87
Views
0
CrossRef citations to date
0
Altmetric
Review

Ruxolitinib: a targeted treatment option for patients with polycythemia vera

, &
Pages 7-19 | Published online: 12 May 2016

Abstract

Polycythemia vera (PV) is a chronic myeloproliferative neoplasm characterized by erythrocytosis and the presence of Janus kinase (JAK) 2V617F or similar mutations. This review summarizes the pathophysiology of PV, the challenges associated with traditional treatment options, and the scientific rationale and supportive clinical evidence for targeted therapy with ruxolitinib. Accumulating evidence indicates that activating mutations in JAK2 drive the PV disease state. Traditional PV treatment strategies, including aspirin, phlebotomy, and cytoreduc­tive agents such as hydroxyurea, provide clinical benefits for some but not all patients and may not adequately treat PV-related symptoms. Furthermore, traditional treatment approaches are associated with potential side effects that may limit their usage and lead some patients to discon­tinue the treatment. Ruxolitinib is an orally available small-molecule tyrosine kinase inhibitor that is a potent and selective inhibitor of JAK1/JAK2. Ruxolitinib is approved in the US for patients with PV with an inadequate response or intolerance to hydroxyurea and in Europe for adults with PV who are resistant to or intolerant of hydroxyurea. In the Phase III RESPONSE registration trial, ruxolitinib was superior to the best available therapy in patients with PV who were resistant to or intolerant of hydroxyurea in controlling hematocrit levels, reducing spleen volume, and improving PV-related symptoms and quality-of-life measures. The most common nonhematologic adverse events in ruxolitinib-treated patients were headache, diarrhea, pruritus, and fatigue in the RESPONSE trial; hematologic adverse events were primarily grade 1 or 2. In the Phase IIIb nonregistration RELIEF trial, there were nonsignificant trends toward an improved symptom control in patients with PV on a stable hydroxyurea dose who were generally well controlled but reported disease-associated symptoms and switched to ruxolitinib vs those who continued hydroxyurea therapy. Updated treatment guidelines will be important for educating physicians about the role of ruxolitinib in the treatment of patients with PV.

Introduction

Polycythemia vera (PV) is a disorder predominantly characterized by erythrocytosis.1 As opposed to secondary erythrocytoses, PV and primary familial congenital poly­cythemia are categorized as primary erythrocytoses, which result from enhanced responses to erythropoietin (EPO).2 In 1951, PV and three other disorders with similar pathophysiologic characteristics (myelofibrosis [MF], essential thrombocythemia [ET], and chronic myeloid leukemia) were characterized as “myeloproliferative disorders” by Dr William Dameshek.3 Subsequent cytogenetic analyses and clonality studies demonstrated that PV is a clonal malignancy acquired through one or more somatic mutations in a pluripotent hematopoietic stem cell, leading to increased myeloid proliferation.Citation4,Citation5 However, a molecular target responsible for PV was not yet identified at that time.Citation6

Early hypotheses regarding the pathologic basis of PV included potential abnormal growth factor signaling pathways,Citation4 transcriptional dysregulation,Citation5 and constitutive activation of signal transducer and activator of transcription (STAT) proteins.Citation5 Polycythemia rubra vera-1 (PRV-1) was once hypothesized as a PV-specific marker that could be important for elucidating the molecular mechanism of the disorder.Citation5,Citation7 The PV diagnostic strategy drastically changed after the discovery of activating mutations in the Janus kinase 2 (JAK2) tyrosine kinase.Citation8Citation11 Following this discovery, the World Health Organization characterized PV as a chronic myeloproliferative neoplasm (MPN) primarily defined by erythrocytosis and the presence of JAK2V617F or similar mutations.Citation1

Patients diagnosed with PV may have a marked disease burden and a higher mortality risk when compared with the general population, primarily driven by cardiovascular/thrombotic events, disease transformation to MF or acute myeloid leukemia (AML), and solid malignancies.Citation12Citation15 Traditional PV treatment strategies include aspirin, phlebotomy, hydroxyurea, and other cytoreductive treatments.Citation16 Such options provide clinical benefits for some patients, including a reduced risk of cardiovascular/thrombotic events.Citation17Citation20 However, traditional treatment options do not provide adequate benefit for some patientsCitation21,Citation22 and may not alleviate PV-related symptoms.Citation23,Citation24 As such, there remains an unmet need for improved clinical outcomes, symptom alleviation, and enhancement of quality-of-life (QoL).

The objective of this review is to provide a summary of the pathobiology of PV, including the evolving understanding of the molecular etiology of PV, the challenges associated with traditional PV treatment options, and the scientific rationale and clinical data for ruxolitinib (Jakafi®, Incyte Corporation, Wilmington, DE, USA; Jakavi®, Novartis AG, Horsham, West Sussex, UK) supporting its role as a new targeted treatment option for patients with PV.

Biology of PV: pre- and post-JAK2V617F era

Pre-JAK2V617F molecular understanding of the etiology of PV

Early studies of erythropoiesis provided important information about the hematopoiesis process in the PV setting.Citation4 In vitro analyses demonstrated that bone marrow progenitor cells isolated from patients with PV (but not control bone marrow samples) were able to form EPO-independent endogenous erythroid colonies (EECs).Citation4 In 1987, a review of nine studies reported that 97% of patients with PV had EECs in the bone marrow and/or peripheral blood.Citation25 Although EECs were also observed in some patients with ET,Citation26 their presence is a hallmark of PV and was used as a clinical diagnostic tool.Citation4

The observed increased proliferative responsiveness of PV progenitor cells to EPO, IGF-1, and other growth factors (eg, interleukin [IL]-3, granulocyte-macrophage colony-stimulating factor [GM-CSF], thrombopoietin, and stem cell factor) implicated abnormal cytokine signaling pathways in the molecular underpinnings of PV.Citation4,Citation5,Citation26,Citation27 However, studies that examined mutations related to cytokine signaling targets (eg, the EPO receptor, IGF-1 receptor, IGF-1–binding proteins, and tyrosine phosphatases) were unsuccessful in elucidating the pathogenesis of PV.Citation4 It was also hypothesized that abnormal cytokine signaling was not necessarily related to a limited number of specific mutations but rather to more general defects in transcriptional regulation that could affect a variety of metabolic pathways that play a role in the pathogenesis of PV.Citation4 The transcriptional dysregulation hypothesis was supported by studies of cells from patients with PV, which reported decreased levels of the thrombopoietin receptor c-MPL in plateletsCitation28 and an increased proportion of erythroid progenitors expressing BCL-x (an antiapoptotic protein).Citation29 Downstream signal transduction molecules important to cytokine receptor signaling, including EPO-mediated pathways, were further studied to identify the potential PV candidate genes.Citation5 It was hypothesized that EPO-mediated activation of the JAK/STAT pathway induced BCL-x expression, which inhibited apoptosis.Citation6 However, data suggested that constitutive STAT3 activation alone was not the primary molecular cause of PV.Citation30 Therefore, it remained uncertain which one of the signal transducers in this pathway accounted for the increased erythropoiesis observed in patients with PV.Citation6

Collectively, available data up to this point suggested that the JAK/STAT transduction pathway might play a critical role in preventing the apoptosis of erythroid progenitors but not necessarily in the proliferation of erythroid cells.Citation6 The search for the molecular cause of PV continued. Using subtractive hybridization techniques, overexpression of PRV-1 mRNA was observed in the granulocytes of patients with PV but not in normal controls.Citation31 However, the specificity of PRV-1 mRNA overexpression to the PV settingCitation31 was later contradicted by data indicating no consistent differences in PRV-1 protein levels between granulocytes from patients with PV and those from normal controls.Citation32 Furthermore, the sensitivity of PRV-1 expression to GM-CSF exposureCitation31 suggested that alterations in cytokine levels (not the PV disease state itself) explained the variability of PRV-1 levels. As such, the technically demanding and time-consuming EEC assay continued to be the most reliable test for the diagnosis of PV,Citation33 and the molecular cause of PV remained elusive.

