3,005
Views
2
CrossRef citations to date
0
Altmetric
Review

Improving symptom burden and quality of life in patients with myelofibrosis: current strategies and future directions

&
Pages 607-619 | Received 16 Apr 2021, Accepted 14 Jun 2021, Published online: 09 Aug 2021

References

  • Delhommeau F, Jeziorowska D, Marzac C, et al. Molecular aspects of myeloproliferative neoplasms. Int J Hematol. 2010;91(2):165–173.
  • Vannucchi AM, Lasho TL, Guglielmelli P, et al. Mutations and prognosis in primary myelofibrosis. Leukemia. 2013;27(9):1861–1869.
  • Lasho TL, Jimma T, Finke CM, et al. SRSF2 mutations in primary myelofibrosis: significant clustering with IDH mutations and independent association with inferior overall and leukemia-free survival. Blood. 2012;120(20):4168–4171.
  • Zhang SJ, Abdel-Wahab O. Disordered epigenetic regulation in the pathophysiology of myeloproliferative neoplasms. Curr Hematol Malig Rep. 2012;7(1):34–42.
  • Vainchenker W, Kralovics R. Genetic basis and molecular pathophysiology of classical myeloproliferative neoplasms. Blood. 2017;129(6):667–679.
  • Lichtman M, Tefferi A. Primary myelofibrosis. In: Kaushansky K, Beutler E, Seligsohn U, et al., editors. Williams hematology. 8th ed. New York:McGraw-Hill; 2011. p.1381–1389.
  • 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(2):401–408.
  • Geyer HL, Dueck AC, Scherber RM, et al. Impact of Inflammation on Myeloproliferative Neoplasm Symptom Development. Mediators Inflamm. 2015;2015:284706.
  • Prakash S, Hoffman R, Barouk S, et al. Splenic extramedullary hematopoietic proliferation in Philadelphia chromosome-negative myeloproliferative neoplasms: heterogeneous morphology and cytological composition. Mod Pathol. 2012;25(6):815–827.
  • Cervantes F. How I treat myelofibrosis. Blood. 2014;124(17):2635–2642.
  • 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(1):68–76.
  • McFarland DC, Polizzi H, Mascarenhas J, et al. Psychological symptoms among patients with BCR-ABL-negative myeloproliferative neoplasms. J Natl Compr Canc Netw. 2016;14(12):1563–1570.
  • Padrnos L, Scherber R, Geyer H, et al. Depressive symptoms and myeloproliferative neoplasms: understanding the confounding factor in a complex condition. Cancer Med. 2020;9(22):8301–8309.
  • Geyer HL, Andreasson B, Kosiorek HE, et al. The role of sexuality symptoms in myeloproliferative neoplasm symptom burden and quality of life: an analysis by the MPN QOL international study group. Cancer. 2016;122(12):1888–1896.
  • Geyer HL, Kosiorek H, Dueck AC, et al. Associations between gender, disease features and symptom burden in patients with myeloproliferative neoplasms: an analysis by the MPN QOL international working group. Haematologica. 2017;102(1):85–93.
  • Hotamisligil GS. Inflammation and metabolic disorders. Nature. 2006;444(7121):860–867.
  • Christensen SF, Scherber RM, Brochmann N, et al. Body mass index and total symptom burden in myeloproliferative neoplasms discovery of a U-shaped association. Cancers (Basel). 2020;12(8):8.
  • Yu J, Paranagama D, Geyer HL, et al. Relationship between symptom burden and disability leave among patients with myeloproliferative neoplasms (MPNs): findings from the living with MPN patient survey. Ann Hematol. 2019;98(5):1119–1125.
  • 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;96(10):1653–1665.
  • 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(13):2895–2901.
  • 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(9):1703–1708.
  • Passamonti F, Giorgino T, Mora B, et al. A clinical-molecular prognostic model to predict survival in patients with post polycythemia vera and post essential thrombocythemia myelofibrosis. Leukemia. 2017;31(12):2726–2731.
  • Tefferi A, Guglielmelli P, Lasho TL, et al. MIPSS70+ version 2.0: mutation and karyotype-enhanced international prognostic scoring system for primary myelofibrosis. J Clin Oncol. 2018;36(17):1769–1770.
  • Miller CB, Komrokji RS, Mesa RA, et al. Practical measures of clinical benefit with ruxolitinib therapy: an exploratory analysis of COMFORT-I. Clin Lymphoma Myeloma Leuk. 2017;17(8):479–487.
  • 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(9):1199–1203.
  • 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(33):4098–4103.
  • Gerds A, Gotlib J, Bose P, et al. National comprehensive cancer network (NCCN) clinical practice guidelines in oncology: myeloproliferative Neoplasms. Version 1.2020 [ cited 2021 Mar 31]. Available from: www.nccn.org
  • Verstovsek S, Mesa RA, Gotlib J, et al., A double-blind, placebo-controlled trial of ruxolitinib for myelofibrosis. N Engl J Med. 2012. 366(9):799–807.
