10.1016/j.clml.2017.11.008

SOHO state-of-the-art update and next questions: MPN

Prithviraj Bose 1, Jason Gotlib 2, Claire N Harrison 3, Srdan Verstovsek 1
PMCID: PMC5915302  NIHMSID: NIHMS926594  PMID: 29277359

Abstract

The discovery of the activating JAK2V617F mutation in 2005 in the majority of patients with the classic Philadelphia chromosome-negative myeloproliferative neoplasms (MPN) spurred intense interest in research into these disorders, culminating in the identification of activating mutations in MPL in 2006 and indels in CALR in 2013, thus providing additional mechanistic explanations for the universal activation of Janus kinase-signal transducer and activator of transcription (JAK-STAT) observed in these conditions, and the success of the JAK1/2 inhibitor ruxolitinib, which first received regulatory approval in 2011. The field has continued to advance rapidly since then, and the last two years have witnessed important changes to the classification of MPN and diagnostic criteria for polycythemia vera (PV), novel insights into the mechanisms of bone marrow fibrosis in primary myelofibrosis (PMF), increasing appreciation of the biologic differences between essential thrombocythemia (ET), prefibrotic and overt PMF and between primary and post-PV/ET myelofibrosis (MF). Additionally, the mechanisms through which mutant calreticulin drives JAK-STAT pathway activation and oncogenic transformation are now better understood. Although mastocytosis is no longer included under the broad heading of MPN in the 2016 revision to the World Health Organization classification, an important milestone in mastocytosis research was reached in 2017 with the regulatory approval of midostaurin for patients with advanced systemic mastocytosis (AdvSM). In this article, we review the major recent developments in the areas of PV, ET and MF, and also briefly summarize the literature on midostaurin and other KIT inhibitors for patients with AdvSM.

Keywords: myeloproliferative neoplasms, polycythemia vera, essential thrombocythemia, myelofibrosis, pre-fibrotic, ruxolitinib, pacritinib, midostaurin

Introduction

The field of myeloproliferative neoplasms (MPN) has come a long way since their original recognition and description as a group of related disorders in William Dameshek’s classic treatise published in 1951.1 Improvements in our understanding of the genetic underpinnings of many of these conditions, as well as refinements in morphologic classification, have led to the creation of a separate major category for mastocytosis, a biologically unique set of related disorders, and the designation of pre-fibrotic primary myelofibrosis, previously a provisional category under primary myelofibrosis, as a distinct entity.2 Elucidation of molecular mechanisms has provided new therapeutic insights, such as the successful use of ruxolitinib for rare MPN such as chronic neutrophilic leukemia (CNL)3 and for the rare myeloid neoplasm associated with t(8;9)(p22;p24.1), that fuses the PCM1 gene to the JAK2 gene.4 The topic of eosinophilia-associated MPN has recently been reviewed.5 In this paper, we summarize the major recent developments in the classic Philadelphia chromosome-negative (Ph) MPN, polycythemia vera (PV), essential thrombocythemia (ET) and myelofibrosis (MF), as well as provide a brief update on the evolving management of advanced systemic mastocytosis (SM).

Polycythemia vera: state of the art update

Diagnosis

The 2016 revision to the World Health Organization (WHO) classification of myeloid neoplasms contains significant changes to the diagnostic criteria for PV (Table 1).2 This stems from the recognition that individuals with so-called “masked” PV have higher rates of progression to MF and inferior leukemia-free survival (LFS) and overall survival (OS) as compared to those with overt PV,6,7 which may reflect, in part, a lower intensity of therapy in these patients because of missed/delayed diagnosis.8 Thus, the hemoglobin and hematocrit thresholds for making a diagnosis of PV in men and women have been lowered to 16.5 g/dl/0.49 and 16 g/dl/0.48, respectively.2 Some experts continue to use Cr51-red cell mass (RCM) measurements to diagnose PV, especially in patients who do not meet the 2008 WHO criteria for elevated hemoglobin or hematocrit values (>18.5 g/dl/0.6 in men and >16.5 g/dl/0.56 in women),9 pointing out that the hematocrit is a derived value sensitive to changes in red cell size;10 however, RCM measurement is not readily available, even at most tertiary care centers. The second major change to the diagnostic criteria for PV concerns the role of bone marrow biopsy. This is now a requirement for diagnosis, except in obvious cases that meet the higher WHO 2008 cutoffs for hemoglobin and hematocrit.2 The enhanced emphasis on bone marrow examination was based on the need to distinguish PV morphologically from cases of JAK2-mutated ET with polycythemia in the context of the lowered hemoglobin and hematocrit thresholds for PV diagnosis.11,12

Table 1.

2016 World Health Organization (WHO) Criteria for the diagnosis of polycythemia vera2

Major criteria
  1. Hemoglobin >16.5 g/dL in men and >16 g/dL in women or, Hematocrit >49% in men and >48% in women or, red cell mass (RCM) >25% above mean normal predicted value
  2. Bone marrow biopsy showing hypercellularity for age with trilineage growth (panmyelosis) including prominent erythroid, granulocytic, and megakaryocytic proliferation with pleiomorphic, mature megakaryocytes (differences in size)
  3. Presence of a JAK2V617F or JAK2 exon 12 mutation
Minor criterion
  Subnormal erythropoietin level

Diagnosis requires meeting all three major criteria, or the first two major criteria and the minor criterion. Bone marrow biopsy may not be required in cases with sustained absolute erythrocytosis (hemoglobin levels >18.5 g/dL in men (hematocrit >55.5%) and >16.5 g/dL in women (hematocrit >49.5%) if major criterion 3 and the minor criterion are present.

Therapy: the resurgence of interferons

Hydroxyurea (HU) is currently the most commonly used first-line drug in patients with PV who require cytoreduction.13 The use of ruxolitinib as second-line therapy after failure of HU (Table 2)14 is supported by two large, randomized controlled trials (RCTs), RESPONSE15,16 and RESPONSE-2.17 Interferons, which are distinguished by their ability to induce molecular remissions,18,19 may also be used as initial therapy, and are increasingly preferred for young patients given their lack of leukemogenicity.20,21 Long-term follow-up of a single-institution trial of pegylated interferon alfa (IFN-α), however, shows that discontinuation rates due to toxicity can be substantial.22 Ropeginterferon alfa-2b is a new monopegylated isoform of IFN-α-2b that has the advantage of requiring administration only every 2 weeks. This agent, which produced an overall response rate (ORR) of 90% (47% complete, 43% partial) in the phase 1/2 PEGINVERA study23 with a complete molecular response (CMR) in 21% of patients and a partial molecular response in 47% of patients, was subsequently compared head-to-head with HU in the phase 3 PROUD-PV RCT.24 Non-inferiority in the 12-month complete hematologic response (CHR) rate was demonstrated in this trial, which enrolled both cytoreductive treatment-naïve patients and those who had been on HU for <3 years without achievement of CHR or development of resistance or intolerance.24 Pegylated IFN-α-2a (Pegasys®) is also being compared to HU in the frontline setting in a large multi-center phase 3 RCT.25 A planned interim analysis of this trial performed after 75 patients had been on study for 12 months did not show a statistically significant difference in ORR between the two arms (69% for HU and 81% for pegylated IFN-α-2a).25

Table 2.

ELN definition of resistance and intolerance to hydroxyurea in polycythemia vera14

1. Need for phlebotomy to keep hematocrit <45% after 3 months of ≥2 g/day of HU, or
2. Uncontrolled myeloproliferation, i.e., platelets >400 × 109/L AND leukocytes >10 × 109/L after 3 months of ≥2 g/day of HU, or
3. Failure to reduce massive (≥10 cm below the left costal margin) splenomegaly by ≥50% as measured by palpation, OR failure to completely relieve symptoms related to splenomegaly, after 3 months of ≥2 g/day of HU, or
4. ANC <1 × 109/L OR platelets <100 × 109/L OR hemoglobin <10 g/dL at the lowest dose of HU required to achieve a complete or partial response (defined below)
5. Presence of leg ulcers or other unacceptable HU-related non-hematologic toxicities, such as mucocutaenous manifestations, gastrointestinal symptoms, pneumonitis or fever at any dose of HU

Complete response: hematocrit <45% without phlebotomy, platelets <400 × 109/L, leukocytes ≤10 × 109/L, and no disease-related symptoms. Partial response: hematocrit <45% without phlebotomy, or response in ≥3 other ELN consensus criteria.203 HU, hydroxyurea; ANC, absolute neutrophil count; ELN, European LeukemiaNet.

PV: areas of controversy and next questions

Although JAK2V617F expression is sufficient to produce the PV phenotype in mouse models,2629 several lines of evidence suggest that additional genetic and/or epigenetic lesions are necessary to sustain the disease.30,31 Patients who acquire JAK2V617F before a TET2 mutation are more likely to present with PV than ET, and have an increased risk of thrombosis.32 Homozygosity for JAK2V617F, strongly correlated with a PV phenotype, is more common in men.33 However, even after removing gender as a potential confounder, gene expression profiling reveals two distinct clinical phenotypes in PV.34 In one series of 133 patients with PV, targeted sequencing revealed one or more sequence variants/mutations (in genes other than JAK2) in 53%; variants/mutations in ASXL1, SRSF2 and IDH2 (combined prevalence 15%) were identified as being prognostically adverse.35 Clearly, there are factors beyond canonical JAK-STAT signaling that are involved in the pathogenesis and maintenance of PV (and other MPN); these are yet to be clearly elucidated. Thrombotic risk stratification in PV continues to rely on two variables: patient age and previous history of thrombosis.21 Whether or not leukocytosis contributes to the risk of thrombosis is still controversial, with some studies finding leukocytosis to independently predict thrombosis,3638 and others reporting no association;39 furthermore, the precise cutoffs are not clear. Leukocytosis ≥15 × 109/L was found to adversely affect both OS and LFS in a large (n = 1,545) study.40 While some evidence suggests that control of leukocytosis with cytoreductive therapy may decrease the risk of thrombosis,41 this remains debatable, and isolation of the effect of leukocytosis reduction from that of hematocrit control is difficult.

An interesting new strategy that has entered the clinic in both high-risk, previously treated PV and JAK2V617F+ ET is that of using small-molecule inhibitors of human double minute 2 (MDM2), which trigger p53-dependent apoptosis in the absence of deletions or inactivating mutations of TP53 (NCT02407080). Based on preclinical studies showing synergism,42,43 pegylated IFN-α can be added in cases of insufficient response. While an attractive laboratory-based concept, whether this class of drugs could gain regulatory approval in these indolent diseases, given their expense and toxicity and the approval of ruxolitinib for second-line therapy of PV and ongoing development for ET (see below) is questionable. Similar considerations apply to histone deacetylase inhibitors (reviewed in ref.44), which are clinically active but have chronic toxicities.

Essential thrombocythemia: state of the art update

CALR mutations: pathophysiology and clinical correlations in ET

In 2013, mutations in exon 9 of the gene encoding calreticulin (CALR), an endoplasmic reticulum (ER) chaperone, were reported in 20–30% of patients with ET and primary MF (PMF), accounting for the majority of JAK2/MPL-unmutated cases.45,46 These mutations, that can broadly be divided into 52-base pair deletions (termed type 1/type-1 like) or 5-base pair insertions (termed type 2/type 2-like), all result in an altered C-terminus of the mutant protein with loss of negative charge and impaired Ca++ binding, loss of the “KDEL” ER retention motif, and activation of the Janus kinase – signal transducer and activator of transcription (JAK-STAT) pathway.45,46 Recent work from several groups has demonstrated that mutant CALR must bind to and activate MPL (the thrombopoietin receptor) to drive MPN pathogenesis, and that the altered C-terminus is required for oncogenic transformation.4750 CALR mutations appear to confer a greater proliferative advantage to the neoplastic clone compared with JAK2 mutations; clonal expansion is faster in CALR-mutated cases than in JAK2-mutated cases, both in ET and in PMF.51

CALR-mutated patients with ET tend to be younger, more frequently male, and have higher platelet counts and lower hemoglobin levels and leukocyte counts than their JAK2-mutated counterparts.5254 Of particular importance, thrombotic risk in CALR-mutant ET appears particularly low, so much so that young patients with CALR-mutated ET and no history of thrombosis (i.e., “very low risk” patients) may forego aspirin.20,55,56 Most studies have found no impact of CALR mutations on OS, leukemic transformation (LT) or the risk of progression of ET to MF,5254 although it was recently reported that CALR-mutated ET patients progress more slowly to MF than JAK2-mutated or triple negative patients.57 Type 1/type 1-like and type 2/type 2-like CALR mutations occur at approximately equal frequencies in ET, and although one study noted a significantly higher risk of progression to MF among ET patients with type 1/type 1-like CALR mutations as opposed to type 2/type 2-like mutations,58 other investigators have not found this to be the case.59

Advances in risk stratification and current and future therapy of ET

ET is the most indolent of the classic MPN,60,61 and treatment is currently based on thrombotic risk.21 The latter is estimated using the revised IPSET (International Prognostic Score for ET)-thrombosis score (Table 3), which takes into account patient age, thrombosis history and presence or absence of JAK2V617F and cardiovascular risk factors.56 This model has been validated in an independent cohort,62 and is not impacted by CALR mutational status.63 Importantly, leukocytosis ≥11 × 109/L, which is not a prognostic variable in the IPSET-thrombosis model, is, however, predictive of worse survival in ET, as are advanced age (≥60 years) and prior thrombosis.64 Furthermore, a number of studies have reported a correlation between leukocytosis, but not thrombocytosis, with the risk of thrombosis in ET.6568 Very recently, a two-center study of 1,494 patients found male sex, age ≥60 years and leukocyte count ≥11 × 109/L to be independent predictors of shortened survival in ET; thrombosis history was not significant upon multivariate analysis in this study.69 An increased serum lactate dehydrogenase level has also been reported to correlate with inferior survival in ET,70 which leads one to speculate whether these patients might, in fact, have had pre-PMF (discussed below).

Table 3.

Revised International Prognostic Score for Thrombosis in Essential Thrombocythemia56

Very low risk No thrombosis history, age ≤60 years, and JAK2 wild type
Low risk No thrombosis history, age ≤60 years, and JAK2 mutation
Intermediate risk No thrombosis history, age >60 years, and JAK2 wild type
High risk Thrombosis history or age >60 years with JAK2 mutation

HU is usually the preferred agent for cytoreduction in ET for patients who need cytoreductive therapy, based on the findings of the PT-1 study, in which HU was compared head-to-head with anagrelide.71 More recently, anagrelide was found to be non-inferior to HU in the ANAHYDRET study;72 nevertheless, this agent is typically used second-line.21 It is important to appreciate that the patient populations studied in the PT-1 and ANAHYDRET studies and their designs were not identical; the definition of ET relied on different criteria (Polycythemia Vera Study Group (PVSG) criteria in PT-1 and WHO in ANAHYDRET), the patients in PT-1 could have received prior therapy, whereas those in ANAHYDRET could not, and the use of aspirin was not mandated in the ANAHYDRET study. As in PV, interferons may be preferred over HU in young patients.21 Formal criteria to define resistance and intolerance to HU in patients with ET have been published (Table 4).73 Not captured in these criteria are the substantial symptom burden that patients with ET can have,74,75 which may not be alleviated by standard cytoreductive therapies even in the presence of hematologic response. In a phase 1/2 study in 39 patients with HU-resistant/intolerant ET, median platelet and leukocyte counts decreased rapidly with ruxolitinib over the first 4 weeks of therapy, and many patients experienced a ≥50% improvement in a variety of symptoms by week 12.76 However, ruxolitinib did not improve the rate of CHR compared with best available therapy (BAT) in HU-resistant/intolerant ET patients in the MAJIC randomized clinical trial; rates of thrombosis, hemorrhage or progression to MF did not significantly differ, either.77 Nevertheless, ruxolitinib is currently being tested as second-line therapy in patients with high risk ET in two trials: the RESET-272 study (NCT03123588) in the US in which it is compared to anagrelide, and in a French study (NCT02962388) in which it is compared to anagrelide or IFN-α. The RUXO-BEAT trial (NCT02577926) in Germany compares ruxolitinib to BAT in patients with high-risk ET who may be treatment-naïve or previously treated; patients with high risk PV with prior exposure to cytoreductive agents for ≤6 weeks may also enroll.

Table 4.

