Skip to main content
Top
Published in: Journal of Hematology & Oncology 1/2020

Open Access 01-12-2020 | Primary Myelofibrosis | Review

Novel therapeutics in myeloproliferative neoplasms

Authors: Sangeetha Venugopal, John Mascarenhas

Published in: Journal of Hematology & Oncology | Issue 1/2020

Login to get access

Abstract

Hyperactive signaling of the Janus-Associated Kinase/Signal Transducers and Activators of Transcription (JAK/STAT) pathway is central to the pathogenesis of Philadelphia-chromosome-negative myeloproliferative neoplasms (MPN), i.e., polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF) which are characterized by inherent biological and clinical heterogeneity. Patients with MPNs suffer from substantial symptom burden and curtailed longevity due to thrombohemorrhagic complications or progression to myelofibrosis or acute myeloid leukemia. Therefore, the management strategies focus on thrombosis risk mitigation in PV/ET, alleviation of symptom burden and improvement in cytopenias and red blood cell transfusion requirements, and disease course alteration in PMF. The United States Food and Drug Administration’s (USFDA) approval of two JAK inhibitors (ruxolitinib, fedratinib) has transformed the therapeutic landscape of MPNs in assuaging the need for frequent therapeutic phlebotomy (PV) and reduction in spleen and symptom burden (PV and PMF). Despite improving biological understanding of these complex clonal hematopoietic stem/progenitor cell neoplasms, none of the currently available therapies appear to modify the proclivity of the disease per se, thereby remaining an urgent unmet clinical need and an ongoing area of intense clinical investigation. This review will highlight the evolving targeted therapeutic agents that are in early- and late-stage MPN clinical development.
Literature
2.
go back to reference Levine RL, Wadleigh M, Cools J, Ebert BL, Wernig G, Huntly BJ, et al. Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. Cancer Cell. 2005;7(4):387–97.CrossRefPubMed Levine RL, Wadleigh M, Cools J, Ebert BL, Wernig G, Huntly BJ, et al. Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. Cancer Cell. 2005;7(4):387–97.CrossRefPubMed
3.
go back to reference Nangalia J, Massie CE, Baxter EJ, Nice FL, Gundem G, Wedge DC, et al. Somatic CALR mutations in myeloproliferative neoplasms with nonmutated JAK2. N Engl J Med. 2013;369(25):2391–405.CrossRefPubMedPubMedCentral Nangalia J, Massie CE, Baxter EJ, Nice FL, Gundem G, Wedge DC, et al. Somatic CALR mutations in myeloproliferative neoplasms with nonmutated JAK2. N Engl J Med. 2013;369(25):2391–405.CrossRefPubMedPubMedCentral
4.
go back to reference Pikman Y, Lee BH, Mercher T, McDowell E, Ebert BL, Gozo M, et al. MPLW515L is a novel somatic activating mutation in myelofibrosis with myeloid metaplasia. PLoS Med. 2006;3(7):e270.CrossRefPubMedPubMedCentral Pikman Y, Lee BH, Mercher T, McDowell E, Ebert BL, Gozo M, et al. MPLW515L is a novel somatic activating mutation in myelofibrosis with myeloid metaplasia. PLoS Med. 2006;3(7):e270.CrossRefPubMedPubMedCentral
6.
go back to reference Policitemia GIS. Polycythemia vera: the natural history of 1213 patients followed for 20 years. Ann Intern Med. 1995;123(9):656–64.CrossRef Policitemia GIS. Polycythemia vera: the natural history of 1213 patients followed for 20 years. Ann Intern Med. 1995;123(9):656–64.CrossRef
7.
go back to reference De Stefano V, Za T, Rossi E, Vannucchi AM, Ruggeri M, Elli E, et al. Recurrent thrombosis in patients with polycythemia vera and essential thrombocythemia: incidence, risk factors, and effect of treatments. Haematologica. 2008;93(3):372–80.CrossRefPubMed De Stefano V, Za T, Rossi E, Vannucchi AM, Ruggeri M, Elli E, et al. Recurrent thrombosis in patients with polycythemia vera and essential thrombocythemia: incidence, risk factors, and effect of treatments. Haematologica. 2008;93(3):372–80.CrossRefPubMed
8.
