0
Views
0
CrossRef citations to date
0
Altmetric
Review Article

The evolving landscape of polycythemia vera therapies

, , &
Received 11 May 2024, Accepted 30 Jul 2024, Accepted author version posted online: 06 Aug 2024
Accepted author version

REFERENCES

  • Baxter EJ, Scott LM, Campbell PJ, et al. Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. The Lancet 2005 Mar 19-25;365(9464):1054–1061. 10.1016/S0140-6736(05)71142-9
  • James C, Ugo V, Le Couédic JP, et al. A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. Nature 2005 Apr 28;434(7037):1144–1148.10.1038/nature03546
  • Kralovics R, Passamonti F, Buser AS, et al. A gain-of-function mutation of JAK2 in myeloproliferative disorders. N Engl J Med 2005 Apr 28;352(17):1779–1790.10.1056/NEJMoa051113
  • Scott LM, Tong W, Levine RL, et al. JAK2 exon 12 mutations in polycythemia vera and idiopathic erythrocytosis. N Engl J Med 2007 Feb 1;356(5):459–468.10.1056/NEJMoa065202
  • Tefferi A, Lavu S, Mudireddy M, et al. JAK2 exon 12 mutated polycythemia vera: Mayo-Careggi MPN Alliance study of 33 consecutive cases and comparison with JAK2V617F mutated disease. Am J Hematol 2018 Aug;93(4):E93–e96.10.1002/ajh.25017
  • Tondeur S, Paul F, Riou J, et al. Long-term follow-up of JAK2 exon 12 polycythemia vera: a French Intergroup of Myeloproliferative Neoplasms (FIM) study. Leukemia 2021 Mar;35(3):871–875.10.1038/s41375-020-0991-x
  • Arber DA, Orazi A, Hasserjian RP, et al. International Consensus Classification of Myeloid Neoplasms and Acute Leukemias: integrating morphologic, clinical, and genomic data. Blood 2022 Sep 15;140(11):1200–1228.10.1182/blood.2022015850
  • Geyer H, Scherber R, Kosiorek H, et al. Symptomatic Profiles of Patients With Polycythemia Vera: Implications of Inadequately Controlled Disease. J Clin Oncol 2016 Jan 10;34(2):151–159. 10.1200/JCO.2015.62.9337
  • Grunwald MR, Burke JM, Kuter DJ, et al. Symptom Burden and Blood Counts in Patients With Polycythemia Vera in the United States: An Analysis From the REVEAL Study. Clin Lymphoma Myeloma Leuk 2019 Sep;19(9):579–84.e1. 10.1016/j.clml.2019.06.001
  • Marchetti M, Vannucchi AM, Griesshammer M, et al. Appropriate management of polycythaemia vera with cytoreductive drug therapy: European LeukemiaNet 2021 recommendations. Lancet Haematol 2022 Apr;9(4):e301–e11.
  • Tefferi A, Rumi E, Finazzi G, et al. Survival and prognosis among 1545 patients with contemporary polycythemia vera: an international study. Leukemia 2013 Sep;27(9):1874–1881.10.1038/leu.2013.163
  • Szuber N, Mudireddy M, Nicolosi M, et al. 3023 Mayo Clinic Patients With Myeloproliferative Neoplasms: Risk-Stratified Comparison of Survival and Outcomes Data Among Disease Subgroups. Mayo Clin Proc 2019 Apr;94(4):599–610. 10.1016/j.mayocp.2018.08.022
  • Griesshammer M, Kiladjian JJ, Besses C Thromboembolic events in polycythemia vera. Ann Hematol 2019 May;98(5):1071–1082.10.1007/s00277-019-03625-x
  • Hultcrantz M, Björkholm M, Dickman PW, et al. Risk for Arterial and Venous Thrombosis in Patients With Myeloproliferative Neoplasms: A Population-Based Cohort Study. Ann Intern Med 2018 Mar 6;168(5):317–325.10.7326/M17-0028
  • Mazza GL, Mead-Harvey C, Mascarenhas J, et al. Symptom burden and quality of life in patients with high-risk essential thrombocythaemia and polycythaemia vera receiving hydroxyurea or pegylated interferon alfa-2a: a post-hoc analysis of the MPN-RC 111 and 112 trials. Lancet Haematol 2022 Jan;9(1):e38–e48. 10.1016/S2352-3026(21)00343-4
  • Mesa RA, Miller CB, Thyne M, et al. Differences in treatment goals and perception of symptom burden between patients with myeloproliferative neoplasms (MPNs) and hematologists/oncologists in the United States: Findings from the MPN Landmark survey. Cancer 2017 Feb 1;123(3):449–458.10.1002/cncr.30325
  • Barbui T, Thiele J, Gisslinger H, et al. The 2016 WHO classification and diagnostic criteria for myeloproliferative neoplasms: document summary and in-depth discussion. Blood Cancer J 2018 Feb 9;8(2):15. 10.1038/s41408-018-0054-y
  • Barbui T, Thiele J, Vannucchi AM, Tefferi A. Rationale for revision and proposed changes of the WHO diagnostic criteria for polycythemia vera, essential thrombocythemia and primary myelofibrosis. Blood Cancer Journal 20152015/08 /01;5(8):e337–e37.
