Background
2016 marks the inception of formal management guidelines from NCCN on the Philadelphia chromosome–negative myeloproliferative neoplasms (MPNs). Guidelines recently released by ESMO1 distinguished between the major goals of therapy in essential thrombocythemia (ET) and polycythemia vera (PV)—the prevention of thrombosis and bleeding—from those in myelofibrosis (MF), in which prolongation of survival and management of anemia, constitutional symptoms, and splenomegaly are most important. Underlying these disparate goals of therapy is the fundamentally different natural history of these MPNs: survival in MF is short (median, 6.5 years in 1996–2007 in one international study),2 whereas ET and PV are each characterized by an indolent clinical course and prolonged survival, although shorter than that of the age- and sex-matched US population.3,4
MPNs are characterized by universal activation of Janus kinase/signal transducer and activator of transcription (JAK-STAT) signaling,5,6 in most cases due to gain-of-function mutations in JAK2,7–11 MPL,12,13 or CALR.14,15 The first-in-class JAK1/2 inhibitor, ruxolitinib, approved for the treatment of intermediate- or high-risk MF16,17 and hydroxyurea-resistant/intolerant PV (see Table 1 for definition of resistance and intolerance to hydroxyurea),18 represents the first example of disease-modifying pharmacologic therapy in MPNs, and the inaugural NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) contain special guidance on the use of ruxolitinib (in this issue; to view the most recent version, visit NCCN.org). Long-term follow-up of the pivotal COMFORT trials has demonstrated
Definition of Resistance/Intolerance to Hydroxyurea in Polycythemia Vera


Drug Development in PV and ET
The indolent natural course of PV and ET, leading to prolonged overall survival,3,4 has meant that drug therapy in these conditions has mostly been limited to the use of aspirin29 and hydroxyurea or anagrelide (in ET) in high-risk patients requiring cytoreduction for prevention of thrombotic complications.30–32 Hematocrit control (to <45%)33 in low-risk patients with PV is usually achieved with phlebotomy alone. However, patients with PV and ET, especially the former, can have a substantial symptom burden.23,34–36 In the RESPONSE trial, which led to the recent approval of ruxolitinib for the treatment of hydroxyurea-resistant/intolerant PV, 49% of patients in the ruxolitinib group achieved a 50% or more reduction in their total symptom score (TSS) at 32 weeks compared with 5% in the standard therapy group.18 Hematocrit control was achieved by 60% versus 20%, respectively, and a 35% or more spleen volume reduction (SVR) by 38% and 1%, respectively.18 Additionally, 24% in the ruxolitinib arm achieved complete hematologic remission (CHR) compared with 9% in the standard therapy arm18; these benefits were maintained in most patients after a minimum of 80 weeks of follow-up.37 No patient lost their spleen response between weeks 32 and 80, and 90% of patients still receiving ruxolitinib at week 32 had no phlebotomies performed during this period. RESPONSE was not designed to evaluate rates of thrombosis, but at week 32, before crossover to ruxolitinib, there were 6 thromboembolic events in the standard therapy arm and only 1 in the ruxolitinib arm.18 At the 80-week analysis, the rates of all-grade and grade 3/4 thromboembolic events per 100 patient-years of exposure were 1.8 and 0.9, respectively, among patients originally randomized to ruxolitinib, versus 4.1 and 2.7 in those receiving ruxolitinib after crossover, and 8.2 and 2.7 in those receiving BAT.37 Because ruxolitinib is immunosuppressive, infections are a concern with this agent. Grade 1/2 herpes zoster infections occurred in 7 patients in the ruxolitinib group (6.4%) compared with none in the standard therapy group through week 32.18 With longer follow-up (minimum 80 weeks), rates remained higher with ruxolitinib: 5.3 and 5.4 per 100 patient-years of exposure among patients originally randomized to and those who crossed over to ruxolitinib, respectively, versus none among patients receiving standard therapy.37 Overall, the rate of infections (any grade) was 41.8% in the ruxolitinib group and 36.9% in the standard-therapy group; the rate of grade 3/4 infection was 3.6% and 2.7%, respectively, in the primary analysis.18 Per 100 patient-years of exposure, the rates of all-grade and grade 3/4 infections were 29.4, 27.8, and 58.4, and 4.0, 5.4, and 4.1 for patients originally randomized to ruxolitinib (n=110), those crossed over to ruxolitinib (n=98), and those originally assigned to BAT (n=111), respectively.37 Evaluation of ruxolitinib in patients with ET is ongoing (ClinicalTrials.gov identifiers: NCT00726232, NCT02577926).
