Skip to main content
Journal of Clinical Oncology logoLink to Journal of Clinical Oncology
. 2022 Jan 27;40(11):1147–1154. doi: 10.1200/JCO.21.02246

Disease Modification in Myelofibrosis: An Elusive Goal?

Pankit Vachhani 1, Srdan Verstovsek 2, Prithviraj Bose 2,
PMCID: PMC8987221  PMID: 35084934

Introduction

Janus kinase (JAK) inhibitors represent the cornerstone of myelofibrosis (MF) management, consistent with the ubiquitous activation of JAK-signal transducer and activator of transcription signaling in the Philadelphia chromosome–negative myeloproliferative neoplasms.1,2 Their broad immunosuppressive and anti-inflammatory actions underlie their efficacy in graft-versus-host disease,3,4 and rheumatoid5,6 and psoriatic arthritis.7,8 However, notwithstanding their undeniable benefits in ameliorating splenomegaly and MF-related symptoms,9,10 they are viewed as being predominantly anti-inflammatory, with limited disease-modifying effects. The latter remain poorly understood, however. Reductions in bone marrow fibrosis (BMF) and driver mutation (JAK2, MPL, and CALR) expressed allele burden, only modestly achieved by ruxolitinib,11,12 have generally been considered to represent disease modification. However, BMF reduction should be accompanied by improvement in cytopenias, progression-free survival (PFS), or overall survival (OS) to be clinically meaningful, and MF is genomically a complex disease not addicted to a single molecular driver. Modulation of cytokines has been advanced as an example of disease modification, but clinical correlation is mostly limited to symptom improvement.13 No therapy has been demonstrated to thwart leukemic transformation. With a plethora of novel agents14 in development (Table 1), several new primary end points (beyond the usual ≥ 35% spleen volume reduction [SVR35] and ≥ 50% reduction in total symptom score [TSS50] at 24 weeks), including red blood cell transfusion independence (RBC-TI) and OS, are being adopted in pivotal trials. There is also much discussion of the disease-modifying potential of the newer agents, sometimes in combination with JAK inhibitors, on the basis of preclinical synergism.15,16

TABLE 1.

Candidate Surrogate Markers for Disease Modification by JAK Inhibitors and Other Novel Therapies in Development for Myelofibrosis

graphic file with name jco-40-1147-g001.jpg

Survival Outcomes in JAK Inhibitor–Naive and JAK Inhibitor–Exposed Patients

An exploratory analysis of pooled 5-year data from the randomized COMFORT trials (N = 528) demonstrated a survival benefit for ruxolitinib, despite crossover.10 An OS advantage for ruxolitinib was also documented in comparisons of trial patients with matched historical controls17,18 and in real-world studies.19,20 Considering the modest impacts of ruxolitinib on BMF and JAK2 V617F expressed allele burden,11,12 survival prolongation by ruxolitinib may be indirect, ie, through improvements in metabolism, organ function, and overall well-being.9 Although spleen size may not be prognostic in MF, multiple studies have demonstrated that the achievement, durability, and degree of spleen response to ruxolitinib correlate with OS.17,21,22 Spleen response, thus, likely serves as a biomarker of JAK inhibition in MF. However, it is not clear that early intervention with ruxolitinib in patients without symptomatic splenomegaly or significant MF-related symptoms improves survival; therefore, consensus guidelines recommend using ruxolitinib on the basis of the presence of symptoms and/or splenomegaly, rather than purely for extending survival.23-25

Long-term follow-up data are lacking for the randomized JAKARTA trial of fedratinib26 because of the temporary full clinical hold (all patients had to stop therapy) that was mandated by the US Food and Drug Administration due to concerns over Wernicke's encephalopathy. Nevertheless, despite short follow-up and censoring, median PFS was significantly better for fedratinib 400 mg/d versus placebo.27 Median OS had not been reached in either group. Importantly, PFS is a relatively novel end point in MF trials, and data to establish the surrogacy of PFS for OS are currently lacking.

The randomized SIMPLIFY-1 and -2 trials evaluated the JAK1/2 and activin receptor type 1 (ACVR1) inhibitor momelotinib in the JAK inhibitor–naive and –pretreated settings, respectively.28,29 In both trials, significantly greater proportions of patients in the momelotinib arms achieved RBC-TI at 24 weeks; interestingly, in SIMPLIFY-1, the achievement of 24-week RBC-TI on momelotinib, but not achievement of SVR35 or TSS50 at week 24, correlated significantly with OS.30 This relationship trended toward statistical significance in SIMPLIFY-2. Requirement for RBC transfusions is a well-established adverse prognostic factor in MF; thus, the correlation of RBC-TI with OS is not unexpected and may not be unique to momelotinib, although the anemia benefit of this drug does appear to be unique among current JAK inhibitors.31