Definition of resistance and intolerance to hydroxyurea in essential thrombocythemia73

1. Platelets >600 × 109/L after 3 months of ≥2 g/day of HU (≥2.5 g/day if body weight >80 kg)
2. Platelets <400 × 109/L and leukocytes <2.5 × 109/L at any dose of HU
3. Platelets <400 × 109/L and hemoglobin <10 g/dL at any dose of HU
4. Presence of leg ulcers or other unacceptable muco-cutaneous manifestations at any dose of HU
5. HU-related fever

ET: areas of controversy and next questions

Although JAK2V617F is classically associated with a PV phenotype,2629 mice expressing this mutation have been shown to develop ET-, PMF- and PV-like disease, and differences in gene dosage/mutant allele burden have been invoked as explanations of how the same mutation can lead to different disease phenotypes.78,79 Indeed, knock-in mouse models of ET with variable rates of progression to MF using all 3 driver mutations (JAK2V617F, MPLW515L and CALR exon 9 mutations) have been generated.50,7982 Furthermore, as noted above, mutation order influences disease phenotype in the Ph MPN: prior mutation of TET2 has been shown to alter the transcriptional signature of JAK2V617F in a cell-intrinsic manner, preventing the latter from up-regulating genes associated with proliferation.32 Experimentally, it has been shown in a mouse model that loss of STAT1 in the presence of JAK2V617F promotes a PV phenotype over an ET phenotype, while activating STAT1 using IFN-γ has the opposite effect.83 Targeted deep sequencing of 183 patients with ET revealed the presence of one or more sequence variants/mutations in “non-driver” genes in 53%; those in SH2B3, SF3B1, U2AF1, TP53, IDH2 and EZH2 (combined prevalence 15%) adversely impacted survival.35 As the mutational landscape of ET and PV is further unraveled, new insights into disease pathogenesis are likely to emerge.

In the clinical setting, although considerable evidence points to a higher risk of thrombosis6568 and worse survival64 in patients with leukocytosis, the cutoffs vary across studies and it remains unknown if control of leukocytosis with cytoreductive therapy will improve outcomes, although some experts emphasize this in their practices.84 At present, leukocytosis is not routinely considered when making therapeutic decisions, which continue to be informed by thrombotic risk, as determined by the revised IPSET-thrombosis score.56 In the case of thrombocytosis, there is no good evidence of a correlation between platelet count and clotting risk; rather, most physicians check for the presence of acquired von Willebrand disease and use cytoreductive drugs to mitigate bleeding risk in patients with “extreme thrombocytosis” (reviewed in ref.85).

Myelofibrosis: state of the art update

Pre-fibrotic primary myelofibrosis

The 2016 WHO classification of myeloid neoplasms recognizes pre-fibrotic PMF (pre-PMF) as a separate entity under the MPN umbrella, an important change from prior versions that included pre-PMF as a “provisional” category within PMF.2 Diagnostic criteria for pre-PMF appear in Table 5, and the pathologic distinction between ET and pre-PMF has been the subject of considerable controversy over the years.86 Nevertheless, it is clear that compared to patients with ET, patients with pre-PMF have worse OS and higher rates of LT and progression to overt MF.8789 Patients with pre-PMF also appear to have higher rates of thrombosis90 and bleeding91 than those with ET and, as in ET, leukocytosis may predict for an increased risk of thrombosis, arterial in particular, among patients with pre-PMF.92,93 Recently, the findings from two large studies totaling over 1000 patients with PMF support the existence of a phenotypic continuum from pre-fibrotic to overt PMF, with overt PMF being associated with a higher incidence of cytopenias, higher circulating blast counts, greater symptom burden and higher incidence of splenomegaly, as well as worse prognostic risk scores and significantly reduced OS compared with pre-PMF.94,95

Table 5.

2016 World Health Organization (WHO) criteria for the diagnosis of pre-fibrotic primary myelofibrosis2

Major criteria
  1. Megakaryocytic proliferation and atypia, without reticulin fibrosis >grade 1, accompanied by increased age-adjusted BM cellularity, granulocytic proliferation and often, decreased erythropoiesis
  2. Not meeting WHO criteria for CML, PV, ET, MDS, or other myeloid neoplasms
  3. Presence of JAK2, CALR, or MPL mutation or, in the absence of these mutations, presence of another clonal marker, e.g., mutations in ASXL1, EZH2, TET2, IDH1, IDH2, SRSF2, SF3B1, or absence of minor reactive bone marrow reticulin fibrosis
Minor criteria
  1. Anemia not attributed to a comorbid condition
  2. Leukocytosis ≥11 × 109/L
  3. Palpable splenomegaly
  4. LDH above institutional upper limit of normal

Diagnosis requires meeting all three major criteria and at least one minor criterion. CML, chronic myeloid leukemia; PV, polycythemia vera; ET, essential thrombocythemia; MDS, myelodysplastic syndromes.

Novel insights into mechanisms of bone marrow fibrosis in PMF

PMF is distinguished clinically from PV and ET by the development of anemia in nearly all patients, a much higher incidence of splenomegaly and a significantly greater symptom burden, along with a substantially higher risk of LT and markedly shortened survival.96 The intrinsic biologic complexity of PMF is considerably greater than that of PV or ET; this is reflected in the mutational burden of the three diseases.97 In fact, PMF has been proposed to be best considered a myelodysplastic/myeloproliferative neoplasm, characterized by a high frequency of “non-driver” mutations affecting epigenetic regulation and the spliceosome machinery.98 Bone marrow fibrosis in PMF has classically been viewed as being a reactive process.10 However, some recent findings have challenged this notion. For example, it has been demonstrated that bone marrow from patients with PMF is rich in clonal, neoplastic monocyte-derived fibrocytes that produce collagen and fibronectin and give rise to a lethal MF-like phenotype when transplanted into immunodeficient mice.99 Other investigators have shown that Gli1+ mesenchymal stromal cells (MSCs) are recruited from endosteal and perivascular niches to become fibrosis-driving myofibroblasts in JAK2V617F+ mouse models and in the bone marrow of MPN patients,100 and proposed that non-canonical modes of activation of the Gli transcription factors may explain the modest effects of therapeutic hedgehog (smoothened) inhibition in MF.101 Overexpression of v-maf avian musculoaponeurotic fibrosarcoma oncogene homolog (MAF) has recently been implicated in the pathogenesis of bone marrow fibrosis in PMF through excessive production of the pro-fibrotic mediator SPP1 and resultant proliferation of fibroblasts and MSCs, leading to collagen production.102

Improvements in prognostication

Although many studies evaluating prognostic factors in patients with PMF have been published over the years, the most frequently used prognostic scoring systems in clinical practice are the International Prognostic Scoring System (IPSS), the Dynamic IPSS (DIPSS) and the DIPSS-plus, which incorporates karyotype together with clinical variables (reviewed in103). Circulating or bone marrow blasts ≥10%, platelets <50 × 109/L and chromosome 17 aberrations have been found to characterize an “accelerated phase” in patients with MF, which appears to be a necessary step in LT of chronic phase MF and portends an extremely poor prognosis.104 Other investigators have reported >80% 2-year mortality in PMF patients with monosomal karyotype, inv(3)/i(17q) abnormalities, or any two of the following: circulating blasts ≥10%, leukocyte count ≥40 × 109/L, or other unfavorable karyotype.105 The demonstration that PMF patients with CALR mutations have the best OS, and so-called “triple negative” patients exhibit the worst OS,106,107 as well as the identification of “high molecular risk” (HMR) mutations in PMF (ASXL1, EZH2, IDH1/2, SRSF2),108,109 has led to efforts to incorporate mutational data into prognostic models for PMF.110 Others have proposed prognostic scoring systems that only take into account age and genomic information.111,112 Unlike in ET, type 1/type 1-like and type 2/type 2-like CALR mutations have been shown to have different prognostic impacts, with the favorable prognosis associated with CALR mutations restricted to the more common type 1 mutations.113,114 Marked separation of the survival curves of PMF patients stratified only by the mutational status of CALR and ASXL1 has also been reported.115 Although not included in the major prognostic models, the grade of bone marrow fibrosis (0–1 versus 2–3) was recently shown to significantly impact OS, independent of IPSS variables and mutational status, with patients with higher grades of fibrosis also being more likely to have cytopenias, constitutional symptoms, larger splenomegaly and HMR mutations.116 Lastly, thrombotic risk in PMF appears largely restricted to patients with JAK2 mutations.117

Progression of PV to MF occurs in 4.9–6% of cases at 10 years and 6–14% at 15 years; the corresponding percentages for ET are 0.8–4.9% and 4–11%, respectively.118 A variety of risk factors for progression to post-PV/ET MF have been identified (reviewed in ref.118). Although managed similarly,119 a number of groups have reported significant differences between the clinical behavior of PMF and post-PV/ET MF, with the latter representing a more indolent disease process characterized by better survival, more so for post-ET MF than for post-PV MF, and found that prognostic models developed from cohorts of patients with PMF, such as the IPSS, DIPSS and DIPSS-plus, do not reliably distinguish between prognostic categories in post-PV/ET MF.120123 Prior studies in small numbers of patients had identified anemia (hemoglobin <10 g/dL), thrombocytopenia (platelets <100 × 109/L) and leukocytosis (WBCs >30 × 109/L) as being prognostically adverse in post-PV MF,124 and unfavorable karyotype in both post-PV and post-ET MF.125 These observations laid the foundation for the large (n = 781) Myelofibrosis Secondary to PV or ET (MYSEC) project, the results of which were recently published.126 The superior survival of patients with post-ET MF compared with those with post-PV MF was confirmed in this study (median, 14.5 versus 8.1 years), as was that of CALR-mutated patients when compared to JAK2-mutated patients, as is the case in PMF.57,126 There was no difference in terms of OS between patients with type 1/type-1 like and type 2/type-2 like CALR mutations.57 Among patients with post-ET MF, rates of LT were significantly higher among those with triple negative or JAK2-mutated disease than those with CALR-mutated disease, while thrombosis risk was not affected by driver mutation status.57 The researchers identified five variables (Table 6) which, when combined with patient age on a nomogram in a prognostic model (the MYSEC-PM), allow allocation to one of four prognostic categories (low, intermediate-1, intermediate-2 and high risk) with significantly different survival times (median, not reached, 9.3, 4.4 and 2 years, respectively).126

Table 6.

The Myelofibrosis Secondary to PV and ET - Prognostic Model (MYSEC-PM)126

Clinical variable Points assigned
Hemoglobin <11 g/dL 2
Circulating blasts ≥3% 2
CALR-unmutated genotype 2
Platelets <150 × 109/L 1
Constitutional symptoms 1

Points total to be used along with patient age on published nomogram to identify risk category.

Update on ruxolitinib

Six years after its approval,127 ruxolitinib remains the only approved agent for the treatment of MF. The final, 5-year updates of the pivotal COMFORT trials were recently published.128,129 Overall, the rates of best response improved over time, no new safety signals emerged, and the median duration of spleen response was about 3 years. Although these trials were not powered for survival, patients originally assigned to ruxolitinib lived longer than those assigned to placebo or BAT, despite near-complete crossover, confirming the superior survival observed at earlier timepoints.130132 The rates of reduction in bone marrow fibrosis grade and mutant JAK2 allele burden remain modest.128,129 Interestingly, comparing across trials and diseases, mutant JAK2 allele burden reduction with ruxolitinib appears more robust in the RESPONSE trial in PV.133 In the COMFORT-1 trial, greater reductions in the mutant JAK2 allele burden occurred in patients with shorter disease duration, potentially arguing for the use of ruxolitinib in less advanced stages of the disease.134 A substantial amount of data supports the use of ruxolitinib in patients with IPSS intermediate-1 risk disease (reviewed in ref.135). The phase 3, placebo-controlled ReTHINK trial, designed to evaluate ruxolitinib in patients with lower risk disease with one or more HMR mutations but no significant symptoms or splenomegaly, had to be closed due to poor accrual.136 Current consensus guidelines recommend the use of ruxolitinib in low risk patients with troublesome symptoms and/or splenomegaly,137 but not for its survival advantage, citing “weak evidence”.138

Other than a study that reported a greater benefit of ruxolitinib in patients with a JAK2V617F allele burden ≥50%,139 no factors have been identified that predict its clinical efficacy in patients with MF.140 Several key insights have been provided by analyses of the ~100 patients enrolled on the phase 1/2 trial of ruxolitinib141 at the MD Anderson Cancer Center. Spleen responses appear dose-dependent and correlate with survival,142 a finding confirmed in a pooled analysis of the COMFORT trials.143 Furthermore, the presence of ≥3 non-driver mutations, mainly affecting epigenetic regulators, is associated with much lower odds of having a spleen response, a shorter time to treatment discontinuation and inferior survival.144 Finally, among patients who discontinued ruxolitinib, declining platelet counts and clonal evolution on ruxolitinib therapy predicted for worse outcomes.145 Anemia, an on-target phenomenon resulting from JAK2 inhibition, often impairs dose optimization of ruxolitinib in clinical practice, and is frequently a cause of premature discontinuation. Ruxolitinib-induced anemia is most pronounced during the first 12–24 weeks of therapy, after which hemoglobin levels return to a new, lower baseline. Importantly, it has been shown that ruxolitinib-induced anemia does not share the adverse prognosis of disease-associated anemia146 and, in fact, that ruxolitinib therapy can overcome the latter.147 Ruxolitinib should be dosed according to platelet counts as outlined in the prescribing information, along with supportive measures for the anemia (discussed further below).

Novel therapeutic strategies

Anemia remains a significant clinical problem in MF, often hindering dose optimization of ruxolitinib. Currently available therapeutic options, i.e., androgens, steroids, erythropoiesis-stimulating agents (ESAs) and immunomodulatory agents (Imids) are unsatisfactory.148 An interesting new class of drugs (“activin receptor type II ligand traps”) consists of fusion proteins that sequester ligands belonging to the transforming growth factor beta (TGF-β) superfamily, thereby abrogating their suppressive effect on terminal erythropoiesis.149 Response rates of ~40% have been reported with sotatercept,150 the first molecule in this class, and evaluation of this agent is ongoing, both alone and in combination with ruxolitinib (NCT01712308). A clinical trial of the related agent luspatercept in anemic patients with MF will soon open to accrual (NCT03194542). The inhibitor-of-apoptosis (IAP) antagonist LCL-161 has also been shown to produce clinical improvement (CI) in anemia,151 but this drug is difficult to combine with ruxolitinib on theoretical grounds because of suppression by ruxolitinib of tumor necrosis factor alfa (TNF-α), believed to be necessary for the biological effect of LCL-161152. The immunomodulatory agents lenalidomide and pomalidomide, while producing anemia responses in 20–30% of patients when administered alone, can be quite myelosuppressive when administered in conjunction with ruxolitinib.153,154 On the other hand, thalidomide is relatively non-myelosuppressive and well-tolerated at low doses (i.e., 50 mg/d).155157 The combination of ruxolitinib and thalidomide is currently being explored in a clinical trial (NCT03069326) that enrolls both ruxolitinib-naïve patients and those who have had an insufficient response to ruxolitinib.

A number of JAK2 inhibitors besides ruxolitinib have been tested in patients with MF but, unfortunately, none has been approved to date (reviewed in ref.158). Most were discontinued because of toxicity; of these, fedratinib, which also inhibits bromodomain extra-terminal (BET) proteins,159 was in the most advanced phase of clinical development.160 Development of this clearly active drug (36–40% spleen volume reduction (SVR) and 34–36% symptom response rates at 24 weeks in JAK inhibitor-naïve patients and a 55% SVR rate in ruxolitinib-exposed patients) was halted due to the occurrence of several cases of suspected Wernicke’s encephalopathy.160,161 Very recently, the development of momelotinib, a JAK1/2 inhibitor that had the unique benefit of improving anemia in patients with MF, was stopped given disappointing results in the SIMPLIFY-1 and SIMPLIFY-2 phase 3 trials in terms of the conventional endpoints of ≥35% SVR and ≥50% reduction in total symptom score (TSS), despite evidence of benefit in anemia-related endpoints.162,163 The JAK2-selective inhibitor pacritinib was superior to BAT (excluding ruxolitinib) in JAK inhibitor-naïve patients in the phase 3 PERSIST-1 trial,164 with somewhat mixed results obtained in another phase 3 randomized trial (PERSIST-2) comparing two doses of pacritinib, 400 mg daily and 200 mg twice daily, against BAT in thrombocytopenic patients (baseline platelets <100 × 109/L) with MF. Of note, the BAT arm included 40–45% of patients who previously had received ruxolitinib, and 44% of patients assigned to the BAT arm received ruxolitinib at some point during the study.165 The primary objective was to compare the efficacy of pacritinib, pooling both the dosing arms, to BAT, and the secondary objectives were to compare twice daily and once daily pacritinib individually to BAT. In the primary comparison, 18% of pooled pacritinib patients versus 3% of BAT patients achieved ≥35% SVR (p=0.001), while for ≥50% TSS reduction, these proportions were 25% and 14%, respectively (p=0.079). In the secondary analyses, twice daily pacritinib beat BAT in terms of both ≥35% SVR (22% versus 3%, p=0.001) and ≥50% TSS reduction (32% versus 14%, p=0.011). Once daily pacritinib was superior to BAT for ≥35% SVR (15% versus 3%, p=0.017) but not for ≥50% TSS reduction (17% versus 14%, p=0.652). More patients receiving pacritinib than BAT experienced a ≥50% reduction in their red blood cell transfusion burden. Both gastrointestinal and hematologic adverse events were generally less frequent in the twice daily dosing group. Concerns over excess mortality in the pacritinib-treated patients in these trials prompted the Food and Drug Administration (FDA) to mandate a dose-finding study (PAC203, NCT03165734) in thrombocytopenic patients with MF failing ruxolitinib. Preliminary results from a phase 2 study of another JAK2-selective inhibitor, NS-018, in ruxolitinib-pretreated patients showed a ≥35% SVR rate of 12% and a ≥50% TSS improvement rate of 35%.166 Finally, the JAK1 inhibitor itacitinib yielded encouraging symptom responses (≥50% TSS reduction in 30–35% of patients at 12–24 weeks) in a phase 2 trial;167 this agent is now being studied in combination with low-dose ruxolitinib, as well as alone in patients who fail ruxolitinib after initial response (NCT03144687).