Although not specifically approved for MPN therapy, high response rates, including molecular responses, have been reported with the use of pegylated interferon-α-2a in both PV and ET.38,39 One study in 40 patients with PV reported a CHR rate of 94.6% and a decrease in the percentage of mutated JAK2 in 89.6% of patients, with improvement over time in the depth of molecular responses.38 A total of 7 patients achieved a complete molecular remission (CMR), which persisted after treatment discontinuation in 5 patients.38 In another study, CHR rates of 70% and 76% were reported among patients with PV and ET, respectively, with molecular response rates of 38% in ET and 54% in PV (CMR in 6% and 14%, respectively).39 After a median follow-up of 82.5 months, 32 of 83 enrolled patients (39%) remained on this study, but therapy was on hold for 8 of these patients.40 The median durations of hematologic and molecular responses were 66 and 53 months, respectively.40 JAK2 status or allele burden had no impact on achievement of response (clinical or molecular), time to response, or duration of therapy, but patients achieving CMR had the longest response duration.40 Pegylated interferon-α-2a also significantly decreased the allelic burden of mutant CALR from a median of 41% at baseline to 26% after treatment in a study of 31 patients with CALR-mutated ET, with 2 patients achieving CMR.41
Recently, a next-generation, mono-PEGylated interferon-α-2b isoform with a long elimination half-life enabling administration every 2 weeks, ropeginterferon α-2b, has been developed for PV.42 In a phase I/II trial in 51 patients, there were no dose-limiting toxicities and the overall response rate was 90%, including CHR in 47%.42 A CMR was obtained in 21% of patients and a partial molecular response in 47%.42 A steady decrease in the allelic burden of JAK2 V617F was also noted with ruxolitinib therapy in the RESPONSE trial in patients with hydroxyurea-resistant/intolerant PV; the mean percentage decrease from baseline was 40% by week 208 (12.2% at week 32; 22% at week 80).37,43 Although decreases in JAK2 V617F allelic burden are often considered to represent evidence of disease-modifying activity, it is important to remember that although it is the most common mutation in MPN, JAK2 V617F is not the disease-initiating mutation44 and can be a late genetic event.45 For example, TET2 mutations can precede JAK2 mutations, and the order of mutation acquisition can affect disease phenotype, thrombosis risk, and in vitro sensitivity of JAK2-mutant progenitors to ruxolitinib, as well as the proliferative response to JAK2 V617F.46 Importantly, notwithstanding its effects on JAK2 V617F–positive clones, pegylated interferon-α-2a appears not to affect TET2-mutant cells.47 In the study of CALR-mutated ET mentioned earlier, molecular responses to pegylated interferon-α were poorer (minor to no molecular responses) in patients who also had TET2, ASXL1, IDH2, and/or TP53 mutations.41 All patients who achieved a CMR or partial molecular response to pegylated interferon-α had no additional mutations besides CALR.41 Achievement of a complete molecular response (in terms of JAK2 V617F but not TET2) to ruxolitinib, though infrequent, has been reported in patients with PV and ET (3 of 22 patients in one study at the 5-year time point).48
The telomerase inhibitor, imetelstat, was evaluated in a small phase II study in 18 patients with previously treated ET, with a primary end point of best hematologic response.49 All patients responded at some point in time, and 16 (89%) had a CHR as their best response. Molecular responses occurred in 7 of 8 patients with JAK2 V617F–positive mutations; CALR and MPL mutant allele burdens also decreased by 15% to 66%. Myelosuppression and hepatotoxicity were the major adverse events.49 Imetelstat is administered intravenously, and was initially administered once weekly in this study (maintenance dosing at a reduced frequency began after achievement of a CHR or partial hematologic response), a route and schedule that may be hard to justify when treating an indolent disease with a long natural history (median survival, 19.8 years even at a large, tertiary care referral center).3 Histone deacetylase inhibitors (HDACIs) are efficacious preclinically in MPN, both in vitro and in vivo,50,51 and are clearly active in patients with PV and ET, but the chronic low-grade toxicities of these agents (eg, fatigue, diarrhea, nausea, myelosuppression) make them difficult to tolerate long-term.52–54
Drug Development Pipeline in MF
The need for disease-modifying therapies is significantly greater in MF than in PV or ET, owing to the substantially more aggressive disease biology in MF. Although current pharmacologic therapy of primary MF centers on ruxolitinib,55 efforts are ongoing to develop novel ruxolitinib-based combinations, new JAK2 inhibitors, and other classes of drugs, such as antifibrotic agents and telomerase inhibitors. Given the survival benefit of ruxolitinib in patients with advanced MF,16,19,56 it is now being studied in patients with earlier stages of MF and one or more prognostically detrimental mutations (ASXL1, EZH2, SRSF2, IDH1, IDH2)57 in the ReTHINK trial (ClinicalTrials.gov identifier: NCT02598297).
Ruxolitinib-Based Combination Strategies
Combinations With Antianemia Medications: Anemia is both a hallmark of MF and an on-target effect of ruxolitinib, because erythropoiesis is critically dependent on JAK-STAT signaling.58 In fact, dose-limiting anemia and thrombocytopenia have been suggested as one potential explanation for the rather modest effect of ruxolitinib on JAK2 V617F allele burden reduction in MF.59 Medications generally used to alleviate the anemia of MF include danazol, erythropoiesis-stimulating agents (ESAs), immunomodulatory drugs (IMiDs; eg, thalidomide, lenalidomide, pomalidomide), and steroids.55 Anemia response rates to IMiDs are in the 20% to 30% range, whereas response rates to ESAs and androgens vary from 20% to 60%.55 Naturally, there is considerable interest in combining these agents with ruxolitinib, monotherapy with which causes an initial decline in the hemoglobin level, followed by the establishment of a new lower baseline.16,17 However, the simultaneous administration of ruxolitinib and lenalidomide is difficult because of excessive myelosuppression.60
POMINC is a trial of ruxolitinib and pomalidomide in MF that is ongoing in Germany (ClinicalTrials.gov identifier: NCT01644110), and one of ruxolitinib and thalidomide is planned in the United States. Although in theory, JAK2 inhibition would be expected to antagonize the effects of ESAs,58 in practice some improvement of anemia may be seen with the addition of ESAs to ruxolitinib. A trial of ruxolitinib plus danazol (n=14) was halted secondary to lack of anemia response61; 3 patients (21.4%) had clinical improvement in spleen size according to IWG-MRT criteria.22 These trials are listed in Table 2.
Finally, a new class of agents termed activin receptor IIA ligand traps have been shown to correct ineffective erythropoiesis and improve anemia in mouse models of α-thalassemia through inactivation of GDF11, a cytokine that blocks terminal erythroid maturation.62 These drugs have shown considerable efficacy in myelodysplastic syndrome (MDS) with ringed sideroblasts,63 and evaluation in MF is ongoing, with plans for combination with ruxolitinib (ClinicalTrials.gov identifier: NCT01712308).
Mechanism-Based Combinations With Other Targeted Agents: A number of different classes of targeted agents continue to be explored in combination with ruxolitinib in MF.25 These include epigenetic therapies such as DNA methyltransferase inhibitors (DNMTI) and HDACIs, phosphatidylinositol-3-kinase (PI3K)/Akt/mTOR inhibitors, and hedgehog inhibitors. Table 2 lists ongoing trials of ruxolitinib-based combinations in MF.