Prognosis after ruxolitinib discontinuation is poor (median OS, 11-14 months).32-35 Ruxolitinib failure (variously defined, and may include patients who exhibit intolerance, suboptimal, lack, or loss of response) is an area of intense new drug development. In a phase II trial of imetelstat (9.4 mg/kg intravenously once every 3 weeks) in patients with MF who had failed JAK inhibitor therapy, the median OS was 29.9 months after a median follow-up of 27.4 months.36 By contrast, the rate of SVR35 at 24 weeks was a modest 10.2%. The apparent OS advantage for imetelstat in the ruxolitinib failure setting was, however, validated in a comparison with a closely matched cohort of patients seen at a major academic center.37 Imetelstat is now being studied in a registrational, phase III trial (IMpactMF, ClinicalTrials.gov identifier: NCT04576156) in the JAK inhibitor failure setting with OS as the primary end point, unprecedented in MF pivotal trials. Interestingly, however, in an Italian study (N = 218), the median OS of patients who remained in chronic phase at ruxolitinib discontinuation (n = 167) was 27.5 months, whereas that of patients whose disease transformed to blast phase was 3.9 months.34 Furthermore, there were no survival differences between the chronic phase patients who discontinued ruxolitinib because of resistance versus intolerance. These observations underscore the likely heterogeneity of ruxolitinib failure populations in various reports. It is possible, for instance, that ruxolitinib is discontinued much later in the disease course at certain centers than at others, eg, those highly experienced in management of this drug. This could significantly affect survival after ruxolitinib discontinuation.

Candidate Biomarkers of Disease Modification: Problems and Pitfalls

Bone marrow fibrosis.

Long considered reactive, there is evidence that the fibrocytes that drive BMF in MF are clonal (neoplastic).38 BMF improvement has been reported in a number of recent studies evaluating novel therapeutics (Table 1); however, the significance of this per se remains doubtful, unless a clear correlation with improvement in cytopenias (often confounded by myelosuppression from the drugs themselves), or prolongation of PFS or OS, can be shown. BMF improvement, although modest in comparison to spleen and symptom responses, has also been observed with JAK inhibitor therapy, particularly in the setting of prolonged treatment.39,40 Of note, successful allogeneic hematopoietic cell transplantation, to date the only potential cure for MF, often results in complete regression of BMF, the speed of attainment of which may predict OS independently of prognosis at transplantation.41 Although current BMF grading systems mainly consider reticulin fibrosis, there is increasing recognition that assessment of collagen and the grade of osteosclerosis provides important additional information.42 With the exception of PRM-151,43,44 most contemporary clinical trials reporting rates of BMF improvement have not separately reported changes in reticulin and collagen. In general, reproducibility of BMF assessment in expert hands is high (approximately 90%),42 emphasizing the importance of central expert pathology review of samples from clinical trial participants.

Driver mutation allele burden reduction.

Reduction in the variant allele frequencies (VAFs) of mutated JAK2, MPL, and CALR has intuitive appeal as a measure of disease modification, but MF is characterized by a high incidence of other, non–phenotype-driving mutations.45,46 In a small study of pegylated interferon alfa-2a in patients with early-stage MF (N = 30), the presence of such mutations predicted poorer responses.47 Conversely, JAK2 V617F, in particular, is one of the most common forms of clonal hematopoiesis of undetermined significance (CHIP).48-50 Thus, the presence or persistence of the mutation at a low level on or after effective therapy may simply reflect background CHIP. Also, the attainment of complete molecular response is itself a moving target. The lower limit of detection of most next-generation sequencing–based mutation panels in use is approximately 5%. More sensitive assays, eg, droplet digital or allele-specific oligonucleotide polymerase chain reaction, may be able to detect much lower VAFs.51 Long-term follow-up of COMFORT-1 demonstrated greater JAK2 V617F VAF reductions in ruxolitinib-treated MF patients with shorter disease duration, suggesting a benefit of earlier treatment,11 as alluded to above. Conversely, the odds of spleen response to ruxolitinib are lower and time to discontinuation shorter in the presence of additional, nondriver mutations.52 However, early intervention trials are challenging to undertake because of the long-term follow-up necessary to demonstrate favorable clinical outcomes. The ReTHINK trial,53 which randomly assigned patients with lower-risk MF without clinically relevant problems but high-risk molecular features (ie, mutations in ASXL1, EZH2, IDH1/2, or SRSF2) to low-dose ruxolitinib (10 mg twice a day) or placebo for the purpose of delaying clinical progression, could not be completed because of poor accrual, likely related in part to the placebo component and also concerns about long-term ruxolitinib adverse events in clinically well patients.

Anemia improvement and transfusion independence.

Anemia is common in MF, and both anemia and RBC transfusion requirement have long been recognized as risk factors for leukemic transformation and death,54-56 featured prominently in prognostic models57,58 and important for transplant decision making.59 The correlation between 24-week RBC-TI and OS in SIMPLIFY-130 lends additional credibility to this end point, linking anemia improvement to survival benefit. However, transfusion decisions in clinical practice are inherently subjective. Ruxolitinib-induced anemia does not carry the adverse prognosis of disease-induced anemia60 and is likely biologically distinct, perhaps explaining the higher RBC-TI rates in the add-on cohorts of the phase II trials of luspatercept61 and the bromodomain and extraterminal protein inhibitor, pelabresib (Table 1).62 Besides RBC-TI, amelioration of anemia in non–RBC-transfusion-dependent individuals may possibly represent biologic evidence of disease modification, as exemplified by pelabresib in the MANIFEST trial,62 where clinical improvement in anemia appeared to be associated with increased bone marrow erythropoiesis. In the context of JAK inhibitor therapy, anemia is predominantly viewed as an adverse effect of on-target JAK2 inhibition and resultant suppression of erythropoiesis.63 However, even with JAK inhibitors other than momelotinib, which improves anemia via ACVR1 inhibition and suppression of hepatic hepcidin production,64 some patients do achieve RBC-TI65; the mechanism behind this (eg, improved BM function, correction of hypersplenism, and others) remains unclear. Nevertheless, this is a small subset of patients, and combinations of traditional, more myelosuppressive JAK inhibitors with agents that improve anemia, eg, pelabresib or luspatercept, will likely continue to be attractive.