go back to reference Hultcrantz M, Andersson TM, Landgren O, Derolf AR, Dickman PW, Björkholm M, et al. Risk of arterial and venous thrombosis in 11,155 patients with myeloproliferative neoplasms and 44,620 matched controls; a population-based study. Washington, DC: American Society of Hematology; 2014. Hultcrantz M, Andersson TM, Landgren O, Derolf AR, Dickman PW, Björkholm M, et al. Risk of arterial and venous thrombosis in 11,155 patients with myeloproliferative neoplasms and 44,620 matched controls; a population-based study. Washington, DC: American Society of Hematology; 2014.
9.
go back to reference Kennedy JA, Atenafu EG, Messner HA, Craddock KJ, Brandwein JM, Lipton JH, et al. Treatment outcomes following leukemic transformation in Philadelphia-negative myeloproliferative neoplasms. Blood J Am Soc Hematol. 2013;121(14):2725–33. Kennedy JA, Atenafu EG, Messner HA, Craddock KJ, Brandwein JM, Lipton JH, et al. Treatment outcomes following leukemic transformation in Philadelphia-negative myeloproliferative neoplasms. Blood J Am Soc Hematol. 2013;121(14):2725–33.
10.
go back to reference Tefferi A, Rumi E, Finazzi G, Gisslinger H, Vannucchi A, Rodeghiero F, et al. Survival and prognosis among 1545 patients with contemporary polycythemia vera: an international study. Leukemia. 2013;27(9):1874–81.CrossRefPubMedPubMedCentral Tefferi A, Rumi E, Finazzi G, Gisslinger H, Vannucchi A, Rodeghiero F, et al. Survival and prognosis among 1545 patients with contemporary polycythemia vera: an international study. Leukemia. 2013;27(9):1874–81.CrossRefPubMedPubMedCentral
11.
go back to reference Barbui T, Finazzi G, Carobbio A, Thiele J, Passamonti F, Rumi E, et al. Development and validation of an International Prognostic Score of thrombosis in World Health Organization-essential thrombocythemia (IPSET-thrombosis). Blood. 2012;120(26):5128–33.CrossRefPubMed Barbui T, Finazzi G, Carobbio A, Thiele J, Passamonti F, Rumi E, et al. Development and validation of an International Prognostic Score of thrombosis in World Health Organization-essential thrombocythemia (IPSET-thrombosis). Blood. 2012;120(26):5128–33.CrossRefPubMed
12.
go back to reference Ali H, Aldoss I, Yang D, Mokhtari S, Khaled S, Aribi A, et al. MIPSS70+ v2.0 predicts long-term survival in myelofibrosis after allogeneic HCT with the Flu/Mel conditioning regimen. Blood Adv. 2019;3(1):83–95.CrossRefPubMedPubMedCentral Ali H, Aldoss I, Yang D, Mokhtari S, Khaled S, Aribi A, et al. MIPSS70+ v2.0 predicts long-term survival in myelofibrosis after allogeneic HCT with the Flu/Mel conditioning regimen. Blood Adv. 2019;3(1):83–95.CrossRefPubMedPubMedCentral
13.
go back to reference Barbui T, Carobbio A, Rumi E, Finazzi G, Gisslinger H, Rodeghiero F, et al. In contemporary patients with polycythemia vera, rates of thrombosis and risk factors delineate a new clinical epidemiology. Blood J Am Soc Hematol. 2014;124(19):3021–3. Barbui T, Carobbio A, Rumi E, Finazzi G, Gisslinger H, Rodeghiero F, et al. In contemporary patients with polycythemia vera, rates of thrombosis and risk factors delineate a new clinical epidemiology. Blood J Am Soc Hematol. 2014;124(19):3021–3.
