209
Views
0
CrossRef citations to date
0
Altmetric
REVIEW

Polycythemia Vera: Barriers to and Strategies for Optimal Management

ORCID Icon, , &
Pages 77-90 | Received 27 Sep 2023, Accepted 16 Dec 2023, Published online: 21 Dec 2023

References

  • Ma X, Vanasse G, Cartmel B, Wang Y, Selinger HA. Prevalence of polycythemia vera and essential thrombocythemia. Am J Hematol. 2008;83(5):359–362. doi:10.1002/ajh.21129
  • Moulard O, Mehta J, Fryzek J, Olivares R, Iqbal U, Mesa RA. Epidemiology of myelofibrosis, essential thrombocythemia, and polycythemia vera in the European Union. Eur J Haematol. 2014;92(4):289–297. doi:10.1111/ejh.12256
  • Ianotto JC, Curto-Garcia N, Lauermannova M, Radia D, Kiladjian JJ, Harrison C. Characteristics and outcomes of patients with essential thrombocythemia or polycythemia vera diagnosed before 20 years of age, a systematic review. Haematologica. 2019;2018:200832. doi:10.3324/haematol.2018.200832
  • Falanga A, Marchetti M. Thrombotic disease in the myeloproliferative neoplasms. Hematology. 2012;2012(1):571–581. doi:10.1182/asheducation.V2012.1.571.3798557
  • Tefferi A, Lasho TL, Guglielmelli P, et al. Targeted deep sequencing in polycythemia vera and essential thrombocythemia. Blood Adv. 2016;1(1):21–30. doi:10.1182/bloodadvances.2016000216
  • Tang G, Hidalgo Lopez JE, Wang SA, et al. Characteristics and clinical significance of cytogenetic abnormalities in polycythemia vera. Haematologica. 2017;102(9):1511–1518. doi:10.3324/haematol.2017.165795
  • Cerquozzi S, Tefferi A. Blast transformation and fibrotic progression in polycythemia vera and essential thrombocythemia: a literature review of incidence and risk factors. Blood Cancer J. 2015;5(11):e366–10. doi:10.1038/bcj.2015.95
  • Tefferi A, Guglielmelli P, Lasho TL, et al. Mutation-enhanced international prognostic systems for essential thrombocythaemia and polycythaemia vera. Br J Haematol. 2020;189(2):291–302. doi:10.1111/BJH.16380
  • McMullin MF, Harrison CN, Ali S, et al. A guideline for the diagnosis and management of polycythaemia vera. A British Society for Haematology Guideline. Br J Haematol. 2019;184(2):176–191. doi:10.1111/bjh.15648
  • Tefferi A, Vannucchi AM, Barbui T. Polycythemia vera: historical oversights, diagnostic details, and therapeutic views. Leukemia. 2021;35(12):3339–3351. doi:10.1038/s41375-021-01401-3
  • Tefferi A, Barbui T. Polycythemia vera: 2024 update on diagnosis, risk-stratification, and management. Am J Hematol. 2023;98(9):1465–1487. doi:10.1002/ajh.27002
  • 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;7(12):662. doi:10.1038/s41408-017-0035-6
  • Mancuso S, Santoro M, Accurso V, et al. Cardiovascular risk in polycythemia vera: thrombotic risk and survival: can cytoreductive therapy be useful in patients with low-risk polycythemia vera with cardiovascular risk factors? Oncol Res Treat. 2020;43(10):526–530. doi:10.1159/000509376
  • Estruch R, Ros E, Salas-Salvadó J, et al. Primary prevention of cardiovascular disease with a Mediterranean diet. N Engl J Med. 2013;368(14):1279–1290. doi:10.1056/NEJMoa1200303
  • Casas R, Urpi-Sardà M, Sacanella E, et al. Anti-inflammatory effects of the Mediterranean diet in the early and late stages of atheroma plaque development. Mediators Inflamm. 2017;2017:3674390. doi:10.1155/2017/3674390
  • Mendez LF, Nguyen H, Nguyen J, et al. The Nutrient Trial (NUTRitional Intervention among myEloproliferative Neoplasms): feasibility Phase. Blood. 2019;134:5380. doi:10.1182/blood-2019-130851
  • Berk PD, Goldberg JD, Donovan PB, Fruchtman S, Berlin NI, Wasserman LR. Therapeutic recommendations in polycythemia vera based on polycythemia vera study group protocols. Semin Hematol. 1986;23:132–143.