Epigenetic abnormalities are frequent in MF, and JAK2 influences nuclear gene transcription in a variety of ways, such that a number of mechanisms64 underlie the synergism observed between JAK2 inhibitors and epigenetic modifiers in MF. For HDACIs, an important concept is the downregulation, through acetylation, by HDAC6 inhibitors of the chaperone protein, heat shock protein 90, of which JAK2 is a client.65 This is a major mechanism through which HDACIs promote the degradation of JAK2 V617F and potentiate the effects of JAK2 inhibitors in MPN cells.66 HDACI monotherapy is definitely active in MF in terms of symptom and spleen responses
Clinical Trials (Nontransplant) of Ruxolitinib-Based Combinations in MF


Although aberrant promoter hypermethylation is characteristic of PV and ET, primary MF is characterized by both aberrant hypermethylation and hypomethylation.72 The DNMTI azacitidine has limited clinical activity on its own in patients with MF,73 but has been combined with ruxolitinib in a phase II trial in these patients.74 Azacitidine was introduced at a dose of 25 mg/m2/d for 5 days in 28-day cycles after 3 months of ruxolitinib alone, with provision for gradual dose increase to 75 mg/m2/d as tolerated. The response rate22 among 28 evaluable patients was 82%, with a median time to response of 1 month; 11 of 13 serially evaluable responders experienced a reduction in their JAK2 V617F allelic burden (2 experienced >50% reduction) and 6 of 22 (27%) had improvement in BM fibrosis grade. Most patients required dose interruptions and/or adjustments.74 In a study of 54 patients with post-MPN MDS or acute myeloid leukemia, azacitidine produced an overall response rate of 52% with a median response duration of 9 months and median overall survival of 11 months.75 Decitabine also appears active in patients with high-risk primary MF and MPN in accelerated or blastic phase,76 and clinical trials of ruxolitinib in combination with decitabine in the latter populations are ongoing (ClinicalTrials.gov identifiers: NCT02076191, NCT02257138).
Activation of the PI3K/Akt/mTOR pathway is an important downstream consequence of JAK-STAT signaling in MPN,77 and the dual PI3K/mTOR inhibitor, BEZ235, synergistically enhances the activity of JAK2 inhibitors against cultured and primary CD34-positive MPN cells, with relative sparing of normal CD34-positive hematopoietic progenitor cells.78 Ruxolitinib was combined with the pan-PI3K inhibitor buparlisib in a 2-arm, phase Ib study in intermediate- or high-risk patients with MF who could either be JAK inhibitor–naïve (arm A, n=22) or previously treated with JAK inhibitors (arm B, n=20).79 The maximum tolerated dose for the combination was determined to be 15 mg twice daily of ruxolitinib, along with 60 mg daily of buparlisib. A total of 55% and 20% of patients in arms A and B, respectively, achieved a 50% or greater reduction in palpable spleen length (considered approximately equivalent to ≥35% SVR) at week 24, but these percentages decreased to 45% and 18%, respectively, when only patients in the expansion phase receiving uniform drug doses were considered. After 24 weeks of treatment at the maximum tolerated dose, only 4 patients (3 in arm A; 1 in arm B) exhibited an improvement in BM fibrosis, whereas 2 in arm A had worsening BM fibrosis.79 Although the IWG-MRT response rate22 to everolimus (mTOR inhibitor) monotherapy in intermediate- or high-risk MF was only 23%, most patients experienced complete resolution of systemic symptoms.80
Excess hedgehog ligand secretion and loss of PTCH2, which drives canonical and noncanonical hedgehog signaling, have been described in MPN.81 The smoothened inhibitor, sonidegib, has been shown to improve leukocytosis and thrombocytosis, reduce the allelic burden of mutant JAK2, and significantly reduce BM fibrosis in a murine model of ET/MF.82 In a phase Ib/II study, the recommended phase II doses for combination ruxolitinib and sonidegib were found to be 20 mg twice daily and 400 mg daily, respectively.83 A total of 27 JAK inhibitor–naïve patients with intermediate- or high-risk MF and palpable splenomegaly were treated at these doses for a median of 28.6 weeks. Seventeen patients (63%) experienced adverse events requiring dose adjustment or interruption. At the end of week 24, 12 patients (44.4%) had a 35% or greater reduction in spleen volume (although 15 [55.6%] achieved ≥50% reduction in palpable spleen length), the mean reduction in JAK2 V617F allelic burden was 9%, and 2 patients had an improvement in BM fibrosis, whereas 3 had worsening.83
Newer JAK2 Inhibitors
A number of investigational JAK2 inhibitors have been discontinued, mostly owing to toxicity concerns, and ruxolitinib remains the only one currently approved for oncologic indications.84 Among the other compounds, only momelotinib, pacritinib, and NS-018 remain in clinical trials.84 Table 3 lists ongoing trials of these agents in MF.
Momelotinib is a JAK1/2 inhibitor that may produce significant anemia responses, in addition to spleen and symptom responses, among patients with MF.85 Some data suggest that momelotinib inhibits ALK2-mediated expression of hepcidin in the liver, which results in increased release of iron from sequestered cellular stores and enhanced erythropoiesis.86 In separate phase I/II studies (n=16687 and n=61,88 respectively) in patients with intermediate-2– or high-risk71 MF, or intermediate-1–risk71 MF with symptomatic organomegaly or unresponsiveness to available therapy, momelotinib produced spleen responses (by physical examination) in 39% and 72%, and anemia
Ongoing Clinical Trials (Nontransplant) of JAK Inhibitors Other Than Ruxolitinib in MF


The JAK2/FMS-like receptor tyrosine kinase 3 (FLT3) inhibitor pacritinib also potently inhibits colony-stimulating factor 1 receptor, and interleukin-1 receptor-associated kinase 1 and does not inhibit JAK1; these actions may explain its lack of myelosuppression.90 Pacritinib was evaluated in a phase II study in 35 patients with MF who were either newly diagnosed with an intermediate- or high-risk Lille score91 and not candidates for standard therapy or had splenomegaly that was poorly controlled with standard therapy.92 Patients with any degree of cytopenia were eligible. A total of 42% of evaluable patients experienced a 50% or greater reduction in spleen size from baseline up to week 24 by physical examination (31% of evaluable patients had ≥35% SVR on imaging) and 48.4% of patients had a 50% or greater reduction in TSS (using the Myelofibrosis Symptom Assessment Form [MFSAF])93 from baseline up to week 24. Grade 1 or 2 diarrhea (69%) and nausea (49%) were the most common adverse events.92 Pacritinib was then compared with BAT (excluding ruxolitinib) in the phase III PERSIST-1 trial in patients with MF.94 SVR rates at week 24 were 19.1% for pacritinib versus 4.7% for BAT in the intention-to-treat (ITT) analysis, and 25% versus 5.9%, respectively, among evaluable patients. A total of 24.5% versus 6.5% (ITT) and 40.9% versus 9.9% (evaluable population) of patients, respectively, had a 50% or greater reduction in their MPN-SAF TSS.23 A total of 25.7% of pacritinib patients became transfusion-independent versus 0% of patients randomized to BAT. Most adverse events in the pacritinib arm were grade 1/2 diarrhea, nausea, or vomiting.94 However, the FDA placed a “full clinical hold” on pacritinib after reports of deaths due to intracranial hemorrhage, cardiac failure, and cardiac arrest in the phase III PERSIST-2 trial (Table 3).