A plethora of novel, targeted agents are under study in patients with MF, either alone or in combination with ruxolitinib (for recent reviews of this subject, see refs.168,169). The telomerase inhibitor imetelstat generated much enthusiasm after complete and partial remissions, reversal of bone marrow fibrosis and molecular responses were reported in a pilot study,170 but this was substantially dampened by updates from the IMBARK™ study in JAK inhibitor-exposed patients with relapsed/refractory intermediate-2/high risk MF. Enrollment to the lower-dose arm has been suspended, and an efficacy and safety analysis of the enrolled higher-dose arm is currently ongoing.171 The anti-fibrotic agent PRM-151 (recombinant pentraxin-2) slowed the development of bone marrow fibrosis in vivo and prolonged the survival of immunodeficient mice transplanted with bone marrow cells from patients with MF.99 Promising findings were presented from a clinical trial, both in patients receiving PRM-151 alone and in combination with ruxolitinib.172,173 Enrollment in a pivotal trial of PRM-151 (NCT01981850) has since been completed, and results are expected soon. Many studies evaluating ruxolitinib in combination with other targeted agents are underway (Table 7); some of these, e.g., those combining ruxolitinib with inhibitors of histone deacetylases, heat shock protein 90 (HSP90) and phosphatidylinositol-3-kinase (PI3K) are backed by sound preclinical data.174179 Thus far, the only combinations that have appeared to yield somewhat better responses than expected with ruxolitinib alone have been those with azacitidine180 and panobinostat.181 Clinical data are awaited on other concepts supported by compelling preclinical findings, e.g., the combination of ruxolitinib with the cyclin-dependent kinase 4/6 inhibitor ribociclib and a PIM kinase inhibitor.182 Yet other novel drug classes, e.g., BH3-mimetics,183 selective inhibitors of nuclear transport184 and BET inhibitors/proteolysis-targeting chimeras (PROTACs)185,186 appear highly promising in the laboratory but are yet to enter the clinic in patients with MF.

Table 7.

Ongoing trials evaluating ruxolitinib combinations in patients with myelofibrosis.

Partner drug class Specific agent Clinicaltrials.gov identifier
Histone deacetylase inhibitor Panobinostat NCT01693601
Panobinostat NCT01433445
Pracinostat NCT02267278
Phosphatidylinositol-3-kinase (delta isoform) inhibitor INCB050465 NCT02718300
Idelalisib NCT02436135
TGR1202 NCT02493530
Immunomodulatory agent Thalidomide NCT03069326
Lenalidomide NCT01375140
Pomalidomide NCT01644110
Janus kinase 1 inhibitor Itacitinib NCT03144687
BH3-mimetic Navitoclax NCT03222609
Hedgehog (smoothened) inhibitor Vismodegib NCT02593760
Sonidegib NCT01787552
Cyclin-dependent kinase 4/6 inhibitor and PIM kinase inhibitor Ribociclib and PIM447 NCT02370706
Androgen Danazol NCT01732445
Interferon Pegylated interferon alfa 2a NCT02742324
Hypomethylating agent Azacitidine NCT01787487
Activin receptor ligand trap Sotatercept NCT01712308
Luspatercept NCT03194542
Erythropoiesis stimulating agent Any (observational study) NCT03208803

Myelofibrosis: next questions

Whether or not ruxolitinib improves OS in patients with MF continues to be debated, with concerns raised over the COMFORT trials not being powered to show differences in survival and comparisons with historical controls being flawed due to imbalances in patient characteristics.187 Some experts have suggested that the survival advantage for ruxolitinib observed in the COMFORT trials may reflect improvements in appetite, weight and overall functionality rather than a true disease-modifying effect.188 Indeed, the limited effects of ruxolitinib on bone marrow fibrosis and driver mutation allele burden suggest that any disease-modifying activity of the drug is likely to be relatively minor. The ReTHINK trial136 sought to answer this question by evaluating the drug in patients with genetically high-risk disease without significant splenomegaly or symptoms, but had to be closed owing to poor accrual. Rational, ruxolitinib-based combinations may be the way forward, but thus far, no clear winner has emerged among the combinations for which clinical data is available. Similarly, the pathogenesis of bone marrow fibrosis remains poorly understood, and although many agents have been investigated, there are no drugs available at present that convincingly improve bone marrow fibrosis in MF.

Mechanisms of resistance to ruxolitinib remain unclear. It has been shown preclinically that MF is intrinsically more resistant to JAK2 inhibition than PV or ET.189 Although resistance-conferring mutations in the kinase domain of JAK2 have been described,190 these are rare and not clinically relevant in most patients. JAK2 inhibitor “persistence” has been described as a mechanism of therapeutic resistance to conventional (type 1) JAK2 inhibitors,191 and has been shown to be overcome by drugs that degrade JAK2, such as HSP90 inhibitors,176 or by “type 2” JAK2 inhibitors that bind to and stabilize the kinase in its inactive conformation;192 however, no such agent is in clinical trials yet. From a clinical drug development perspective, there continues to be a major unmet need for a JAK2 inhibitor that is effective after ruxolitinib failure and/or that can be used safely in severely thrombocytopenic (platelets <50 × 109/L) subjects. Whether pacritinib or NS-018 will fulfill this need remains to be seen. The future role, if any, of JAK1-selective inhibitors such as itacitinib is unclear at this time, given the understandably low rate of SVR. Equally, the data available in the public domain at this time do not suggest that any of the other classes of drugs used as single agents, e.g., imetelstat, PRM-151 or LCL-161 are close to regulatory approval. Finally, the quest for an effective agent for MF-associated anemia, particularly for use in conjunction with ruxolitinib, continues. Enrollment of patients on clinical trials continues to be of paramount importance.

Recent developments in advanced systemic mastocytosis

The multi-kinase inhibitor, midostaurin, was recently approved by the FDA for the treatment of patients with aggressive systemic mastocytosis (ASM), systemic mastocytosis with an antecedent hematologic neoplasm (SM-AHN) and mast cell leukemia (MCL) based on the findings of a single-arm non-comparative trial (n = 116) in which midostaurin produced an ORR of 60% with a major response (complete resolution of at least one type of SM-related organ damage) in 45% of patients.193 The median OS was 28.7 months, and the median progression-free survival, 14.1 months. As midostaurin also inhibits fms-like tyrosine kinase 3 (FLT3), the most common toxicities were gastrointestinal in nature.193 Midostaurin was effective regardless of the presence or absence of KITD816V, advanced SM subtype, or exposure to prior therapies. Although SM is a mutant KIT-driven disorder, most patients with advanced SM (especially patients with SM-AHN) harbor additional mutations.194 Mutations in SRSF2, ASXL1 and RUNX1 (S/A/R) have recently been identified as being associated with inferior OS in advanced SM, as has the number of mutated genes in the so-called S/A/R panel.195,196 Furthermore, these and other mutations (e.g., TET2) have been shown to precede the acquisition of KITD816V.197 Mutational profiling of patients receiving midostaurin on the aforementioned pivotal trial showed that reduction of the KITD816V allele burden by ≥25% at six months predicted for improved OS, while the S/A/R+ genotype and clonal evolution on midostaurin were associated with worse survival and disease progression, respectively.198 The success of midostaurin has spurred the development of other KIT inhibitors for patients with advanced SM. BLU-285 is a potent and highly selective inhibitor of KIT exon 17 mutants, including the D816V mutant found in >80% of patients with SM;199 it also inhibits the common D842V mutant of platelet-derived growth factor receptor alfa (PDGFRA).200 Promising data from an ongoing phase 1 trial (NCT02561988) of this agent in patients with advanced SM have been presented, with improvements in symptoms and C-findings, as well as in objective measures of mast cell burden.201 DCC-2618 is a potent pan-KIT and PDGFR “switch control” inhibitor that is also being studied (NCT02571036) in patients with advanced SM;202 clinical data with this agent are not available yet.