  • Marchioli R, Finazzi G, Specchia G, et al. Cardiovascular events and intensity of treatment in polycythemia vera. N Engl J Med. 2012;368(1):22–33. doi:10.1056/NEJMoa1208500
  • Barbui T, Tefferi A, Vannucchi AM, et al. Philadelphia chromosome-negative classical myeloproliferative neoplasms: revised management recommendations from European LeukemiaNet. Leukemia. 2018;32(5):1057–1069. doi:10.1038/s41375-018-0077-1
  • Assi TB, Baz E. Current applications of therapeutic phlebotomy. Blood Transfus. 2014;12(Suppl 1):s75–83. doi:10.2450/2013.0299-12
  • Ohnishi H. Side effects of phlebotomy: pathophysiology, diagnosis, treatment and prophylaxis. Rinsho Byori. 2005;53(10):904–910.
  • Barton JC, Bottomley SS. Iron deficiency due to excessive therapeutic phlebotomy in hemochromatosis. Am J Hematol. 2000;65(3):223–226. doi:10.1002/1096-8652(200011)65:3<223::aid-ajh8>3.0.co;2-9
  • Padmanabhan A, Connelly-Smith L, Aqui N, et al. Guidelines on the use of therapeutic apheresis in clinical practice – evidence-based approach from the writing committee of the American society for apheresis: the eighth special issue. J Clin Apher. 2019;34(3):171–354. doi:10.1002/jca.21705
  • Barbui T, Passamonti F, Accorsi P, et al. Evidence- and consensus-based recommendations for phlebotomy in polycythemia vera. Leukemia. 2018;32(9):2077–2081. doi:10.1038/s41375-018-0199-5
  • Tartaglia AP, Goldberg JD, Berk PD, Wasserman LR. Adverse effects of antiaggregating platelet therapy in the treatment of polycythemia vera. Semin Hematol. 1986;23(3):172–176.
  • Landolfi R, Marchioli R, Kutti J, et al. Efficacy and safety of low-dose aspirin in polycythemia vera. N Engl J Med. 2004;350(2):114–124. doi:10.1056/NEJMoa035572
  • Landolfi R, Di Gennaro L. Prevention of thrombosis in polycythemia vera and essential thrombocythemia. Haematologica. 2008;93(3 SE–Editorials):331–335. doi:10.3324/haematol.12604
  • Ashley C, Eric S, Bruce AW. Direct oral anticoagulant use: a practical guide to common clinical challenges. J Am Heart Assoc. 2020;9(13):e017559. doi:10.1161/JAHA.120.017559
  • Raskob GE, van Es N, Verhamme P, et al. Edoxaban for the Treatment of Cancer-Associated Venous Thromboembolism. N Engl J Med. 2017;378(7):615–624. doi:10.1056/NEJMoa1711948
  • Young AM, Marshall A, Thirlwall J, et al. Comparison of an oral factor xa inhibitor with low molecular weight heparin in patients with cancer with venous thromboembolism: results of a randomized trial (SELECT-D). J Clin Oncol. 2018;36(20):2017–2023. doi:10.1200/JCO.2018.78.8034
  • Ianotto JC, Couturier MA, Galinat H, et al. Administration of direct oral anticoagulants in patients with myeloproliferative neoplasms. Int J Hematol. 2017;106(4):517–521. doi:10.1007/S12185-017-2282-5
  • Curto-Garcia N, Doyle AJ, Breen KA, et al. Outcomes of patients receiving direct oral anticoagulants for myeloproliferative neoplasm-associated venous thromboembolism in a large tertiary centre in the UK. Br J Haematol. 2020;189. doi:10.1111/bjh.16485
  • How J, Story C, Ren S, et al. Practice patterns and outcomes of direct oral anticoagulant use in myeloproliferative neoplasm patients. Blood Cancer J. 2021;11(11). doi:10.1038/S41408-021-00566-5
  • Barbui T, De Stefano V, Carobbio A, et al. Direct oral anticoagulants for myeloproliferative neoplasms: results from an international study on 442 patients. Leukemia. 2021;35(10):2989. doi:10.1038/S41375-021-01279-1
  • Yarbro JW. Mechanism of action of hydroxyurea. Semin Oncol. 1992;19:3).