14.
go back to reference Marchioli R, Finazzi G, Specchia G, Cacciola R, Cavazzina R, Cilloni D, et al. Cardiovascular events and intensity of treatment in polycythemia vera. N Engl J Med. 2013;368(1):22–33.CrossRefPubMed Marchioli R, Finazzi G, Specchia G, Cacciola R, Cavazzina R, Cilloni D, et al. Cardiovascular events and intensity of treatment in polycythemia vera. N Engl J Med. 2013;368(1):22–33.CrossRefPubMed
15.
go back to reference Patrono C, Rocca B, De Stefano V. Platelet activation and inhibition in polycythemia vera and essential thrombocythemia. Blood. 2013;121(10):1701–11.CrossRefPubMed Patrono C, Rocca B, De Stefano V. Platelet activation and inhibition in polycythemia vera and essential thrombocythemia. Blood. 2013;121(10):1701–11.CrossRefPubMed
16.
go back to reference 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.PubMed 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.PubMed
17.
go back to reference Cortelazzo S, Finazzi G, Ruggeri M, Vestri O, Galli M, Rodeghiero F, et al. Hydroxyurea for patients with essential thrombocythemia and a high risk of thrombosis. N Engl J Med. 1995;332(17):1132–6.CrossRefPubMed Cortelazzo S, Finazzi G, Ruggeri M, Vestri O, Galli M, Rodeghiero F, et al. Hydroxyurea for patients with essential thrombocythemia and a high risk of thrombosis. N Engl J Med. 1995;332(17):1132–6.CrossRefPubMed
18.
go back to reference Kiladjian JJ, Cassinat B, Chevret S, Turlure P, Cambier N, Roussel M, et al. Pegylated interferon-alfa-2a induces complete hematologic and molecular responses with low toxicity in polycythemia vera. Blood. 2008;112(8):3065–72.CrossRefPubMed Kiladjian JJ, Cassinat B, Chevret S, Turlure P, Cambier N, Roussel M, et al. Pegylated interferon-alfa-2a induces complete hematologic and molecular responses with low toxicity in polycythemia vera. Blood. 2008;112(8):3065–72.CrossRefPubMed
19.
go back to reference Verger E, Cassinat B, Chauveau A, Dosquet C, Giraudier S, Schlageter M-H, et al. Clinical and molecular response to interferon-α therapy in essential thrombocythemia patients with CALR mutations. Blood J Am Soc Hematol. 2015;126(24):2585–91. Verger E, Cassinat B, Chauveau A, Dosquet C, Giraudier S, Schlageter M-H, et al. Clinical and molecular response to interferon-α therapy in essential thrombocythemia patients with CALR mutations. Blood J Am Soc Hematol. 2015;126(24):2585–91.
20.
go back to reference Vannucchi AM, Kiladjian JJ, Griesshammer M, Masszi T, Durrant S, Passamonti F, et al. Ruxolitinib versus standard therapy for the treatment of polycythemia vera. N Engl J Med. 2015;372(5):426–35.CrossRefPubMedPubMedCentral Vannucchi AM, Kiladjian JJ, Griesshammer M, Masszi T, Durrant S, Passamonti F, et al. Ruxolitinib versus standard therapy for the treatment of polycythemia vera. N Engl J Med. 2015;372(5):426–35.CrossRefPubMedPubMedCentral
21.
go back to reference Passamonti F, Griesshammer M, Palandri F, Egyed M, Benevolo G, Devos T, 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.CrossRefPubMed Passamonti F, Griesshammer M, Palandri F, Egyed M, Benevolo G, Devos T, 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.CrossRefPubMed
22.