  • Khoury JD, Solary E, Abla O, et al. The 5th edition of the World Health Organization Classification of Haematolymphoid Tumours: Myeloid and Histiocytic/Dendritic Neoplasms. Leukemia 2022 Jul;36(7):1703–1719.10.1038/s41375-022-01613-1
  • Marchioli R, Finazzi G, Landolfi R, et al. Vascular and neoplastic risk in a large cohort of patients with polycythemia vera. J Clin Oncol 2005 Apr 1;23(10):2224–2232. 10.1200/JCO.2005.07.062
  • Barbui T, Tefferi A, Vannucchi AM, et al. Philadelphia chromosome-negative classical myeloproliferative neoplasms: revised management recommendations from European LeukemiaNet. Leukemia 2018 May;32(5):1057–1069.10.1038/s41375-018-0077-1
  • Barbui T, Passamonti F, Accorsi P, et al. Evidence- and consensus-based recommendations for phlebotomy in polycythemia vera. Leukemia 2018 Sep;32(9):2077–2081.10.1038/s41375-018-0199-5
  • Tefferi A, Guglielmelli P, Lasho TL, et al. Mutation-enhanced international prognostic systems for essential thrombocythaemia and polycythaemia vera. Br J Haematol 2020 Apr;189(2):291–302.10.1111/bjh.16380
  • Ronner L, Podoltsev N, Gotlib J, et al. Persistent leukocytosis in polycythemia vera is associated with disease evolution but not thrombosis. Blood 2020 May 7;135(19):1696–1703.10.1182/blood.2019003347
  • Landolfi R, Di Gennaro L, Barbui T, et al. Leukocytosis as a major thrombotic risk factor in patients with polycythemia vera. Blood 2007 Mar 15;109(6):2446–2452.10.1182/blood-2006-08-042515
  • Krecak I, Lekovic D, Arsenovic I, et al. The triple A model (age, absolute neutrophil count, absolute lymphocyte count-AAA) predicts survival and thrombosis in polycythemia vera. Am J Hematol 2024 May;99(5):989–992.10.1002/ajh.27261
  • Zhang Y, Zhou Y, Wang Y, et al. Thrombosis among 1537 patients with JAK2(V617F) -mutated myeloproliferative neoplasms: Risk factors and development of a predictive model. Cancer Med 2020 Mar;9(6):2096–105.
  • Guglielmelli P, Loscocco GG, Mannarelli C, et al. JAK2V617F variant allele frequency >50% identifies patients with polycythemia vera at high risk for venous thrombosis. Blood Cancer J 2021 Dec 11;11(12):199.
  • Silver RT, Vandris K, Wang YL, et al. JAK2V617F allele burden in polycythemia vera correlates with grade of myelofibrosis, but is not substantially affected by therapy. Leukemia Research 20112011/02 /01/;35(2):177–82.