NS-018 is a JAK2-selective inhibitor currently undergoing phase II testing in patients with intermediate- or high-risk MF (ClinicalTrials.gov identifier: NCT01423851; Table 3). The recommended phase II dose based on data from the phase I portion of this trial is 300 mg/d.95 A total of 23 of the 48 patients enrolled on the phase I portion had received prior JAK2 inhibitor therapy. A 50% or greater reduction in palpable spleen size was achieved in 56% of patients, including 47% of patients with prior JAK inhibitor treatment.95 After 3 cycles of treatment, reductions in MFSAF scores93 were observed for all symptoms, and 11 of 30 (37%) evaluable patients had a reduction in BM fibrosis by 1 grade or more, although the JAK2 V617F allelic burden was largely unchanged.95 There was a hint from this study that NS-018 may elicit an early improvement in BM fibrosis in some patients, and that this might correlate with improvements in anemia and thrombocytopenia, as well as with spleen responses; however, the numbers were small, precluding firm conclusions. The cytokine regulation profile for NS-018 appears to be slightly different from that of ruxolitinib.95
Antifibrotic Agents
PRM-151 is an intravenously administered recombinant form of human pentraxin-2 (serum amyloid P, a highly conserved, naturally circulating plasma protein and soluble pattern recognition receptor of the innate immune system that may localize specifically to sites of injury and function to aid in the removal of damaged tissue)96 that was studied on weekly and every-4-week schedules, either alone or in conjunction with ruxolitinib, in 27 patients with intermediate- or high-risk97 MF and grade 2 or greater BM fibrosis.98 More than half had received a prior JAK inhibitor. In the first stage of the trial (24 weeks of therapy), 9 of 26 (35%) evaluable patients experienced response, 4 had symptoms of clinical improvement, and 6 had BM fibrosis responses.22 PRM-151 was well-tolerated in all 4 arms.98 A total of 18 patients continued onto an extension phase (stage 2), and data on 13 who had completed at least 72 weeks of treatment were presented last year.99 Symptom and spleen responses continued to improve over time, as did anemia and thrombocytopenia, with most transfusion-dependent patients becoming transfusion-independent. A total of 54% had a BM morphologic response, and 85% showed a response via computer-assisted image analysis; 69% and 38% of patients experienced a 50% or greater and 100% reductions from baseline, respectively, in their TSS23 between 24 and 72 weeks.99 In 5 patients with baseline hemoglobin level less than 10 g/dL, the median hemoglobin level increased from 8.6 g/dL at baseline to 10.7 g/dL at week 72. Three of these patients who were receiving transfusions at baseline achieved transfusion independence, with durations of 32 to 60 weeks.99 Similarly, of 9 patients with baseline platelet counts of less than 100 x 109/L, the median platelet count increased from 38 x 109/L at baseline to 52 x 109/L. Four of these patients were receiving platelet transfusions at baseline, and all became transfusion-independent.99 No serious adverse events related to PRM-151 occurred.99
Imetelstat
Imetelstat is an intravenously administered lipid-conjugated oligonucleotide telomerase inhibitor that was studied in 33 patients with intermediate-2– or high-risk100 MF, 48% of whom had received prior JAK inhibitor therapy101; 7 patients (21%) achieved a complete or partial response.22 BM fibrosis was reversed in all 4 patients with a complete response, 3 of whom also experienced a molecular response. Responses did not correlate with baseline telomere length. Responses were restricted to patients with mutated JAK2, and were not seen in the presence of ASXL1 mutations.101 Furthermore, responses appeared to correlate with the presence of mutations in the RNA splicing genes, SF3B1 or U2AF1. Grade 3/4 myelosuppression and grade 1/2 hepatotoxicity were frequent. However, none of the abnormalities in liver-enzyme levels were linked to clinically overt liver damage and most were reversible.101 Imetelstat, 9.4 or 4.7 mg/kg every 3 weeks, is currently being evaluated in patients with intermediate-2– and high-risk MF that failed to respond to therapy with a JAK inhibitor in a phase II study (ClinicalTrials.gov identifier: NCT02426086). In a recent press release, Geron Corporation (Menlo Park, CA) announced that the 4.7-mg/kg dose will not be evaluated further due to insufficient activity.102
Conclusions
This review summarizes the drugs currently in clinical development for MPNs, with a focus on those most likely to influence MPN management guidelines over the next several years, as has occurred with ruxolitinib. It is possible that ruxolitinib will be used in lower-risk patients with MF, particularly those with adverse genomic features, based on findings from the ReTHINK study. Ruxolitinib-based combinations, particularly with DNMTI and HDACIs, as well as with antianemia medications, are likely to gain widespread acceptance. Regulatory approval of one or more JAK inhibitors beyond ruxolitinib is anticipated, although the discontinuation of many of these agents tells a cautionary tale. The experience with PRM-151 and imetelstat is limited, but these drugs are attractive by virtue of their ability to improve or reverse BM fibrosis.
Despite the success of ruxolitinib, there remain many unmet needs in terms of drug development for patients with MF. First and foremost, the survival of these patients remains short, and the modest gains in survival achieved with ruxolitinib may be more attributable to improvement in appetite, weight gain, correction of hypocholesterolemia, and reversal of cachexia than to actual targeting of the neoplastic clone.103 Other areas where novel therapeutics are needed include anemia of MF, whether from the disease or worsened by ruxolitinib. The infectious risks of long-term JAK inhibitor therapy remain an important concern. Ruxolitinib's approval is a welcome advancement in the treatment of PV, as is the advent of ropeginterferon-α. Approval of these or other agents is awaited by patients with ET who have troublesome symptoms. Finally, no current therapy reliably prevents progression of PV or ET to MF or acute myeloid leukemia.
Dr. Bose has disclosed that he has received honorarium from Incyte Corporation. Dr. Verstovsek has disclosed that he has received research funding from Incyte Corporation, AstraZeneca, Lilly Oncology, Roche, Geron, NS Pharma, Bristol Myers Squibb, Celgene Corporation, Gilead Sciences, Seattle Genetics, Promedior, CTI Biopharma Corporation, Galena Biopharma, Pfizer, Inc., Genentech, and Blueprint Pharmaceuticals.