Cytokines and symptoms/SVR.

A wide array of proinflammatory cytokines are elevated in patients with MF, and some, like interleukins 8, 12, and 15 and the circulating interleukin-2 receptor, retain independent prognostic significance.66 In the phase I and II study of ruxolitinib, the drug broadly suppressed cytokines, which correlated with symptom benefit.13 Achievement of SVR35 at 24 weeks on both ruxolitinib67 and fedratinib68 has also been correlated with reductions in tumor necrosis factor alpha, and increases in leptin and adiponectin. Several novel agents, both alone69,70 and in combination with ruxolitinib,71-73 have demonstrated cytokine modulation. However, the lack of strong correlation coefficients and validation across multiple large studies precludes the use of one or more cytokines as end points. Specific cytokine levels and/or changes that may correlate with SVR, improvement in TSS and/or individual symptoms, OS, and other disease parameters, eg, cytopenias, need further study. TSS50 at 24 weeks has been a coprimary or key secondary end point of nearly all phase III trials of JAK inhibitors in MF, and is the sole primary end point of the pivotal MOMENTUM trial of momelotinib.74 Although of obvious importance to patients, there is likely a subjective component to this questionnaire-based end point, unlike SVR35. With a number of novel agents, for example, there is a noticeable discrepancy between rates of spleen and symptom response, eg, itacitinib, imetelstat, navtemadlin, parsaclisib, bomedemstat, and PRM-151 (Table 1).36,44,75-78 Given the limitations of cytokines as disease biomarkers, it is likely that patient-reported outcomes are here to stay for the foreseeable future as important clinical trial end points.

In conclusion, cure and/or improvement in survival remains the holy grail of MF treatment.79 The profusion of novel agents under investigation and the acceptance of new end points in trials, including OS, are welcome developments. However, the candidacy of biomarker-driven end points as surrogates for OS has yet to be convincingly demonstrated. By contrast, clinical end points, such as SVR35 and RBC-TI, have been shown to correlate with OS. Furthermore, end points tailored to JAK inhibitors may not lend themselves well to other novel agents—this is now being recognized in trial design. It is critical to harmonize inclusion criteria and response assessment across trials, particularly those being conducted in the ruxolitinib failure and suboptimal response settings, to allow for a fair appraisal of investigational agents. For now, it is important not to lose sight of hard clinical outcomes in our quest for disease modification.

Pankit Vachhani

Consulting or Advisory Role: Blueprint Medicines, Incyte, AbbVie, Jazz Pharmaceuticals, CTI BioPharma Corp, Novartis, Amgen, Pfizer, Genentech

Speakers' Bureau: Incyte

Research Funding: Seattle Genetics (Inst), Amgen (Inst), Astex Pharmaceuticals (Inst), Incyte (Inst), Blueprint Medicines (Inst), Kartos Therapeutics (Inst), Gilead/Forty Seven (Inst), Constellation Pharmaceuticals (Inst), AbbVie (Inst), CTI BioPharma Corp (Inst), Takeda (Inst)

Srdan Verstovsek

This author is a member of the Journal of Clinical Oncology Editorial Board. Journal policy recused the author from having any role in the peer review of this manuscript.

Consulting or Advisory Role: Constellation Pharmaceuticals, Sierra Oncology, Incyte, Novartis, Celgene

Research Funding: Incyte (Inst), Celgene (Inst), Protagonist Therapeutics (Inst), Sierra Oncology (Inst), PharmaEssentia (Inst), Telios (Inst), Constellation Pharmaceuticals (Inst), AbbVie (Inst), Geron (Inst), Galecto Biotech (Inst), Kartos Therapeutics (Inst)

Prithviraj Bose

Honoraria: Incyte, CTI BioPharma Corp, Blueprint Medicines, BMS, Novartis, Karyopharm Therapeutics, Pharmaessentia, AbbVie

Consulting or Advisory Role: Sierra Oncology

Research Funding: Celgene (Inst), CTI BioPharma Corp (Inst), Incyte (Inst), Blueprint Medicines (Inst), NS Pharma (Inst), Promedior (Inst), Constellation Pharmaceuticals (Inst), Kartos Therapeutics (Inst), Astellas Pharma (Inst), BMS (Inst), Pfizer (Inst), Cogent (Inst)

No other potential conflicts of interest were reported.

SUPPORT

MD Anderson Cancer Center Support Grant from the National Cancer Institute (National Institutes of Health), P30 CA016672, awarded to Dr Peter W.T. Pisters.

O'Neal Comprehensive Cancer Center at UAB Core Support Grant from the National Cancer Institute (National Institutes of Health), P30 CA013148, awarded to Dr Barry P Sleckman.