go back to reference Casu C, Oikonomidou PR, Chen H, Nandi V, Ginzburg Y, Prasad P, et al. Minihepcidin peptides as disease modifiers in mice affected by beta-thalassemia and polycythemia vera. Blood. 2016;128(2):265–76.CrossRefPubMedPubMedCentral Casu C, Oikonomidou PR, Chen H, Nandi V, Ginzburg Y, Prasad P, et al. Minihepcidin peptides as disease modifiers in mice affected by beta-thalassemia and polycythemia vera. Blood. 2016;128(2):265–76.CrossRefPubMedPubMedCentral
23.
go back to reference King KY, Matatall KA, Shen C-C, Goodell MA, Swierczek SI, Prchal JT. Comparative long-term effects of interferon α and hydroxyurea on human hematopoietic progenitor cells. Exp Hematol. 2015;43(10):912–8.CrossRefPubMedPubMedCentral King KY, Matatall KA, Shen C-C, Goodell MA, Swierczek SI, Prchal JT. Comparative long-term effects of interferon α and hydroxyurea on human hematopoietic progenitor cells. Exp Hematol. 2015;43(10):912–8.CrossRefPubMedPubMedCentral
24.
go back to reference Kiladjian J-J, Cassinat B, Turlure P, Cambier N, Roussel M, Bellucci S, et al. High molecular response rate of polycythemia vera patients treated with pegylated interferon α–2a. Blood. 2006;108(6):2037–40.CrossRefPubMed Kiladjian J-J, Cassinat B, Turlure P, Cambier N, Roussel M, Bellucci S, et al. High molecular response rate of polycythemia vera patients treated with pegylated interferon α–2a. Blood. 2006;108(6):2037–40.CrossRefPubMed
25.
go back to reference Quintás-Cardama A, Kantarjian H, Manshouri T, Luthra R, Estrov Z, Pierce S, 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.CrossRefPubMedPubMedCentral Quintás-Cardama A, Kantarjian H, Manshouri T, Luthra R, Estrov Z, Pierce S, 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.CrossRefPubMedPubMedCentral
26.
go back to reference Yacoub A, Mascarenhas J, Kosiorek H, Prchal JT, Berenzon D, Baer MR, et al. Pegylated interferon alfa-2a for polycythemia vera or essential thrombocythemia resistant or intolerant to hydroxyurea. Blood. 2019;134(18):1498–509.CrossRefPubMedPubMedCentral Yacoub A, Mascarenhas J, Kosiorek H, Prchal JT, Berenzon D, Baer MR, et al. Pegylated interferon alfa-2a for polycythemia vera or essential thrombocythemia resistant or intolerant to hydroxyurea. Blood. 2019;134(18):1498–509.CrossRefPubMedPubMedCentral
27.
go back to reference Them NC, Bagienski K, Berg T, Gisslinger B, Schalling M, Chen D, et al. Molecular responses and chromosomal aberrations in patients with polycythemia vera treated with peg-proline-interferon alpha-2b. Am J Hematol. 2015;90(4):288–94.CrossRefPubMedPubMedCentral Them NC, Bagienski K, Berg T, Gisslinger B, Schalling M, Chen D, et al. Molecular responses and chromosomal aberrations in patients with polycythemia vera treated with peg-proline-interferon alpha-2b. Am J Hematol. 2015;90(4):288–94.CrossRefPubMedPubMedCentral
28.
go back to reference Gisslinger H, Zagrijtschuk O, Buxhofer-Ausch V, Thaler J, Schloegl E, Gastl GA, et al. Ropeginterferon alfa-2b, a novel IFNα-2b, induces high response rates with low toxicity in patients with polycythemia vera. Blood J Am Soc Hematol. 2015;126(15):1762–9. Gisslinger H, Zagrijtschuk O, Buxhofer-Ausch V, Thaler J, Schloegl E, Gastl GA, et al. Ropeginterferon alfa-2b, a novel IFNα-2b, induces high response rates with low toxicity in patients with polycythemia vera. Blood J Am Soc Hematol. 2015;126(15):1762–9.