  • Moliterno AR, Kaizer H, Reeves BN JAK2 V617F allele burden in polycythemia vera: burden of proof. Blood 2023 Apr 20;141(16):1934–1942.10.1182/blood.2022017697
  • Tang G, Hidalgo Lopez JE, Wang SA, et al. Characteristics and clinical significance of cytogenetic abnormalities in polycythemia vera. Haematologica 2017 Sep;102(9):1511–1518.10.3324/haematol.2017.165795
  • Barraco D, Cerquozzi S, Hanson CA, et al. Cytogenetic findings in WHO-defined polycythaemia vera and their prognostic relevance. Br J Haematol 2018 Aug;182(3):437–440.10.1111/bjh.14798
  • National Comprehensive Cancer Network. Myeloproliferative Neoplasms (version 1.2024). 2024 [cited March 30, 2024]; Available from: https://www.nccn.org/professionals/physician_gls/pdf/mpn.pdf
  • Cerquozzi S, Barraco D, Lasho T, et al. Risk factors for arterial versus venous thrombosis in polycythemia vera: a single center experience in 587 patients. Blood Cancer J 2017 Dec 27;7(12):662. 10.1038/s41408-017-0035-6
  • Barbui T, Vannucchi AM, Carobbio A, et al. The effect of arterial hypertension on thrombosis in low-risk polycythemia vera. Am J Hematol 2017 Jan;92(1):E5–e6.10.1002/ajh.24583
  • Lucijanic M, Lekovic D, Bogdanovic A, et al. Being John Plasma Volumovich, pecularities of plasma volume estimation in patients with polycythemia vera. Thromb Res 2024 Jul;239:109039. 10.1016/j.thromres.2024.109039
  • Marchioli R, Finazzi G, Specchia G, et al. Cardiovascular events and intensity of treatment in polycythemia vera. N Engl J Med 2013 Jan 3;368(1):22–33.10.1056/NEJMoa1208500
  • Landolfi R, Marchioli R, Kutti J, et al. Efficacy and safety of low-dose aspirin in polycythemia vera. N Engl J Med 2004 Jan 8;350(2):114–124.10.1056/NEJMoa035572
  • Rocca B, Tosetto A, Betti S, et al. A randomized double-blind trial of 3 aspirin regimens to optimize antiplatelet therapy in essential thrombocythemia. Blood. 2020;136(2):171–182. doi: 10.1182/blood.2019004596
  • Benevolo G, Marchetti M, Melchio R, et al. Diagnosis and Management of Cardiovascular Risk in Patients with Polycythemia Vera. Vasc Health Risk Manag 2023;19:765–778. 10.2147/VHRM.S429995
  • Kimpton M, Cerquozzi S, Toupin D, et al. Assessing the Feasibility of Thromboprophylaxis with Apixaban in JAK2-Positive Myeloproliferative Neoplasm Patients: The Airport-MPN Trial. Blood. 2023;142(Supplement 1):1823–23. doi: 10.1182/blood-2023-175012
  • Berk PD, Goldberg JD, Silverstein MN, et al. Increased incidence of acute leukemia in polycythemia vera associated with chlorambucil therapy. N Engl J Med 1981 Feb 19;304(8):441–447.10.1056/NEJM198102193040801
  • Harrison CN, Koschmieder S, Foltz L, et al. The impact of myeloproliferative neoplasms (MPNs) on patient quality of life and productivity: results from the international MPN Landmark survey. Ann Hematol 2017 Oct;96(10):1653–1665.10.1007/s00277-017-3082-y
  • Mesa R, Miller CB, Thyne M, et al. Myeloproliferative neoplasms (MPNs) have a significant impact on patients’ overall health and productivity: the MPN Landmark survey. BMC Cancer 2016 Feb 27;16:167. 1 10.1186/s12885-016-2208-2
  • Heidel FH, Al-Ali HK, Hirt C, et al. Questions arising on phlebotomy in polycythemia vera: prophylactic measures to reduce thromboembolic events require patient-focused decisions. Leukemia 2018 Sep;32(9):2085–2087.10.1038/s41375-018-0214-x
  • Tibes R, Mesa RA Emerging drugs for polycythemia vera. Expert Opin Emerg Drugs 2013 Sep;18(3):393–404.10.1517/14728214.2013.832754
  • Miles LA, Bowman RL, Merlinsky TR, et al. Single-cell mutation analysis of clonal evolution in myeloid malignancies. Nature 2020 Nov;587(7834):477–482.10.1038/s41586-020-2864-x
  • Tefferi A, Barbui T Polycythemia vera: 2024 update on diagnosis, risk-stratification, and management. Am J Hematol 2023 Sep;98(9):1465–1487.10.1002/ajh.27002
  • Evstatiev R, Bukaty A, Jimenez K, et al. Iron deficiency alters megakaryopoiesis and platelet phenotype independent of thrombopoietin. Am J Hematol 2014 May;89(5):524–529.10.1002/ajh.23682
  • Barbui T, Vannucchi AM, De Stefano V, et al. Ropeginterferon alfa-2b versus phlebotomy in low-risk patients with polycythaemia vera (Low-PV study): a multicentre, randomised phase 2 trial. Lancet Haematol 2021 Mar;8(3):e175–e84.