AUTHOR CONTRIBUTIONS

Manuscript writing: All authors

Final approval of manuscript: All authors

Accountable for all aspects of the work: All authors

AUTHORS' DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST

Disease Modification in Myelofibrosis: An Elusive Goal?

The following represents disclosure information provided by authors of this manuscript. All relationships are considered compensated unless otherwise noted. Relationships are self-held unless noted. I = Immediate Family Member, Inst = My Institution. Relationships may not relate to the subject matter of this manuscript. For more information about ASCO's conflict of interest policy, please refer to www.asco.org/rwc or ascopubs.org/jco/authors/author-center.

Open Payments is a public database containing information reported by companies about payments made to US-licensed physicians (Open Payments).

Pankit Vachhani

Consulting or Advisory Role: Blueprint Medicines, Incyte, AbbVie, Jazz Pharmaceuticals, CTI BioPharma Corp, Novartis, Amgen, Pfizer, Genentech

Speakers' Bureau: Incyte

Research Funding: Seattle Genetics (Inst), Amgen (Inst), Astex Pharmaceuticals (Inst), Incyte (Inst), Blueprint Medicines (Inst), Kartos Therapeutics (Inst), Gilead/Forty Seven (Inst), Constellation Pharmaceuticals (Inst), AbbVie (Inst), CTI BioPharma Corp (Inst), Takeda (Inst)

Srdan Verstovsek

This author is a member of the Journal of Clinical Oncology Editorial Board. Journal policy recused the author from having any role in the peer review of this manuscript.

Consulting or Advisory Role: Constellation Pharmaceuticals, Sierra Oncology, Incyte, Novartis, Celgene

Research Funding: Incyte (Inst), Celgene (Inst), Protagonist Therapeutics (Inst), Sierra Oncology (Inst), PharmaEssentia (Inst), Telios (Inst), Constellation Pharmaceuticals (Inst), AbbVie (Inst), Geron (Inst), Galecto Biotech (Inst), Kartos Therapeutics (Inst)

Prithviraj Bose

Honoraria: Incyte, CTI BioPharma Corp, Blueprint Medicines, BMS, Novartis, Karyopharm Therapeutics, Pharmaessentia, AbbVie

Consulting or Advisory Role: Sierra Oncology

Research Funding: Celgene (Inst), CTI BioPharma Corp (Inst), Incyte (Inst), Blueprint Medicines (Inst), NS Pharma (Inst), Promedior (Inst), Constellation Pharmaceuticals (Inst), Kartos Therapeutics (Inst), Astellas Pharma (Inst), BMS (Inst), Pfizer (Inst), Cogent (Inst)

No other potential conflicts of interest were reported.