29.
go back to reference Tiziano Barbui AMV, Valerio De Stefano, Arianna M, Alessandra C, Arianna G, Fabio C, Massimiliano B, Alessandra I, Francesca P, Giulia B, Fabrizio P, Alessandra R, Giuseppe C, Marianna C, Davide R, Caterina M, Sergio S, Elisa R, Andrea P, Nicola C, Barbara M, Emma C, Giuseppe GL, Paola G, Elena R, Silvia B, Francesca L, Luigi S, Cristina B, Nicola V, Marta B, Alessandro R. Phase II Randomized clinical trial comparing ropeginterferon versus phlebotomy in low-risk patients with polycythemia Vera. Results of the Pre-planned Interim Analysis. EHA. EHA Library. Barbui T. 06/14/20; 303391; LB26022020. Tiziano Barbui AMV, Valerio De Stefano, Arianna M, Alessandra C, Arianna G, Fabio C, Massimiliano B, Alessandra I, Francesca P, Giulia B, Fabrizio P, Alessandra R, Giuseppe C, Marianna C, Davide R, Caterina M, Sergio S, Elisa R, Andrea P, Nicola C, Barbara M, Emma C, Giuseppe GL, Paola G, Elena R, Silvia B, Francesca L, Luigi S, Cristina B, Nicola V, Marta B, Alessandro R. Phase II Randomized clinical trial comparing ropeginterferon versus phlebotomy in low-risk patients with polycythemia Vera. Results of the Pre-planned Interim Analysis. EHA. EHA Library. Barbui T. 06/14/20; 303391; LB26022020.
30.
go back to reference Mascarenhas J, Kosiorek HE, Prchal JT, Rambaldi A, Berenzon D, Yacoub A, et al. Results of the myeloproliferative neoplasms-research consortium (MPN-RC) 112 randomized trial of pegylated interferon alfa-2a (PEG) versus hydroxyurea (HU) therapy for the treatment of high risk polycythemia vera (PV) and high risk essential thrombocythemia (ET). Blood. 2018;132(Supplement 1):577.CrossRef Mascarenhas J, Kosiorek HE, Prchal JT, Rambaldi A, Berenzon D, Yacoub A, et al. Results of the myeloproliferative neoplasms-research consortium (MPN-RC) 112 randomized trial of pegylated interferon alfa-2a (PEG) versus hydroxyurea (HU) therapy for the treatment of high risk polycythemia vera (PV) and high risk essential thrombocythemia (ET). Blood. 2018;132(Supplement 1):577.CrossRef
31.
go back to reference Gisslinger H, Klade C, Georgiev P, Krochmalczyk D, Gercheva-Kyuchukova L, Egyed M, 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):e196–208.CrossRefPubMed Gisslinger H, Klade C, Georgiev P, Krochmalczyk D, Gercheva-Kyuchukova L, Egyed M, 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):e196–208.CrossRefPubMed
32.
go back to reference Guerini V, Barbui V, Spinelli O, Salvi A, Dellacasa C, Carobbio A, et al. The histone deacetylase inhibitor ITF2357 selectively targets cells bearing mutated JAK2V617F. Leukemia. 2008;22(4):740–7.CrossRefPubMed Guerini V, Barbui V, Spinelli O, Salvi A, Dellacasa C, Carobbio A, et al. The histone deacetylase inhibitor ITF2357 selectively targets cells bearing mutated JAK2V617F. Leukemia. 2008;22(4):740–7.CrossRefPubMed
33.
go back to reference Rambaldi A, Dellacasa CM, Finazzi G, Carobbio A, Ferrari ML, Guglielmelli P, 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):446–55.PubMed Rambaldi A, Dellacasa CM, Finazzi G, Carobbio A, Ferrari ML, Guglielmelli P, 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):446–55.PubMed
34.