Conclusion

These are exciting times in MPN research. In just the last two years, the field has witnessed major changes such as important modifications to the WHO diagnostic criteria for PV, elucidation of how CALR mutations activate the JAK-STAT pathway in ET and PMF, establishment of a prognostic scoring system specifically for patients with post-PV/ET MF, and the first-ever drug approval for patients with advanced SM. These achievements have been the result of both astute clinical observations and elegant preclinical work. There remain, however, many unanswered questions regarding the biology of MPN and major unmet clinical needs. Hopefully, the coming years will see an even more accelerated pace of discovery for the benefit of our patients.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.DAMESHEK W. Some speculations on the myeloproliferative syndromes. Blood. 1951;6:372–5. [PubMed] [Google Scholar]
  • 2.Arber DA, Orazi A, Hasserjian R, Thiele J, Borowitz MJ, Le Beau MM, Bloomfield CD, Cazzola M, Vardiman JW. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood. 2016;127:2391–405. doi: 10.1182/blood-2016-03-643544. [DOI] [PubMed] [Google Scholar]
  • 3.Maxson JE, Gotlib J, Pollyea DA, Fleischman AG, Agarwal A, Eide CA, Bottomly D, Wilmot B, McWeeney SK, Tognon CE, Pond JB, Collins RH, Goueli B, Oh ST, Deininger MW, Chang BH, Loriaux MM, Druker BJ, Tyner JW. Oncogenic CSF3R mutations in chronic neutrophilic leukemia and atypical CML. N Engl J Med. 2013;368:1781–90. doi: 10.1056/NEJMoa1214514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Rumi E, Milosevic JD, Casetti I, Dambruoso I, Pietra D, Boveri E, Boni M, Bernasconi P, Passamonti F, Kralovics R, Cazzola M. Efficacy of ruxolitinib in chronic eosinophilic leukemia associated with a PCM1-JAK2 fusion gene. J Clin Oncol. 2013;31:e269–71. doi: 10.1200/JCO.2012.46.4370. [DOI] [PubMed] [Google Scholar]
  • 5.Reiter A, Gotlib J. Myeloid neoplasms with eosinophilia. Blood. 2017;129:704–14. doi: 10.1182/blood-2016-10-695973. [DOI] [PubMed] [Google Scholar]
  • 6.Barbui T, Thiele J, Gisslinger H, Finazzi G, Carobbio A, Rumi E, Luigia Randi M, Betozzi I, Vannucchi AM, Pieri L, Carrai V, Gisslinger B, Mullauer L, Ruggeri M, Rambaldi A, Tefferi A. Masked polycythemia vera (mPV): results of an international study. Am J Hematol. 2014;89:52–4. doi: 10.1002/ajh.23585. [DOI] [PubMed] [Google Scholar]
  • 7.Barbui T, Thiele J, Carobbio A, Gisslinger H, Finazzi G, Rumi E, Luigia Randi M, Vannucchi AM, Gisslinger B, Mullauer L, Ruggeri M, Rambaldi A, Tefferi A. Masked polycythemia vera diagnosed according to WHO and BCSH classification. Am J Hematol. 2014;89:199–202. doi: 10.1002/ajh.23617. [DOI] [PubMed] [Google Scholar]
  • 8.Lussana F, Carobbio A, Randi ML, Elena C, Rumi E, Finazzi G, Bertozzi I, Pieri L, Ruggeri M, Palandri F, Polverelli N, Elli E, Tieghi A, Iurlo A, Ruella M, Cazzola M, Rambaldi A, Vannucchi AM, Barbui T. A lower intensity of treatment may underlie the increased risk of thrombosis in young patients with masked polycythaemia vera. Br J Haematol. 2014;167:541–6. doi: 10.1111/bjh.13080. [DOI] [PubMed] [Google Scholar]
  • 9.Silver RT, Chow W, Orazi A, Arles SP, Goldsmith SJ. Evaluation of WHO criteria for diagnosis of polycythemia vera: a prospective analysis. Blood. 2013;122:1881–6. doi: 10.1182/blood-2013-06-508416. [DOI] [PubMed] [Google Scholar]
  • 10.Spivak JL. Myeloproliferative Neoplasms. N Engl J Med. 2017;376:2168–81. doi: 10.1056/NEJMra1406186. [DOI] [PubMed] [Google Scholar]
  • 11.Barbui T, Thiele J, Carobbio A, Guglielmelli P, Rambaldi A, Vannucchi AM, Tefferi A. Discriminating between essential thrombocythemia and masked polycythemia vera in JAK2 mutated patients. Am J Hematol. 2014;89:588–90. doi: 10.1002/ajh.23694. [DOI] [PubMed] [Google Scholar]
  • 12.Barbui T, Thiele J, Vannucchi AM, Tefferi A. Rethinking the diagnostic criteria of polycythemia vera. Leukemia. 2014;28:1191–5. doi: 10.1038/leu.2013.380. [DOI] [PubMed] [Google Scholar]
  • 13.Vannucchi AM. How I treat polycythemia vera. Blood. 2014;124:3212–20. doi: 10.1182/blood-2014-07-551929. [DOI] [PubMed] [Google Scholar]
  • 14.Barosi G, Birgegard G, Finazzi G, Griesshammer M, Harrison C, Hasselbalch H, Kiladijan JJ, Lengfelder E, Mesa R, Mc Mullin MF, Passamonti F, Reilly JT, Vannucchi AM, Barbui T. A unified definition of clinical resistance and intolerance to hydroxycarbamide in polycythaemia vera and primary myelofibrosis: results of a European LeukemiaNet (ELN) consensus process. Br J Haematol. 2010;148:961–3. doi: 10.1111/j.1365-2141.2009.08019.x. [DOI] [PubMed] [Google Scholar]
  • 15.Vannucchi AM, Kiladjian JJ, Griesshammer M, Masszi T, Durrant S, Passamonti F, Harrison CN, Pane F, Zachee P, Mesa R, He S, Jones MM, Garrett W, Li J, Pirron U, Habr D, Verstovsek S. Ruxolitinib versus standard therapy for the treatment of polycythemia vera. N Engl J Med. 2015;372:426–35. doi: 10.1056/NEJMoa1409002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Verstovsek S, Vannucchi AM, Griesshammer M, Masszi T, Durrant S, Passamonti F, Harrison CN, Pane F, Zachee P, Kirito K, Besses C, Hino M, Moiraghi B, Miller CB, Cazzola M, Rosti V, Blau I, Mesa R, Jones MM, Zhen H, Li J, Francillard N, Habr D, Kiladjian JJ. Ruxolitinib versus best available therapy in patients with polycythemia vera: 80-week follow-up from the RESPONSE trial. Haematologica. 2016;101:821–9. doi: 10.3324/haematol.2016.143644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Passamonti F, Griesshammer M, Palandri F, Egyed M, Benevolo G, Devos T, Callum J, Vannucchi AM, Sivgin S, Bensasson C, Khan M, Mounedji N, Saydam G. Ruxolitinib for the treatment of inadequately controlled polycythaemia vera without splenomegaly (RESPONSE-2): a randomised, open-label, phase 3b study. Lancet Oncol. 2017;18:88–99. doi: 10.1016/S1470-2045(16)30558-7. [DOI] [PubMed] [Google Scholar]
  • 18.Mullally A, Bruedigam C, Poveromo L, Heidel FH, Purdon A, Vu T, Austin R, Heckl D, Breyfogle LJ, Kuhn CP, Kalaitzidis D, Armstrong SA, Williams DA, Hill GR, Ebert BL, Lane SW. Depletion of Jak2V617F myeloproliferative neoplasm-propagating stem cells by interferon-alpha in a murine model of polycythemia vera. Blood. 2013;121:3692–702. doi: 10.1182/blood-2012-05-432989. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Kiladjian JJ, Cassinat B, Chevret S, Turlure P, Cambier N, Roussel M, Bellucci S, Grandchamp B, Chomienne C, Fenaux P. Pegylated interferon-alfa-2a induces complete hematologic and molecular responses with low toxicity in polycythemia vera. Blood. 2008;112:3065–72. doi: 10.1182/blood-2008-03-143537. [DOI] [PubMed] [Google Scholar]
  • 20.Mesa RA, Jamieson C, Bhatia R, Deininger MW, Fletcher CD, Gerds AT, Gojo I, Gotlib J, Gundabolu K, Hobbs G, McMahon B, Mohan SR, Oh S, Padron E, Papadantonakis N, Pancari P, Podoltsev N, Rampal R, Ranheim E, Reddy V, Rein LAM, Scott B, Snyder DS, Stein BL, Talpaz M, Verstovsek S, Wadleigh M, Wang ES, Bergman MA, Gregory KM, Sundar H. NCCN Guidelines Insights: Myeloproliferative Neoplasms, Version 2.2018. J Natl Compr Canc Netw. 2017;15:1193–207. doi: 10.6004/jnccn.2017.0157. [DOI] [PubMed] [Google Scholar]
  • 21.Vannucchi AM, Barbui T, Cervantes F, Harrison C, Kiladjian JJ, Kroger N, Thiele J, Buske C ESMO Guidelines Committee. Philadelphia chromosome-negative chronic myeloproliferative neoplasms: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2015;26(Suppl 5):v85–99. doi: 10.1093/annonc/mdv203. [DOI] [PubMed] [Google Scholar]
  • 22.Masarova L, Patel KP, Newberry KJ, Cortes J, Borthakur G, Konopleva M, Estrov Z, Kantarjian H, Verstovsek S. Pegylated interferon alfa-2a in patients with essential thrombocythaemia or polycythaemia vera: a post-hoc, median 83 month follow-up of an open-label, phase 2 trial. Lancet Haematol. 2017 doi: 10.1016/S2352-3026(17)30030-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Gisslinger H, Zagrijtschuk O, Buxhofer-Ausch V, Thaler J, Schloegl E, Gastl GA, Wolf D, Kralovics R, Gisslinger B, Strecker K, Egle A, Melchardt T, Burgstaller S, Willenbacher E, Schalling M, Them NC, Kadlecova P, Klade C, Greil R. Ropeginterferon alfa-2b, a novel IFNalpha-2b, induces high response rates with low toxicity in patients with polycythemia vera. Blood. 2015;126:1762–9. doi: 10.1182/blood-2015-04-637280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Gisslinger H, Klade C, Georgiev P, Skotnicki A, Gercheva-Kyuchukova L, Egyed M, Rossiev V, Dulicek P, Illes A, Pylypenko H, Sivcheva L, Mayer J, Grohmann-Izay B, Hasselbalch H, Kralovics R, Kiladjian J. Final Results from PROUD-PV a Randomized Controlled Phase 3 Trial Comparing Ropeginterferon Alfa-2b to Hydroxyurea in Polycythemia Vera Patients. Blood. 2016;128:475-. [Google Scholar]
  • 25.Mascarenhas JO, Prchal JT, Rambaldi A, Mesa RA, Berenzon D, Yacoub A, Harrison CN, McMullin MF, Vannucchi AM, Ewing JC, O'Connell C, Kiladjian J, Mead A, Winton EF, Leibowitz DS, De Stefano V, Arcasoy MO, Kessler CM, Catchatorian R, Rondelli D, Silver RT, Ritchie EK, Nagler A, Kremyanskaya M, Schlenk RF, Weinberg RS, Salama ME, Tognoni G, Prosperini G, Di Lelio A, Serone E, Marfisi L, Kleczko J, Kosiorek HE, Barbui T, Dueck AC, Hoffman R. Interim Analysis of the Myeloproliferative Disorders Research Consortium (MPD-RC) 112 Global Phase III Trial of Front Line Pegylated Interferon Alpha-2a Vs. Hydroxyurea in High Risk Polycythemia Vera and Essential Thrombocythemia. Blood. 2016;128:479-. [Google Scholar]
  • 26.Wernig G, Mercher T, Okabe R, Levine RL, Lee BH, Gilliland DG. Expression of Jak2V617F causes a polycythemia vera-like disease with associated myelofibrosis in a murine bone marrow transplant model. Blood. 2006;107:4274–81. doi: 10.1182/blood-2005-12-4824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Akada H, Yan D, Zou H, Fiering S, Hutchison RE, Mohi MG. Conditional expression of heterozygous or homozygous Jak2V617F from its endogenous promoter induces a polycythemia vera-like disease. Blood. 2010;115:3589–97. doi: 10.1182/blood-2009-04-215848. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Marty C, Lacout C, Martin A, Hasan S, Jacquot S, Birling MC, Vainchenker W, Villeval JL. Myeloproliferative neoplasm induced by constitutive expression of JAK2V617F in knock-in mice. Blood. 2010;116:783–7. doi: 10.1182/blood-2009-12-257063. [DOI] [PubMed] [Google Scholar]
  • 29.Mullally A, Lane SW, Ball B, Megerdichian C, Okabe R, Al-Shahrour F, Paktinat M, Haydu JE, Housman E, Lord AM, Wernig G, Kharas MG, Mercher T, Kutok JL, Gilliland DG, Ebert BL. Physiological Jak2V617F expression causes a lethal myeloproliferative neoplasm with differential effects on hematopoietic stem and progenitor cells. Cancer Cell. 2010;17:584–96. doi: 10.1016/j.ccr.2010.05.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Li J, Kent DG, Godfrey AL, Manning H, Nangalia J, Aziz A, Chen E, Saeb-Parsy K, Fink J, Sneade R, Hamilton TL, Pask DC, Silber Y, Zhao X, Ghevaert C, Liu P, Green AR. JAK2V617F homozygosity drives a phenotypic switch in myeloproliferative neoplasms, but is insufficient to sustain disease. Blood. 2014;123:3139–51. doi: 10.1182/blood-2013-06-510222. [DOI] [PubMed] [Google Scholar]
  • 31.Godfrey AL, Chen E, Pagano F, Ortmann CA, Silber Y, Bellosillo B, Guglielmelli P, Harrison CN, Reilly JT, Stegelmann F, Bijou F, Lippert E, McMullin MF, Boiron JM, Dohner K, Vannucchi AM, Besses C, Campbell PJ, Green AR. JAK2V617F homozygosity arises commonly and recurrently in PV and ET, but PV is characterized by expansion of a dominant homozygous subclone. Blood. 2012;120:2704–7. doi: 10.1182/blood-2012-05-431791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Ortmann CA, Kent DG, Nangalia J, Silber Y, Wedge DC, Grinfeld J, Baxter EJ, Massie CE, Papaemmanuil E, Menon S, Godfrey AL, Dimitropoulou D, Guglielmelli P, Bellosillo B, Besses C, Dohner K, Harrison CN, Vassiliou GS, Vannucchi A, Campbell PJ, Green AR. Effect of mutation order on myeloproliferative neoplasms. N Engl J Med. 2015;372:601–12. doi: 10.1056/NEJMoa1412098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Godfrey AL, Chen E, Pagano F, Silber Y, Campbell PJ, Green AR. Clonal analyses reveal associations of JAK2V617F homozygosity with hematologic features, age and gender in polycythemia vera and essential thrombocythemia. Haematologica. 2013;98:718–21. doi: 10.3324/haematol.2012.079129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Spivak JL, Considine M, Williams DM, Talbot CC, Jr, Rogers O, Moliterno AR, Jie C, Ochs MF. Two clinical phenotypes in polycythemia vera. N Engl J Med. 2014;371:808–17. doi: 10.1056/NEJMoa1403141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Tefferi A, Lasho TL, Guglielmelli P, Finke CM, Rotunno G, Elala Y, Pacilli A, Hanson CA, Pancrazzi A, Ketterling RP, Mannarelli C, Barraco D, Fanelli T, Pardanani A, Gangat N, Vannucchi AM. Targeted deep sequencing in polycythemia vera and essential thrombocythemia. Blood Advances. 2016;1:21–30. doi: 10.1182/bloodadvances.2016000216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Barbui T, Masciulli A, Marfisi MR, Tognoni G, Finazzi G, Rambaldi A, Vannucchi A. White blood cell counts and thrombosis in polycythemia vera: a subanalysis of the CYTO-PV study. Blood. 2015;126:560–1. doi: 10.1182/blood-2015-04-638593. [DOI] [PubMed] [Google Scholar]
  • 37.Gangat N, Strand J, Li CY, Wu W, Pardanani A, Tefferi A. Leucocytosis in polycythaemia vera predicts both inferior survival and leukaemic transformation. Br J Haematol. 2007;138:354–8. doi: 10.1111/j.1365-2141.2007.06674.x. [DOI] [PubMed] [Google Scholar]
  • 38.Landolfi R, Di Gennaro L, Barbui T, De Stefano V, Finazzi G, Marfisi R, Tognoni G, Marchioli R European Collaboration on Low-Dose Aspirin in Polycythemia Vera (ECLAP) Leukocytosis as a major thrombotic risk factor in patients with polycythemia vera. Blood. 2007;109:2446–52. doi: 10.1182/blood-2006-08-042515. [DOI] [PubMed] [Google Scholar]
  • 39.Passamonti F, Rumi E, Pietra D, Elena C, Boveri E, Arcaini L, Roncoroni E, Astori C, Merli M, Boggi S, Pascutto C, Lazzarino M, Cazzola M. A prospective study of 338 patients with polycythemia vera: the impact of JAK2 (V617F) allele burden and leukocytosis on fibrotic or leukemic disease transformation and vascular complications. Leukemia. 2010;24:1574–9. doi: 10.1038/leu.2010.148. [DOI] [PubMed] [Google Scholar]
  • 40.Tefferi A, Rumi E, Finazzi G, Gisslinger H, Vannucchi AM, Rodeghiero F, Randi ML, Vaidya R, Cazzola M, Rambaldi A, Gisslinger B, Pieri L, Ruggeri M, Bertozzi I, Sulai NH, Casetti I, Carobbio A, Jeryczynski G, Larson DR, Mullauer L, Pardanani A, Thiele J, Passamonti F, Barbui T. Survival and prognosis among 1545 patients with contemporary polycythemia vera: an international study. Leukemia. 2013;27:1874–81. doi: 10.1038/leu.2013.163. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Caramazza D, Caracciolo C, Barone R, Malato A, Saccullo G, Cigna V, Berretta S, Schinocca L, Quintini G, Abbadessa V, Di Raimondo F, Siragusa S. Correlation between leukocytosis and thrombosis in Philadelphia-negative chronic myeloproliferative neoplasms. Ann Hematol. 2009;88:967–71. doi: 10.1007/s00277-009-0706-x. [DOI] [PubMed] [Google Scholar]