REFERENCES

  • 1. 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. doi: 10.1182/blood-2011-02-335042. [DOI] [PubMed] [Google Scholar]
  • 2. 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:e123–33. doi: 10.1182/blood-2014-02-554634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Zeiser R, von Bubnoff N, Butler J, et al. Ruxolitinib for glucocorticoid-refractory acute graft-versus-host disease. N Engl J Med. 2020;382:1800–1810. doi: 10.1056/NEJMoa1917635. [DOI] [PubMed] [Google Scholar]
  • 4. Zeiser R, Polverelli N, Ram R, et al. Ruxolitinib for glucocorticoid-refractory chronic graft-versus-host disease. N Engl J Med. 2021;385:228–238. doi: 10.1056/NEJMoa2033122. [DOI] [PubMed] [Google Scholar]
  • 5. van Vollenhoven RF, Fleischmann R, Cohen S, et al. Tofacitinib or adalimumab versus placebo in rheumatoid arthritis. N Engl J Med. 2012;367:508–519. doi: 10.1056/NEJMoa1112072. [DOI] [PubMed] [Google Scholar]
  • 6. Taylor PC, Keystone EC, van der Heijde D, et al. Baricitinib versus placebo or adalimumab in rheumatoid arthritis. N Engl J Med. 2017;376:652–662. doi: 10.1056/NEJMoa1608345. [DOI] [PubMed] [Google Scholar]
  • 7. Gladman D, Rigby W, Azevedo VF, et al. Tofacitinib for psoriatic arthritis in patients with an inadequate response to TNF inhibitors. N Engl J Med. 2017;377:1525–1536. doi: 10.1056/NEJMoa1615977. [DOI] [PubMed] [Google Scholar]
  • 8. Mease P, Hall S, FitzGerald O, et al. Tofacitinib or adalimumab versus placebo for psoriatic arthritis. N Engl J Med. 2017;377:1537–1550. doi: 10.1056/NEJMoa1615975. [DOI] [PubMed] [Google Scholar]
  • 9. 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. doi: 10.1182/blood-2013-02-482232. [DOI] [PubMed] [Google Scholar]
  • 10. Verstovsek S, Gotlib J, Mesa RA, et al. Long-term survival in patients treated with ruxolitinib for myelofibrosis: COMFORT-I and -II pooled analyses. J Hematol Oncol. 2017;10:156. doi: 10.1186/s13045-017-0527-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Deininger M, Radich J, Burn TC, et al. The effect of long-term ruxolitinib treatment on JAK2p.V617F allele burden in patients with myelofibrosis. Blood. 2015;126:1551–1554. doi: 10.1182/blood-2015-03-635235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. 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. doi: 10.1038/leu.2016.148. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Verstovsek S, Kantarjian H, Mesa RA, et al. Safety and efficacy of INCB018424, a JAK1 and JAK2 inhibitor, in myelofibrosis. N Engl J Med. 2010;363:1117–1127. doi: 10.1056/NEJMoa1002028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Bose P, Verstovsek S. SOHO state of the art updates and next questions: Identifying and treating "progression" in myelofibrosis. Clin Lymphoma Myeloma Leuk. 2021;21:641–649. doi: 10.1016/j.clml.2021.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Bose P, Verstovsek S. JAK inhibition for the treatment of myelofibrosis: Limitations and future perspectives. Hemasphere. 2020;4:e424. doi: 10.1097/HS9.0000000000000424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Kuykendall A, Horvat NP, Pandey G, et al. Finding a jill for JAK: Assessing past, present and future JAK inhibitor combination approaches in myelofibrosis. Cancers (Basel) 2020;12:2278. doi: 10.3390/cancers12082278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Verstovsek S, Kantarjian HM, Estrov Z, et al. Long-term outcomes of 107 patients with myelofibrosis receiving JAK1/JAK2 inhibitor ruxolitinib: Survival advantage in comparison to matched historical controls. Blood. 2012;120:1202–1209. doi: 10.1182/blood-2012-02-414631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Passamonti F, Maffioli M, Cervantes F, et al. Impact of ruxolitinib on the natural history of primary myelofibrosis: A comparison of the DIPSS and the COMFORT-2 cohorts. Blood. 2014;123:1833–1835. doi: 10.1182/blood-2013-12-544411. [DOI] [PubMed] [Google Scholar]
  • 19. Guglielmelli P, Ghirardi A, Carobbio A, et al. Impact of ruxolitinib on survival of patients with myelofibrosis in real world—Update of ERNEST (European Registry for Myeloproliferative Neoplasms) study. Hemasphere. 2021;5 (suppl; abstr S158) [Google Scholar]
  • 20. Verstovsek S, Parasuraman S, Yu J, et al. Real-world survival of US patients with intermediate- to high-risk myelofibrosis: Impact of ruxolitinib approval. Ann Hematol. 2022;101:131–137. doi: 10.1007/s00277-021-04682-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Vannucchi AM, Kantarjian HM, Kiladjian JJ, et al. A pooled analysis of overall survival in COMFORT-I and COMFORT-II, 2 randomized phase 3 trials of ruxolitinib for the treatment of myelofibrosis. Haematologica. 2015;100:1139–1145. doi: 10.3324/haematol.2014.119545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Palandri F, Palumbo GA, Bonifacio M, et al. Durability of spleen response affects the outcome of ruxolitinib-treated patients with myelofibrosis: Results from a multicentre study on 284 patients. Leuk Res. 2018;74:86–88. doi: 10.1016/j.leukres.2018.10.001. [DOI] [PubMed] [Google Scholar]