  • Cabibbo S, Manenti GO, Antolino A, et al. Evaluation of erythrocytapheresis compared to phlebotomy in polycythaemia vera patients. Hematol Transfus Cell Ther 2023 Aug 24. 10.1016/j.htct.2023.07.003
  • Choe WH, Park BG, Lee KH, et al. Automated double red-cell phlebotomy for the treatment of erythrocytosis. J Clin Apher 2012 Nov;27(5):255–259. 10.1002/jca.21241
  • Hensley B, Geyer H, Mesa R Polycythemia vera: current pharmacotherapy and future directions. Expert Opin Pharmacother 2013 Apr;14(5):609–617. 10.1517/14656566.2013.779671
  • Spivak JL Polycythemia vera: myths, mechanisms, and management. Blood 2002 Dec 15;100(13):4272–4290. Blood 10.1182/blood-2001-12-0349
  • Verstovsek S, Pemmaraju N, Reaven NL, et al. Real-world treatments and thrombotic events in polycythemia vera patients in the USA. Ann Hematol 2023 Mar;102(3):571–581.10.1007/s00277-023-05089-6
  • Bewersdorf JP, How J, Masarova L, et al. Moving toward disease modification in polycythemia vera. Blood 2023 Nov 30;142(22):1859–1870.10.1182/blood.2023021503
  • Tremblay D. Cytoreduction for ET and PV: who, what, when, and how? Hematology Am Soc Hematol Educ Program 2023 Dec 8;2023(1):660–666. 2023 10.1182/hematology.2023000451
  • Ferrari A, Carobbio A, Masciulli A, et al. Clinical outcomes under hydroxyurea treatment in polycythemia vera: a systematic review and meta-analysis. Haematologica 2019 Dec;104(12):2391–2399.10.3324/haematol.2019.221234
  • Barbui T, Vannucchi AM, Finazzi G, et al. A reappraisal of the benefit-risk profile of hydroxyurea in polycythemia vera: A propensity-matched study. Am J Hematol 2017 Nov;92(11):1131–1136.10.1002/ajh.24851
  • Knudsen TA, Hansen DL, Ocias LF, et al. Final Analysis of the Daliah Trial: A Randomized Phase III Trial of Interferon-α Versus Hydroxyurea in Patients with MPN. Blood 20232023/11 /02/;142:746.