Discovery of JAK2V617F and the importance of JAK2 in the pathogenesis of PV

In 2005, four separate groups identified the somatic gain-of-function JAK2V617F mutation,Citation8Citation11 a constitutively active allele associated with activation of downstream signaling components, including STAT3 and STAT5.Citation8,Citation10,Citation34 In murine models, JAK2V617F bone marrow cells are associated with PV disease features, including erythrocytosis, leukocytosis, and enlarged spleen.Citation10,Citation35Citation38 Nearly all patients with PV have an activating mutation in JAK2, most often JAK2V617F (95% of patients) or an activating mutation in exon 12 (4% of patients).Citation37Citation39 Furthermore, patients with elevated JAK2 allele burden (percentage of mutant allele relative to the total [wild type + mutant]) are at increased risk of developing post-PV MF.Citation39

The discovery of the JAK2V617F mutation was critical for understanding the pathobiology of PV and dovetailed with other preclinical data concerning the JAK/STAT signaling pathway. For example, GM-CSF activates JAK1Citation40 and drives granulopoiesis in a pathway that includes JAK2 in cell culture systems,Citation41 and IL-12–driven T-cell proliferation requires JAK2 activity.Citation42 EPO signaling through the EPO receptor activates JAK2,Citation43 which in turn activates STAT1, STAT3, and possibly STAT5 in cell culture systems.Citation44,Citation45 Mice lacking JAK2 are embryonic lethal because of extreme anemia.Citation46,Citation47 Unlike wild-type mice, hematopoietic progenitor cells from JAK2−/− mice do not proliferate and form megakaryocytic colonies in response to thrombopoietin.Citation47 In addition, conditional knockout of JAK2 in adult hematopoietic progenitor cells is associated with reduced viability and lowered platelet counts.Citation48 GM-CSF antiapoptotic activity in human eosinophils requires JAK2 activation.Citation49 Finally, JAK2 is activated in response to thrombopoietin binding to the Mpl receptor and is required for the downstream activation of STAT3.Citation50 With the knowledge that activating mutations in JAK2 were driving the PV disease state, researchers and clinicians had the rationale for the development of new, targeted treatment options, which was the impetus for testing ruxolitinib, a potent JAK1/JAK2 inhibitor, in patients with PV.

Clinical presentation of PV

Elevated blood counts

Elevated blood counts are one of the key diagnostic criteria associated with PV. In a large international study evaluating prognosis and survival among 1,545 patients with PV, many patients presented with elevated hemoglobin (median, 18.4 g/dL) and hematocrit values (median, 55%) at diagnosis.Citation14 White blood cell (WBC) and platelet counts were also elevated (median, 10.4×109/L and 466×109/L, respectively).Citation14 In addition, approximately one half of the patients presented with leukocytosis (WBC count >10.5×109/L; 49%) and thrombocytosis (platelet count ≥450×109/L; 53%).Citation14

Patients with PV who have a high JAK2 allele burden may experience further elevated blood counts compared with those who have less allele burden. A prospective study including 173 patients with PV demonstrated that there was a correlation between the JAK2V617F allele burden and the increased risk of erythropoiesis and myelopoiesis.Citation51 Patients with higher JAK2V617F burden at diagnosis also presented with higher hematocrit levels (regression coefficient [r] =0.67; P<0.001) and higher WBC counts (r=0.54; P<0.001).Citation51

Thromboembolic and mortality risk

Although elevated blood counts are important diagnostic markers of PV, thromboembolic events are often responsible for the initial clinical presentation of the disease. Patients with PV are at an increased risk of cardiovascular/thromboembolic events and mortality compared with the general population.Citation12,Citation14,Citation15,Citation52 Results from a large Swedish registry database study (n=11,155 patients with PV and n=44,620 matched controls) indicated that the risk of arterial and venous thrombosis in patients with PV was five- and ninefold higher, respectively, in patients with PV compared with the general population.Citation53

In a large, randomized, controlled trial testing cytoreductive therapy in patients with PV (CYTO-PV), elevated hematocrit levels of 45%–50% (n=183) were associated with a fourfold increase in cardiovascular complications and a significantly higher rate of cardiovascular death compared with those who had hematocrit level of <45% (n=182).Citation20 Data from this trial also indicated that patients with a WBC count of ≥11×109/L were nearly four times more likely to experience major thrombosis when compared with those who had a WBC count of <7×109/L.Citation54 However, available data do not suggest that thrombocytosis is a significant risk factor for cardiovascular/thromboembolic events in patients with PV. The European Collaboration on Low-Dose Aspirin in Polycythemia Vera prospective trial demonstrated that the rates of thromboembolic events and mortality were not significantly different between patients with baseline platelet counts >400×109/L vs those with lower platelet counts.Citation55 The molecular pathway responsible for the increased incidence of thromboembolic events in patients with PV is unclear. However, data suggest that constitutive JAK2 signaling may contribute to several PV features associated with the development of thromboembolic events, including erythrocytosis,Citation10,Citation20 increased adhesive qualities of JAK2V617F erythrocytes,Citation56 leukocytosis,Citation36,Citation54 systemic inflammation,Citation57 and activation of blood cell types.Citation58Citation65

Signs and symptoms

Patients with PV may experience a variety of disease-related symptoms, reduced QoL,Citation52 and reduced work productivity.Citation66 Fatigue, pruritus, difficulty in sleeping, day or night sweats, and dizziness are among the most frequently reported symptoms of PV.Citation66 Patients with PV may also develop splenomegaly,Citation14 which can be uncomfortable or painful in some patients.Citation67

The specific pathways underlying PV-related symptoms are unknown, but some evidence suggests that cytokine signaling plays an important role. Notably, serum levels of inflammatory cytokines, which signal through JAK1 and/or JAK2,Citation68 are elevated in patients with PV.Citation69 A cytokine profiling study in patients with MF found several clinical correlations between cytokine levels, constitutional symptoms, and splenomegaly.Citation70

Some symptoms may stem from increased blood counts caused by the constitutive activation of JAK2. The JAK2V617F mutation is associated with elevated hematocrit levels in murine modelsCitation10,Citation35,Citation36 and in patients with PV,Citation51 which may be associated with increased blood viscosity and symptoms that result from an impaired cerebral blood flow, including headache and dizziness.Citation71 Patients with PV who have higher JAK2V617F allele burden may be at increased risk of developing splenomegaly and pruritus and are more frequently in need of cytoreductive therapy.Citation39,Citation51

The effects of PV on symptoms and QoL, as commonly measured by the MPN Symptom Assessment Form (MPN-SAF) and the European Organisation for Research and Treatment of Cancer Quality-of-Life Questionnaire Core 30, are similar to those of other MPNs.Citation13 Some patients report that burdensome PV-related symptoms have negative effects on their QoL, productivity, and activities of daily living.Citation66 In a recent US survey of 380 patients with PV, 66% experienced symptoms that reduced their QoL, 22% were sick from work for ≥1 day in the preceding 30 days, and 48% experienced PV-related interference with daily activities.Citation66

Limitations of traditional treatment options

Treatment overview

Traditional treatment options are effective for many patients with PV but do not target the molecular underpinnings of the disease and are associated with several limitations, suggesting that improved treatment options are warranted. The goals of treatment in PV are to reduce the risk of cardiovascular/thrombotic events and control disease-related symptoms.Citation16 Maintaining a hematocrit level of <45% is associated with a reduced risk of cardiovascular/thrombotic events and related death.Citation20 For patients with splenomegaly, treatment should also aim to reduce spleen size.Citation72 Aspirin, phlebotomy, and cytoreduction are the three main elements of traditional PV treatment strategies.Citation16,Citation72