References
- 1.↑
Vannucchi AM, Barbui T, Cervantes F et al.. Philadelphia chromosome-negative chronic myeloproliferative neoplasms: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2015;26(Suppl 5):v85–99.
- 2.↑
Cervantes F, Dupriez B, Passamonti F et al.. Improving survival trends in primary myelofibrosis: an international study. J Clin Oncol 2012;30:2981–2987.
- 3.↑
Tefferi A, Guglielmelli P, Larson DR et al.. Long-term survival and blast transformation in molecularly annotated essential thrombocythemia, polycythemia vera, and myelofibrosis. Blood 2014;124:2507–2513; quiz 2615.
- 4.↑
Tefferi A, Rumi E, Finazzi G et al.. Survival and prognosis among 1545 patients with contemporary polycythemia vera: an international study. Leukemia 2013;27:1874–1881.
- 5.↑
Anand S, Stedham F, Gudgin E et al.. Increased basal intracellular signaling patterns do not correlate with JAK2 genotype in human myeloproliferative neoplasms. Blood 2011;118:1610–1621.
- 6.↑
Rampal R, Al-Shahrour F, Abdel-Wahab O et al.. Integrated genomic analysis illustrates the central role of JAK-STAT pathway activation in myeloproliferative neoplasm pathogenesis. Blood 2014;123:e123–133.
- 7.↑
Baxter EJ, Scott LM, Campbell PJ et al.. Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet 2005;365:1054–1061.
- 8.
James C, Ugo V, Le Couedic JP et al.. A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. Nature 2005;434:1144–1148.
- 9.
Kralovics R, Passamonti F, Buser AS et al.. A gain-of-function mutation of JAK2 in myeloproliferative disorders. N Engl J Med 2005;352:1779–1790.
- 10.
Levine RL, Wadleigh M, Cools J et al.. Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. Cancer Cell 2005;7:387–397.
- 11.↑
Scott LM, Tong W, Levine RL et al.. JAK2 exon 12 mutations in polycythemia vera and idiopathic erythrocytosis. N Engl J Med 2007;356:459–468.
- 12.↑
Pardanani AD, Levine RL, Lasho T et al.. MPL515 mutations in myeloproliferative and other myeloid disorders: a study of 1182 patients. Blood 2006;108:3472–3476.
- 13.↑
Pikman Y, Lee BH, Mercher T et al.. MPLW515L is a novel somatic activating mutation in myelofibrosis with myeloid metaplasia. PLoS Med 2006;3:e270.
- 14.↑
Klampfl T, Gisslinger H, Harutyunyan AS et al.. Somatic mutations of calreticulin in myeloproliferative neoplasms. N Engl J Med 2013;369:2379–2390.
- 15.↑
Nangalia J, Massie CE, Baxter EJ et al.. Somatic CALR mutations in myeloproliferative neoplasms with nonmutated JAK2. N Engl J Med 2013;369:2391–2405.
- 16.↑
Verstovsek S, Mesa RA, Gotlib J et al.. A double-blind, placebo-controlled trial of ruxolitinib for myelofibrosis. N Engl J Med 2012;366:799–807.
- 17.↑
Harrison C, Kiladjian JJ, Al-Ali HK et al.. JAK inhibition with ruxolitinib versus best available therapy for myelofibrosis. N Engl J Med 2012;366:787–798.
- 18.↑
Vannucchi AM, Kiladjian JJ, Griesshammer M et al.. Ruxolitinib versus standard therapy for the treatment of polycythemia vera. N Engl J Med 2015;372:426–435.
- 19.↑
Verstovsek S, Mesa RA, Gotlib JR et al.. Long-term outcomes of ruxolitinib (RUX) therapy in patients (pts) with myelofibrosis (MF): 5-year final efficacy and safety analysis from COMFORT-I [abstract]. J Clin Oncol 2016;35(Supp):Abstract 7012.
- 20.↑
Harrison CN, Vannucchi AM, Kiladjian JJ et al.. Long-term findings from COMFORT-II, a phase 3 study of ruxolitinib vs best available therapy for myelofibrosis. Leukemia 2016;30:1701–1707.
- 21.↑
Mesa RA, Gotlib J, Gupta V et al.. Effect of ruxolitinib therapy on myelofibrosis-related symptoms and other patient-reported outcomes in COMFORT-I: a randomized, double-blind, placebo-controlled trial. J Clin Oncol 2013;31:1285–1292.
- 22.↑
Tefferi A, Cervantes F, Mesa R et al.. Revised response criteria for myelofibrosis: International Working Group-Myeloproliferative Neoplasms Research and Treatment (IWG-MRT) and European LeukemiaNet (ELN) consensus report. Blood 2013;122:1395–1398.
- 23.↑
Emanuel RM, Dueck AC, Geyer HL et al.. Myeloproliferative neoplasm (MPN) symptom assessment form total symptom score: prospective international assessment of an abbreviated symptom burden scoring system among patients with MPNs. J Clin Oncol 2012;30:4098–4103.
- 24.↑
Guglielmelli P, Rotunno G, Bogani C et al.. Ruxolitinib is an effective treatment for CALR-positive patients with myelofibrosis. Br J Haematol 2016;173:938–940.
- 25.↑
Stein BL, Swords R, Hochhaus A, Giles F. Novel myelofibrosis treatment strategies: potential partners for combination therapies. Leukemia 2014;28:2139–2147.
- 26.↑
Kroger NM, Deeg JH, Olavarria E et al.. Indication and management of allogeneic stem cell transplantation in primary myelofibrosis: a consensus process by an EBMT/ELN international working group. Leukemia 2015;29:2126–2133.
- 27.↑
Spoerl S, Mathew NR, Bscheider M et al.. Activity of therapeutic JAK 1/2 blockade in graft-versus-host disease. Blood 2014;123:3832–3842.
- 28.↑
Zeiser R, Burchert A, Lengerke C et al.. Ruxolitinib in corticosteroid-refractory graft-versus-host disease after allogeneic stem cell transplantation: a multicenter survey. Leukemia 2015;29:2062–2068.
- 29.↑
Landolfi R, Marchioli R, Kutti J et al.. Efficacy and safety of low-dose aspirin in polycythemia vera. N Engl J Med 2004;350:114–124.