  • 42.Lu M, Wang X, Li Y, Tripodi J, Mosoyan G, Mascarenhas J, Kremyanskaya M, Najfeld V, Hoffman R. Combination treatment in vitro with Nutlin, a small-molecule antagonist of MDM2, and pegylated interferon-alpha 2a specifically targets JAK2V617F-positive polycythemia vera cells. Blood. 2012;120:3098–105. doi: 10.1182/blood-2012-02-410712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Lu M, Xia L, Li Y, Wang X, Hoffman R. The orally bioavailable MDM2 antagonist RG7112 and pegylated interferon alpha 2a target JAK2V617F-positive progenitor and stem cells. Blood. 2014;124:771–9. doi: 10.1182/blood-2013-11-536854. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Bose P, Verstovsek S. Investigational histone deacetylase inhibitors (HDACi) in myeloproliferative neoplasms. Expert Opin Investig Drugs. 2016;25:1393–403. doi: 10.1080/13543784.2016.1250882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Klampfl T, Gisslinger H, Harutyunyan AS, Nivarthi H, Rumi E, Milosevic JD, Them NC, Berg T, Gisslinger B, Pietra D, Chen D, Vladimer GI, Bagienski K, Milanesi C, Casetti IC, Sant'Antonio E, Ferretti V, Elena C, Schischlik F, Cleary C, Six M, Schalling M, Schonegger A, Bock C, Malcovati L, Pascutto C, Superti-Furga G, Cazzola M, Kralovics R. Somatic mutations of calreticulin in myeloproliferative neoplasms. N Engl J Med. 2013;369:2379–90. doi: 10.1056/NEJMoa1311347. [DOI] [PubMed] [Google Scholar]
  • 46.Nangalia J, Massie CE, Baxter EJ, Nice FL, Gundem G, Wedge DC, Avezov E, Li J, Kollmann K, Kent DG, Aziz A, Godfrey AL, Hinton J, Martincorena I, Van Loo P, Jones AV, Guglielmelli P, Tarpey P, Harding HP, Fitzpatrick JD, Goudie CT, Ortmann CA, Loughran SJ, Raine K, Jones DR, Butler AP, Teague JW, O'Meara S, McLaren S, Bianchi M, Silber Y, Dimitropoulou D, Bloxham D, Mudie L, Maddison M, Robinson B, Keohane C, Maclean C, Hill K, Orchard K, Tauro S, Du MQ, Greaves M, Bowen D, Huntly BJ, Harrison CN, Cross NC, Ron D, Vannucchi AM, Papaemmanuil E, Campbell PJ, Green AR. Somatic CALR mutations in myeloproliferative neoplasms with nonmutated JAK2. N Engl J Med. 2013;369:2391–405. doi: 10.1056/NEJMoa1312542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Araki M, Yang Y, Masubuchi N, Hironaka Y, Takei H, Morishita S, Mizukami Y, Kan S, Shirane S, Edahiro Y, Sunami Y, Ohsaka A, Komatsu N. Activation of the thrombopoietin receptor by mutant calreticulin in CALR-mutant myeloproliferative neoplasms. Blood. 2016;127:1307–16. doi: 10.1182/blood-2015-09-671172. [DOI] [PubMed] [Google Scholar]
  • 48.Chachoua I, Pecquet C, El-Khoury M, Nivarthi H, Albu RI, Marty C, Gryshkova V, Defour JP, Vertenoeil G, Ngo A, Koay A, Raslova H, Courtoy PJ, Choong ML, Plo I, Vainchenker W, Kralovics R, Constantinescu SN. Thrombopoietin receptor activation by myeloproliferative neoplasm associated calreticulin mutants. Blood. 2016;127:1325–35. doi: 10.1182/blood-2015-11-681932. [DOI] [PubMed] [Google Scholar]
  • 49.Elf S, Abdelfattah NS, Chen E, Perales-Paton J, Rosen EA, Ko A, Peisker F, Florescu N, Giannini S, Wolach O, Morgan EA, Tothova Z, Losman JA, Schneider RK, Al-Shahrour F, Mullally A. Mutant Calreticulin Requires Both Its Mutant C-terminus and the Thrombopoietin Receptor for Oncogenic Transformation. Cancer Discov. 2016;6:368–81. doi: 10.1158/2159-8290.CD-15-1434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Marty C, Pecquet C, Nivarthi H, El-Khoury M, Chachoua I, Tulliez M, Villeval JL, Raslova H, Kralovics R, Constantinescu SN, Plo I, Vainchenker W. Calreticulin mutants in mice induce an MPL-dependent thrombocytosis with frequent progression to myelofibrosis. Blood. 2016;127:1317–24. doi: 10.1182/blood-2015-11-679571. [DOI] [PubMed] [Google Scholar]
  • 51.Cavalloni C, Rumi E, Ferretti VV, Pietra D, Roncoroni E, Bellini M, Ciboddo M, Casetti IC, Landini B, Fugazza E, Troletti D, Astori C, Cazzola M. Sequential evaluation of CALR mutant burden in patients with myeloproliferative neoplasms. Oncotarget. 2017;8:33416–21. doi: 10.18632/oncotarget.16797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Rotunno G, Mannarelli C, Guglielmelli P, Pacilli A, Pancrazzi A, Pieri L, Fanelli T, Bosi A, Vannucchi AM Associazione Italiana per la Ricerca sul Cancro Gruppo Italiano Malattie Mieloproliferative Investigators. Impact of calreticulin mutations on clinical and hematological phenotype and outcome in essential thrombocythemia. Blood. 2014;123:1552–5. doi: 10.1182/blood-2013-11-538983. [DOI] [PubMed] [Google Scholar]
  • 53.Rumi E, Pietra D, Ferretti V, Klampfl T, Harutyunyan AS, Milosevic JD, Them NC, Berg T, Elena C, Casetti IC, Milanesi C, Sant'antonio E, Bellini M, Fugazza E, Renna MC, Boveri E, Astori C, Pascutto C, Kralovics R, Cazzola M Associazione Italiana per la Ricerca sul Cancro Gruppo Italiano Malattie Mieloproliferative Investigators. JAK2 or CALR mutation status defines subtypes of essential thrombocythemia with substantially different clinical course and outcomes. Blood. 2014;123:1544–51. doi: 10.1182/blood-2013-11-539098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Tefferi A, Wassie EA, Lasho TL, Finke C, Belachew AA, Ketterling RP, Hanson CA, Pardanani A, Gangat N, Wolanskyj AP. Calreticulin mutations and long-term survival in essential thrombocythemia. Leukemia. 2014;28:2300–3. doi: 10.1038/leu.2014.148. [DOI] [PubMed] [Google Scholar]
  • 55.Alvarez-Larran A, Pereira A, Guglielmelli P, Hernandez-Boluda JC, Arellano-Rodrigo E, Ferrer-Marin F, Samah A, Griesshammer M, Kerguelen A, Andreasson B, Burgaleta C, Schwarz J, Garcia-Gutierrez V, Ayala R, Barba P, Gomez-Casares MT, Paoli C, Drexler B, Zweegman S, McMullin MF, Samuelsson J, Harrison C, Cervantes F, Vannucchi AM, Besses C. Antiplatelet therapy versus observation in low-risk essential thrombocythemia with a CALR mutation. Haematologica. 2016;101:926–31. doi: 10.3324/haematol.2016.146654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Barbui T, Vannucchi AM, Buxhofer-Ausch V, De Stefano V, Betti S, Rambaldi A, Rumi E, Ruggeri M, Rodeghiero F, Randi ML, Bertozzi I, Gisslinger H, Finazzi G, Carobbio A, Thiele J, Passamonti F, Falcone C, Tefferi A. Practice-relevant revision of IPSET-thrombosis based on 1019 patients with WHO-defined essential thrombocythemia. Blood Cancer J. 2015;5:e369. doi: 10.1038/bcj.2015.94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Passamonti F, Mora B, Giorgino T, Guglielmelli P, Cazzola M, Maffioli M, Rambaldi A, Caramella M, Komrokji R, Gotlib J, Kiladjian JJ, Cervantes F, Devos T, Palandri F, De Stefano V, Ruggeri M, Silver R, Benevolo G, Albano F, Caramazza D, Rumi E, Merli M, Pietra D, Casalone R, Barbui T, Pieri L, Vannucchi AM. Driver mutations' effect in secondary myelofibrosis: an international multicenter study based on 781 patients. Leukemia. 2017;31:970–3. doi: 10.1038/leu.2016.351. [DOI] [PubMed] [Google Scholar]
  • 58.Pietra D, Rumi E, Ferretti VV, Di Buduo CA, Milanesi C, Cavalloni C, Sant'Antonio E, Abbonante V, Moccia F, Casetti IC, Bellini M, Renna MC, Roncoroni E, Fugazza E, Astori C, Boveri E, Rosti V, Barosi G, Balduini A, Cazzola M. Differential clinical effects of different mutation subtypes in CALR-mutant myeloproliferative neoplasms. Leukemia. 2016;30:431–8. doi: 10.1038/leu.2015.277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Elala YC, Lasho TL, Gangat N, Finke C, Barraco D, Haider M, Abou Hussein AK, Hanson CA, Ketterling RP, Pardanani A, Tefferi A. Calreticulin variant stratified driver mutational status and prognosis in essential thrombocythemia. Am J Hematol. 2016;91:503–6. doi: 10.1002/ajh.24338. [DOI] [PubMed] [Google Scholar]
  • 60.Passamonti F, Rumi E, Pungolino E, Malabarba L, Bertazzoni P, Valentini M, Orlandi E, Arcaini L, Brusamolino E, Pascutto C, Cazzola M, Morra E, Lazzarino M. Life expectancy and prognostic factors for survival in patients with polycythemia vera and essential thrombocythemia. Am J Med. 2004;117:755–61. doi: 10.1016/j.amjmed.2004.06.032. [DOI] [PubMed] [Google Scholar]
  • 61.Tefferi A, Guglielmelli P, Larson DR, Finke C, Wassie EA, Pieri L, Gangat N, Fjerza R, Belachew AA, Lasho TL, Ketterling RP, Hanson CA, Rambaldi A, Finazzi G, Thiele J, Barbui T, Pardanani A, Vannucchi AM. Long-term survival and blast transformation in molecularly annotated essential thrombocythemia, polycythemia vera, and myelofibrosis. Blood. 2014;124:2507–13. doi: 10.1182/blood-2014-05-579136. quiz 2615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Haider M, Gangat N, Lasho T, Abou Hussein AK, Elala YC, Hanson C, Tefferi A. Validation of the revised International Prognostic Score of Thrombosis for Essential Thrombocythemia (IPSET-thrombosis) in 585 Mayo Clinic patients. Am J Hematol. 2016;91:390–4. doi: 10.1002/ajh.24293. [DOI] [PubMed] [Google Scholar]
  • 63.Finazzi G, Carobbio A, Guglielmelli P, Cavalloni C, Salmoiraghi S, Vannucchi AM, Cazzola M, Passamonti F, Rambaldi A, Barbui T. Calreticulin mutation does not modify the IPSET score for predicting the risk of thrombosis among 1150 patients with essential thrombocythemia. Blood. 2014;124:2611–2. doi: 10.1182/blood-2014-08-596676. [DOI] [PubMed] [Google Scholar]
  • 64.Passamonti F, Thiele J, Girodon F, Rumi E, Carobbio A, Gisslinger H, Kvasnicka HM, Ruggeri M, Randi ML, Gangat N, Vannucchi AM, Gianatti A, Gisslinger B, Mullauer L, Rodeghiero F, d'Amore ES, Bertozzi I, Hanson CA, Boveri E, Marino F, Maffioli M, Caramazza D, Antonioli E, Carrai V, Buxhofer-Ausch V, Pascutto C, Cazzola M, Barbui T, Tefferi A. A prognostic model to predict survival in 867 World Health Organization-defined essential thrombocythemia at diagnosis: a study by the International Working Group on Myelofibrosis Research and Treatment. Blood. 2012;120:1197–201. doi: 10.1182/blood-2012-01-403279. [DOI] [PubMed] [Google Scholar]
  • 65.Campbell PJ, MacLean C, Beer PA, Buck G, Wheatley K, Kiladjian JJ, Forsyth C, Harrison CN, Green AR. Correlation of blood counts with vascular complications in essential thrombocythemia: analysis of the prospective PT1 cohort. Blood. 2012;120:1409–11. doi: 10.1182/blood-2012-04-424911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Carobbio A, Antonioli E, Guglielmelli P, Vannucchi AM, Delaini F, Guerini V, Finazzi G, Rambaldi A, Barbui T. Leukocytosis and risk stratification assessment in essential thrombocythemia. J Clin Oncol. 2008;26:2732–6. doi: 10.1200/JCO.2007.15.3569. [DOI] [PubMed] [Google Scholar]
  • 67.Carobbio A, Finazzi G, Antonioli E, Vannucchi AM, Barosi G, Ruggeri M, Rodeghiero F, Delaini F, Rambaldi A, Barbui T. Hydroxyurea in essential thrombocythemia: rate and clinical relevance of responses by European LeukemiaNet criteria. Blood. 2010;116:1051–5. doi: 10.1182/blood-2010-03-272179. [DOI] [PubMed] [Google Scholar]
  • 68.Carobbio A, Thiele J, Passamonti F, Rumi E, Ruggeri M, Rodeghiero F, Randi ML, Bertozzi I, Vannucchi AM, Antonioli E, Gisslinger H, Buxhofer-Ausch V, Finazzi G, Gangat N, Tefferi A, Barbui T. Risk factors for arterial and venous thrombosis in WHO-defined essential thrombocythemia: an international study of 891 patients. Blood. 2011;117:5857–9. doi: 10.1182/blood-2011-02-339002. [DOI] [PubMed] [Google Scholar]
  • 69.Tefferi A, Betti S, Barraco D, Mudireddy M, Shah S, Hanson CA, Ketterling RP, Pardanani A, Gangat N, Coltro G, Guglielmelli P, Vannucchi AM. Gender and survival in essential thrombocythemia: A two-center study of 1,494 patients. Am J Hematol. 2017 doi: 10.1002/ajh.24882. [DOI] [PubMed] [Google Scholar]
  • 70.Mudireddy M, Barraco D, Hanson CA, Pardanani A, Gangat N, Tefferi A. The prognostic relevance of serum lactate dehydrogenase and mild bone marrow reticulin fibrosis in essential thrombocythemia. Am J Hematol. 2017;92:454–9. doi: 10.1002/ajh.24689. [DOI] [PubMed] [Google Scholar]
  • 71.Harrison CN, Campbell PJ, Buck G, Wheatley K, East CL, Bareford D, Wilkins BS, van der Walt JD, Reilly JT, Grigg AP, Revell P, Woodcock BE, Green AR United Kingdom Medical Research Council Primary Thrombocythemia 1 Study. Hydroxyurea compared with anagrelide in high-risk essential thrombocythemia. N Engl J Med. 2005;353:33–45. doi: 10.1056/NEJMoa043800. [DOI] [PubMed] [Google Scholar]
  • 72.Gisslinger H, Gotic M, Holowiecki J, Penka M, Thiele J, Kvasnicka HM, Kralovics R, Petrides PE ANAHYDRET Study Group. Anagrelide compared with hydroxyurea in WHO-classified essential thrombocythemia: the ANAHYDRET Study, a randomized controlled trial. Blood. 2013;121:1720–8. doi: 10.1182/blood-2012-07-443770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Barosi G, Besses C, Birgegard G, Briere J, Cervantes F, Finazzi G, Gisslinger H, Griesshammer M, Gugliotta L, Harrison C, Hasselbalch H, Lengfelder E, Reilly JT, Michiels JJ, Barbui T. A unified definition of clinical resistance/intolerance to hydroxyurea in essential thrombocythemia: results of a consensus process by an international working group. Leukemia. 2007;21:277–80. doi: 10.1038/sj.leu.2404473. [DOI] [PubMed] [Google Scholar]
  • 74.Mesa RA, Niblack J, Wadleigh M, Verstovsek S, Camoriano J, Barnes S, Tan AD, Atherton PJ, Sloan JA, Tefferi A. The burden of fatigue and quality of life in myeloproliferative disorders (MPDs): an international Internet-based survey of 1179 MPD patients. Cancer. 2007;109:68–76. doi: 10.1002/cncr.22365. [DOI] [PubMed] [Google Scholar]
  • 75.Harrison CN, Koschmieder S, Foltz L, Guglielmelli P, Flindt T, Koehler M, Mathias J, Komatsu N, Boothroyd RN, Spierer A, Perez Ronco J, Taylor-Stokes G, Waller J, Mesa RA. The impact of myeloproliferative neoplasms (MPNs) on patient quality of life and productivity: results from the international MPN Landmark survey. Ann Hematol. 2017 doi: 10.1007/s00277-017-3082-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Verstovsek S, Passamonti F, Rambaldi A, Barosi G, Rumi E, Gattoni E, Pieri L, Zhen H, Granier M, Assad A, Cazzola M, Kantarjian HM, Barbui T, Vannucchi AM. Ruxolitinib for essential thrombocythemia refractory to or intolerant of hydroxyurea: long-term phase 2 study results. Blood. 2017;130:1768–71. doi: 10.1182/blood-2017-02-765032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Harrison CN, Mead AJ, Panchal A, Fox S, Yap C, Gbandi E, Houlton A, Alimam S, Ewing J, Wood M, Chen F, Coppell J, Panoskaltsis N, Knapper S, Ali S, Hamblin A, Scherber R, Dueck AC, Cross NCP, Mesa R, McMullin MF. Ruxolitinib versus best available therapy for ET intolerant or resistant to hydroxycarbamide in a randomized trial. Blood. 2017 doi: 10.1182/blood-2017-05-785790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Shide K, Shimoda HK, Kumano T, Karube K, Kameda T, Takenaka K, Oku S, Abe H, Katayose KS, Kubuki Y, Kusumoto K, Hasuike S, Tahara Y, Nagata K, Matsuda T, Ohshima K, Harada M, Shimoda K. Development of ET, primary myelofibrosis and PV in mice expressing JAK2 V617F. Leukemia. 2008;22:87–95. doi: 10.1038/sj.leu.2405043. [DOI] [PubMed] [Google Scholar]