  • 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(9):787–798.
  • Verstovsek S, Mesa RA, Gotlib J, et al. Long-term treatment with ruxolitinib for patients with myelofibrosis: 5-year update from the randomized, double-blind, placebo-controlled, phase 3 COMFORT-I trial. J Hematol Oncol. 2017;10(1):55.
  • 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(8):1701–1707.
  • 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(1):156.
  • 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(5):643–651.
  • Harrison CN, Schaap N, Vannucchi AM, et al., Janus kinase-2 inhibitor fedratinib in patients with myelofibrosis previously treated with ruxolitinib (JAKARTA-2): a single-arm, open-label, non-randomised, phase 2, multicentre study. Lancet Haematol. 2017. 4(7):e317–24.
  • 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(6):594–603.
  • 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(5):e225–36.
  • 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(5):652–659.
  • 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(34):3844–3850.
  • 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. 2018;5(2):e73–81.
  • Harrison C, Mesa RA, Jamieson C, et al. Case series of potential Wernicke’s encephalopathy in patients treated with fedratinib. Blood. 2017;130(supplement 1):4197.
  • Huang B, Yang XD, Zhou MM, et al. Brd4 coactivates transcriptional activation of NF-kappaB via specific binding to acetylated RelA. Mol Cell Biol. 2009;29(5):1375–1387.
  • Mascarenhas J, Harrsion C, Patriarca A, et al. CPI-0610, a Bromodomain and Extraterminal Domain Protein (BET) Inhibitor, in Combination with Ruxolitinib, in JAK-Inhibitor-Naïve Myelofibrosis Patients: update of MANIFEST Phase 2 Study. Blood. 2020;136(Supplement 1):43.
  • Yacoub A, Pettit K, Bradley TJ, et al. A Phase 2 Study of the LSD1 Inhibitor IMG7289 (bomedemstat) for the Treatment of Advanced Myelofibrosis. Blood. 2020;136. Abstract 51.
  • 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(16):1664–1674.
  • Odenike O, Godwin J, Van Besien K, et al. Phase II trial of low dose, subcutaneous decitabine in myelofibrosis. Blood. 2008;111(11):2809.
  • Abdelouahab H, Zhang Y, Wittner M, et al. CXCL12/CXCR4 pathway is activated by oncogenic JAK2 in a PI3K-dependent manner. Oncotarget. 2017 Aug 15;8(33):54082–54085.
  • Stivala S, Codilupi T, Brkic S, et al. Targeting compensatory MEK/ERK activation increases JAK inhibitor efficacy in myeloproliferative neoplasms. J Clin Invest. 2019;129(4):1596–1611.
  • Santos FPS, Getta B, Masarova L, et al. Prognostic impact of RAS-pathway mutations in patients with myelofibrosis. Leukemia. 2020;34(3):799–810.
  • Mascarenhas J, Vannucchi AM, Mean AJ, et al. An open-label, global, multicenter, phase 1b/2 study of KRT-232, a first-in-class, oral small-molecule inhibitor of murine double minute 2 (MDM2), combined with ruxolitinib in patients who have myelofibrosis and a suboptimal response to ruxolitinib. Blood. 2020;136(supplement 1):44–45.
  • 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(Supplement 1):49–50.
  • Gerds AT, Vannucchi AM, Passamonti M, et al. A phase 2 study of Luspatercept in patients with myelofibrosis-associated Anemia. Blood. 2019;134(Supplement 1):557.
  • Mascarenhas J, Komrokji RS, Cavo M, et al. Imetelstat Is effective treatment for patients with intermediate-2 or high-risk myelofibrosis who have relapsed on or are refractory to janus kinase inhibitor therapy: results of a phase 2 randomized study of two dose levels. Blood. 2018;132(Supplement 1):685.
  • Cramp F, Byron-Daniel J. Exercise for the management of cancer-related fatigue in adults. Cochrane Database Syst Rev. 2012;11:CD006145.
  • Tolstrup Larsen R, Tang LH, Brochmann N, et al. Associations between fatigue, physical activity, and QoL in patients with myeloproliferative neoplasms. Eur J Haematol. 2018;100(6):550–559.
  • Pedersen KM, Zangger G, Brochmann N, et al. The effectiveness of exercise-based rehabilitation to patients with myeloproliferative neoplasms-An explorative study. Eur J Cancer Care (Engl). 2018 Sep;27(5):e12865.
  • Huberty J, Eckert R, Larkey L, et al. Perceptions of myeloproliferative neoplasm patients participating in an online yoga intervention: a qualitative study. Integr Cancer Ther. 2018;17(4):1150–1162.
  • Huberty J, Eckert R, Dueck A, et al. Online yoga in myeloproliferative neoplasm patients: results of a randomized pilot trial to inform future research. BMC Complement Altern Med. 2019;19(1):121.