  • Fruchtman SM, Mack K, Kaplan ME, Peterson P, Berk PD, Wasserman LR. From efficacy to safety: a Polycythemia Vera Study group report on hydroxyurea in patients with polycythemia vera. Semin Hematol. 1997;34(1):17–23.
  • Najean Y, Rain JD. Treatment of polycythemia vera: the use of hydroxyurea and pipobroman in 292 patients under the age of 65 years. Blood. 1997;90(9):3370–3377. doi:10.1182/blood.V90.9.3370
  • Kiladjian JJ, Chevret S, Dosquet C, Chomienne C, Rain JD. Treatment of polycythemia vera with hydroxyurea and pipobroman: final results of a randomized trial initiated in 1980. J Clin Oncol. 2011;29(29):3907–3913. doi:10.1200/JCO.2011.36.0792
  • 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;92(11):1131–1136. doi:10.1002/ajh.24851
  • 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;139(19):2931–2941. doi:10.1182/blood.2021012743
  • Gisslinger H, Klade C, Georgiev P, et al. Final results from PROUD-PV a randomized controlled phase 3 trial comparing ropeginterferon alfa-2b to hydroxyurea in polycythemia vera patients; 2016.
  • 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;7(3):e196–e208. doi:10.1016/S2352-3026(19)30236-4
  • Berthaut I, Guignedoux G, Kirsch-Noir F, et al. Influence of sickle cell disease and treatment with hydroxyurea on sperm parameters and fertility of human males. Haematologica. 2008;93(7):988–993. doi:10.3324/haematol.11515
  • Antonioli E, Guglielmelli P, Pieri L, et al. Hydroxyurea-related toxicity in 3411 patients with Ph’-negative MPN. Am J Hematol. 2012;87(5):552–554. doi:10.1002/ajh.23160
  • Gavini DR, Salvi DJ, Shah PH, Uma D, Lee JH, Hamid P. Non-melanoma skin cancers in patients on hydroxyurea for Philadelphia chromosome-negative myeloproliferative neoplasms: a systematic review. Cureus. 2021;13(8):e16978. doi:10.7759/cureus.16978
  • Barbui T, Ghirardi A, Masciulli A, et al. Second cancer in Philadelphia negative myeloproliferative neoplasms (MPN-K). A nested case-control study. Leukemia. 2019;33(8):1996–2005. doi:10.1038/S41375-019-0487-8
  • 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;119(6):1363–1369. doi:10.1182/blood-2011-10-387787
  • 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(20):4829–4833. doi:10.1182/blood-2008-09-176818
  • Steinberg MH, McCarthy WF, Castro O, et al. The risks and benefits of long-term use of hydroxyurea in sickle cell anemia: a 17.5 year follow-up. Am J Hematol. 2010;85(6):403. doi:10.1002/AJH.21699
  • Kiladjian JJ, Giraudier S, Cassinat B. Interferon-alpha for the therapy of myeloproliferative neoplasms: targeting the malignant clone. Leukemia. 2016;30(4):776–781. doi:10.1038/leu.2015.326
  • Kiladjian JJ, Klade C, Georgiev P, et al. Long-term outcomes of polycythemia vera patients treated with ropeginterferon Alfa-2b. Leukemia. 2022;36(5):1408–1411. doi:10.1038/s41375-022-01528-x
  • 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;126(15):1762–1769. doi:10.1182/blood-2015-04-637280
  • Kiladjian JJ, Chomienne C, Fenaux P. Interferon-alpha therapy in bcr-abl-negative myeloproliferative neoplasms. Leukemia. 2008;22(11):1990–1998. doi:10.1038/leu.2008.280
  • 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;27(32):5418–5424. doi:10.1200/JCO.2009.23.6075
  • Duminuco A, Nardo A, Giuffrida G, et al. Myelofibrosis and survival prognostic models: a journey between past and future. J Clin Med. 2023;12(6):2188. doi:10.3390/JCM12062188
  • Duminuco A, Vetro C, Giallongo C, Palumbo GA. The pharmacotherapeutic management of patients with myelofibrosis: looking beyond JAK inhibitors. Expert Opin Pharmacother. 2023;24:1449–1461. doi:10.1080/14656566.2023.2228695