- 30.↑
Cortelazzo S, Finazzi G, Ruggeri M et al.. Hydroxyurea for patients with essential thrombocythemia and a high risk of thrombosis. N Engl J Med 1995;332:1132–1136.
- 31.
Harrison CN, Campbell PJ, Buck G et al.. Hydroxyurea compared with anagrelide in high-risk essential thrombocythemia. N Engl J Med 2005;353:33–45.
- 32.↑
Gisslinger H, Gotic M, Holowiecki J et al.. Anagrelide compared with hydroxyurea in WHO-classified essential thrombocythemia: the ANAHYDRET Study, a randomized controlled trial. Blood 2013;121:1720–1728.
- 33.↑
Marchioli R, Finazzi G, Specchia G et al.. Cardiovascular events and intensity of treatment in polycythemia vera. N Engl J Med 2013;368:22–33.
- 34.↑
Geyer HL, Scherber RM, Dueck AC et al.. Distinct clustering of symptomatic burden among myeloproliferative neoplasm patients: retrospective assessment in 1470 patients. Blood 2014;123:3803–3810.
- 35.
Mesa RA, Niblack J, Wadleigh M et al.. 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.
- 36.↑
Scherber R, Dueck AC, Johansson P et al.. The Myeloproliferative Neoplasm Symptom Assessment Form (MPN-SAF): international prospective validation and reliability trial in 402 patients. Blood 2011;118:401–408.
- 37.↑
Verstovsek S, Vannucchi AM, Griesshammer M et al.. Ruxolitinib versus best available therapy in patients with polycythemia vera: 80-week follow-up from the RESPONSE trial. Haematologica 2016;101:821–829.
- 38.↑
Kiladjian JJ, Cassinat B, Chevret S et al.. Pegylated interferon-alfa-2a induces complete hematologic and molecular responses with low toxicity in polycythemia vera. Blood 2008;112:3065–3072.
- 39.↑
Quintas-Cardama A, Kantarjian H, Manshouri T et al.. Pegylated interferon alfa-2a yields high rates of hematologic and molecular response in patients with advanced essential thrombocythemia and polycythemia vera. J Clin Oncol 2009;27:5418–5424.
- 40.↑
Masarova L, Verstovsek S, Patel KP et al.. Efficacy and safety of pegylated interferon alpha-2a in patients with essential thrombocythemia and polycythemia vera: results after a median 7-year follow-up of a phase 2 study [abstract]. Blood 2015;126:Abstract 60.
- 41.↑
Verger E, Cassinat B, Chauveau A et al.. Clinical and molecular response to interferon-alpha therapy in essential thrombocythemia patients with CALR mutations. Blood 2015;126:2585–2591.
- 42.↑
Gisslinger H, Zagrijtschuk O, Buxhofer-Ausch V et al.. Ropeginterferon alfa-2b, a novel IFNalpha-2b, induces high response rates with low toxicity in patients with polycythemia vera. Blood 2015;126:1762–1769.
- 43.↑
Vannucchi AM, Verstovsek S, Guglielmelli P et al.. Ruxolitinib (RUX) reduces JAK2V617F allele burden (AB) in patients (pts) with polycythemia vera (PV) enrolled in the response study. Presented at the 21st Congress of the European Hematology Association; June 9–12, 2016; Copenhagen, Denmark.
- 44.↑
Vainchenker W, Delhommeau F, Constantinescu SN, Bernard OA. New mutations and pathogenesis of myeloproliferative neoplasms. Blood 2011;118:1723–1735.
- 45.↑
Kralovics R, Teo SS, Li S et al.. Acquisition of the V617F mutation of JAK2 is a late genetic event in a subset of patients with myeloproliferative disorders. Blood 2006;108:1377–1380.
- 46.↑
Ortmann CA, Kent DG, Nangalia J et al.. Effect of mutation order on myeloproliferative neoplasms. N Engl J Med 2015;372:601–612.
- 47.↑
Kiladjian JJ, Masse A, Cassinat B et al.. Clonal analysis of erythroid progenitors suggests that pegylated interferon alpha-2a treatment targets JAK2V617F clones without affecting TET2 mutant cells. Leukemia 2010;24:1519–1523.
- 48.↑
Pieri L, Pancrazzi A, Pacilli A et al.. JAK2V617F complete molecular remission in polycythemia vera/essential thrombocythemia patients treated with ruxolitinib. Blood 2015;125:3352–3353.
- 49.↑
Baerlocher GM, Oppliger Leibundgut E, Ottmann OG et al.. Telomerase inhibitor imetelstat in patients with essential thrombocythemia. N Engl J Med 2015;373:920–928.
- 50.↑
Guerini V, Barbui V, Spinelli O et al.. The histone deacetylase inhibitor ITF2357 selectively targets cells bearing mutated JAK2(V617F). Leukemia 2008;22:740–747.
- 51.↑
Akada H, Akada S, Gajra A et al.. Efficacy of vorinostat in a murine model of polycythemia vera. Blood 2012;119:3779–3789.
- 52.↑
Andersen CL, McMullin MF, Ejerblad E et al.. A phase II study of vorinostat (MK-0683) in patients with polycythaemia vera and essential thrombocythaemia. Br J Haematol 2013;162:498–508.
- 53.
Rambaldi A, Dellacasa CM, Finazzi G et al.. A pilot study of the histone-deacetylase inhibitor givinostat in patients with JAK2V617F positive chronic myeloproliferative neoplasms. Br J Haematol 2010;150:446–455.
- 54.↑
Finazzi G, Vannucchi AM, Martinelli V et al.. A phase II study of Givinostat in combination with hydroxycarbamide in patients with polycythaemia vera unresponsive to hydroxycarbamide monotherapy. Br J Haematol 2013;161:688–694.
- 56.↑
Harrison CN, Vannucchi AM, Kiladjian J et al.. Long-term efficacy and safety in COMFORT-II, a phase 3 study comparing ruxolitinib with best available therapy for the treatment of myelofibrosis: 5-year final study results. Blood 2015;126:59.
- 57.↑
Vannucchi AM, Lasho TL, Guglielmelli P et al.. Mutations and prognosis in primary myelofibrosis. Leukemia 2013;27:1861–1869.
- 58.↑
Parganas E, Wang D, Stravopodis D et al.. Jak2 is essential for signaling through a variety of cytokine receptors. Cell 1998;93:385–395.