  • Scherber RM, Langlais BT, Geyer H, et al. Nutrition and supplement use characteristics in the myeloproliferative neoplasms: results from the nutrient survey. Blood. 2017;130(Supplement 1):2193.
  • Witt CM, Balneaves LG, Cardoso MJ, et al. A comprehensive definition for integrative oncology. J Natl Cancer Inst Monogr. 2017;2017(52):3–7.
  • Gowin K, Langlais BT, Kosiorek HE, et al. The SIMM study: survey of integrative medicine in myeloproliferative neoplasms. Cancer Med. 2020;9(24):9445–9453.
  • Minton O, Richardson A, Sharpe M, et al. Drug therapy for the management of cancer-related fatigue. Cochrane Database Syst Rev. 2010; (7):CD006704. https://doi.org/10.1002/14651858.CD006704.pub3.
  • Given C, Given B, Rahbar M, et al. Effect of a cognitive behavioral intervention on reducing symptom severity during chemotherapy. J Clin Oncol. 2004;22(3):507–516.
  • Gerds A, Su D, Martynova A, et al. Ruxolitinib rechallenge can improve constitutional symptoms and splenomegaly in patients with myelofibrosis: a case series. Clin Lymphoma Myeloma Leuk. 2018;18(11):e463–8.
  • Sankar K, Pettit K. Non-pharmacologic management of splenomegaly for patients with myelofibrosis: is there any role for splenectomy or splenic radiation in 2020? Curr Hematol Malig Rep. 2020;15(5):391–400.
  • Tefferi A, Mesa RA, Nagorney DM, et al. Splenectomy in myelofibrosis with myeloid metaplasia: a single-institution experience with 223 patients. Blood. 2000;95(7):2226–2233.
  • Mesa RA, Tefferi A. Palliative splenectomy in myelofibrosis with myeloid metaplasia. Leuk Lymphoma. 2001;42(5):901–911.
  • Pardanani A, Brown P, Neben-Wittich M, et al. Effective management of accelerated phase myelofibrosis with low-dose splenic radiotherapy. Am J Hematol. 2010;85(9):715–716.
  • Elliott MA, Chen MG, Silverstein MN, et al. Splenic irradiation for symptomatic splenomegaly associated with myelofibrosis with myeloid metaplasia. Br J Haematol. 1998;103(2):505–511.
  • 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.
  • Vaa BE, Wolanskyj AP, Roeker L, et al. Pruritus in primary myelofibrosis: clinical and laboratory correlates. Am J Hematol. 2012;87(2):136–138.
  • Fjellner B, Hägermark O. Pruritus in polycythemia vera: treatment with aspirin and possibility of platelet involvement. Acta Derm Venereol. 1979;59(6):505–512.
  • Steinman HK, Kobza-Black A, Lotti TM, et al. Polycythaemia rubra vera and water-induced pruritus: blood histamine levels and cutaneous fibrinolytic activity before and after water challenge. Br J Dermatol. 1987;116(3):329–333.
  • Ishii T, Wang J, Zhang W, et al. Pivotal role of mast cells in pruritogenesis in patients with myeloproliferative disorders. Blood. 2009;113(23):5942–5950.
  • Tefferi A, Lasho TL, Schwager SM, et al. The clinical phenotype of wild-type, heterozygous, and homozygous JAK2V617F in polycythemia vera. Cancer. 2006;106(3):631–635.
  • Vannucchi AM, Antonioli E, Guglielmelli P, et al. Clinical profile of homozygous JAK2 617V>F mutation in patients with polycythemia vera or essential thrombocythemia. Blood. 2007;110(3):840–846.
  • Pardanani A, Lim KH, Lasho TL, et al. Prognostically relevant breakdown of 123 patients with systemic mastocytosis associated with other myeloid malignancies. Blood. 2009;114(18):3769–3772.
  • Lim KH, Tefferi A, Lasho TL, et al. Systemic mastocytosis in 342 consecutive adults: survival studies and prognostic factors. Blood. 2009;113(23):5727–5736.
  • Hermans MAW, Schrijver B, van Holten-Neelen CCPA, et al. The JAK1/JAK2- inhibitor ruxolitinib inhibits mast cell degranulation and cytokine release. Clin Exp Allergy. 2018;48(11):1412–1420.
  • Diehn F, Tefferi A. Pruritus in polycythaemia vera: prevalence, laboratory correlates and management. Br J Haematol. 2001;115(3):619–621.
  • Steinman HK, Greaves MW. Aquagenic pruritus. J Am Acad Dermatol. 1985;13(1):91–96.
  • 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.
  • Tefferi A, Fonseca R. Selective serotonin reuptake inhibitors are effective in the treatment of polycythemia vera-associated pruritus. Blood. 2002;99(7):2627.
  • Baldo A, Sammarco E, Plaitano R, et al. Narrowband (TL-01) ultraviolet B phototherapy for pruritus in polycythaemia vera. Br J Dermatol. 2002;147(5):979–981.