go back to reference Finazzi G, Vannucchi AM, Martinelli V, Ruggeri M, Nobile F, Specchia G, et al. A phase II study of Givinostat in combination with hydroxycarbamide in patients with polycythaemia vera unresponsive to hydroxycarbamide monotherapy. Br J Haematol. 2013;161(5):688–94.CrossRefPubMed Finazzi G, Vannucchi AM, Martinelli V, Ruggeri M, Nobile F, Specchia G, et al. A phase II study of Givinostat in combination with hydroxycarbamide in patients with polycythaemia vera unresponsive to hydroxycarbamide monotherapy. Br J Haematol. 2013;161(5):688–94.CrossRefPubMed
35.
go back to reference Rambaldi A, Iurlo A, Vannucchi AM, Noble R, von Bubnoff N, Guarini A, et al. Safety and efficacy of the maximum tolerated dose of givinostat in polycythemia vera: a two-part Phase Ib/II study. Leukemia. 2020;34(8):2234–7.CrossRefPubMedPubMedCentral Rambaldi A, Iurlo A, Vannucchi AM, Noble R, von Bubnoff N, Guarini A, et al. Safety and efficacy of the maximum tolerated dose of givinostat in polycythemia vera: a two-part Phase Ib/II study. Leukemia. 2020;34(8):2234–7.CrossRefPubMedPubMedCentral
36.
go back to reference Lu M, Xia L, Li Y, Wang X, Hoffman R. The orally bioavailable MDM2 antagonist RG7112 and pegylated interferon α 2a target JAK2V617F-positive progenitor and stem cells. Blood. 2014;124(5):771–9.CrossRefPubMedPubMedCentral Lu M, Xia L, Li Y, Wang X, Hoffman R. The orally bioavailable MDM2 antagonist RG7112 and pegylated interferon α 2a target JAK2V617F-positive progenitor and stem cells. Blood. 2014;124(5):771–9.CrossRefPubMedPubMedCentral
37.
go back to reference Mascarenhas J, Lu M, Kosiorek H, Virtgaym E, Xia L, Sandy L, et al. Oral idasanutlin in patients with polycythemia vera. Blood J Am Soc Hematol. 2019;134(6):525–33. Mascarenhas J, Lu M, Kosiorek H, Virtgaym E, Xia L, Sandy L, et al. Oral idasanutlin in patients with polycythemia vera. Blood J Am Soc Hematol. 2019;134(6):525–33.
38.
go back to reference Niebel D, Kirfel J, Janzen V, Höller T, Majores M, Gütgemann I. Lysine-specific demethylase 1 (LSD1) in hematopoietic and lymphoid neoplasms. Blood. 2014;124(1):151–2.CrossRefPubMed Niebel D, Kirfel J, Janzen V, Höller T, Majores M, Gütgemann I. Lysine-specific demethylase 1 (LSD1) in hematopoietic and lymphoid neoplasms. Blood. 2014;124(1):151–2.CrossRefPubMed
39.
go back to reference Jutzi JS, Kleppe M, Dias J, Staehle HF, Shank K, Teruya-Feldstein J, et al. LSD1 inhibition prolongs survival in mouse models of MPN by selectively targeting the disease clone. HemaSphere. 2018;2(3):e54.CrossRefPubMedPubMedCentral Jutzi JS, Kleppe M, Dias J, Staehle HF, Shank K, Teruya-Feldstein J, et al. LSD1 inhibition prolongs survival in mouse models of MPN by selectively targeting the disease clone. HemaSphere. 2018;2(3):e54.CrossRefPubMedPubMedCentral
40.
go back to reference Theocharides AP, Lundberg P, Lakkaraju AK, Lysenko V, Myburgh R, Aguzzi A, et al. Homozygous calreticulin mutations in patients with myelofibrosis lead to acquired myeloperoxidase deficiency. Blood J Am Soc Hematol. 2016;127(25):3253–9. Theocharides AP, Lundberg P, Lakkaraju AK, Lysenko V, Myburgh R, Aguzzi A, et al. Homozygous calreticulin mutations in patients with myelofibrosis lead to acquired myeloperoxidase deficiency. Blood J Am Soc Hematol. 2016;127(25):3253–9.