  • 23. Barbui T, Tefferi A, Vannucchi AM, et al. Philadelphia chromosome-negative classical myeloproliferative neoplasms: Revised management recommendations from european LeukemiaNet. Leukemia. 2018;32:1057–1069. doi: 10.1038/s41375-018-0077-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Marchetti M, Barosi G, Cervantes F, et al. Which patients with myelofibrosis should receive ruxolitinib therapy? ELN-SIE evidence-based recommendations. Leukemia. 2016;31:882–888. doi: 10.1038/leu.2016.283. [DOI] [PubMed] [Google Scholar]
  • 25. Mesa R, Jamieson C, Bhatia R, et al. Myeloproliferative neoplasms, version 2.2017, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 2016;14:1572–1611. doi: 10.6004/jnccn.2016.0169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Pardanani A, Harrison C, Cortes JE, et al. Safety and efficacy of fedratinib in patients with primary or secondary myelofibrosis: A randomized clinical trial. JAMA Oncol. 2015;1:643–651. doi: 10.1001/jamaoncol.2015.1590. [DOI] [PubMed] [Google Scholar]
  • 27. Harrison CN, Kiladjian JJ, Verstovsek S, et al. Overall and progression-free survival in patients treated with fedratinib as first-line myelofibrosis (MF) therapy and after prior ruxolitinib (RUX): Results from the JAKARTA and JAKARTA2 trials. Hemasphere. 2021;5 supp; abstr S203. [Google Scholar]
  • 28. Mesa RA, Kiladjian JJ, Catalano JV, et al. SIMPLIFY-1: A phase III randomized trial of momelotinib versus ruxolitinib in janus kinase inhibitor-naive patients with myelofibrosis. J Clin Oncol. 2017;35:3844–3850. doi: 10.1200/JCO.2017.73.4418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Harrison CN, Vannucchi AM, Platzbecker U, et al. Momelotinib versus best available therapy in patients with myelofibrosis previously treated with ruxolitinib (SIMPLIFY 2): A randomised, open-label, phase 3 trial. Lancet Haematol. 2017;5:e73–e81. doi: 10.1016/S2352-3026(17)30237-5. [DOI] [PubMed] [Google Scholar]
  • 30. Mesa RA, Oh S, Gerds AT, et al. Transfusion independence is associated with improved overall survival in myelofibrosis patients receiving momelotinib. Hemasphere. 2021;5 suppl; abstr S202. [Google Scholar]
  • 31. Sureau L, Orvain C, Ianotto JC, et al. Efficacy and tolerability of janus kinase inhibitors in myelofibrosis: A systematic review and network meta-analysis. Blood Cancer J. 2021;11:135. doi: 10.1038/s41408-021-00526-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Mascarenhas J, Mehra M, He J, et al. Patient characteristics and outcomes after ruxolitinib discontinuation in patients with myelofibrosis. J Med Econ. 2020;23:721–727. doi: 10.1080/13696998.2020.1741381. [DOI] [PubMed] [Google Scholar]
  • 33. Newberry KJ, Patel K, Masarova L, et al. Clonal evolution and outcomes in myelofibrosis after ruxolitinib discontinuation. Blood. 2017;130:1125–1131. doi: 10.1182/blood-2017-05-783225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Palandri F, Breccia M, Bonifacio M, et al. Life after ruxolitinib: Reasons for discontinuation, impact of disease phase, and outcomes in 218 patients with myelofibrosis. Cancer. 2019;126:1243–1252. doi: 10.1002/cncr.32664. [DOI] [PubMed] [Google Scholar]
  • 35. Kuykendall AT, Shah S, Talati C, et al. Between a rux and a hard place: Evaluating salvage treatment and outcomes in myelofibrosis after ruxolitinib discontinuation. Ann Hematol. 2018;97:435–441. doi: 10.1007/s00277-017-3194-4. [DOI] [PubMed] [Google Scholar]
  • 36. Mascarenhas J, Komrokji RS, Palandri F, et al. Randomized, single-blind, multicenter phase II study of two doses of imetelstat in relapsed or refractory myelofibrosis. J Clin Oncol. 2021;39:2881–2892. doi: 10.1200/JCO.20.02864. [DOI] [PubMed] [Google Scholar]
  • 37. Kuykendall AT, Sun L, Mascarenhas J, et al. Favorable overall survival with imetelstat in relapsed/refractory myelofibrosis patients compared with real-world data. Ann Hematol. 2022;101:139–146. doi: 10.1007/s00277-021-04683-w. [DOI] [PubMed] [Google Scholar]
  • 38. Verstovsek S, Manshouri T, Pilling D, et al. Role of neoplastic monocyte-derived fibrocytes in primary myelofibrosis. J Exp Med. 2016;213:1723–1740. doi: 10.1084/jem.20160283. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Jamieson C, Hasserjian R, Gotlib J, et al. Effect of treatment with a JAK2-selective inhibitor, fedratinib, on bone marrow fibrosis in patients with myelofibrosis. J Transl Med. 2015;13:294. doi: 10.1186/s12967-015-0644-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Kvasnicka HM, Thiele J, Bueso-Ramos CE, et al. Long-term effects of ruxolitinib versus best available therapy on bone marrow fibrosis in patients with myelofibrosis. J Hematol Oncol. 2018;11:42. doi: 10.1186/s13045-018-0585-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Kroger N, Zabelina T, Alchalby H, et al. Dynamic of bone marrow fibrosis regression predicts survival after allogeneic stem cell transplantation for myelofibrosis. Biol Blood Marrow Transpl. 2014;20:812–815. doi: 10.1016/j.bbmt.2014.02.019. [DOI] [PubMed] [Google Scholar]
  • 42. Kvasnicka HM, Beham-Schmid C, Bob R, et al. Problems and pitfalls in grading of bone marrow fibrosis, collagen deposition and osteosclerosis - a consensus-based study. Histopathology. 2016;68:905–915. doi: 10.1111/his.12871. [DOI] [PubMed] [Google Scholar]
  • 43. Verstovsek S, Hasserjian RP, Pozdnyakova O, et al. PRM-151 in myelofibrosis: Efficacy and safety in an open label extension study. Blood. 2018;132 suppl; abstr 686. [Google Scholar]