  • 79.Li J, Spensberger D, Ahn JS, Anand S, Beer PA, Ghevaert C, Chen E, Forrai A, Scott LM, Ferreira R, Campbell PJ, Watson SP, Liu P, Erber WN, Huntly BJ, Ottersbach K, Green AR. JAK2 V617F impairs hematopoietic stem cell function in a conditional knock-in mouse model of JAK2 V617F-positive essential thrombocythemia. Blood. 2010;116:1528–38. doi: 10.1182/blood-2009-12-259747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Hobbs CM, Manning H, Bennett C, Vasquez L, Severin S, Brain L, Mazharian A, Guerrero JA, Li J, Soranzo N, Green AR, Watson SP, Ghevaert C. JAK2V617F leads to intrinsic changes in platelet formation and reactivity in a knock-in mouse model of essential thrombocythemia. Blood. 2013;122:3787–97. doi: 10.1182/blood-2013-06-501452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Koppikar P, Abdel-Wahab O, Hedvat C, Marubayashi S, Patel J, Goel A, Kucine N, Gardner JR, Combs AP, Vaddi K, Haley PJ, Burn TC, Rupar M, Bromberg JF, Heaney ML, de Stanchina E, Fridman JS, Levine RL. Efficacy of the JAK2 inhibitor INCB16562 in a murine model of MPLW515L-induced thrombocytosis and myelofibrosis. Blood. 2010;115:2919–27. doi: 10.1182/blood-2009-04-218842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Shide K, Kameda T, Yamaji T, Sekine M, Inada N, Kamiunten A, Akizuki K, Nakamura K, Hidaka T, Kubuki Y, Shimoda H, Kitanaka A, Honda A, Sawaguchi A, Abe H, Miike T, Iwakiri H, Tahara Y, Sueta M, Hasuike S, Yamamoto S, Nagata K, Shimoda K. Calreticulin mutant mice develop essential thrombocythemia that is ameliorated by the JAK inhibitor ruxolitinib. Leukemia. 2016 doi: 10.1038/leu.2016.308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Duek A, Lundberg P, Shimizu T, Grisouard J, Karow A, Kubovcakova L, Hao-Shen H, Dirnhofer S, Skoda RC. Loss of Stat1 decreases megakaryopoiesis and favors erythropoiesis in a JAK2-V617F-driven mouse model of MPNs. Blood. 2014;123:3943–50. doi: 10.1182/blood-2013-07-514208. [DOI] [PubMed] [Google Scholar]
  • 84.Rumi E, Cazzola M. How I treat essential thrombocythemia. Blood. 2016;128:2403–14. doi: 10.1182/blood-2016-05-643346. [DOI] [PubMed] [Google Scholar]
  • 85.Falchi L, Bose P, Newberry KJ, Verstovsek S. Approach to patients with essential thrombocythaemia and very high platelet counts: what is the evidence for treatment? Br J Haematol. 2017;176:352–64. doi: 10.1111/bjh.14443. [DOI] [PubMed] [Google Scholar]
  • 86.Barbui T, Thiele J, Vannucchi AM, Tefferi A. Problems and pitfalls regarding WHO-defined diagnosis of early/prefibrotic primary myelofibrosis versus essential thrombocythemia. Leukemia. 2013;27:1953–8. doi: 10.1038/leu.2013.74. [DOI] [PubMed] [Google Scholar]
  • 87.Barbui T, Thiele J, Passamonti F, Rumi E, Boveri E, Ruggeri M, Rodeghiero F, d'Amore ES, Randi ML, Bertozzi I, Marino F, Vannucchi AM, Antonioli E, Carrai V, Gisslinger H, Buxhofer-Ausch V, Mullauer L, Carobbio A, Gianatti A, Gangat N, Hanson CA, Tefferi A. Survival and disease progression in essential thrombocythemia are significantly influenced by accurate morphologic diagnosis: an international study. J Clin Oncol. 2011;29:3179–84. doi: 10.1200/JCO.2010.34.5298. [DOI] [PubMed] [Google Scholar]
  • 88.Barbui T, Thiele J, Carobbio A, Passamonti F, Rumi E, Randi ML, Bertozzi I, Vannucchi AM, Gisslinger H, Gisslinger B, Finazzi G, Ruggeri M, Rodeghiero F, Rambaldi A, Gangat N, Tefferi A. Disease characteristics and clinical outcome in young adults with essential thrombocythemia versus early/prefibrotic primary myelofibrosis. Blood. 2012;120:569–71. doi: 10.1182/blood-2012-01-407981. [DOI] [PubMed] [Google Scholar]
  • 89.Thiele J, Kvasnicka HM, Mullauer L, Buxhofer-Ausch V, Gisslinger B, Gisslinger H. Essential thrombocythemia versus early primary myelofibrosis: a multicenter study to validate the WHO classification. Blood. 2011;117:5710–8. doi: 10.1182/blood-2010-07-293761. [DOI] [PubMed] [Google Scholar]
  • 90.Rupoli S, Goteri G, Picardi P, Micucci G, Canafoglia L, Scortechini AR, Federici I, Giantomassi F, Da Lio L, Zizzi A, Honorati E, Leoni P. Thrombosis in essential thrombocytemia and early/prefibrotic primary myelofibrosis: the role of the WHO histological diagnosis. Diagn Pathol. 2015;10 doi: 10.1186/s13000-015-0269-1. 29,015-0269-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Finazzi G, Carobbio A, Thiele J, Passamonti F, Rumi E, Ruggeri M, Rodeghiero F, Randi ML, Bertozzi I, Vannucchi AM, Antonioli E, Gisslinger H, Buxhofer-Ausch V, Gangat N, Rambaldi A, Tefferi A, Barbui T. Incidence and risk factors for bleeding in 1104 patients with essential thrombocythemia or prefibrotic myelofibrosis diagnosed according to the 2008 WHO criteria. Leukemia. 2012;26:716–9. doi: 10.1038/leu.2011.258. [DOI] [PubMed] [Google Scholar]
  • 92.Buxhofer-Ausch V, Gisslinger H, Thiele J, Gisslinger B, Kvasnicka HM, Mullauer L, Frantal S, Carobbio A, Passamonti F, Rumi E, Ruggeri M, Rodeghiero F, Randi ML, Bertozzi I, Vannucchi AM, Antonioli E, Finazzi G, Gangat N, Tefferi A, Barbui T. Leukocytosis as an important risk factor for arterial thrombosis in WHO-defined early/prefibrotic myelofibrosis: an international study of 264 patients. Am J Hematol. 2012;87:669–72. doi: 10.1002/ajh.23217. [DOI] [PubMed] [Google Scholar]
  • 93.Buxhofer-Ausch V, Gisslinger B, Schalling M, Gleiss A, Schiefer AI, Mullauer L, Thiele J, Kralovics R, Gisslinger H. Impact of white blood cell counts at diagnosis and during follow-up in patients with essential thrombocythaemia and prefibrotic primary myelofibrosis. Br J Haematol. 2016 doi: 10.1111/bjh.14202. [DOI] [PubMed] [Google Scholar]
  • 94.Guglielmelli P, Pacilli A, Rotunno G, Rumi E, Rosti V, Delaini F, Maffioli M, Fanelli T, Pancrazzi A, Pietra D, Salmoiraghi S, Mannarelli C, Franci A, Paoli C, Rambaldi A, Passamonti F, Barosi G, Barbui T, Cazzola M, Vannucchi AM AGIMM Group. Presentation and outcome of patients with 2016 WHO diagnosis of prefibrotic and overt primary myelofibrosis. Blood. 2017;129:3227–36. doi: 10.1182/blood-2017-01-761999. [DOI] [PubMed] [Google Scholar]
  • 95.Mudireddy M, Shah S, Lasho T, Barraco D, Hanson CA, Ketterling RP, Gangat N, Pardanani A, Tefferi A. Prefibrotic versus overtly fibrotic primary myelofibrosis: clinical, cytogenetic, molecular and prognostic comparisons. Br J Haematol. 2017 doi: 10.1111/bjh.14838. [DOI] [PubMed] [Google Scholar]
  • 96.Tefferi A. Myelofibrosis with myeloid metaplasia. N Engl J Med. 2000;342:1255–65. doi: 10.1056/NEJM200004273421706. [DOI] [PubMed] [Google Scholar]
  • 97.Delic S, Rose D, Kern W, Nadarajah N, Haferlach C, Haferlach T, Meggendorfer M. Application of an NGS-based 28-gene panel in myeloproliferative neoplasms reveals distinct mutation patterns in essential thrombocythaemia, primary myelofibrosis and polycythaemia vera. Br J Haematol. 2016;175:419–26. doi: 10.1111/bjh.14269. [DOI] [PubMed] [Google Scholar]
  • 98.Vainchenker W, Kralovics R. Genetic basis and molecular pathophysiology of classical myeloproliferative neoplasms. Blood. 2017;129:667–79. doi: 10.1182/blood-2016-10-695940. [DOI] [PubMed] [Google Scholar]
  • 99.Verstovsek S, Manshouri T, Pilling D, Bueso-Ramos CE, Newberry KJ, Prijic S, Knez L, Bozinovic K, Harris DM, Spaeth EL, Post SM, Multani AS, Rampal RK, Ahn J, Levine RL, Creighton CJ, Kantarjian HM, Estrov Z. Role of neoplastic monocyte-derived fibrocytes in primary myelofibrosis. J Exp Med. 2016;213:1723–40. doi: 10.1084/jem.20160283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Schneider RK, Mullally A, Dugourd A, Peisker F, Hoogenboezem R, Van Strien PMH, Bindels EM, Heckl D, Busche G, Fleck D, Muller-Newen G, Wongboonsin J, Ventura Ferreira M, Puelles VG, Saez-Rodriguez J, Ebert BL, Humphreys BD, Kramann R. Gli1+ Mesenchymal Stromal Cells Are a Key Driver of Bone Marrow Fibrosis and an Important Cellular Therapeutic Target. Cell Stem Cell. 2017;20:785,800.e8. doi: 10.1016/j.stem.2017.03.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Sasaki K, Gotlib JR, Mesa RA, Newberry KJ, Ravandi F, Cortes JE, Kelly P, Kutok JL, Kantarjian HM, Verstovsek S. Phase II evaluation of IPI-926, an oral Hedgehog inhibitor, in patients with myelofibrosis. Leuk Lymphoma. 2015;56:2092–7. doi: 10.3109/10428194.2014.984703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Ruberti S, Bianchi E, Guglielmelli P, Rontauroli S, Barbieri G, Tavernari L, Fanelli T, Norfo R, Pennucci V, Fattori GC, Mannarelli C, Bartalucci N, Mora B, Elli L, Avanzini MA, Rossi C, Salmoiraghi S, Zini R, Salati S, Prudente Z, Rosti V, Passamonti F, Rambaldi A, Ferrari S, Tagliafico E, Vannucchi AM, Manfredini R. Involvement of MAF/SPP1 axis in the development of bone marrow fibrosis in PMF patients. Leukemia. 2017 doi: 10.1038/leu.2017.220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Bose P, Verstovsek S. The evolution and clinical relevance of prognostic classification systems in myelofibrosis. Cancer. 2016;122:681–92. doi: 10.1002/cncr.29842. [DOI] [PubMed] [Google Scholar]
  • 104.Tam CS, Kantarjian H, Cortes J, Lynn A, Pierce S, Zhou L, Keating MJ, Thomas DA, Verstovsek S. Dynamic model for predicting death within 12 months in patients with primary or post-polycythemia vera/essential thrombocythemia myelofibrosis. J Clin Oncol. 2009;27:5587–93. doi: 10.1200/JCO.2009.22.8833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Tefferi A, Jimma T, Gangat N, Vaidya R, Begna KH, Hanson CA, Van Dyke DL, Caramazza D, Pardanani A. Predictors of greater than 80% 2-year mortality in primary myelofibrosis: a Mayo Clinic study of 884 karyotypically annotated patients. Blood. 2011;118:4595–8. doi: 10.1182/blood-2011-08-371096. [DOI] [PubMed] [Google Scholar]
  • 106.Rumi E, Pietra D, Pascutto C, Guglielmelli P, Martinez-Trillos A, Casetti I, Colomer D, Pieri L, Pratcorona M, Rotunno G, Sant'Antonio E, Bellini M, Cavalloni C, Mannarelli C, Milanesi C, Boveri E, Ferretti V, Astori C, Rosti V, Cervantes F, Barosi G, Vannucchi AM, Cazzola M Associazione Italiana per la Ricerca sul Cancro Gruppo Italiano Malattie Mieloproliferative Investigators. Clinical effect of driver mutations of JAK2, CALR, or MPL in primary myelofibrosis. Blood. 2014;124:1062–9. doi: 10.1182/blood-2014-05-578435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Tefferi A, Lasho TL, Finke CM, Knudson RA, Ketterling R, Hanson CH, Maffioli M, Caramazza D, Passamonti F, Pardanani A. CALR vs JAK2 vs MPL-mutated or triple-negative myelofibrosis: clinical, cytogenetic and molecular comparisons. Leukemia. 2014;28:1472–7. doi: 10.1038/leu.2014.3. [DOI] [PubMed] [Google Scholar]
  • 108.Vannucchi AM, Lasho TL, Guglielmelli P, Biamonte F, Pardanani A, Pereira A, Finke C, Score J, Gangat N, Mannarelli C, Ketterling RP, Rotunno G, Knudson RA, Susini MC, Laborde RR, Spolverini A, Pancrazzi A, Pieri L, Manfredini R, Tagliafico E, Zini R, Jones A, Zoi K, Reiter A, Duncombe A, Pietra D, Rumi E, Cervantes F, Barosi G, Cazzola M, Cross NC, Tefferi A. Mutations and prognosis in primary myelofibrosis. Leukemia. 2013;27:1861–9. doi: 10.1038/leu.2013.119. [DOI] [PubMed] [Google Scholar]
  • 109.Guglielmelli P, Lasho TL, Rotunno G, Score J, Mannarelli C, Pancrazzi A, Biamonte F, Pardanani A, Zoi K, Reiter A, Duncombe A, Fanelli T, Pietra D, Rumi E, Finke C, Gangat N, Ketterling RP, Knudson RA, Hanson CA, Bosi A, Pereira A, Manfredini R, Cervantes F, Barosi G, Cazzola M, Cross NC, Vannucchi AM, Tefferi A. The number of prognostically detrimental mutations and prognosis in primary myelofibrosis: an international study of 797 patients. Leukemia. 2014;28:1804–10. doi: 10.1038/leu.2014.76. [DOI] [PubMed] [Google Scholar]
  • 110.Vannucchi AM, Guglielmelli P, Rotunno G, Pascutto C, Pardanani A, Ferretti V, Pacilli A, Pancrazzi A, Lasho T, Hanson CA, Ketterling R, Gangat N, Tagliafico E, Manfredini R, Artusi V, Bernardis I, Pietra D, Rumi E, Maffioli M, Rosti V, Salmoiraghi S, Delaini F, Bosi A, Cilloni D, Cervantes F, Passamonti F, Barosi G, Rambaldi A, Barbui T, Cazzola M, Tefferi A. Mutation-Enhanced International Prognostic Scoring System (MIPSS) for Primary Myelofibrosis: An AGIMM & IWG-MRT Project. Blood. 2014;124:405. [Google Scholar]
  • 111.Rozovski U, Verstovsek S, Manshouri T, Dembitz V, Bozinovic K, Newberry K, Zhang Y, Bove JE, 4th, Pierce S, Kantarjian H, Estrov Z. An accurate, simple prognostic model consisting of age, JAK2, CALR, and MPL mutation status for patients with primary myelofibrosis. Haematologica. 2017;102:79–84. doi: 10.3324/haematol.2016.149765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Tefferi A, Guglielmelli P, Finke C, Lasho TL, Gangat N, Ketterling R, Hanson CA, Pardanani A, Vannucchi AM. Integration of Mutations and Karyotype Towards a Genetics-Based Prognostic Scoring System (GPSS) for Primary Myelofibrosis. Blood. 2014;124:406. [Google Scholar]
  • 113.Tefferi A, Lasho TL, Finke C, Belachew AA, Wassie EA, Ketterling RP, Hanson CA, Pardanani A. Type 1 vs type 2 calreticulin mutations in primary myelofibrosis: differences in phenotype and prognostic impact. Leukemia. 2014;28:1568–70. doi: 10.1038/leu.2014.83. [DOI] [PubMed] [Google Scholar]
  • 114.Guglielmelli P, Rotunno G, Fanelli T, Pacilli A, Brogi G, Calabresi L, Pancrazzi A, Vannucchi AM. Validation of the differential prognostic impact of type 1/type 1-like versus type 2/type 2-like CALR mutations in myelofibrosis. Blood Cancer J. 2015;5:e360. doi: 10.1038/bcj.2015.90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Tefferi A, Guglielmelli P, Lasho TL, Rotunno G, Finke C, Mannarelli C, Belachew AA, Pancrazzi A, Wassie EA, Ketterling RP, Hanson CA, Pardanani A, Vannucchi AM. CALR and ASXL1 mutations-based molecular prognostication in primary myelofibrosis: an international study of 570 patients. Leukemia. 2014;28:1494–500. doi: 10.1038/leu.2014.57. [DOI] [PubMed] [Google Scholar]
  • 116.Guglielmelli P, Rotunno G, Pacilli A, Rumi E, Rosti V, Delaini F, Maffioli M, Fanelli T, Pancrazzi A, Pieri L, Fjerza R, Pietra D, Cilloni D, Sant'Antonio E, Salmoiraghi S, Passamonti F, Rambaldi A, Barosi G, Barbui T, Cazzola M, Vannucchi AM. Prognostic impact of bone marrow fibrosis in primary myelofibrosis. A study of the AGIMM group on 490 patients. Am J Hematol. 2016;91:918–22. doi: 10.1002/ajh.24442. [DOI] [PubMed] [Google Scholar]
  • 117.Finazzi MC, Carobbio A, Cervantes F, Isola IM, Vannucchi AM, Guglielmelli P, Rambaldi A, Finazzi G, Barosi G, Barbui T. CALR mutation, MPL mutation and triple negativity identify patients with the lowest vascular risk in primary myelofibrosis. Leukemia. 2015;29:1209–10. doi: 10.1038/leu.2014.343. [DOI] [PubMed] [Google Scholar]
  • 118.Cerquozzi S, Tefferi A. Blast transformation and fibrotic progression in polycythemia vera and essential thrombocythemia: a literature review of incidence and risk factors. Blood Cancer J. 2015;5:e366. doi: 10.1038/bcj.2015.95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Cervantes F. How I treat myelofibrosis. Blood. 2014;124:2635–42. doi: 10.1182/blood-2014-07-575373. [DOI] [PubMed] [Google Scholar]