- 59.↑
Meyer SC, Levine RL. Molecular pathways: molecular basis for sensitivity and resistance to JAK kinase inhibitors. Clin Cancer Res 2014;20:2051–2059.
- 60.↑
Daver N, Cortes J, Newberry K et al.. Ruxolitinib in combination with lenalidomide as therapy for patients with myelofibrosis. Haematologica 2015;100:1058–1063.
- 61.↑
Gowin KM, Kosiorek HE, Dueck AC et al.. Final analysis of a multicenter pilot phase 2 study of ruxolitinib and danazol in patients with myelofibrosis [abstract]. Blood 2015;126:Abstract 1618.
- 62.↑
Dussiot M, Maciel TT, Fricot A et al.. An activin receptor IIA ligand trap corrects ineffective erythropoiesis in beta-thalassemia. Nat Med 2014;20:398–407.
- 63.↑
Giagounidis A, Platzbecker U, Germing U et al.. Luspatercept treatment leads to long term increases in hemoglobin and reductions in transfusion burden in patients with low or intermediate-1 risk myelodysplastic syndromes (MDS): preliminary results from the phase 2 PACE-MDS Extension Study [abstract]. Blood 2015;126:Abstract 92.
- 64.↑
Mascarenhas J, Roper N, Chaurasia P, Hoffman R. Epigenetic abnormalities in myeloproliferative neoplasms: a target for novel therapeutic strategies. Clin Epigenetics 2011;2:197–212.
- 65.↑
Bali P, Pranpat M, Bradner J et al.. Inhibition of histone deacetylase 6 acetylates and disrupts the chaperone function of heat shock protein 90: a novel basis for antileukemia activity of histone deacetylase inhibitors. J Biol Chem 2005;280:26729–26734.
- 66.↑
Wang Y, Fiskus W, Chong DG et al.. Cotreatment with panobinostat and JAK2 inhibitor TG101209 attenuates JAK2V617F levels and signaling and exerts synergistic cytotoxic effects against human myeloproliferative neoplastic cells. Blood 2009;114:5024–5033.
- 67.↑
Mascarenhas J, Lu M, Li T et al.. A phase I study of panobinostat (LBH589) in patients with primary myelofibrosis (PMF) and post-polycythaemia vera/essential thrombocythaemia myelofibrosis (post-PV/ET MF). Br J Haematol 2013;161:68–75.
- 68.
DeAngelo DJ, Mesa RA, Fiskus W et al.. Phase II trial of panobinostat, an oral pan-deacetylase inhibitor in patients with primary myelofibrosis, post-essential thrombocythaemia, and post-polycythaemia vera myelofibrosis. Br J Haematol 2013;162:326–335.
- 69.↑
Quintas-Cardama A, Kantarjian H, Estrov Z et al.. Therapy with the histone deacetylase inhibitor pracinostat for patients with myelofibrosis. Leuk Res 2012;36:1124–1127.
- 70.↑
Harrison CN, Kiladjian JJ, Heidel FH et al.. 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) [abstract]. Blood 2015;126:Abstract 4060.
- 71.↑
Cervantes F, Dupriez B, Pereira A et al.. New prognostic scoring system for primary myelofibrosis based on a study of the International Working Group for Myelofibrosis Research and Treatment. Blood 2009;113:2895–2901.
- 72.↑
Nischal S, Bhattacharyya S, Christopeit M et al.. Methylome profiling reveals distinct alterations in phenotypic and mutational subgroups of myeloproliferative neoplasms. Cancer Res 2013;73:1076–1085.
- 73.↑
Quintas-Cardama A, Tong W, Kantarjian H et al.. A phase II study of 5-azacitidine for patients with primary and post-essential thrombocythemia/polycythemia vera myelofibrosis. Leukemia 2008;22:965–970.
- 74.↑
Daver NG, Cortes JE, Zhou L et al.. Ruxolitinib (RUX) in combination with 5-azacytidine (AZA) as therapy for patients (pts) with myelofibrosis (MF) [abstract]. Haematologica 2015;15(Suppl 2):Abstract 808.
- 75.↑
Thepot S, Itzykson R, Seegers V et al.. Treatment of progression of Philadelphia-negative myeloproliferative neoplasms to myelodysplastic syndrome or acute myeloid leukemia by azacitidine: a report on 54 cases on the behalf of the Groupe Francophone des Myelodysplasies (GFM). Blood 2010;116:3735–3742.
- 76.↑
Badar T, Kantarjian HM, Ravandi F et al.. Therapeutic benefit of decitabine, a hypomethylating agent, in patients with high-risk primary myelofibrosis and myeloproliferative neoplasm in accelerated or blastic/acute myeloid leukemia phase. Leuk Res 2015;39:950–956.
- 77.↑
Kamishimoto J, Tago K, Kasahara T, Funakoshi-Tago M. Akt activation through the phosphorylation of erythropoietin receptor at tyrosine 479 is required for myeloproliferative disorder-associated JAK2 V617F mutant-induced cellular transformation. Cell Signal 2011;23:849–856.
- 78.↑
Fiskus W, Verstovsek S, Manshouri T et al.. 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;12:577–588.
- 79.↑
Durrant S, Nagler A, Vannucchi AM et al.. An open-label, multicenter, 2-arm, dose-finding, phase 1b study of the combination of ruxolitinib and buparlisib (BKM120) in patients with myelofibrosis: results from HARMONY study [abstract]. Blood 2015;126:Abstract 827.
- 80.↑
Guglielmelli P, Barosi G, Rambaldi A et al.. Safety and efficacy of everolimus, a mTOR inhibitor, as single agent in a phase 1/2 study in patients with myelofibrosis. Blood 2011;118:2069–2076.
- 81.↑
Klein C, Zwick A, Kissel S et al.. Ptch2 loss drives myeloproliferation and myeloproliferative neoplasm progression. J Exp Med 2016;213:273–290.
- 82.↑
Bhagwat N, Keller MD, Rampal R et al.. Improved efficacy of combination of JAK2 and hedgehog inhibitors in myelofibrosis [abstract]. Blood 2013;122:Abstract 666.
- 83.↑
Gupta V, Harrison CN, Hasselbalch HC et al.. Phase 1b/2 study of the efficacy and safety of sonidegib (LDE225) in combination with ruxolitinib (INC424) in patients with myelofibrosis [abstract]. Blood 2015;126:Abstract 825.