Aspirin

The European Collaboration on Low-Dose Aspirin in Polycythemia Vera clinical trial demonstrated that treatment with low-dose aspirin was associated with a reduced risk of cardiovascular/thrombotic events.Citation19 Subsequently, a Cochrane meta-analysis reported that low-dose aspirin was associated with a nonsignificant reduction in the risk of fatal thrombotic events.Citation73 However, patients receiving regular aspirin treatment (≥2×325 mg/wk) may be at a dose-dependent increased risk of gastrointestinal bleeding.Citation74 Aspirin is contraindicated in patients with acquired von Willebrand syndrome and/or platelet count of >1,000×109/L, which are associated with an increased risk of bleeding.Citation75,Citation76

Phlebotomy

Phlebotomy with or without cytoreductive treatment to maintain a hematocrit level of <45% was associated with reduced cardiovascular death and major thrombotic events in patients with PV.Citation20 However, hematocrit maintenance at <45% with phlebotomy and other traditional treatment options can be difficult. The CYTO-PV study required hematocrit control within predefined limits with phlebotomy and/or other traditional treatments (eg, aspirin and cytoreductive therapy); however, >25% of the patients did not maintain hematocrit levels within their target range.Citation77

Phlebotomy procedures may be poorly tolerated by some patients, as evidenced by the observation that 28% of patients in the high-hematocrit arm of the CYTO-PV trial discontinued phlebotomy treatment.Citation20 In a large study of phlebotomy patients undergoing venipuncture (N=3,315), 15% of patients reported that they feared the phlebotomy procedure and 3% reported that they avoided the procedure because of that fear.Citation78 In addition, frequent phlebotomies can cause iron deficiency, which may be associated with restless leg syndrome and impairments in cognitive functioning and mental health in some patients.Citation79Citation81

Cytoreductive agents

Cytoreductive treatment with hydroxyurea is associated with increased survival and may be associated with reduced thromboembolic risk in patients with PV.Citation17,Citation18 However, one retrospective study indicated that 11% and 13% of patients treated with hydroxyurea become resistant and intolerant, respectively.Citation21 In a multivariate analysis, resistance to hydroxyurea was associated with a 6.8-fold higher risk of transformation to AML or MF and a 5.6-fold higher risk of death.Citation21 Hydroxyurea may also be associated with several hematologic and nonhematologic side effects that could limit treatment, including leg ulcers and other mucocutaneous manifestations, gastrointestinal toxicity, and fever.Citation21

Some data suggest that hydroxyurea may increase the risk of transformation to leukemia, especially in younger patients, although further analyses are required to confirm this hypothesis.Citation82,Citation83 The PV Study Group-08 trial reported that the incidence of AML at a median follow-up of 8.6 years was 5.9% in 51 hydroxyurea-treated patients compared with 1.5% in a historical control group of 134 patients from the phlebotomy-only arm of an earlier study; however, the difference between treatment groups was not statistically significant.Citation18 In an analysis of 1,638 patients with PV enrolled in a prospective observational cohort study with a median follow-up of 2.8 years, the incidence of AML/myelodysplas-tic syndrome (MDS) in patients treated with hydroxyurea was not significantly different from that of patients receiving no treatment, phlebotomy, or interferon.Citation84 The French Polycythemia Study Group was a large, randomized trial with a median follow-up of 16.3 years that compared hydroxyurea with pipobroman as first-line treatment of PV in 285 patients <65 years of age.Citation17 The cumulative incidences of progression to AML/MDS at 10 years, 15 years, and 20 years were 6.6%, 16.5%, and 24.2%, respectively, in the hydroxyurea group and 13.1%, 34.1%, and 52.1%, respectively, in the pipobroman group. The authors acknowledged that the incidence of transformation to AML/MDS in hydroxyurea-treated patients was higher than previously reported. However, they empha-sized that the natural evolution of PV should be considered when interpreting the study results. The leukemogenic risk of hydroxyurea appears to be increased when treatment is preceded or followed by alkylating agents (eg, busulfan).Citation83,Citation85 Because hydroxyurea may modestly increase the risk of AML development, it has been suggested that this drug should be used with caution in younger patients (,40 years of age) with MPNs.Citation82

Interferon-α (IFN-α) and pegylated (PEG)-IFN-α variants are not currently indicated by the US Food and Drug Administration (FDA) or the European Medicines Agency for the treatment of patients with PV. However, IFN-α was recommended by the European LeukemiaNet in 2011 as second-line cytoreductive therapy for patients with PV who become resistant to or intolerant of hydroxyurea.Citation86 Treatment with IFN-α and PEG-IFN-α variants has been associated with hematocrit control without phlebotomy, normalization of blood cell counts, and reductions in enlarged spleen size.Citation87Citation89 In a review article that summarized clinical trial experience with different IFN-α variants in patients with PV published between 1991 and 2008, 60% (182/303) of patients achieved freedom from phlebotomies.Citation90 An objective response was observed in ∼80% of patients, although response criteria were heterogenous among these studies.Citation90 In addition, some data suggest that IFN-α treatment may be associated with improvements in some PV-related signs and symptoms, including pruritusCitation91,Citation92 and splenomegaly.Citation87,Citation88,Citation93,Citation94 However, the route of administration (injection) and safety concerns about recombinant IFN-α formulations suggest that IFN-α may not be an ideal treatment option for some patients. A multicenter observational study reported that nonadherence to IFN-α primarily resulted from patients forgetting to administer the injection (50.2%) and other injection-related reasons (32.0%).Citation95 Side effects of IFN-α, including influenza-like symptoms,Citation96 autoimmune disorders, depression, cardiovascular disease, and ocular disease,Citation16 may lead some patients to discontinue the treatment.Citation97 These safety concerns are primarily associated with recombinant IFN-α variants, and it is important to note that PEG-IFN-α variants are associated with fewer toxicity-related treatment discontinuations.Citation97 PEG-IFN-α2a has also demonstrated decreases in JAK2V617F allele burden over time and is associated with preferable hematologic responses in patients with PV.Citation97,Citation98 In two Phase II clinical trials evaluating PEG-IFN-α in patients with PV, complete hematologic response was observed in 70% and 94.6% of patients (median follow-up time, 21 months and 31.4 months, respectively), and complete molecular response (undetectable JAK2V617F) was observed in 14% and 24.1% of evaluable patients, respectively.Citation89,Citation97 Discontinuations because of PEG-IFN-α-related adverse events were 10% and 24.3% in these studies.Citation89,Citation97 Further analyses from ongoing Phase III trials (eg, ClinicalTrials.gov identifiers: NCT01259856, NCT02218047, NCT02523638, NCT01949805, NCT01387763) will be important for determining the role of PEG-IFN-α in treating patients with PV.

Challenges of PV-related symptom alleviation

Traditional treatment options may not alleviate the PV-related symptom burden.Citation23,Citation24 In a survey-based study, patients with MPNs treated with hydroxyurea, IFN-α, busulfan, or 32P reported similar MPN-SAF symptom severity scores as patients who did not receive myelosuppressive agents.Citation23 Furthermore, a prospective analysis of patients with PV reported that treatment with hydroxyurea, aspirin, IFN-α, or phlebotomy was not associated with significant improvements in the MPN-SAF total symptom score (TSS).Citation24 Col lectively, available data suggest that a targeted treatment approach aimed at the molecular pathway associated with the pathogenesis of PV could be more effective than traditional treatment options for ameliorating PV-related symptoms.