  • 44. Verstovsek S, Talpaz M, Wadleigh M, et al. A randomized, double blind phase 2 study of 3 different doses of PRM-151 in patients with myelofibrosis who were previously treated with or ineligible for ruxolitinib. Hemasphere. 2019;3 suppl; abstr S828. [Google Scholar]
  • 45. Vannucchi AM, Lasho TL, Guglielmelli P, et al. Mutations and prognosis in primary myelofibrosis. Leukemia. 2013;27:1861–1869. doi: 10.1038/leu.2013.119. [DOI] [PubMed] [Google Scholar]
  • 46. Tefferi A, Lasho TL, Finke CM, et al. Targeted deep sequencing in primary myelofibrosis. Blood Adv. 2016;1:105–111. doi: 10.1182/bloodadvances.2016000208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Silver RT, Barel AC, Lascu E, et al. The effect of initial molecular profile on response to recombinant interferon-alpha (rIFNalpha) treatment in early myelofibrosis. Cancer. 2017;123:2680–2687. doi: 10.1002/cncr.30679. [DOI] [PubMed] [Google Scholar]
  • 48. Nielsen C, Birgens HS, Nordestgaard BG, et al. The JAK2 V617F somatic mutation, mortality and cancer risk in the general population. Haematologica. 2011;96:450–453. doi: 10.3324/haematol.2010.033191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Nielsen C, Birgens HS, Nordestgaard BG, et al. Diagnostic value of JAK2 V617F somatic mutation for myeloproliferative cancer in 49 488 individuals from the general population. Br J Haematol. 2013;160:70–79. doi: 10.1111/bjh.12099. [DOI] [PubMed] [Google Scholar]
  • 50. Nielsen C, Bojesen SE, Nordestgaard BG, et al. JAK2V617F somatic mutation in the general population: Myeloproliferative neoplasm development and progression rate. Haematologica. 2014;99:1448–1455. doi: 10.3324/haematol.2014.107631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. George TI, Hoehn G, Lin H, et al. Increased detection of KIT D816V mutation in peripheral blood samples from patients with indolent systemic mastocytosis (ISM) in the phase 2 pioneer study using a high sensitivity droplet digital (dd) PCR assay compared with next generation sequencing (NGS) Blood. 2020;136 suppl; abstr 3004. [Google Scholar]
  • 52. Patel KP, Newberry KJ, Luthra R, et al. Correlation of mutation profile and response in patients with myelofibrosis treated with ruxolitinib. Blood. 2015;126:790–797. doi: 10.1182/blood-2015-03-633404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Passamonti F, Kiladjian J, Vannucchi AM, et al. ReTHINK: A randomized, double-blind, placebo-controlled, multicenter, phase 3 study of ruxolitinib in early myelofibrosis patients. J Clin Oncol. 2016;34 suppl; abstr TPS7080. [Google Scholar]
  • 54. Tefferi A, Mesa RA, Pardanani A, et al. Red blood cell transfusion need at diagnosis adversely affects survival in primary myelofibrosis-increased serum ferritin or transfusion load does not. Am J Hematol. 2009;84:265–267. doi: 10.1002/ajh.21391. [DOI] [PubMed] [Google Scholar]
  • 55. Elena C, Passamonti F, Rumi E, et al. Red blood cell transfusion-dependency implies a poor survival in primary myelofibrosis irrespective of IPSS and DIPSS. Haematologica. 2011;96:167–170. doi: 10.3324/haematol.2010.031831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Nicolosi M, Mudireddy M, Lasho TL, et al. Sex and degree of severity influence the prognostic impact of anemia in primary myelofibrosis: Analysis based on 1109 consecutive patients. Leukemia. 2018;32:1254–1258. doi: 10.1038/s41375-018-0028-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Bose P, Verstovsek S. Mutational profiling in myelofibrosis: Implications for management. Int J Hematol. 2019;111:192–199. doi: 10.1007/s12185-019-02758-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Bose P, Verstovsek S. The evolution and clinical relevance of prognostic classification systems in myelofibrosis. Cancer. 2016;122:681–692. doi: 10.1002/cncr.29842. [DOI] [PubMed] [Google Scholar]
  • 59. 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. doi: 10.1038/leu.2015.233. [DOI] [PubMed] [Google Scholar]
  • 60. Gupta V, Harrison C, Hexner EO, et al. The impact of anemia on overall survival in patients with myelofibrosis treated with ruxolitinib in the COMFORT studies. Haematologica. 2016;101:e482–e484. doi: 10.3324/haematol.2016.151449. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Gerds AT, Vannucchi AM, Passamonti F, et al. Duration of response to luspatercept in patients (pts) requiring red blood cell (RBC) transfusions with myelofibrosis (MF)-updated data from the phase 2 ACE-536-MF-001 study. Blood. 2020;136 suppl; abstr 2992. [Google Scholar]
  • 62. Verstovsek S, Kremyanskaya M, Mascarenhas JO, et al. Pelabresib (CPI-0610) improved anemia associated with myelofibrosis: Interim results from manifest phase 2 study. Hemasphere. 2021;5 suppl; abstr EP1077. [Google Scholar]
  • 63. Parganas E, Wang D, Stravopodis D, et al. Jak2 is essential for signaling through a variety of cytokine receptors. Cell. 1998;93:385–395. doi: 10.1016/s0092-8674(00)81167-8. [DOI] [PubMed] [Google Scholar]