- 84.↑
Geyer HL, Mesa RA. Therapy for myeloproliferative neoplasms: when, which agent, and how? Blood 2014;124:3529–3537.
- 85.↑
Pardanani A, Laborde RR, Lasho TL et al.. Safety and efficacy of CYT387, a JAK1 and JAK2 inhibitor, in myelofibrosis. Leukemia 2013;27:1322–1327.
- 86.↑
Asshoff M, Warr M, Haschka D et al.. The Jak1/Jak2 inhibitor momelotinib inhibits Alk2, decreases hepcidin production and ameliorates anemia of chronic disease (ACD) in rodents [abstract]. Blood 2015;126:Abstract 538.
- 87.↑
Pardanani A, Gotlib JR, Gupta V et al.. Update on the long-term efficacy and safety of momelotinib, a JAK1 and JAK2 inhibitor, for the treatment of myelofibrosis [abstract]. Blood 2013;122:Abstract 108.
- 88.↑
Gupta V, Mesa RA, Deininger MW et al.. A phase 1/2, open-label study evaluating twice-daily administration of momelotinib in myelofibrosis [published online ahead of print September 15, 2016]. Haematologica, pii: haematol.2016.148924.
- 89.↑
Abdelrahman RA, Begna KH, Al-Kali A et al.. Momelotinib treatment-emergent neuropathy: prevalence, risk factors and outcome in 100 patients with myelofibrosis. Br J Haematol 2015;169:77–80.
- 90.↑
Singer JW, Al-Fayoumi S, Ma H et al.. Comprehensive kinase profile of pacritinib, a nonmyelosuppressive Janus kinase 2 inhibitor. J Exp Pharmacol 2016;8:11–19.
- 91.↑
Dupriez B, Morel P, Demory JL et al.. Prognostic factors in agnogenic myeloid metaplasia: a report on 195 cases with a new scoring system. Blood 1996;88:1013–1018.
- 92.↑
Komrokji RS, Seymour JF, Roberts AW et al.. Results of a phase 2 study of pacritinib (SB1518), a JAK2/JAK2(V617F) inhibitor, in patients with myelofibrosis. Blood 2015;125:2649–2655.
- 93.↑
Mesa RA, Schwager S, Radia D et al.. The Myelofibrosis Symptom Assessment Form (MFSAF): an evidence-based brief inventory to measure quality of life and symptomatic response to treatment in myelofibrosis. Leuk Res 2009;33:1199–1203.
- 94.↑
Mesa RA, Egyed M, Szoke A et al.. Pacritinib (PAC) vs best available therapy (BAT) in myelofibrosis (MF): 60 week follow-up of the phase III PERSIST-1 trial [abstract]. J Clin Oncol 2016;34:Abstract 7065.
- 95.↑
Verstovsek S, Talpaz M, Ritchie E et al.. A Phase I, open-label, dose-escalation, multicenter study of the JAK2 inhibitor NS-018 in patients with myelofibrosis [published online ahead of print September 2, 2016]. Leukemia, doi: 10.1038/leu.2016.215.
- 96.↑
Duffield JS, Lupher ML Jr. PRM-151 (recombinant human serum amyloid P/pentraxin 2) for the treatment of fibrosis. Drug News Perspect 2010;23:305–315.
- 97.↑
Passamonti F, Cervantes F, Vannucchi AM et al.. A dynamic prognostic model to predict survival in primary myelofibrosis: a study by the IWG-MRT (International Working Group for Myeloproliferative Neoplasms Research and Treatment). Blood 2010;115:1703–1708.
- 98.↑
Verstovsek S, Mesa RA, Foltz LM et al.. Phase 2 trial of PRM-151, an anti-fibrotic agent, in patients with myelofibrosis: stage 1 results [abstract]. Blood 2014;124:Abstract 713.
- 99.↑
Verstovsek S, Mesa RA, Foltz LM et al.. PRM-151 in myelofibrosis: durable efficacy and safety at 72 weeks [abstract]. Blood 2015;126:Abstract 56.
- 100.↑
Gangat N, Caramazza D, Vaidya R et al.. DIPSS plus: a refined Dynamic International Prognostic Scoring System for primary myelofibrosis that incorporates prognostic information from karyotype, platelet count, and transfusion status. J Clin Oncol 2011;29:392–397.
- 101.↑
Tefferi A, Lasho TL, Begna KH et al.. A pilot study of the telomerase inhibitor imetelstat for myelofibrosis. N Engl J Med 2015;373:908–919.
- 102.↑
Geron Provides Update on Imetelstat Trials Being Conducted by Janssen. Geron Web site. Available at: http://ir.geron.com/phoenix.zhtml?c=67323&p=irol-newsArticle&ID=2201055. Accessed September 18, 2016.
- 103.↑
Mascarenhas J, Hoffman R. A comprehensive review and analysis of the effect of ruxolitinib therapy on the survival of patients with myelofibrosis. Blood 2013;121:4832–4837.
- 104.
Barosi G, Birgegard G, Finazzi G et al.. 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;148:961–963.
- 105.
Barosi G, Birgegard G, Finazzi G et al.. Response criteria for essential thrombocythemia and polycythemia vera: result of a European LeukemiaNet consensus conference. Blood 2009;113:4829–4833.
- 106.
Tefferi A, Verstovsek S, Barosi G et al.. Pomalidomide is active in the treatment of anemia associated with myelofibrosis. J Clin Oncol 2009;27:4563–4569.
- 107.
Tefferi A, Passamonti F, Barbui T et al.. Phase 3 study of pomalidomide in myeloproliferative neoplasm (MPN)-associated myelofibrosis with RBC-transfusion-dependence [abstract]. Blood 2013;122:Abstract 394.
- 108.
Radin AI, Kim HT, Grant BW et al.. Phase II study of alpha2 interferon in the treatment of the chronic myeloproliferative disorders (E5487): a trial of the Eastern Cooperative Oncology Group. Cancer 2003;98:100–109.
- 109.
Jabbour E, Kantarjian H, Cortes J et al.. PEG-IFN-alpha-2b therapy in BCR-ABL-negative myeloproliferative disorders: final result of a phase 2 study. Cancer 2007;110:2012–2018.
- 110.
Silver RT, Vandris K, Goldman JJ. Recombinant interferon-alpha may retard progression of early primary myelofibrosis: a preliminary report. Blood 2011;117:6669–6672.