  • Mascarenhas J, Kosiorek HE, Prchal JT, et al. A randomized phase 3 trial of interferon-α vs hydroxyurea in polycythemia vera and essential thrombocythemia. Blood 2022 May 12;139(19):2931–2941.10.1182/blood.2021012743
  • Gisslinger H, Klade C, Georgiev P, et al. Ropeginterferon alfa-2b versus standard therapy for polycythaemia vera (PROUD-PV and CONTINUATION-PV): a randomised, non-inferiority, phase 3 trial and its extension study. Lancet Haematol 2020 Mar;7(3):e196–e208. 10.1016/S2352-3026(19)30236-4
  • Podoltsev NA, Zhu M, Zeidan AM, et al. The impact of phlebotomy and hydroxyurea on survival and risk of thrombosis among older patients with polycythemia vera. Blood Adv 2018 Oct 23;2(20):2681–2690. 10.1182/bloodadvances.2018021436
  • Barbui T, De Stefano V, Ghirardi A, et al. Different effect of hydroxyurea and phlebotomy on prevention of arterial and venous thrombosis in Polycythemia Vera. Blood Cancer J 2018 Nov 26;8(12):124. 10.1038/s41408-018-0161-9
  • Mesa RA, Miller CB, Mascarenhas JO, et al. Hydroxyurea Treatment History and Quality of Life in Patients with Polycythemia Vera: Results from the MPN Landmark Survey in the United States. Blood. 2015;126(23):4077–77. doi: 10.1182/blood.V126.23.4077.4077
  • Antonioli E, Carobbio A, Pieri L, et al. Hydroxyurea does not appreciably reduce JAK2 V617F allele burden in patients with polycythemia vera or essential thrombocythemia. Haematologica 2010 Aug;95(8):1435–1438.10.3324/haematol.2009.021444
  • Gangat N, Tefferi A Myeloproliferative neoplasms and pregnancy: Overview and practice recommendations. Am J Hematol 2021 Mar 1;96(3):354–366.10.1002/ajh.26067
  • Alvarez-Larrán A, Garrote M, Ferrer-Marín F, et al. Real-world analysis of main clinical outcomes in patients with polycythemia vera treated with ruxolitinib or best available therapy after developing resistance/intolerance to hydroxyurea. Cancer 2022 Jul 1;128(13):2441–2448.10.1002/cncr.34195
  • Sharon R, Tatarsky I, Ben-Arieh Y. Treatment of polycythemia vera with hydroxyurea. Cancer. 1986;57(4):718–720. doi: 10.1002/1097-0142(19860215)57:4<718:AID-CNCR2820570406>3.0.CO;2-P
  • Wang R, Shallis RM, Stempel JM, et al. Second malignancies among older patients with classical myeloproliferative neoplasms treated with hydroxyurea. Blood Adv 2023 Mar 14;7(5):734–743. 10.1182/bloodadvances.2022008259
  • Stegelmann F, Wille K, Busen H, et al. Significant association of cutaneous adverse events with hydroxyurea: results from a prospective non-interventional study in BCR-ABL1-negative myeloproliferative neoplasms (MPN) - on behalf of the German Study Group-MPN. Leukemia 2021 Feb;35(2):628–631.10.1038/s41375-020-0945-3
  • Lu M, Zhang W, Li Y, et al. Interferon-alpha targets JAK2V617F-positive hematopoietic progenitor cells and acts through the p38 MAPK pathway. Exp Hematol 2010 Jun;38(6):472–80.
  • Mullally A, Bruedigam C, Poveromo L, et al. Depletion of Jak2V617F myeloproliferative neoplasm-propagating stem cells by interferon-α in a murine model of polycythemia vera. Blood 2013 May 2;121(18):3692–3702.10.1182/blood-2012-05-432989
  • Chen X, Regn S, Raffegerst S, et al. Interferon alpha in combination with GM-CSF induces the differentiation of leukaemic antigen-presenting cells that have the capacity to stimulate a specific anti-leukaemic cytotoxic T-cell response from patients with chronic myeloid leukaemia. Br J Haematol 2000 Nov;111(2):596–607.10.1111/j.1365-2141.2000.02361.x
  • Riley CH, Brimnes MK, Hansen M, et al. Interferon-α induces marked alterations in circulating regulatory T cells, NK cell subsets, and dendritic cells in patients with JAK2V617F-positive essential thrombocythemia and polycythemia vera. Eur J Haematol 2016 Jul;97(1):83–92. 10.1111/ejh.12687
  • Skov V, Riley CH, Thomassen M, et al. The impact of interferon-alpha2 on HLA genes in patients with polycythemia vera and related neoplasms. Leuk Lymphoma 2017 Aug;58(8):1914–1921.10.1080/10428194.2016.1262032
  • PharmaEssentia USA Corporation. BESREMi (ropeginterferon alfa-2b-njft) injection fsuIUSA. Burlington, MA: PharmaEssentia. 2021.