  • 64. Asshoff M, Petzer V, Warr MR, et al. Momelotinib inhibits ACVR1/ALK2, decreases hepcidin production and ameliorates anemia of chronic disease in rodents. Blood. 2017;129:1823–1830. doi: 10.1182/blood-2016-09-740092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Mesa RA, Vannucchi AM, Mead A, et al. Pacritinib versus best available therapy for the treatment of myelofibrosis irrespective of baseline cytopenias (PERSIST-1): An international, randomised, phase 3 trial. Lancet Haematol. 2017;4:e225–e236. doi: 10.1016/S2352-3026(17)30027-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Tefferi A, Vaidya R, Caramazza D, et al. Circulating interleukin (IL)-8, IL-2R, IL-12, and IL-15 levels are independently prognostic in primary myelofibrosis: A comprehensive cytokine profiling study. J Clin Oncol. 2011;29:1356–1363. doi: 10.1200/JCO.2010.32.9490. [DOI] [PubMed] [Google Scholar]
  • 67. Harrison CN, Kiladjian J, Gisslinger H, et al. Association of cytokine levels and reductions in spleen size in COMFORT-II, a phase III study comparing ruxolitinib to best available therapy (BAT) J Clin Oncol. 2012;30 suppl; abstr 6625. [Google Scholar]
  • 68. Pardanani A, Tefferi A, Jamieson C, et al. A phase 2 randomized dose-ranging study of the JAK2-selective inhibitor fedratinib (SAR302503) in patients with myelofibrosis. Blood Cancer J. 2015;5:e335. doi: 10.1038/bcj.2015.63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Kremyanskaya M, Mascarenhas J, Palandri F, et al. Pelabresib (CPI-0610) monotherapy in patients with myelofibrosis – Update of clinical and translational data from the ongoing Manifest trial. Blood. 2021;138 suppl; abstr 141. [Google Scholar]
  • 70. Vachhani P, Lange A, Delgado RG, et al. Potential disease-modifying activity of navtemadlin (KRT-232), a first-in-class MDM2 inhibitor, correlates with clinical benefits in Relapsed/Refractory myelofibrosis (MF) Blood. 2021;138 suppl; abstr 3581. [Google Scholar]
  • 71. Verstovsek S, Salama ME, Mascarenhas J, et al. Disease-modifying potential of BET inhibitor pelabresib (CPI-0610) as demonstrated by improvements in bone marrow function and clinical activity in patients with myelofibrosis – Preliminary data. Blood. 2021;138 suppl; abstr 2568. [Google Scholar]
  • 72. Pemmaraju N, Garcia JS, Potluri J, et al. The addition of navitoclax to ruxolitinib demonstrates efficacy within different high-risk populations in patients with Relapsed/Refractory myelofibrosis. Blood. 2020;136 suppl; abstr 52. [Google Scholar]
  • 73. Kremyanskaya M, Mascarenhas JO, Patriarca A, et al. Clinical benefit of pelabresib (CPI-0610) in combination with ruxolitinib in JAK inhibitor treatment naïve myelofibrosis patients: Interim efficacy subgroup analysis from arm 3 of manifest PH2 study. Hemasphere. 2021;5 suppl; abstr EP1085. [Google Scholar]
  • 74. Verstovsek S, Chen CC, Egyed M, et al. MOMENTUM: Momelotinib vs danazol in patients with myelofibrosis previously treated with JAKi who are symptomatic and anemic. Future Oncol. 2021;17:1449–1458. doi: 10.2217/fon-2020-1048. [DOI] [PubMed] [Google Scholar]
  • 75. Mascarenhas JO, Talpaz M, Gupta V, et al. Primary analysis of a phase II open-label trial of INCB039110, a selective JAK1 inhibitor, in patients with myelofibrosis. Haematologica. 2016;102:327–335. doi: 10.3324/haematol.2016.151126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76. Al-Ali H, Delgado RG, Lange A, et al. KRT-232, A first-in-class, murine double minute 2 inhibitor, for myelofibrosis relapsed or refractory to janus-associated kinase inhibitor treatment. Hemasphere. 2020;4 suppl; abstr S215. [Google Scholar]
  • 77. Yacoub A, Borate U, Rampal RK, et al. Add-on parsaclisib (A PI3K-DELTA INHIBITOR) in patients with myelofibrosis and suboptimal response to ruxolitinib: Interim analysis from a phase 2 study. Hemasphere. 2021;5 suppl; abstr EP1075. [Google Scholar]
  • 78. Gill H, Yacoub A, Pettit KM, et al. A phase 2 study of the LSD1 inhibitor Img-7289 (bomedemstat) for the treatment of advanced myelofibrosis. Blood. 2021;138 suppl; abstr 139. [Google Scholar]
  • 79. Verstovsek S. Changing myelofibrosis's natural course at last. Blood. 2014;123:1776–1777. doi: 10.1182/blood-2014-01-551267. [DOI] [PubMed] [Google Scholar]
  • 80. 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. doi: 10.1056/NEJMoa1110557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81. 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. doi: 10.1056/NEJMoa1110556. [DOI] [PubMed] [Google Scholar]
  • 82. Harrison CN, Schaap N, Vannucchi AM, et al. Fedratinib in patients with myelofibrosis previously treated with ruxolitinib: An updated analysis of the JAKARTA2 study using stringent criteria for ruxolitinib failure. Am J Hematol. 2020;95:594–603. doi: 10.1002/ajh.25777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Mascarenhas J, Hoffman R, Talpaz M, et al. Pacritinib vs best available therapy, including ruxolitinib, in patients with myelofibrosis: A randomized clinical trial. JAMA Oncol. 2018;4:652–659. doi: 10.1001/jamaoncol.2017.5818. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Gerds AT, Savona MR, Scott BL, et al. Determining the recommended dose of pacritinib: Results from the PAC203 dose-finding trial in advanced myelofibrosis. Blood Adv. 2020;4:5825–5835. doi: 10.1182/bloodadvances.2020003314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. Masarova L, Verstovsek S, Hidalgo-Lopez JE, et al. A phase 2 study of ruxolitinib in combination with azacitidine in patients with myelofibrosis. Blood. 2018;132:1664–1674. doi: 10.1182/blood-2018-04-846626. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Journal of Clinical Oncology are provided here courtesy of American Society of Clinical Oncology

RESOURCES