41.
go back to reference Elf S, Abdelfattah NS, Chen E, Perales-Patón J, Rosen EA, Ko A, et al. Mutant calreticulin requires both its mutant C-terminus and the thrombopoietin receptor for oncogenic transformation. Cancer Discov. 2016;6(4):368–81.CrossRefPubMedPubMedCentral Elf S, Abdelfattah NS, Chen E, Perales-Patón J, Rosen EA, Ko A, et al. Mutant calreticulin requires both its mutant C-terminus and the thrombopoietin receptor for oncogenic transformation. Cancer Discov. 2016;6(4):368–81.CrossRefPubMedPubMedCentral
42.
go back to reference Holmström MO, Ahmad SM, Klausen U, Bendtsen SK, Martinenaite E, Riley CH, et al. High frequencies of circulating memory T cells specific for calreticulin exon 9 mutations in healthy individuals. Blood Cancer J. 2019;9(2):1–14.CrossRef Holmström MO, Ahmad SM, Klausen U, Bendtsen SK, Martinenaite E, Riley CH, et al. High frequencies of circulating memory T cells specific for calreticulin exon 9 mutations in healthy individuals. Blood Cancer J. 2019;9(2):1–14.CrossRef
43.
go back to reference Holmström M, Martinenaite E, Ahmad S, Met Ö, Friese C, Kjaer L, et al. The calreticulin (CALR) exon 9 mutations are promising targets for cancer immune therapy. Leukemia. 2018;32(2):429–37.CrossRefPubMed Holmström M, Martinenaite E, Ahmad S, Met Ö, Friese C, Kjaer L, et al. The calreticulin (CALR) exon 9 mutations are promising targets for cancer immune therapy. Leukemia. 2018;32(2):429–37.CrossRefPubMed
44.
go back to reference Cimen Bozkus C, Roudko V, Finnigan JP, Mascarenhas J, Hoffman R, Iancu-Rubin C, et al. Immune Checkpoint Blockade Enhances Shared Neoantigen-Induced T-cell Immunity Directed against Mutated Calreticulin in Myeloproliferative Neoplasms. Cancer Discov. 2019;9(9):1192–207.CrossRefPubMed Cimen Bozkus C, Roudko V, Finnigan JP, Mascarenhas J, Hoffman R, Iancu-Rubin C, et al. Immune Checkpoint Blockade Enhances Shared Neoantigen-Induced T-cell Immunity Directed against Mutated Calreticulin in Myeloproliferative Neoplasms. Cancer Discov. 2019;9(9):1192–207.CrossRefPubMed
45.
go back to reference Verstovsek S, Mesa RA, Gotlib J, Levy RS, Gupta V, DiPersio JF, et al. A double-blind, placebo-controlled trial of ruxolitinib for myelofibrosis. N Engl J Med. 2012;366(9):799–807.CrossRefPubMedPubMedCentral Verstovsek S, Mesa RA, Gotlib J, Levy RS, Gupta V, DiPersio JF, et al. A double-blind, placebo-controlled trial of ruxolitinib for myelofibrosis. N Engl J Med. 2012;366(9):799–807.CrossRefPubMedPubMedCentral
46.
go back to reference Harrison CN, Schaap N, Vannucchi AM, Kiladjian JJ, Tiu RV, Zachee P, 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.CrossRefPubMed Harrison CN, Schaap N, Vannucchi AM, Kiladjian JJ, Tiu RV, Zachee P, 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.CrossRefPubMed
47.
go back to reference Newberry KJ, Patel K, Masarova L, Luthra R, Manshouri T, Jabbour E, et al. Clonal evolution and outcomes in myelofibrosis after ruxolitinib discontinuation. Blood J Am Soc Hematol. 2017;130(9):1125–31. Newberry KJ, Patel K, Masarova L, Luthra R, Manshouri T, Jabbour E, et al. Clonal evolution and outcomes in myelofibrosis after ruxolitinib discontinuation. Blood J Am Soc Hematol. 2017;130(9):1125–31.