Ruxolitinib: mode of action, pharmacodynamics, and pharmacokinetics

Mode of action and pharmacodynamics

Ruxolitinib phosphate – (R)-3-(4-(7H-pyrrolo[2,3-D]pyrim-idin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile phosphate – is a small-molecule tyrosine kinase inhibitor approved by the FDA for the treatment of patients with PV who have had an inadequate response to or are intolerant of hydroxyurea.Citation99 In Europe, ruxolitinib is indicated for the treatment of adult patients with PV who are resistant to or intolerant of hydroxyurea.Citation100 In the US, ruxolitinib is also indicated for the treatment of patients with intermediate- or high-risk MF, including primary MF, post-PV MF, and post-ET MF;Citation99,Citation100 ruxolitinib is approved for a similar population of patients with MF in Europe.Citation100

In preclinical models, ruxolitinib effectively inhibits the JAK/STAT signaling pathway as a potent and selective inhibitor of JAK1 (mean [SD] 50% inhibitory concentration [IC50], 3.3 [1.2] nM) and JAK2 (mean [SD] IC50, 2.8 [1.2] nM).Citation101 Additionally, ruxolitinib has modest selectivity against Tyk2 (mean [SD] IC50, 19 [3.2] nM) and marked selectivity against JAK3 (mean [SD] IC50, 428 [243] nM).Citation101 Ruxolitinib showed no significant inhibition against 26 additional kinases at concentrations 100-fold the IC50 of JAK1 and JAK2, suggesting high specificity for JAK1 and JAK2.Citation101 The effectiveness of ruxolitinib in this pathway has also been demonstrated in cytokine-stimulated whole-blood assays in which preincubation with ruxolitinib inhibited IL-6– and thrombopoietin-mediated STAT3 phosphorylation (mean [SD] IC50, 282 [54] nM and 281 [62] nM, respectively).Citation101

Several preclinical experiments demonstrated that ruxolitinib inhibits JAK pathway signaling of both wild-type and mutant JAK2.Citation101 In cell lines expressing a JAK2V617F mutation, ruxolitinib effectively inhibited the phosphorylation of JAK2 downstream targets (eg, STAT3, STAT5, and ERK1/2) and induced apoptosis in a dose-dependent fashion.Citation101 Ruxolitinib also inhibited erythroid and myeloid progenitor cell proliferation in primary cultures from healthy individuals (burst-forming unit-erythroid IC50, 407 nM; colony-forming unit-erythroid IC50, 551 nM) and patients with PV carrying the JAK2V617F mutation (burst-forming unit-erythroid IC50, 223 nM; colony-forming unit-erythroid IC50, 444 nM).Citation101 In a murine model, ruxolitinib prolonged the survival, reduced the JAK2V617F allele burden, ameliorated splenomegaly, and normalized the elevated levels of circulating proinflammatory cytokines associated with debilitating constitutional symptoms of PV (eg, IL-6 and tumor necrosis factor-α).Citation101

Pharmacokinetics

Ruxolitinib pharmacokinetics supports an oral route of administration.Citation102 Ruxolitinib has a dose-proportional systemic exposure profile, with minimal accumulation following multiple doses.Citation102 In addition, ruxolitinib is rapidly absorbed, with 95% oral bioavailability.Citation103,Citation104 The maximum tolerated dose of ruxolitinib was established at 25 mg twice daily and 100 mg once daily,Citation102 and the average terminal half-life was ∼3 hours.Citation102

Ruxolitinib is metabolized primarily by the cytochrome P450 3A4 (CYP3A4) enzyme.Citation103 In healthy human subjects, >99% of ruxolitinib doses are metabolized in a pathway that includes oxidation to single and multiple hydroxylated products, some of which then undergo O-glucuronidation.Citation104 Unmetabolized ruxolitinib is the predominant plasma drug entity until 6 hours postdose (58%).Citation104 Most (96%) of the total dose is excreted within 24 hours postdose, primarily via urine (74%) and feces (22%).Citation104 Patients who receive concomitant potent CYP3A4 inhibitors may require a 50% reduction in the ruxolitinib starting dose; however, data suggest that no dosage change is required when ruxolitinib is coadministered with inducers or mild to moderate inhibitors of CYP3A4.Citation103

Ruxolitinib dose reduction is recommended in patients with hepatic or renal impairment.Citation99 In patients with mild and severe hepatic impairment, exposure to ruxolitinib (area under the curve) was increased because of the reduced clearance, although the magnitude of increase in ruxolitinib exposure was not correlated with the degree of hepatic impairment.Citation105 In contrast, an increasing severity of renal impairment was associated with increased exposure to ruxolitinib’s active metabolites and consequently with increased ruxolitinib pharmacologic activity.Citation105

Ruxolitinib: clinical outcomes

Efficacy

The randomized, open-label Phase III RESPONSE registration trial demonstrated that ruxolitinib was superior to the best available therapy in adult patients with PV who were phlebotomy dependent, had splenomegaly, and were resistant to or intolerant of hydroxyurea.Citation22 Patients were randomized to receive ruxolitinib (n=110) or the best available therapy (n=112). Pharmacological treatment in the best available therapy was chosen at the discretion of the treating physician, primarily hydroxyurea (58.9%) and IFN (11.6%); 15.2% of patients received no medication (except aspirin).Citation22 Several limitations of the RESPONSE trial, including the open-label design and the nonstandardized determination of treatment in the best available therapy arm, may have influenced the study treatment compliance and precluded the study from comparing specific treatments with ruxolitinib. However, the study was not powered for such comparisons, and allowing treating physicians to determine the course of treatment was representative of real-world clinical settings. It is not uncommon for patients to continue treatment with hydroxyurea despite the evidence of resistance or intolerance.

In the primary analysis, a higher proportion of patients receiving ruxolitinib vs the best available therapy achieved the composite primary endpoint of hematocrit control and ≥35% reduction in spleen volume from baseline at Week 32 (20.9% vs 0.9%, P<0.001).Citation22 Higher proportions of patients receiving ruxolitinib also achieved individual components of the primary endpoint (hematocrit control, 60.0% vs 19.6%; ≥35% reduction in spleen volume from baseline, 38.2% vs 0.9%)Citation22 and complete hematologic response (23.6% vs 8.9%, P=0.003)Citation22 at Week 32 (). A post hoc analysis demonstrated that the degree of splenomegaly at baseline did not influence the achievement of hematocrit control or spleen size reduction with ruxolitinib treatment.Citation106 Data also suggest that ruxolitinib conferred benefits in patients who did not meet the primary study endpoint. In patients who did not achieve hematocrit control at Week 32, ruxolitinib prolonged the median time to subsequent phlebotomy eligibility compared with the best available therapy (52 weeks vs 21 weeks, respectively).Citation107

Table 1 RESPONSE Phase III trial efficacy resultsTable Footnotea

A total of 96 patients (85.7%) initially randomized to receive the best available therapy crossed over to the ruxolitinib arm because of lack of efficacy; most did so at or soon after the Week 32 visit.Citation22 After cross over to ruxolitinib, a greater proportion of patients achieved ≥35% reduction in spleen size (crossover, 38.5%; best available therapy during randomized treatment, 1.8%), and phlebotomy treatment rates were lower (crossover, 38.5/100 patient-years; best available therapy during randomized treatment, 196.8/100 patient-years).Citation108

A preplanned analysis of RESPONSE conducted 80 weeks after the last patient received his/her first dose demonstrated a durable response with ruxolitinib. This analysis indicated that of the 23 patients randomized to ruxolitinib who achieved the primary endpoint response at Week 32, only one patient lost the response.Citation109 The primary endpoint components were also durable; the probability of maintaining hematocrit control without phlebotomy was 89%, and no patients who achieved ≥35% spleen volume reduction at Week 32 lost their response.Citation109 Furthermore, patients who achieved a complete hematologic response in the primary analysis had a 69% probability of maintaining this response for ≥80 weeks after their initial response.Citation109

Ruxolitinib treatment has also been associated with a decrease in JAK2V617F allele burden in patients with PV. In RESPONSE, the mean JAK2V617F allele burden changes from baseline in the ruxolitinib arm were −12.2% and −34.7% at Weeks 32 and 112, respectively.Citation22 In contrast, patients randomized to receive the best available therapy had a mean 1.2% increase in JAK2V617F allele burden at Week 32.Citation22 A single-center study of 22 patients with PV (n=11) or ET (n=11) reported mean changes in JAK2V617F allele burden of −19% and −28% after 36 months and 60 months of treatment with ruxolitinib, respectively.Citation110