  • 120.Hernandez-Boluda JC, Pereira A, Gomez M, Boque C, Ferrer-Marin F, Raya JM, Garcia-Gutierrez V, Kerguelen A, Xicoy B, Barba P, Martinez J, Luno E, Alvarez-Larran A, Martinez-Lopez J, Arbelo E, Besses C Grupo Espanol de Enfermedades Mieloproliferativas Filadelfia Negativas. The International Prognostic Scoring System does not accurately discriminate different risk categories in patients with post-essential thrombocythemia and post-polycythemia vera myelofibrosis. Haematologica. 2014;99:e55–7. doi: 10.3324/haematol.2013.101733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Gowin K, Coakley M, Kosiorek H, Mesa R. Discrepancies of applying primary myelofibrosis prognostic scores for patients with post polycythemia vera/essential thrombocytosis myelofibrosis. Haematologica. 2016;101:e405–6. doi: 10.3324/haematol.2016.149013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Masarova L, Bose P, Daver N, Pemmaraju N, Newberry KJ, Manshouri T, Cortes J, Kantarjian HM, Verstovsek S. Patients with post-essential thrombocythemia and post-polycythemia vera differ from patients with primary myelofibrosis. Leuk Res. 2017;59:110–6. doi: 10.1016/j.leukres.2017.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Tefferi A, Saeed L, Hanson CA, Ketterling RP, Pardanani A, Gangat N. Application of current prognostic models for primary myelofibrosis in the setting of post-polycythemia vera or post-essential thrombocythemia myelofibrosis. Leukemia. 2017 doi: 10.1038/leu.2017.268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Passamonti F, Rumi E, Caramella M, Elena C, Arcaini L, Boveri E, Del Curto C, Pietra D, Vanelli L, Bernasconi P, Pascutto C, Cazzola M, Morra E, Lazzarino M. A dynamic prognostic model to predict survival in post-polycythemia vera myelofibrosis. Blood. 2008;111:3383–7. doi: 10.1182/blood-2007-11-121434. [DOI] [PubMed] [Google Scholar]
  • 125.Dingli D, Schwager SM, Mesa RA, Li CY, Dewald GW, Tefferi A. Presence of unfavorable cytogenetic abnormalities is the strongest predictor of poor survival in secondary myelofibrosis. Cancer. 2006;106:1985–9. doi: 10.1002/cncr.21868. [DOI] [PubMed] [Google Scholar]
  • 126.Passamonti F, Giorgino T, Mora B, Guglielmelli P, Rumi E, Maffioli M, Rambaldi A, Caramella M, Komrokji R, Gotlib J, Kiladjian JJ, Cervantes F, Devos T, Palandri F, De Stefano V, Ruggeri M, Silver RT, Benevolo G, Albano F, Caramazza D, Merli M, Pietra D, Casalone R, Rotunno G, Barbui T, Cazzola M, Vannucchi AM. A clinical-molecular prognostic model to predict survival in patients with post polycythemia vera and post essential thrombocythemia myelofibrosis. Leukemia. 2017 doi: 10.1038/leu.2017.169. [DOI] [PubMed] [Google Scholar]
  • 127.Deisseroth A, Kaminskas E, Grillo J, Chen W, Saber H, Lu HL, Rothmann MD, Brar S, Wang J, Garnett C, Bullock J, Burke LB, Rahman A, Sridhara R, Farrell A, Pazdur R. U.S. Food and Drug Administration approval: ruxolitinib for the treatment of patients with intermediate and high-risk myelofibrosis. Clin Cancer Res. 2012;18:3212–7. doi: 10.1158/1078-0432.CCR-12-0653. [DOI] [PubMed] [Google Scholar]
  • 128.Verstovsek S, Mesa RA, Gotlib J, Gupta V, DiPersio JF, Catalano JV, Deininger MW, Miller CB, Silver RT, Talpaz M, Winton EF, Harvey JH, Jr, Arcasoy MO, Hexner EO, Lyons RM, Paquette R, Raza A, Jones M, Kornacki D, Sun K, Kantarjian H COMFORT-I investigators. Long-term treatment with ruxolitinib for patients with myelofibrosis: 5-year update from the randomized, double-blind, placebo-controlled, phase 3 COMFORT-I trial. J Hematol Oncol. 2017;10 doi: 10.1186/s13045-017-0417-z. 55,017-0417-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Harrison CN, Vannucchi AM, Kiladjian JJ, Al-Ali HK, Gisslinger H, Knoops L, Cervantes F, Jones MM, Sun K, McQuitty M, Stalbovskaya V, Gopalakrishna P, Barbui T. Long-term findings from COMFORT-II, a phase 3 study of ruxolitinib vs best available therapy for myelofibrosis. Leukemia. 2016 doi: 10.1038/leu.2016.148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Verstovsek S, Mesa RA, Gotlib J, Levy RS, Gupta V, DiPersio JF, Catalano JV, Deininger M, Miller C, Silver RT, Talpaz M, Winton EF, Harvey JH, Jr, Arcasoy MO, Hexner E, Lyons RM, Paquette R, Raza A, Vaddi K, Erickson-Viitanen S, Koumenis IL, Sun W, Sandor V, Kantarjian HM. A double-blind, placebo-controlled trial of ruxolitinib for myelofibrosis. N Engl J Med. 2012;366:799–807. doi: 10.1056/NEJMoa1110557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Verstovsek S, Mesa RA, Gotlib J, Levy RS, Gupta V, DiPersio JF, Catalano JV, Deininger MW, Miller CB, Silver RT, Talpaz M, Winton EF, Harvey JH, Jr, Arcasoy MO, Hexner EO, Lyons RM, Paquette R, Raza A, Vaddi K, Erickson-Viitanen S, Sun W, Sandor V, Kantarjian HM. Efficacy, safety and survival with ruxolitinib in patients with myelofibrosis: results of a median 2-year follow-up of COMFORT-I. Haematologica. 2013;98:1865–71. doi: 10.3324/haematol.2013.092155. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Cervantes F, Vannucchi AM, Kiladjian JJ, Al-Ali HK, Sirulnik A, Stalbovskaya V, McQuitty M, Hunter DS, Levy RS, Passamonti F, Barbui T, Barosi G, Harrison CN, Knoops L, Gisslinger H COMFORT-II investigators. Three-year efficacy, safety, and survival findings from COMFORT-II, a phase 3 study comparing ruxolitinib with best available therapy for myelofibrosis. Blood. 2013;122:4047–53. doi: 10.1182/blood-2013-02-485888. [DOI] [PubMed] [Google Scholar]
  • 133.Vannucchi AM, Verstovsek S, Guglielmelli P, Griesshammer M, Burn TC, Naim A, Paranagama D, Marker M, Gadbaw B, Kiladjian JJ. Ruxolitinib reduces JAK2 p.V617F allele burden in patients with polycythemia vera enrolled in the RESPONSE study. Ann Hematol. 2017 doi: 10.1007/s00277-017-2994-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Deininger M, Radich J, Burn TC, Huber R, Paranagama D, Verstovsek S. The effect of long-term ruxolitinib treatment on JAK2p.V617F allele burden in patients with myelofibrosis. Blood. 2015;126:1551–4. doi: 10.1182/blood-2015-03-635235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Harrison CN, Talpaz M, Mead AJ. Ruxolitinib is effective in patients with intermediate-1 risk myelofibrosis: a summary of recent evidence. Leuk Lymphoma. 2016;57:2259–67. doi: 10.1080/10428194.2016.1195501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Passamonti F, Kiladjian J, Vannucchi AM, Reiter A, Bharathy S, Iommazzo D, Stalbovskaya V, Gopalakrishna P, Mesa RA. ReTHINK: A randomized, double-blind, placebo-controlled, multicenter, phase 3 study of ruxolitinib in early myelofibrosis patients. J Clin Oncol. 2016;34:TPS7080. [Google Scholar]
  • 137.Mesa R, Jamieson C, Bhatia R, Deininger MW, Gerds AT, Gojo I, Gotlib J, Gundabolu K, Hobbs G, Klisovic RB, Kropf P, Mohan SR, Oh S, Padron E, Podoltsev N, Pollyea DA, Rampal R, Rein LA, Scott B, Snyder DS, Stein BL, Verstovsek S, Wadleigh M, Wang ES, Bergman MA, Gregory KM, Sundar H. Myeloproliferative Neoplasms, Version 2.2017, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2016;14:1572–611. doi: 10.6004/jnccn.2016.0169. [DOI] [PubMed] [Google Scholar]
  • 138.Marchetti M, Barosi G, Cervantes F, Birgegard G, Griesshammer M, Harrison C, Hehlmann R, Kiladjian JJ, Kroger N, McMullin MF, Passamonti F, Vannucchi A, Barbui T. Which patients with myelofibrosis should receive ruxolitinib therapy? ELN-SIE evidence-based recommendations. Leukemia. 2016 doi: 10.1038/leu.2016.283. [DOI] [PubMed] [Google Scholar]
  • 139.Barosi G, Klersy C, Villani L, Bonetti E, Catarsi P, Poletto V, Campanelli R, Impera S, Latagliata R, Viarengo G, Carolei A, Massa M, Musso M, Crescimanno A, Gale RP, Rosti V. JAK2(V617F) allele burden 50% is associated with response to ruxolitinib in persons with MPN-associated myelofibrosis and splenomegaly requiring therapy. Leukemia. 2016;30:1772–5. doi: 10.1038/leu.2016.45. [DOI] [PubMed] [Google Scholar]
  • 140.Verstovsek S, Mesa RA, Gotlib J, Levy RS, Gupta V, DiPersio JF, Catalano JV, Deininger M, Miller C, Silver RT, Talpaz M, Winton EF, Harvey JH, Jr, Arcasoy MO, Hexner E, Lyons RM, Paquette R, Raza A, Vaddi K, Erickson-Viitanen S, Sun W, Sandor V, Kantarjian HM. The clinical benefit of ruxolitinib across patient subgroups: analysis of a placebo-controlled, Phase III study in patients with myelofibrosis. Br J Haematol. 2013;161:508–16. doi: 10.1111/bjh.12274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Verstovsek S, Kantarjian H, Mesa RA, Pardanani AD, Cortes-Franco J, Thomas DA, Estrov Z, Fridman JS, Bradley EC, Erickson-Viitanen S, Vaddi K, Levy R, Tefferi A. Safety and efficacy of INCB018424, a JAK1 and JAK2 inhibitor, in myelofibrosis. N Engl J Med. 2010;363:1117–27. doi: 10.1056/NEJMoa1002028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Verstovsek S, Kantarjian HM, Estrov Z, Cortes JE, Thomas DA, Kadia T, Pierce S, Jabbour E, Borthakur G, Rumi E, Pungolino E, Morra E, Caramazza D, Cazzola M, Passamonti F. Long-term outcomes of 107 patients with myelofibrosis receiving JAK1/JAK2 inhibitor ruxolitinib: survival advantage in comparison to matched historical controls. Blood. 2012;120:1202–9. doi: 10.1182/blood-2012-02-414631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Vannucchi AM, Kantarjian HM, Kiladjian JJ, Gotlib J, Cervantes F, Mesa RA, Sarlis NJ, Peng W, Sandor V, Gopalakrishna P, Hmissi A, Stalbovskaya V, Gupta V, Harrison C, Verstovsek S. A pooled analysis of overall survival in COMFORT-I and COMFORT-II, 2 randomized phase 3 trials of ruxolitinib for the treatment of myelofibrosis. Haematologica. 2015 doi: 10.3324/haematol.2014.119545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Patel KP, Newberry KJ, Luthra R, Jabbour E, Pierce S, Cortes J, Singh R, Mehrotra M, Routbort MJ, Luthra M, Manshouri T, Santos FP, Kantarjian H, Verstovsek S. Correlation of mutation profile and response in patients with myelofibrosis treated with ruxolitinib. Blood. 2015;126:790–7. doi: 10.1182/blood-2015-03-633404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Newberry KJ, Patel K, Masarova L, Luthra R, Manshouri T, Jabbour E, Bose P, Daver N, Cortes J, Kantarjian H, Verstovsek S. Clonal evolution and outcomes in myelofibrosis after ruxolitinib discontinuation. Blood. 2017;130:1125–31. doi: 10.1182/blood-2017-05-783225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Gupta V, Harrison C, Hexner EO, Al-Ali HK, Foltz L, Montgomery M, Sun W, Gopalakrishna P, Kantarjian H, Verstovsek S. The impact of anemia on overall survival in patients with myelofibrosis treated with ruxolitinib in the COMFORT studies. Haematologica. 2016;101:e482–4. doi: 10.3324/haematol.2016.151449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Al-Ali HK, Stalbovskaya V, Gopalakrishna P, Perez-Ronco J, Foltz L. Impact of ruxolitinib treatment on the hemoglobin dynamics and the negative prognosis of anemia in patients with myelofibrosis. Leuk Lymphoma. 2016;57:2464–7. doi: 10.3109/10428194.2016.1146950. [DOI] [PubMed] [Google Scholar]
  • 148.Birgegard G. Does anything work for anaemia in myelofibrosis? Best Pract Res Clin Haematol. 2014;27:175–85. doi: 10.1016/j.beha.2014.07.011. [DOI] [PubMed] [Google Scholar]
  • 149.Iancu-Rubin C, Mosoyan G, Wang J, Kraus T, Sung V, Hoffman R. Stromal cellmediated inhibition of erythropoiesis can be attenuated by Sotatercept (ACE-011), an activin receptor type II ligand trap. Exp Hematol. 2013;41:155–166.e17. doi: 10.1016/j.exphem.2012.12.002. [DOI] [PubMed] [Google Scholar]
  • 150.Bose P, Daver N, Jabbour EJ, Pike A, Newberry KJ, Zhou L, Pierce S, Wang X, Kantarjian HM, Verstovsek S. Phase-2 Study of Sotatercept (ACE-011) in Myeloproliferative Neoplasm-Associated Myelofibrosis and Anemia. Blood. 2016;128:478-. [Google Scholar]
  • 151.Pemmaraju N, Carter BZ, Kantarjian HM, Cortes JE, Kadia TM, Garcia-Manero G, DiNardo CD, Bose P, Pierce S, Zhou L, Estrov Z, Tuttle CK, Salinas K, Mak PY, Verstovsek S. Results for Phase II Clinical Trial of LCL161, a SMAC Mimetic, in Patients with Primary Myelofibrosis (PMF), Post-Polycythemia Vera Myelofibrosis (post-PV MF) or Post-Essential Thrombocytosis Myelofibrosis (post-ET MF) Blood. 2016;128:3105-. [Google Scholar]
  • 152.Fleischman AG, Aichberger KJ, Luty SB, Bumm TG, Petersen CL, Doratotaj S, Vasudevan KB, LaTocha DH, Yang F, Press RD, Loriaux MM, Pahl HL, Silver RT, Agarwal A, O'Hare T, Druker BJ, Bagby GC, Deininger MW. TNFalpha facilitates clonal expansion of JAK2V617F positive cells in myeloproliferative neoplasms. Blood. 2011;118:6392–8. doi: 10.1182/blood-2011-04-348144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Daver N, Cortes J, Newberry K, Jabbour E, Zhou L, Wang X, Pierce S, Kadia T, Sasaki K, Borthakur G, Ravandi F, Pemmaraju N, Kantarjian H, Verstovsek S. Ruxolitinib in combination with lenalidomide as therapy for patients with myelofibrosis. Haematologica. 2015;100:1058–63. doi: 10.3324/haematol.2015.126821. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Stegelmann F, Bangerter M, Heidel FH, Griesshammer M, Hebart H, Hochhaus A, Koschmieder S, Möhle R, Reiter A, Scheid C, Kirschbaum R, Reim R, Sutter U, Vetter K, Dohner H, Schlenk RF, Dohner K. A phase-Ib/II study of ruxolitinib plus pomalidomide in myelofibrosis. Blood. 2015;126:826-. [Google Scholar]
  • 155.Mesa RA, Steensma DP, Pardanani A, Li CY, Elliott M, Kaufmann SH, Wiseman G, Gray LA, Schroeder G, Reeder T, Zeldis JB, Tefferi A. A phase 2 trial of combination low-dose thalidomide and prednisone for the treatment of myelofibrosis with myeloid metaplasia. Blood. 2003;101:2534–41. doi: 10.1182/blood-2002-09-2928. [DOI] [PubMed] [Google Scholar]
  • 156.Marchetti M, Barosi G, Balestri F, Viarengo G, Gentili S, Barulli S, Demory JL, Ilariucci F, Volpe A, Bordessoule D, Grossi A, Le Bousse-Kerdiles MC, Caenazzo A, Pecci A, Falcone A, Broccia G, Bendotti C, Bauduer F, Buccisano F, Dupriez B. Low-dose thalidomide ameliorates cytopenias and splenomegaly in myelofibrosis with myeloid metaplasia: a phase II trial. J Clin Oncol. 2004;22:424–31. doi: 10.1200/JCO.2004.08.160. [DOI] [PubMed] [Google Scholar]
  • 157.Weinkove R, Reilly JT, McMullin MF, Curtin NJ, Radia D, Harrison CN. Low-dose thalidomide in myelofibrosis. Haematologica. 2008;93:1100–1. doi: 10.3324/haematol.12416. [DOI] [PubMed] [Google Scholar]
  • 158.Bose P, Abou Zahr A, Verstovsek S. Investigational Janus kinase inhibitors in development for myelofibrosis. Expert Opin Investig Drugs. 2017 doi: 10.1080/13543784.2017.1323871. [DOI] [PubMed] [Google Scholar]
  • 159.Ciceri P, Muller S, O'Mahony A, Fedorov O, Filippakopoulos P, Hunt JP, Lasater EA, Pallares G, Picaud S, Wells C, Martin S, Wodicka LM, Shah NP, Treiber DK, Knapp S. Dual kinase-bromodomain inhibitors for rationally designed polypharmacology. Nat Chem Biol. 2014;10:305–12. doi: 10.1038/nchembio.1471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Pardanani A, Harrison C, Cortes JE, Cervantes F, Mesa RA, Milligan D, Masszi T, Mishchenko E, Jourdan E, Vannucchi AM, Drummond MW, Jurgutis M, Kuliczkowski K, Gheorghita E, Passamonti F, Neumann F, Patki A, Gao G, Tefferi A. Safety and Efficacy of Fedratinib in Patients With Primary or Secondary Myelofibrosis: A Randomized Clinical Trial. JAMA Oncol. 2015;1:643–51. doi: 10.1001/jamaoncol.2015.1590. [DOI] [PubMed] [Google Scholar]