  • Duminuco A, Torre E, Palumbo GA, Harrison C. A Journey Through JAK inhibitors for the treatment of myeloproliferative diseases. Curr Hematol Malignancy Rep. 2023;18:176–189. doi:10.1007/S11899-023-00702-X
  • Vannucchi AM, Kiladjian JJ, Griesshammer M, et al. Ruxolitinib versus standard therapy for the treatment of polycythemia vera. N Engl J Med. 2015;372(5):426–435. doi:10.1056/NEJMOA1409002/SUPPL_FILE/NEJMOA1409002_DISCLOSURES.PDF
  • Passamonti F, Griesshammer M, Palandri F, et al. Ruxolitinib for the treatment of inadequately controlled polycythaemia vera without splenomegaly (RESPONSE-2): a randomised, open-label, phase 3b study. Lancet Oncol. 2017;18(1):88–99. doi:10.1016/S1470-2045(16)30558-7
  • 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;41(19):3534–3544. doi:10.1200/jco.22.01935
  • Vannucchi AM, Te Boekhorst PAW, Harrison CN, et al. EXPAND, a dose-finding study of ruxolitinib in patients with myelofibrosis and low platelet counts: 48-week follow-up analysis. Haematologica. 2019;104(5):947–954. doi:10.3324/HAEMATOL.2018.204602
  • Lussana F, Cattaneo M, Rambaldi A, Squizzato A. Ruxolitinib-associated infections: a systematic review and meta-analysis. Am J Hematol. 2018;93(3):339–347. doi:10.1002/AJH.24976
  • Duminuco A, Scarso S, Cupri A, et al. Leishmania infection during ruxolitinib treatment: the cytokines-based immune response in the setting of immunocompromised patients. J Clin Med. 2023;12(2):578. doi:10.3390/JCM12020578
  • Palumbo GA, Cambria D, La Spina E, et al. Ruxolitinib treatment in myelofibrosis and polycythemia vera causes suboptimal humoral immune response following standard and booster vaccination with BNT162b2 mRNA COVID-19 vaccine. Front Oncol. 2023:13. doi:10.3389/FONC.2023.1117815
  • Duminuco A, Nardo A, Orofino A, et al. Efficacy and safety of tixagevimab-cilgavimab versus SARS-CoV-2 breakthrough infection in the hematological conditions. Cancer. 2023. doi:10.1002/CNCR.35005
  • Lin JQ, Li SQ, Li S, et al. A 10-year retrospective cohort study of ruxolitinib and association with nonmelanoma skin cancer in patients with polycythemia vera and myelofibrosis. J Am Acad Dermatol. 2022;86(2):339–344. doi:10.1016/J.JAAD.2021.10.004
  • Mesa RA, Verstovsek S, Gupta V, et al. Effects of ruxolitinib treatment on metabolic and nutritional parameters in patients with myelofibrosis from COMFORT-I. Clin Lymphoma Myeloma Leuk. 2015;15(4):214–221.e1. doi:10.1016/J.CLML.2014.12.008
  • Maffioli M, Mora B, Ball S, et al. A prognostic model to predict survival after 6 months of ruxolitinib in patients with myelofibrosis. Blood Adv. 2022;6(6):1855–1864. doi:10.1182/BLOODADVANCES.2021006889
  • Duminuco A, Nardo A, Garibaldi B, et al. Prediction of survival and prognosis migration from gold-standard scores in myelofibrosis patients treated with ruxolitinib applying the rr6 prognostic model in a monocentric real-life setting. J Clin Med. 2022;11(24):7418. doi:10.3390/JCM11247418
  • Marchetti M, Vannucchi AM, Griesshammer M, et al. Appropriate management of polycythaemia vera with cytoreductive drug therapy: European LeukemiaNet 2021 recommendations. Lancet Haematol. 2022;9(4):e301–e311. doi:10.1016/S2352-3026(22)00046-1
  • Björkholm M, Derolf ÅR, Hultcrantz M, et al. Treatment-related risk factors for transformation to acute myeloid leukemia and myelodysplastic syndromes in myeloproliferative neoplasms. J Clin Oncol. 2011;29(17):2410. doi:10.1200/JCO.2011.34.7542