Patient-reported outcomes

Patient-reported outcomes favored ruxolitinib over the best available therapy in Phase III RESPONSE trial, with consistent improvements in the MPN-SAF, European Organisation for Research and Treatment of Cancer Quality-of-Life Questionnaire Core 30, Pruritus Symptom Impact Scale, and Patient Global Impression of Change instruments, compared with little improvement or worsening observed with the best available therapy ().Citation22 Notably, even patients who did not achieve hematocrit control with ruxolitinib at Week 32 reported a higher degree of symptom improvement as evaluated by the MPN-SAF compared with patients who received the best available therapy (38% vs 4%, respectively).Citation107

Table 2 Patient-reported outcomes in the RESPONSE trialTable Footnotea

The randomized, multicenter, double-blind, double-dummy, Phase IIIb RELIEF trial was conducted in patients receiving a stable dose of hydroxyurea who were generally well controlled but reported disease-associated symptoms, comparing the change in PV-related symptom burden in patients continuing their hydroxyurea therapy with those switching to ruxolitinib treatment.Citation111 There was a nonsignificant trend toward a greater improvement in the MPN-SAF cytokine cluster TSS (TSS-C) with ruxolitinib compared with hydroxyurea (proportion of patients achieving ≥50% reduction from baseline in TSS-C at Week 16: 43.4% vs 29.6%, respectively; primary endpoint).Citation111 Additionally, there was a nonsignificant trend toward improvement in individual TSS-C symptoms with ruxolitinib compared with continued hydroxyurea in the RELIEF trial.Citation111

Safety and tolerability

The most common nonhematologic adverse events at Week 32 in patients who received ruxolitinib in the Phase III RESPONSE trial were headache, diarrhea, pruritus, and fatigue ().Citation22,Citation112 The most frequent grade 3 or 4 nonhematologic adverse events in the ruxolitinib arm were dyspnea, asthenia, abdominal pain, headache, muscle spasms, and pruritus.Citation22 Long-term treatment data indicated that the incidences of the most common nonhematologic adverse events were similar at Week 48 and in the 80-week analysis (headache, 20.9% and 21.8%; diarrhea, 20.0% and 19.1%; pruritus, 20.0% and 17.3%; fatigue, 17.3% and 17.3%, respectively).Citation112

Table 3 RESPONSE Phase III trial adverse eventsTable Footnotea

Hematologic adverse events were primarily grade 1 or 2 at both the primary analysis () and the 80-week analysis in the RESPONSE trial.Citation22,Citation112 The most common grade 3 or 4 hematologic adverse events in both randomized treatment arms were lymphopenia and thrombocytopenia. The US label for ruxolitinib indicates that thrombocytopenia, anemia, and neutropenia should be managed by dose reduction/interruption or transfusion.Citation99

At Week 32, the rate of any grade 3 or 4 adverse events per 100 patient-years was lower in the ruxolitinib arm (28.8) than in the best available therapy arm (44.0) in the RESPONSE trial.Citation22

Given the risks associated with thromboembolic events and disease transformation in patients with PV,Citation14 it is important to consider such events when evaluating any new treatment option. The incidence of all-grade thromboembolic events at Week 32 was 0.9% in the ruxolitinib arm and 5.4% in the best available therapy arm (grade 3 or 4, 0.9% vs 1.8%, respectively).Citation22 At the time of data cutoff in the primary analysis of RESPONSE, three patients in the ruxolitinib arm and one patient in the best available therapy arm developed MF after randomization; one patient in the ruxolitinib arm also developed AML. Two additional patients randomized to best available therapy experienced MF after cross over to ruxolitinib, one of whom progressed to AML.Citation22

Other important adverse events of interest for patients being treated with ruxolitinib are nonmelanoma skin cancer and infections. At Week 32 in the RESPONSE trial, four patients in the ruxolitinib arm when compared with two patients in the best available therapy arm experienced newly diagnosed nonmelanoma skin cancer. All but one of these patients (best available therapy arm) had a history of precancerous skin lesions or nonmelanoma skin cancer.Citation22 Melanoma skin cancer was diagnosed in zero patients in the ruxolitinib arm and one patient in the best available therapy arm during randomized treatment. The US prescribing information recommends periodic skin examinations for patients treated with ruxolitinib.Citation99 In the RESPONSE trial, the incidence of any infection was similar in the ruxolitinib arm and the best available therapy arm at Week 32 (any grade, 41.8% vs 36.9%, respectively; grade 3 or 4, 3.6% vs 2.7%). However, herpes zoster infection was observed in 6.4% of patients in the ruxolitinib arm (all grade 1 or 2) at Week 32 compared to 0% of patients in the best available therapy arm.Citation22 The US prescribing information states that patients should be advised about the early signs and symptoms of herpes zoster infection and to seek treatment as early as possible if herpes zoster infection is suspected.Citation99

A long-term safety and tolerability evaluation of ruxolitinib was conducted in 241 patients (457 patient-years) who were resistant to or intolerant of hydroxyurea pooled from a Phase II studyCitation113 and RESPONSE (randomized and crossover patients).Citation22,Citation114 The most common nonhematologic adverse events were diarrhea, headache, and pruritus.Citation114 Frequent grade 3 or 4 nonhematologic adverse events included dyspnea, herpes zoster, abdominal pain, back pain, headache, fatigue, and pyrexia.Citation114 Hematologic adverse events were primarily grade 1 or 2; grade 3 or 4 anemia and thrombocytopenia were each reported in 3.7% of patients.Citation114 Overall, seven patients had disease transformation to MF, and two patients had disease transformation to AML,Citation114 a rate consistent with prior publications in similar patient populations with PV.Citation21,Citation84,Citation115 The rate of thromboembolic events in the ruxolitinib group of this pooled analysis was 2.2/100 patient-years, whereas the rate in RESPONSE patients during randomized treatment with best available therapy was 8.2/100 patient-years.Citation114

In Phase IIIb RELIEF trial, adverse events in the ruxolitinib arm were primarily grade 1 or 2.Citation111 The most frequent nonhematologic adverse events were fatigue (20.4% [ruxolitinib arm] vs 10.7% [hydroxyurea arm]), headache (16.7% vs 5.4%), and dizziness (13.0% vs 8.9%). Two patients receiving ruxolitinib had grade 3 or 4 neutropenia.Citation111

Place in therapy and ongoing trials

Ruxolitinib is approved by the FDA for the treatment of patients with PV who have had an inadequate response to or are intolerant of hydroxyurea.Citation99 Further research will be important for identifying other populations of patients with PV who may benefit from treatment with ruxolitinib. RESPONSE 2 (ClinicalTrials.gov identifier: NCT02038036) is an ongoing, randomized, open-label, Phase IIIb clinical trial designed to evaluate the efficacy and safety of ruxolitinib compared with the best available therapy in patients with PV without splenomegaly who are resistant to or intolerant of hydroxyurea and require phlebotomy.Citation116 The primary endpoint is achievement of hematocrit control at Week 16 that is maintained through Week 28, together with no phlebotomy eligibility from Weeks 4 to 28.Citation116 Patients who are randomized to the best available therapy and do not meet the primary endpoint are allowed to cross over to ruxolitinib at or after Week 28.Citation116 Safety and durability of response will be evaluated through Week 52.Citation116

Conclusion

Ruxolitinib is the only approved treatment option designed to target the constitutively active JAK/STAT signaling pathway in patients with PV. Ruxolitinib improves hematocrit control without phlebotomy, improves blood cell counts, and reduces the enlarged spleen size.Citation22,Citation107 Accumulating evidence suggests that ruxolitinib may also ameliorate PV-related symptoms in patients who are resistant to and/or intolerant of hydroxyurea. Most adverse events were grade 1 or 2, and 82.7% of patients continued on treatment for ≥80 weeks in Phase III RESPONSE trial. Nonmelanoma skin cancer has been observed with ruxolitinib treatment, and periodic skin examinations should be performed. Herpes zoster infection rates (all grade 1 or 2) were higher with ruxolitinib compared with the best available therapy in the RESPONSE trial,Citation22 and patients receiving ruxolitinib should be advised about the early signs and symptoms of infection and instructed to seek treatment as early as possible if suspected. Rates of disease transformation to MF and AML observed with ruxolitinibCitation22 are similar to those previously published in similar patient populations with PV.Citation84,Citation115

Collectively, clinical trial data indicate that ruxolitinib is an effective treatment option for many patients with PV who are resistant to and/or intolerant of hydroxyurea. Furthermore, the potential benefits of ruxolitinib are unique when compared with the traditional treatment options because ruxolitinib may alleviate the PV-related symptom burden and improve the QoL. Updated treatment guidelines will be important for educating physicians about using ruxolitinib for the treatment of patients with PV.