  • Vachhani P, Mascarenhas J, Bose P, et al. Interferons in the treatment of myeloproliferative neoplasms. Ther Adv Hematol 2024;15:20406207241229588. 10.1177/20406207241229588
  • Gisslinger H, Zagrijtschuk O, Buxhofer-Ausch V, et al. Ropeginterferon alfa-2b, a novel IFNα-2b, induces high response rates with low toxicity in patients with polycythemia vera. Blood 2015 Oct 8;126(15):1762–1769.10.1182/blood-2015-04-637280
  • 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 Oct 15;112(8):3065–3072.10.1182/blood-2008-03-143537
  • Quintás-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 Nov 10;27(32):5418–5424. 10.1200/JCO.2009.23.6075
  • Yacoub A, Mascarenhas J, Kosiorek H, et al. Pegylated interferon alfa-2a for polycythemia vera or essential thrombocythemia resistant or intolerant to hydroxyurea. Blood 2019 Oct 31;134(18):1498–1509.10.1182/blood.2019000428
  • Jin J, Qin A, Zhang L, et al. A phase II trial to assess the efficacy and safety of ropeginterferon α-2b in Chinese patients with polycythemia vera. Future Oncol 2023 Apr;19(11):753–761. 10.2217/fon-2022-1141
  • Lee S-E, Yoon S-S, Yang D-H, et al. A Single-Arm, Open-Label, Multicenter Study to Assess Molecular Response of P1101 Therapy in Patients with Polycythemia Vera and Elevated Hematocrit: Results from 12-Month Core Study. Blood. 2023;142(Supplement 1):4575–75. doi: 10.1182/blood-2023-184431
  • Kiladjian JJ, Klade C, Georgiev P, et al. Long-term outcomes of polycythemia vera patients treated with ropeginterferon Alfa-2b. Leukemia 2022 May;36(5):1408–1411.10.1038/s41375-022-01528-x
  • Gisslinger H, Klade C, Georgiev P, et al. Event-free survival in patients with polycythemia vera treated with ropeginterferon alfa-2b versus best available treatment. Leukemia 2023 Oct;37(10):2129–2132.10.1038/s41375-023-02008-6
  • Barbui T, Vannucchi AM, De Stefano V, et al. Ropeginterferon versus Standard Therapy for Low-Risk Patients with Polycythemia Vera. NEJM Evid 2023 Jun;2(6):EVIDoa2200335.
  • Beauverd Y, Ianotto J-C, Thaw KH, et al. Impact of Cytoreductive Drugs upon Outcomes in a Contemporary Cohort of Adolescent and Young Adults with Essential Thrombocythemia and Polycythemia Vera. Blood. 2023;142(Supplement 1):748–48. doi: 10.1182/blood-2023-185108
  • Vannucchi AM, Kiladjian JJ, Griesshammer M, et al. Ruxolitinib versus standard therapy for the treatment of polycythemia vera. N Engl J Med 2015 Jan 29;372(5):426–435.10.1056/NEJMoa1409002
  • Kiladjian JJ, Zachee P, Hino M, et al. Long-term efficacy and safety of ruxolitinib versus best available therapy in polycythaemia vera (RESPONSE): 5-year follow up of a phase 3 study. Lancet Haematol 2020 Mar;7(3):e226–e37.
  • Passamonti F, Palandri F, Saydam G, et al. Ruxolitinib versus best available therapy in inadequately controlled polycythaemia vera without splenomegaly (RESPONSE-2): 5-year follow up of a randomised, phase 3b study. Lancet Haematol 2022 Jul;9(7):e480–e92.