  • Siegel FP, Tauscher J, Petrides PE. Aquagenic pruritus in polycythemia vera: characteristics and influence on quality of life in 441 patients. Am J Hematol. 2013;88(8):665–669. doi:10.1002/AJH.23474
  • Gangat N, Strand JJ, Lasho TL, Li C-Y, Pardanani A, Tefferi A. Pruritus in polycythemia vera is associated with a lower risk of arterial thrombosis. Am J Hematol. 2008;83(6):451–453. doi:10.1002/AJH.21156
  • Weick JK, Donovan PB, Najean Y, et al. The use of cimetidine for the treatment of pruritus in polycythemia vera. Arch Intern Med. 1982;142(2):241–242. doi:10.1001/ARCHINTE.1982.003401500410010
  • Steinman HK, Greaves MW. Aquagenic pruritus. J Am Acad Dermatol. 1985;13(1):91–96. doi:10.1016/S0190-9622(85)70149-1
  • Tefferi A, Fonseca R. Selective serotonin reuptake inhibitors are effective in the treatment of polycythemia vera-associated pruritus. Blood. 2002;99(7):2627. doi:10.1182/BLOOD.V99.7.2627
  • Rivard J, Lim HW. Ultraviolet phototherapy for pruritus. Dermatol Ther. 2005;18(4):344–354. doi:10.1111/J.1529-8019.2005.00032.X
  • Finazzi G, De Stefano V, Barbui T. Splanchnic vein thrombosis in myeloproliferative neoplasms: treatment algorithm 2018. Blood Cancer J. 2018;8(7):1–6. doi:10.1038/s41408-018-0100-9
  • De Stefano V, Za T, Rossi E, et al. Recurrent thrombosis in patients with polycythemia vera and essential thrombocythemia: incidence, risk factors, and effect of treatments. Haematologica. 2008;93(3):372–380. doi:10.3324/HAEMATOL.12053
  • McMullin MFF, Mead AJ, Ali S, et al. A guideline for the management of specific situations in polycythaemia vera and secondary erythrocytosis. Br J Haematol. 2019;184(2):161–175. doi:10.1111/BJH.15647
  • Smith DA, Lilie CJ. Acute arterial occlusion. StatPearls; 2023. Available from: https://www.ncbi.nlm.nih.gov/books/NBK441851/. Accessed September 18, 2023.
  • Ulivi L, Squitieri M, Cohen H, Cowley P, Werring DJ. Cerebral venous thrombosis: a practical guide. Pract Neurol. 2020;20(5):356–367. doi:10.1136/PRACTNEUROL-2019-002415
  • Wille K, Huenerbein K, Jagenberg E, et al. Bleeding complications in bcr-abl-negative myeloproliferative neoplasms (MPN): a retrospective single-center study of 829 MPN patients. Eur J Haematol. 2022;108(2):154–162. doi:10.1111/EJH.13721
  • Casu C, Nemeth E, Rivella S. Hepcidin agonists as therapeutic tools. Blood. 2018;131(16):1790. doi:10.1182/BLOOD-2017-11-737411
  • Gerds AT, Gotlib J, Palmer JM, et al. Rusfertide for polycythemia vera: similar dosing in patients receiving therapeutic phlebotomy alone or in combination with cytoreductive treatment. Blood. 2022;140(Supplement 1):12241–12243. doi:10.1182/BLOOD-2022-163847
  • Verstovsek S, Kuykendall A, Hoffman R, et al. Verify: a phase 3 study of the hepcidin mimetic rusfertide (PTG-300) in patients with polycythemia vera. Blood. 2022;140(Supplement 1):3929–3931. doi:10.1182/BLOOD-2022-163755
  • 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(4):446–455. doi:10.1111/J.1365-2141.2010.08266.X
  • Finazzi G, Iurlo A, Martino B, et al. A long-term safety and efficacy study of givinostat in patients with polycythemia vera: the first 4 years of treatment. Blood. 2017;130(Supplement 1):1648. doi:10.1182/BLOOD.V130.SUPPL_1.1648.1648
  • 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;11(3):1–7. doi:10.1038/s41408-021-00445-z
  • 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(4):498–508. doi:10.1111/BJH.12416
  • ClinicalTrials.gov. Home. Available from: https://clinicaltrials.gov/. Accessed November 23, 2023.