Acknowledgments

Writing assistance was provided by Phuong Tran, PharmD, MBA (Complete Healthcare Communications, LLC, Chadds Ford, PA, USA, an ICON plc company), whose work was funded by Incyte Corporation, Wilmington, DE, USA.

Disclosure

KV is an employee of Incyte Corporation. SV participated in advisory boards and received research funding from Incyte Corporation. JJK served as a consultant for Incyte Corporation and Novartis and received a travel grant and research funding paid by Novartis to Hôpital Saint-Louis. The authors report no other conflicts of interest in this work.

References

  • TefferiAVardimanJWClassification and diagnosis of myelopro-liferative neoplasms: the 2008 World Health Organization criteria and point-of-care diagnostic algorithmsLeukemia2008221142217882280
  • McMullinMFDiagnosis and management of congenital and idiopathic erythrocytosisTher Adv Hematol20123639139823606940
  • DameshekWSome speculations on the myeloproliferative syndromesBlood19516437237514820991
  • PrchalJFPrchalJTMolecular basis for polycythemiaCurr Opin Hematol19996210010910088640
  • PahlHLTowards a molecular understanding of polycythemia rubra veraEur J Biochem2000267123395340110848954
  • Fernandez-LunaJLSilvaMRichardCSanzCBenitoAPathogenesis of polycythemia veraHaematologica19988321501589549927
  • SpivakJLBarosiGTognoniGChronic myeloproliferative disordersASH Education Program Book200320031200224
  • LevineRLWadleighMCoolsJActivating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosisCancer Cell20057438739715837627
  • KralovicsRPassamontiFBuserASA gain-of-function mutation of JAK2 in myeloproliferative disordersN Engl J Med2005352171779179015858187
  • JamesCUgoVLe CouedicJPA unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia veraNature200543470371144114815793561
  • BaxterEJScottLMCampbellPJAcquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disordersLancet200536594641054106115781101
  • HultcrantzMKristinssonSYAnderssonTMPatterns of survival among patients with myeloproliferative neoplasms diagnosed in Sweden from 1973 to 2008: a population-based studyJ Clin Oncol201230242995300122802311
  • SteinBLMoliternoARTiuRVPolycythemia vera disease burden: contributing factors, impact on quality-of-life, and emerging treatment optionsAnn Hematol201493121965197625270596
  • TefferiARumiEFinazziGSurvival and prognosis among 1545 patients with contemporary polycythemia vera: an international studyLeukemia20132791874188123739289
  • MarchioliRFinazziGLandolfiRVascular and neoplastic risk in a large cohort of patients with polycythemia veraJ Clin Oncol200523102224223215710945
  • VannucchiAMHow I treat polycythemia veraBlood2014124223212322025278584
  • KiladjianJJChevretSDosquetCChomienneCRainJDTreatment of polycythemia vera with hydroxyurea and pipobroman: final results of a randomized trial initiated in 1980J Clin Oncol201129293907391321911721
  • FruchtmanSMMackKKaplanMEPetersonPBerkPDWassermanLRFrom efficacy to safety: a Polycythemia Vera Study Group report on hydroxyurea in patients with polycythemia veraSemin Hematol199734117239025158
  • LandolfiRMarchioliRKuttiJEfficacy and safety of low-dose aspirin in polycythemia veraN Engl J Med2004350211412414711910
  • MarchioliRFinazziGSpecchiaGCardiovascular events and intensity of treatment in polycythemia veraN Engl J Med20133681223323216616
  • Alvarez-LarranAPereiraACervantesFAssessment and prognostic value of the European LeukemiaNet criteria for clinicohematologic response, resistance, and intolerance to hydroxyurea in polycythemia veraBlood201211961363136922160617
  • VannucchiAMKiladjianJJGriesshammerMRuxolitinib versus standard therapy for the treatment of polycythemia veraN Engl J Med2015372542643525629741
  • JohanssonPMesaRScherberRAssociation between quality-of-life and clinical parameters in patients with myeloproliferative neoplasmsLeuk Lymphoma201253344144421883029
  • EmanuelRDueckACKiladjianJJConventional therapeutic options have limited impact on MPN symptoms: insights from a prospective analysis of the MPN-SAF [abstract 366]Paper Presented at: European Hematology AssociationJune 14–17Amsterdam, the Netherlands2012
  • ReidCDThe significance of endogenous erythroid colonies (EEC) in haematological disordersBlood Rev1987121331403332094
  • HinshelwoodSBenchAJGreenARPathogenesis of polycythaemia veraBlood Rev19971142242329481451
  • DaiCHKrantzSBDessyprisENMeansRTJrHornSTGilbertHSPolycythemia vera. II. Hypersensitivity of bone marrow erythroid, granulocyte-macrophage, and megakaryocyte progenitor cells to interleukin-3 and granulocyte-macrophage colony-stimulating factorBlood19928048918991498332
  • MoliternoARHankinsWDSpivakJLImpaired expression of the thrombopoietin receptor by platelets from patients with polycythemia veraN Engl J Med199833895725809475764
  • SilvaMRichardCBenitoASanzCOlallaIFernandez-LunaJLExpression of Bcl-x in erythroid precursors from patients with poly-cythemia veraN Engl J Med199833895645719475763
  • RoderSSteimleCMeinhardtGPahlHLSTAT3 is constitutively active in some patients with polycythemia rubra veraExp Hematol200129669470211378264
  • TemerinacSKlippelSStrunckECloning of PRV-1, a novel member of the uPAR receptor superfamily, which is overexpressed in polycythemia rubra veraBlood20009582569257610753836
  • KlippelSStrunckEBusseCEBehringerDPahlHLBiochemical characterization of PRV-1, a novel hematopoietic cell surface receptor, which is overexpressed in polycythemia rubra veraBlood200210072441244812239154
  • KralovicsRSkodaRCMolecular pathogenesis of Philadelphia chromosome negative myeloproliferative disordersBlood Rev200519111315572213
  • WolfAEulenfeldRGablerKJAK2-V617F-induced MAPK activity is regulated by PI3K and acts synergistically with PI3K on the proliferation of JAK2-V617F-positive cellsJAKSTAT201323e2457424069558
  • BummTGElseaCCorbinASCharacterization of murine JAK2V617F-positive myeloproliferative diseaseCancer Res20066623111561116517145859
  • TiedtRHao-ShenHSobasMARatio of mutant JAK2-V617F to wild-type Jak2 determines the MPD phenotypes in transgenic miceBlood200811183931394018160670
  • PasquierFCabagnolsXSecardinLPloIVainchenkerWMyelopro-liferative neoplasms: JAK2 signaling pathway as a central target for therapyClin Lymphoma Myeloma Leuk201414supplS23S3525486952
  • MilosevicJDKralovicsRGenetic and epigenetic alterations of myelo-proliferative disordersInt J Hematol201397218319723233154
  • PassamontiFRumiEPietraDA 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 complicationsLeukemia20102491574157920631743
  • NicholsonSEOatesACHarpurAGZiemieckiAWilksAFLaytonJETyrosine kinase JAK1 is associated with the granulocyte-colony-stimulating factor receptor and both become tyrosine-phosphorylated after receptor activationProc Natl Acad Sci U S A1994918298529887512720
  • WangLXueJZadoroznyEVRobinsonLJG-CSF stimulates Jak2-dependent Gab2 phosphorylation leading to Erk1/2 activation and cell proliferationCell Signal200820101890189918644434