  • Harrison CN, Nangalia J, Boucher R, et al. Ruxolitinib Versus Best Available Therapy for Polycythemia Vera Intolerant or Resistant to Hydroxycarbamide in a Randomized Trial. J Clin Oncol 2023 Jul 1;41(19):3534–3544. 10.1200/JCO.22.01935
  • Hernández-Boluda JC, Alvarez-Larrán A, Gómez M, et al. Clinical evaluation of the European LeukaemiaNet criteria for clinicohaematological response and resistance/intolerance to hydroxycarbamide in essential thrombocythaemia. Br J Haematol 2011 Jan;152(1):81–88.10.1111/j.1365-2141.2010.08430.x
  • Alvarez-Larrán A, Pereira A, Cervantes F, et al. Assessment and prognostic value of the European LeukemiaNet criteria for clinicohematologic response, resistance, and intolerance to hydroxyurea in polycythemia vera. Blood 2012 Feb 9;119(6):1363–1369.10.1182/blood-2011-10-387787
  • Tremblay D, Srisuwananukorn A, Ronner L, et al. European LeukemiaNet Response Predicts Disease Progression but Not Thrombosis in Polycythemia Vera. Hemasphere 2022 Jun;6(6):e721.10.1097/HS9.0000000000000721
  • Koschmieder S, Isfort S, Wolf D, et al. Efficacy and safety of ruxolitinib in patients with newly-diagnosed polycythemia vera: futility analysis of the RuxoBEAT clinical trial of the GSG-MPN study group. Ann Hematol 2023 Feb;102(2):349–358.10.1007/s00277-022-05080-7
  • Mikkelsen SU, Kjaer L, Bjørn ME, et al. Safety and efficacy of combination therapy of interferon-α2 and ruxolitinib in polycythemia vera and myelofibrosis. Cancer Med 2018 Aug;7(8):3571–3581. 10.1002/cam4.1619
  • Koschmieder S, Mughal TI, Hasselbalch HC, et al. Myeloproliferative neoplasms and inflammation: whether to target the malignant clone or the inflammatory process or both. Leukemia 2016 May;30(5):1018–1024.10.1038/leu.2016.12
  • Bjørn ME, Hasselbalch HC Minimal residual disease or cure in MPNs? Rationales and perspectives on combination therapy with interferon-alpha2 and ruxolitinib. Expert Rev Hematol 2017 May;10(5):393–404. 10.1080/17474086.2017.1284583
  • Sørensen AL, Mikkelsen SU, Knudsen TA, et al. Ruxolitinib and interferon-α2 combination therapy for patients with polycythemia vera or myelofibrosis: a phase II study. Haematologica 2020 Sep 1;105(9):2262–2272.10.3324/haematol.2019.235648
  • Gowin K, Jain T, Kosiorek H, et al. Pegylated interferon alpha - 2a is clinically effective and tolerable in myeloproliferative neoplasm patients treated off clinical trial. Leuk Res 2017 Mar;54:73–77.
  • Handa S, Ginzburg Y, Hoffman R, et al. Hepcidin mimetics in polycythemia vera: resolving the irony of iron deficiency and erythrocytosis. Curr Opin Hematol 2023 Mar 1;30(2):45–52. 10.1097/MOH.0000000000000747
  • Ginzburg YZ, Feola M, Zimran E, et al. Dysregulated iron metabolism in polycythemia vera: etiology and consequences. Leukemia 2018 Oct;32(10):2105–2116.10.1038/s41375-018-0207-9
  • Ruchala P, Nemeth E The pathophysiology and pharmacology of hepcidin. Trends Pharmacol Sci 2014 Mar;35(3):155–161. 10.1016/j.tips.2014.01.004
  • Liu J, Sun B, Yin H, Liu S. Hepcidin: A Promising Therapeutic Target for Iron Disorders: A Systematic Review. Medicine (Baltimore) 2016 Apr;95(14):e3150.
  • Kubovcakova L, Manolova V., Nyffenegger N. Efficacy of oral Ferroportin Inhibitor in a mouse model of polycythemia vera. In Abstract, European Iron Club Meeting, Zurich, Switzerland February 08–11, 2018.