  • 161.Harrison CN, Schaap N, Vannucchi AM, Kiladjian JJ, Tiu RV, Zachee P, Jourdan E, Winton E, Silver RT, Schouten HC, Passamonti F, Zweegman S, Talpaz M, Lager J, Shun Z, Mesa RA. 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:e317–24. doi: 10.1016/S2352-3026(17)30088-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Mesa RA, Kiladjian J, Catalano JV, Devos T, Egyed M, Hellmann A, McLornan D, Shimoda K, Winton EF, Dong H, Dubowy R, Maltzman JD, Cervantes F, Gotlib J. Phase 3 trial of momelotinib(MMB) vs ruxolitinib (RUX) in JAK inhibitor (JAKi) naive patients with myelofibrosis (MF) J Clin Oncol. 2017;35:7000-. doi: 10.1200/JCO.2017.73.4418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Harrison CN, Vannucchi AM, Platzbecker U, Cervantes F, Gupta V, Lavie D, Passamonti F, Winton EF, Dong H, Kawashima J, Maltzman JD, Kiladjian J, Verstovsek S. Phase 3 randomized trial of momelotinib (MMB) versus best available therapy (BAT) in patients with myelofibrosis (MF) previously treated with ruxolitinib (RUX) J Clin Oncol. 2017;35:7001-. [Google Scholar]
  • 164.Mesa RA, Vannucchi AM, Mead A, Egyed M, Szoke A, Suvorov A, Jakucs J, Perkins A, Prasad R, Mayer J, Demeter J, Ganly P, Singer JW, Zhou H, Dean JP, Te Boekhorst PA, Nangalia J, Kiladjian JJ, Harrison CN. 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 doi: 10.1016/S2352-3026(17)30027-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Mascarenhas J, Hoffman R, Talpaz M, Gerds A, Stein B, Gupta V, Szoke A, Drummond M, Pristupa A, Granston T, Daly R, Dean JP, Al-Fayoumi S, Callahan JA, Singer JW, Gotlib J, Jamieson C, Harrison C, Mesa R, Verstovsek S. Results of the Persist-2 Phase 3 Study of Pacritinib (PAC) Versus Best Available Therapy (BAT), Including Ruxolitinib (RUX), in Patients (pts) with Myelofibrosis (MF) and Platelet Counts <100,000/µl. Blood. 2016;128:LBA-5. [Google Scholar]
  • 166.Verstovsek S, Talpaz M, Ritchie EK, Wadleigh M, Odenike O, Jamieson C, Stein B, Rivera CE, Uno T, Mesa RA. Phase 1/2 Study of NS-018, an Oral JAK2 Inhibitor, in Patients with Primary Myelofibrosis (PMF), Post-Polycythemia Vera Myelofibrosis (postPV MF), or Post-Essential Thrombocythemia Myelofibrosis (postET MF) Blood. 2016;128:1936-. [Google Scholar]
  • 167.Mascarenhas JO, Talpaz M, Gupta V, Foltz LM, Savona MR, Paquette R, Turner AR, Coughlin P, Winton E, Burn TC, O'Neill P, Clark J, Hunter D, Assad A, Hoffman R, Verstovsek S. Primary analysis of a phase II open-label trial of INCB039110, a selective JAK1 inhibitor, in patients with myelofibrosis. Haematologica. 2016 doi: 10.3324/haematol.2016.151126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Vannucchi AM, Harrison CN. Emerging treatments for classical myeloproliferative neoplasms. Blood. 2017;129:693–703. doi: 10.1182/blood-2016-10-695965. [DOI] [PubMed] [Google Scholar]
  • 169.Bose P, Verstovsek S. JAK2 inhibitors for myeloproliferative neoplasms: what is next? Blood. 2017;130:115–25. doi: 10.1182/blood-2017-04-742288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Tefferi A, Lasho TL, Begna KH, Patnaik MM, Zblewski DL, Finke CM, Laborde RR, Wassie E, Schimek L, Hanson CA, Gangat N, Wang X, Pardanani A. A Pilot Study of the Telomerase Inhibitor Imetelstat for Myelofibrosis. N Engl J Med. 2015;373:908–19. doi: 10.1056/NEJMoa1310523. [DOI] [PubMed] [Google Scholar]
  • 171.Geron. Geron Announces Completion of Second Internal Data Reviews for Imetelstat Trials Being Conducted by Janssen. [Accessed: June/7 2017];2017 https://globenewswire.com/news-release/2017/04/10/958245/0/en/Geron-Announces-Completion-of-Second-Internal-Data-Reviews-for-Imetelstat-Trials-Being-Conducted-by-Janssen.html.
  • 172.Verstovsek S, Mesa RA, Foltz LM, Gupta V, Mascarenhas JO, Ritchie EK, Hoffman R, Silver RT, Kremyanskaya M, Pozdnyakova O, Hasserjian RP, Trehu E, Kantarjian HM, Gotlib JR. Phase 2 Trial of PRM-151, an Anti-Fibrotic Agent, in Patients with Myelofibrosis: Stage 1 Results. Blood. 2014;124:713-. [Google Scholar]
  • 173.Verstovsek S, Mesa RA, Foltz LM, Gupta V, Mascarenhas JO, Ritchie EK, Hoffman R, Silver RT, Kremyanskaya M, Pozdnyakova O, Hasserjian RP, Trehu E, Salama ME, Kantarjian HM, Gotlib JR. PRM-151 in Myelofibrosis: Durable Efficacy and Safety at 72 Weeks. Blood. 2015;126:56. [Google Scholar]
  • 174.Evrot E, Ebel N, Romanet V, Roelli C, Andraos R, Qian Z, Dolemeyer A, Dammassa E, Sterker D, Cozens R, Hofmann F, Murakami M, Baffert F, Radimerski T. JAK1/2 and Pan-deacetylase inhibitor combination therapy yields improved efficacy in preclinical mouse models of JAK2V617F-driven disease. Clin Cancer Res. 2013;19:6230–41. doi: 10.1158/1078-0432.CCR-13-0905. [DOI] [PubMed] [Google Scholar]
  • 175.Wang Y, Fiskus W, Chong DG, Buckley KM, Natarajan K, Rao R, Joshi A, Balusu R, Koul S, Chen J, Savoie A, Ustun C, Jillella AP, Atadja P, Levine RL, Bhalla KN. Cotreatment with panobinostat and JAK2 inhibitor TG101209 attenuates JAK2V617F levels and signaling and exerts synergistic cytotoxic effects against human myeloproliferative neoplastic cells. Blood. 2009;114:5024–33. doi: 10.1182/blood-2009-05-222133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Bhagwat N, Koppikar P, Keller M, Marubayashi S, Shank K, Rampal R, Qi J, Kleppe M, Patel HJ, Shah SK, Taldone T, Bradner JE, Chiosis G, Levine RL. Improved targeting of JAK2 leads to increased therapeutic efficacy in myeloproliferative neoplasms. Blood. 2014;123:2075–83. doi: 10.1182/blood-2014-01-547760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Fiskus W, Verstovsek S, Manshouri T, Rao R, Balusu R, Venkannagari S, Rao NN, Ha K, Smith JE, Hembruff SL, Abhyankar S, McGuirk J, Bhalla KN. Heat shock protein 90 inhibitor is synergistic with JAK2 inhibitor and overcomes resistance to JAK2-TKI in human myeloproliferative neoplasm cells. Clin Cancer Res. 2011;17:7347–58. doi: 10.1158/1078-0432.CCR-11-1541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Bartalucci N, Tozzi L, Bogani C, Martinelli S, Rotunno G, Villeval JL, Vannucchi AM. Co-targeting the PI3K/mTOR and JAK2 signalling pathways produces synergistic activity against myeloproliferative neoplasms. J Cell Mol Med. 2013;17:1385–96. doi: 10.1111/jcmm.12162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Fiskus W, Verstovsek S, Manshouri T, Smith JE, Peth K, Abhyankar S, McGuirk J, Bhalla KN. Dual PI3K/AKT/mTOR inhibitor BEZ235 synergistically enhances the activity of JAK2 inhibitor against cultured and primary human myeloproliferative neoplasm cells. Mol Cancer Ther. 2013;12:577–88. doi: 10.1158/1535-7163.MCT-12-0862. [DOI] [PubMed] [Google Scholar]
  • 180.Daver N, Cortes JE, Pemmaraju N, Jabbour EJ, Bose P, Zhou L, Pierce S, Van Derbur S, Borthakur G, Estrov Z, Garcia-Manero G, Kantarjian HM, Verstovsek S. Ruxolitinib (RUX) in Combination with 5-Azacytidine (AZA) As Therapy for Patients (pts) with Myelofibrosis (MF) Blood. 2016;128:1127-. [Google Scholar]
  • 181.Harrison CN, Kiladjian JJ, Heidel FH, Vannucchi AM, Passamonti F, Hayat A, Conneally E, Martino B, Kindler T, Lipka DB, Acharyya S, Gopalakrishna P, Ide S, Mu S, Ribrag V. Efficacy, Safety, and Confirmation of the Recommended Phase 2 Starting Dose of the Combination of Ruxolitinib (RUX) and Panobinostat (PAN) in Patients (Pts) with Myelofibrosis (MF) Blood. 2015;126:4060-. [Google Scholar]
  • 182.Rampal R, Pinzon-Ortiz M, Varshini HSA, Levine RL, Cao A. Synergistic Therapeutic Efficacy of Combined JAK1/2, Pan-PIM, and CDK4/6 Inhibition in Myeloproliferative Neoplasms. Blood. 2016;128:634-. [Google Scholar]
  • 183.Waibel M, Solomon VS, Knight DA, Ralli RA, Kim SK, Banks KM, Vidacs E, Virely C, Sia KC, Bracken LS, Collins-Underwood R, Drenberg C, Ramsey LB, Meyer SC, Takiguchi M, Dickins RA, Levine R, Ghysdael J, Dawson MA, Lock RB, Mullighan CG, Johnstone RW. 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;5:1047–59. doi: 10.1016/j.celrep.2013.10.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Yan D, Tantravahi SK, Pomicter AD, Senina A, Gantz KC, Redwine HM, Prchal JT, Swierczek S, Clair PM, Eiring AM, Baloglu E, O'Hare T, Deininger MW. Selective Inhibition of Nuclear Cytoplasmic Transport As a New Treatment Paradigm in Myelofibrosis. Blood. 2016;128:636-. [Google Scholar]
  • 185.Saenz DT, Fiskus W, Manshouri T, Rajapakshe K, Krieger S, Sun B, Mill CP, DiNardo C, Pemmaraju N, Kadia T, Parmar S, Sharma S, Coarfa C, Qiu P, Verstovsek S, Bhalla KN. BET protein bromodomain inhibitor-based combinations are highly active against post-myeloproliferative neoplasm secondary AML cells. Leukemia. 2016 doi: 10.1038/leu.2016.260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Saenz DT, Fiskus W, Qian Y, Manshouri T, Rajapakshe K, Raina K, Coleman KG, Crew AP, Shen A, Mill CP, Sun B, Qiu P, Kadia TM, Pemmaraju N, DiNardo C, Kim MS, Nowak AJ, Coarfa C, Crews CM, Verstovsek S, Bhalla KN. Novel BET protein proteolysis-targeting chimera exerts superior lethal activity than bromodomain inhibitor (BETi) against post-myeloproliferative neoplasm secondary (s) AML cells. Leukemia. 2017 doi: 10.1038/leu.2016.393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Cervantes F, Pereira A. Does ruxolitinib prolong the survival of patients with myelofibrosis? Blood. 2017;129:832–7. doi: 10.1182/blood-2016-11-731604. [DOI] [PubMed] [Google Scholar]
  • 188.Mascarenhas J, Hoffman R. A comprehensive review and analysis of the effect of ruxolitinib therapy on the survival of patients with myelofibrosis. Blood. 2013;121:4832–7. doi: 10.1182/blood-2013-02-482232. [DOI] [PubMed] [Google Scholar]
  • 189.Kalota A, Jeschke GR, Carroll M, Hexner EO. Intrinsic resistance to JAK2 inhibition in myelofibrosis. Clin Cancer Res. 2013;19:1729–39. doi: 10.1158/1078-0432.CCR-12-1907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Weigert O, Lane AA, Bird L, Kopp N, Chapuy B, van Bodegom D, Toms AV, Marubayashi S, Christie AL, McKeown M, Paranal RM, Bradner JE, Yoda A, Gaul C, Vangrevelinghe E, Romanet V, Murakami M, Tiedt R, Ebel N, Evrot E, De Pover A, Regnier CH, Erdmann D, Hofmann F, Eck MJ, Sallan SE, Levine RL, Kung AL, Baffert F, Radimerski T, Weinstock DM. Genetic resistance to JAK2 enzymatic inhibitors is overcome by HSP90 inhibition. J Exp Med. 2012;209:259–73. doi: 10.1084/jem.20111694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Koppikar P, Bhagwat N, Kilpivaara O, Manshouri T, Adli M, Hricik T, Liu F, Saunders LM, Mullally A, Abdel-Wahab O, Leung L, Weinstein A, Marubayashi S, Goel A, Gonen M, Estrov Z, Ebert BL, Chiosis G, Nimer SD, Bernstein BE, Verstovsek S, Levine RL. Heterodimeric JAK-STAT activation as a mechanism of persistence to JAK2 inhibitor therapy. Nature. 2012;489:155–9. doi: 10.1038/nature11303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Meyer SC, Keller MD, Chiu S, Koppikar P, Guryanova OA, Rapaport F, Xu K, Manova K, Pankov D, O'Reilly RJ, Kleppe M, McKenney AS, Shih AH, Shank K, Ahn J, Papalexi E, Spitzer B, Socci N, Viale A, Mandon E, Ebel N, Andraos R, Rubert J, Dammassa E, Romanet V, Dolemeyer A, Zender M, Heinlein M, Rampal R, Weinberg RS, Hoffman R, Sellers WR, Hofmann F, Murakami M, Baffert F, Gaul C, Radimerski T, Levine RL. CHZ868, a Type II JAK2 Inhibitor, Reverses Type I JAK Inhibitor Persistence and Demonstrates Efficacy in Myeloproliferative Neoplasms. Cancer Cell. 2015;28:15–28. doi: 10.1016/j.ccell.2015.06.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Gotlib J, Kluin-Nelemans HC, George TI, Akin C, Sotlar K, Hermine O, Awan FT, Hexner E, Mauro MJ, Sternberg DW, Villeneuve M, Huntsman Labed A, Stanek EJ, Hartmann K, Horny HP, Valent P, Reiter A. Efficacy and Safety of Midostaurin in Advanced Systemic Mastocytosis. N Engl J Med. 2016;374:2530–41. doi: 10.1056/NEJMoa1513098. [DOI] [PubMed] [Google Scholar]
  • 194.Schwaab J, Schnittger S, Sotlar K, Walz C, Fabarius A, Pfirrmann M, Kohlmann A, Grossmann V, Meggendorfer M, Horny HP, Valent P, Jawhar M, Teichmann M, Metzgeroth G, Erben P, Ernst T, Hochhaus A, Haferlach T, Hofmann WK, Cross NC, Reiter A. Comprehensive mutational profiling in advanced systemic mastocytosis. Blood. 2013;122:2460–6. doi: 10.1182/blood-2013-04-496448. [DOI] [PubMed] [Google Scholar]
  • 195.Jawhar M, Schwaab J, Schnittger S, Meggendorfer M, Pfirrmann M, Sotlar K, Horny HP, Metzgeroth G, Kluger S, Naumann N, Haferlach C, Haferlach T, Valent P, Hofmann WK, Fabarius A, Cross NC, Reiter A. Additional mutations in SRSF2, ASXL1 and/or RUNX1 identify a high-risk group of patients with KIT D816V(+) advanced systemic mastocytosis. Leukemia. 2016;30:136–43. doi: 10.1038/leu.2015.284. [DOI] [PubMed] [Google Scholar]
  • 196.Jawhar M, Schwaab J, Hausmann D, Clemens J, Naumann N, Henzler T, Horny HP, Sotlar K, Schoenberg SO, Cross NC, Fabarius A, Hofmann WK, Valent P, Metzgeroth G, Reiter A. Splenomegaly, elevated alkaline phosphatase and mutations in the SRSF2/ASXL1/RUNX1 gene panel are strong adverse prognostic markers in patients with systemic mastocytosis. Leukemia. 2016;30:2342–50. doi: 10.1038/leu.2016.190. [DOI] [PubMed] [Google Scholar]
  • 197.Jawhar M, Schwaab J, Schnittger S, Sotlar K, Horny HP, Metzgeroth G, Muller N, Schneider S, Naumann N, Walz C, Haferlach T, Valent P, Hofmann WK, Cross NC, Fabarius A, Reiter A. Molecular profiling of myeloid progenitor cells in multi-mutated advanced systemic mastocytosis identifies KIT D816V as a distinct and late event. Leukemia. 2015;29:1115–22. doi: 10.1038/leu.2015.4. [DOI] [PubMed] [Google Scholar]
  • 198.Jawhar M, Schwaab J, Naumann N, Horny HP, Sotlar K, Haferlach T, Metzgeroth G, Fabarius A, Valent P, Hofmann WK, Cross NCP, Meggendorfer M, Reiter A. Response and progression on midostaurin in advanced systemic mastocytosis: KIT D816V and other molecular markers. Blood. 2017;130:137–45. doi: 10.1182/blood-2017-01-764423. [DOI] [PubMed] [Google Scholar]
  • 199.Evans E, Gardino A, Hodous B, Davis A, Zhu J, Kohl NE, Lengauer C. Blu-285, a Potent and Selective Inhibitor for Hematologic Malignancies with KIT Exon 17 Mutations. Blood. 2015;126:568-. [Google Scholar]
  • 200.Gebreyohannes YK, Zhai M, Wozniak A, Wellens J, Cornillie J, Evans E, Gardino AK, Kohl NE, Debiec-Rychter M, Sciot R, Schoffski P. Efficacy of BLU-285, a novel, potent inhibitor of Exon 17 Mutant KIT and PDGFRA D842V, in patient-derived xenograft model of gastrointestinal stromal tumor (GIST) J Clin Oncol. 2016;34:11030-. [Google Scholar]
  • 201.Drummond MW, DeAngelo DJ, Deininger MW, Radia D, Quiery AT, Hexner EO, Shi H, Alvarez-Diez T, Evans EK, Healy ME, Wolf BB, Verstovsek S. Preliminary Safety and Clinical Activity in a Phase 1 Study of Blu-285, a Potent, Highly-Selective Inhibitor of KIT D816V in Advanced Systemic Mastocytosis (SM) Blood. 2016;128:477-. [Google Scholar]
  • 202.Schneeweiss MA, Peter B, Blatt K, Berger D, Stefanzl G, Hadzijusufovic E, Gleixner KV, Valent P. The Multi-Kinase Inhibitor DCC-2618 Inhibits Proliferation and Survival of Neoplastic Mast Cells and Other Cell Types Involved in Systemic Mastocytosis. Blood. 2016;128:1965-. [Google Scholar]
  • 203.Barosi G, Birgegard G, Finazzi G, Griesshammer M, Harrison C, Hasselbalch HC, Kiladjian JJ, Lengfelder E, McMullin MF, Passamonti F, Reilly JT, Vannucchi AM, Barbui T. Response criteria for essential thrombocythemia and polycythemia vera: result of a European LeukemiaNet consensus conference. Blood. 2009;113:4829–33. doi: 10.1182/blood-2008-09-176818. [DOI] [PubMed] [Google Scholar]

RESOURCES