  • SugimotoNNakahiraMAhnHJDifferential requirements for JAK2 and TYK2 in T cell proliferation and IFN-gamma production induced by IL-12 alone or together with IL-18Eur J Immunol200333124325112594853
  • WitthuhnBAQuelleFWSilvennoinenOJAK2 associates with the erythropoietin receptor and is tyrosine phosphorylated and activated following stimulation with erythropoietinCell19937422272368343951
  • PentaKSawyerSTErythropoietin induces the tyrosine phosphorylation, nuclear translocation, and DNA binding of STAT1 and STAT5 in erythroid cellsJ Biol Chem19952705231282312878537396
  • KiritoKNakajimaKWatanabeTIdentification of the human erythropoietin receptor region required for Stat1 and Stat3 activationBlood200299110211011756159
  • GrebienFKerenyiMAKovacicBStat5 activation enables erythropoiesis in the absence of EpoR and Jak2Blood200811194511452218239084
  • ParganasEWangDStravopodisDJak2 is essential for signaling through a variety of cytokine receptorsCell19989333853959590173
  • GrisouardJHao-ShenHDirnhoferSWagnerKUSkodaRCSelective deletion of Jak2 in adult mouse hematopoietic cells leads to lethal anemia and thrombocytopeniaHaematologica2014994e52e5424510341
  • SimonHUYousefiSDibbertBLevi-SchafferFBlaserKAnti-apoptotic signals of granulocyte-macrophage colony-stimulating factor are transduced via Jak2 tyrosine kinase in eosinophilsEur J Immunol19972712353635399464845
  • DrachmanJGMillettKMKaushanskyKThrombopoietin signal transduction requires functional JAK2, not TYK2J Biol Chem199927419134801348410224114
  • VannucchiAMAntonioliEGuglielmelliPProspective identification of high-risk polycythemia vera patients based on JAK2(V617F) allele burdenLeukemia20072191952195917625606
  • SteinBLOhSTBerenzonDPolycythemia vera: an appraisal of the biology and management 10 years after the discovery of JAK2 V617FJ Clin Oncol201533333953396026324368
  • HultcrantzMAnderssonTM-LLandgrenORisk of arterial and venous thrombosis in 11,155 patients with myeloproliferative neoplasms and 44,620 matched controls; a population-based studyBlood2014124 abstract632. (ASH Annual Meeting Abstracts)
  • BarbuiTMasciulliAMarfisiMRWhite blood cell counts and thrombosis in polycythemia vera: a subanalysis of the CYTO-PV studyBlood2015126456056126206947
  • Di NisioMBarbuiTDi GennaroLThe haematocrit and platelet target in polycythemia veraBr J Haematol2007136224925917156406
  • De GrandisMCambotMWautierMPJAK2V617F activates Lu/BCAM-mediated red cell adhesion in polycythemia vera through an EpoR-independent Rap1/Akt pathwayBlood2013121465866523160466
  • BarbuiTCarobbioAFinazziGInflammation and thrombosis in essential thrombocythemia and polycythemia vera: different role of C-reactive protein and pentraxin 3Haematologica201196231531821173097
  • NieswandtBPleinesIBenderMPlatelet adhesion and activation mechanisms in arterial thrombosis and ischaemic strokeJ Thromb Haemost20119suppl 192104
  • HarlanJMKillenPDHarkerLAStrikerGEWrightDGNeutrophil-mediated endothelial injury in vitro mechanisms of cell detachmentJ Clin Invest1981686139414037033282
  • WekslerBBJaffeEABrowerMSColeOFHuman leukocyte cathepsin G and elastase specifically suppress thrombin-induced prostacyclin production in human endothelial cellsBlood1989745162716342477083
  • CeliAPellegriniGLorenzetRP-selectin induces the expression of tissue factor on monocytesProc Natl Acad Sci U S A19949119876787717522321
  • FalangaAMarchettiMEvangelistaVNeutrophil activation and hemostatic changes in healthy donors receiving granulocyte colony-stimulating factorBlood19999382506251410194429
  • LipGYBlannAvon Willebrand factor: a marker of endothelial dysfunction in vascular disorders?Cardiovasc Res19973422552659205537
  • EvangelistaVRajtarGde GaetanoGWhiteJGCerlettiCPlatelet activation by fMLP-stimulated polymorphonuclear leukocytes: the activity of cathepsin G is not prevented by antiproteinasesBlood19917711237923881645603
  • HobbsCMManningHBennettCJAK2V617F leads to intrinsic changes in platelet formation and reactivity in a knock-in mouse model of essential thrombocythemiaBlood2013122233787379724085768
  • MesaRMillerCBThyneMImpact of myeloproliferative neoplasms (MPNs) on patients’ overall health and productivity: results from the MPN LANDMARK SURVEY in the United States [abstract]Blood2014124 abstract3183. (ASH Annual Meeting Abstracts)
  • MesaRANiblackJWadleighMThe burden of fatigue and quality-of-life in myeloproliferative disorders (MPDs): an international Internet-based survey of 1179 MPD patientsCancer20071091687617123268
  • Quintas-CardamaAKantarjianHCortesJVerstovsekSJanus kinase inhibitors for the treatment of myeloproliferative neoplasias and beyondNat Rev Drug Discov201110212714021283107
  • PourcelotETrocmeCMondetJBaillySToussaintBMossuzPCytokine profiles in polycythemia vera and essential thrombocythemia patients: clinical implicationsExp Hematol201442536036824463275
  • TefferiAVaidyaRCaramazzaDFinkeCLashoTPardananiACirculating interleukin (IL)-8, IL-2R, IL-12, and IL-15 levels are independently prognostic in primary myelofibrosis: a comprehensive cytokine profiling studyJ Clin Oncol201129101356136321300928
  • KwaanHCWangJHyperviscosity in polycythemia vera and other red cell abnormalitiesSemin Thromb Hemost200329545145814631544
  • PassamontiFHow I treat polycythemia veraBlood2012120227528422611155
  • SquizzatoARomualdiEPassamontiFMiddeldorpSAntiplatelet drugs for polycythaemia vera and essential thrombocythaemiaCochrane Database Syst Rev20134CD006503
  • HuangESStrateLLHoWWLeeSSChanATLong-term use of aspirin and the risk of gastrointestinal bleedingAm J Med2011124542643321531232
  • TefferiAPolycythemia vera and essential thrombocythemia: 2013 update on diagnosis, risk-stratification, and managementAm J Hematol201388650751623695894
  • MichielsJJBernemanZSchroyensWFinazziGBuddeUvan VlietHHThe paradox of platelet activation and impaired function: platelet-von Willebrand factor interactions, and the etiology of thrombotic and hemorrhagic manifestations in essential thrombocythemia and polycythemia veraSemin Thromb Hemost200632658960416977569
  • MarchioliRFinazziGSpecchiaGMasciulliAMennittoMRBarbuiTTheCYTO-PVa large-scale trial testing the intensity of CYTOreductive therapy to prevent cardiovascular events in patients with polycythemia veraThrombosis2011201179424022084668
  • DeaconBAbramowitzJFear of needles and vasovagal reactions among phlebotomy patientsJ Anxiety Disord200620794696016460906
  • TobiassonMAlyassBSoderlundSBirgegardGHigh prevalence of restless legs syndrome among patients with polycytemia vera treated with venesectioMed Oncol201027110510719225914
  • GreigAJPattersonAJCollinsCEChalmersKAIron deficiency, cognition, mental health and fatigue in women of childbearing age: a systematic reviewJ Nutr Sci20132e1425191562
  • KimJWessling-ResnickMIron and mechanisms of emotional behaviorJ Nutr Biochem201425111101110725154570