  • Casu C, Oikonomidou PR, Chen H, et al. Minihepcidin peptides as disease modifiers in mice affected by β-thalassemia and polycythemia vera. Blood 2016 Jul 14;128(2):265–276.10.1182/blood-2015-10-676742
  • Casu C, Liu A, De Rosa G, et al. Tmprss6-ASO as a tool for the treatment of polycythemia vera mice. PLoS One. 2021;16(12):e0251995. doi: 10.1371/journal.pone.0251995
  • Kremyanskaya M, Kuykendall AT, Pemmaraju N, et al. Rusfertide, a Hepcidin Mimetic, for Control of Erythrocytosis in Polycythemia Vera. N Engl J Med 2024 Feb 22;390(8):723–735.10.1056/NEJMoa2308809
  • Ritchie EK, Pettit KM, Kuykendall AT, et al. Durability of Hematocrit Control in Polycythemia Vera with the First-in-Class Hepcidin Mimetic Rusfertide: Two-Year Follow up Results from the Revive Study. Blood 20232023/11 /02/;142:745.
  • Ginzburg Y, Chew LP, Modi N, et al. Rusfertide Improves Markers of Iron Deficiency in Patients with Polycythemia Vera. Blood. 2023;142(Supplement 1):3208–08. doi: 10.1182/blood-2023-178334
  • Mascarenhas J, Lu M, Kosiorek H, et al. Oral idasanutlin in patients with polycythemia vera. Blood 2019 Aug 8;134(6):525–533.10.1182/blood.2018893545
  • Marubayashi S, Koppikar P, Taldone T, et al. HSP90 is a therapeutic target in JAK2-dependent myeloproliferative neoplasms in mice and humans. J Clin Invest 2010 Oct;120(10):3578–3593. 10.1172/JCI42442
  • Guerini V, Barbui V, Spinelli O, et al. The histone deacetylase inhibitor ITF2357 selectively targets cells bearing mutated JAK2(V617F). Leukemia 2008 Apr;22(4):740–7.
  • Rambaldi A, Iurlo A, Vannucchi AM, et al. Long-term safety and efficacy of givinostat in polycythemia vera: 4-year mean follow up of three phase 1/2 studies and a compassionate use program. Blood Cancer J 2021 Mar 6;11(3):53. 10.1038/s41408-021-00445-z
  • Pettit KM, Gill H, Yacoub A, et al. A phase 2 study of the LSD1 inhibitor bomedemstat (IMG-7289) for the treatment of advanced myelofibrosis (MF): updated results and genomic analyses. Blood. 2022;140(Supplement 1):9717–9720. doi: 10.1182/blood-2022-167122
  • Gill H, Au L, Leung GMK, et al. Phase 2 Study to Assess the Safety and Efficacy of Bomedemstat (MK3543) in Combination with Ruxolitinib in Patients with Myelofibrosis. Blood. 2023;142(Supplement 1):621–21. doi: 10.1182/blood-2023-181971
  • Triguero A, Pedraza A, Pérez-Encinas M, et al. Low-risk polycythemia vera treated with phlebotomies: clinical characteristics, hematologic control and complications in 453 patients from the Spanish Registry of Polycythemia Vera. Ann Hematol 2022 Oct;101(10):2231–2239.10.1007/s00277-022-04963-z
  • Guglielmelli P, Mora B, Gesullo F, et al. Clinical impact of mutated JAK2 allele burden reduction in polycythemia vera and essential thrombocythemia. Am J Hematol 2024 Aug;99(8):1550–1559.10.1002/ajh.27400
  • Daltro De Oliveira R, Soret-Dulphy J, Zhao L-P, et al. Interferon-Alpha (IFN) Therapy Discontinuation Is Feasible in Myeloproliferative Neoplasm (MPN) Patients with Complete Hematological Remission. Blood 2020;136(Supplement 1):35–36.
  • Tefferi A, Lasho TL, Guglielmelli P, et al. Targeted deep sequencing in polycythemia vera and essential thrombocythemia. Blood Adv 2016 Nov 29;1(1):21–30. 10.1182/bloodadvances.2016000216
  • Kiladjian JJ, Massé 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 Aug;24(8):1519–23.
  • Knudsen TA, Skov V, Stevenson K, et al. Genomic profiling of a randomized trial of interferon-α vs hydroxyurea in MPN reveals mutation-specific responses. Blood Adv 2022 Apr 12;6(7):2107–2119. 10.1182/bloodadvances.2021004856

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.