  • BjorkholmMHultcrantzMDerolfARLeukemic transformation in myeloproliferative neoplasms: therapy-related or unrelated?Best Pract Res Clin Haematol201427214115325189725
  • BarbuiTThe leukemia controversy in myeloproliferative disorders: is it a natural progression of disease, a secondary sequela of therapy, or a combination of both?Semin Hematol2004412 suppl 31517
  • FinazziGCarusoVMarchioliRAcute leukemia in polycythemia vera: an analysis of 1638 patients enrolled in a prospective observational studyBlood200510572664267015585653
  • NielsenIHasselbalchHCAcute leukemia and myelodysplasia in patients with a Philadelphia chromosome negative chronic myeloproliferative disorder treated with hydroxyurea alone or with hydroxyurea after busulphanAm J Hematol2003741263112949887
  • BarbuiTBarosiGBirgegardGPhiladelphia-negative classical myeloproliferative neoplasms: critical concepts and management recommendations from European LeukemiaNetJ Clin Oncol201129676177021205761
  • SilverRTLong-term effects of the treatment of polycythemia vera with recombinant interferon-alphaCancer2006107345145816804923
  • SacchiSLeoniPLiberatiMA prospective comparison between treatment with phlebotomy alone and with interferon-alpha in patients with polycythemia veraAnn Hematol19946852472508018766
  • Quintas-CardamaAKantarjianHManshouriTPegylated interferon alfa-2a yields high rates of hematologic and molecular response in patients with advanced essential thrombocythemia and polycythemia veraJ Clin Oncol200927325418542419826111
  • KiladjianJJChomienneCFenauxPInterferon-alpha therapy in bcr-abl-negative myeloproliferative neoplasmsLeukemia200822111990199818843285
  • TaylorPCDolanGNgJPPaulBCollinRReillyJTEfficacy of recombinant interferon-alpha (rIFN-alpha) in polycythaemia vera: a study of 17 patients and an analysis of published dataBr J Haematol199692155598562411
  • MullerEWde WolfJTEggerRLong-term treatment with interferon-alpha 2b for severe pruritus in patients with polycythaemia veraBr J Haematol19958923133187873381
  • StasiRVendittiADel PoetaGRole of human leukocyte interferon-alpha in the treatment of patients with polycythemia veraAm J Med Sci199831542372419537637
  • TurriDMitraMEDi TrapaniRLipariMGPerriconeRCajozzoAAlpha-interferon in polycythemia vera and essential thrombocythemiaHaematologica19917617577
  • DevonshireVLapierreYMacdonellRThe Global Adherence Project (GAP): a multicenter observational study on adherence to disease-modifying therapies in patients with relapsing-remitting multiple sclerosisEur J Neurol2011181697720561039
  • HasselbalchHCA new era for IFN-alpha in the treatment of Phila-delphia-negative chronic myeloproliferative neoplasmsExpert Rev Hematol20114663765522077528
  • KiladjianJJCassinatBChevretSPegylated interferon-alfa-2a induces complete hematologic and molecular responses with low toxicity in polycythemia veraBlood200811283065307218650451
  • Quintas-CardamaAAbdel-WahabOManshouriTMolecular analysis of patients with polycythemia vera or essential thrombocythemia receiving pegylated interferon alpha-2aBlood2013122689390123782935
  • Jakafi® (ruxolitinib)Full Prescribing InformationWilmington, DEIncyte Corporation2014
  • Jakavi® (ruxolitinib)Summary of Product CharacteristicsHorsham, West Sussex, UKNovartis AG2015
  • Quintas-CardamaAVaddiKLiuPPreclinical characterization of the selective JAK1/2 inhibitor INCB018424: therapeutic implications for the treatment of myeloproliferative neoplasmsBlood2010115153109311720130243
  • ShiJGChenXMcGeeRFThe pharmacokinetics, pharmacody-namics, and safety of orally dosed INCB018424 phosphate in healthy volunteersJ Clin Pharmacol201151121644165421257798
  • ShiJGChenXEmmTThe effect of CYP3A4 inhibition or induction on the pharmacokinetics and pharmacodynamics of orally administered ruxolitinib (INCB018424 phosphate) in healthy volunteersJ Clin Pharmacol201252680981821602517
  • ShillingADNedzaFMEmmTMetabolism, excretion, and pharmacokinetics of [14C]INCB018424, a selective Janus tyrosine kinase 1/2 inhibitor, in humansDrug Metab Dispos201038112023203120699411
  • ChenXShiJGEmmTPharmacokinetics and pharmacodynamics of orally administered ruxolitinib (INCB018424 phosphate) in renal and hepatic impairment patientsClin Pharmacol Drug Dev201331344227128228
  • VannucchiAVerstovsekSJonesMEfficacy of ruxolitinib by baseline spleen volume in patients with polycythemia vera resistant to or intolerant of hydroxyureaBlood201412421 abstract1840. (ASH Annual Meeting Abstracts)
  • VerstovsekSKiladjianJJMesaRRuxolitinib efficacy by hematocrit control in patients with polycythemia vera: an analysis of the RESPONSE trialBlood201412421 abstract3201. (ASH Annual Meeting Abstracts)
  • KiladjianJJVannucchiAGriesshammerMClinical benefit of ruxolitinib treatment after crossover from best available therapy in patients with polycythemia vera: analysis of the RESPONSE trialBlood201412421 abstract3181. (ASH Annual Meeting Abstracts)
  • VerstovsekSVannucchiAMGriesshammerMRuxolitinib in polycythemia vera: follow-up from the RESPONSE trialJ Clin Oncol20153315_suppl abstract7087. (ASCO Annual Meeting Abstracts)
  • PieriLPancrazziAPacilliAJAK2V617F complete molecular remission in polycythemia vera/essential thrombocythemia patients treated with ruxolitinibBlood2015125213352335325999444
  • MesaRVannucchiAMYacoubAThe efficacy and safety of continued hydroxyurea therapy versus switching to ruxolitinib in patients with polycythemia vera: a randomized, double-blind, double-dummy, symptom study (RELIEF)Blood201412421 abstract3168. (ASH Annual Meeting Abstracts)
  • KiladjianJJVannucchiAGriesshammerMRuxolitinib versus best available therapy in patients with polycythemia vera: 80-week follow-up from the RESPONSE trialHaematologica2015100s1 abstractS447. (EHA Annual Meeting Abstracts)
  • VerstovsekSPassamontiFRambaldiAA phase 2 study of ruxolitinib, an oral JAK1 and JAK2 inhibitor, in patients with advanced polycythemia vera who are refractory or intolerant to hydroxyureaCancer2014120451352024258498
  • VerstovsekSHarrisonCNKiladjianJ-JEffect of ruxolitinib on markers of iron deficiency: an analysis of the RESPONSE trialHaema-tologica2015 100s1 abstract672. (EHA Annual Meeting Abstracts)
  • PassamontiFRumiEPungolinoELife expectancy and prognostic factors for survival in patients with polycythemia vera and essential thrombocythemiaAm J Med20041171075576115541325
  • PassamontiFSaydamGLimLKhanMHMounedjiNGriesshammerMRESPONSE 2: a phase 3b study evaluating the efficacy and safety of ruxolitinib in patients with hydroxyurea (HU)-resistant/intolerant polycythemia vera (PV) versus best available therapy (BAT)J Clin Oncol2014325s abstractTPS7128. (ASCO Annual Meeting Abstracts)
  • AaronsonNKAhmedzaiSBergmanBThe European Organization for Research and Treatment of Cancer QLQ-C30: a quality-of-life instrument for use in international clinical trials in oncologyJ Natl Cancer Inst19938553653768433390