48.
go back to reference Verstovsek S, Gotlib J, Mesa RA, Vannucchi AM, Kiladjian J-J, Cervantes F, 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.CrossRefPubMedPubMedCentral Verstovsek S, Gotlib J, Mesa RA, Vannucchi AM, Kiladjian J-J, Cervantes F, 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.CrossRefPubMedPubMedCentral
49.
go back to reference Kuykendall AT, Shah S, Talati C, Al Ali N, Sweet K, Padron E, et al. Between a rux and a hard place: evaluating salvage treatment and outcomes in myelofibrosis after ruxolitinib discontinuation. Ann Hematol. 2018;97(3):435–41.CrossRefPubMed Kuykendall AT, Shah S, Talati C, Al Ali N, Sweet K, Padron E, et al. Between a rux and a hard place: evaluating salvage treatment and outcomes in myelofibrosis after ruxolitinib discontinuation. Ann Hematol. 2018;97(3):435–41.CrossRefPubMed
50.
go back to reference Mesa RA, Vannucchi AM, Mead A, Egyed M, Szoke A, Suvorov 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.CrossRefPubMed Mesa RA, Vannucchi AM, Mead A, Egyed M, Szoke A, Suvorov 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.CrossRefPubMed
51.
go back to reference 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):652–9.CrossRefPubMed 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):652–9.CrossRefPubMed
52.
go back to reference Gerds AT, Savona MR, Scott BL, Talpaz M, Egyed M, Harrison CN, et al. Results of PAC203: a randomized phase 2 dose-finding study and determination of the recommended dose of pacritinib. Blood. 2019;134(1):667.CrossRef Gerds AT, Savona MR, Scott BL, Talpaz M, Egyed M, Harrison CN, et al. Results of PAC203: a randomized phase 2 dose-finding study and determination of the recommended dose of pacritinib. Blood. 2019;134(1):667.CrossRef
53.
go back to reference Harrison CN, Gerds AT, Kiladjian J-J, Döhner K, Buckley SA, Smith JA, et al. Pacifica: a randomized, controlled phase 3 study of pacritinib vs. Physician's choice in patients with primary myelofibrosis, post polycythemia vera myelofibrosis, or post essential thrombocytopenia myelofibrosis with severe thrombocytopenia (platelet count <50,000/mL). Blood 2019;134(Supplement_1):4175. Harrison CN, Gerds AT, Kiladjian J-J, Döhner K, Buckley SA, Smith JA, et al. Pacifica: a randomized, controlled phase 3 study of pacritinib vs. Physician's choice in patients with primary myelofibrosis, post polycythemia vera myelofibrosis, or post essential thrombocytopenia myelofibrosis with severe thrombocytopenia (platelet count <50,000/mL). Blood 2019;134(Supplement_1):4175.
54.
go back to reference Mascarenhas JO, Talpaz M, Gupta V, Foltz LM, Savona MR, Paquette R, et al. Primary analysis of a phase II open-label trial of INCB039110, a selective JAK1 inhibitor, in patients with myelofibrosis. Haematologica. 2017;102(2):327–35.CrossRefPubMedPubMedCentral Mascarenhas JO, Talpaz M, Gupta V, Foltz LM, Savona MR, Paquette R, et al. Primary analysis of a phase II open-label trial of INCB039110, a selective JAK1 inhibitor, in patients with myelofibrosis. Haematologica. 2017;102(2):327–35.CrossRefPubMedPubMedCentral
55.
go back to reference Masarova L, Verstovsek S, Hidalgo-Lopez JE, Pemmaraju N, Bose P, Estrov Z, et al. A phase 2 study of ruxolitinib in combination with azacitidine in patients with myelofibrosis. Blood. 2018;132(16):1664–74.CrossRefPubMedPubMedCentral Masarova L, Verstovsek S, Hidalgo-Lopez JE, Pemmaraju N, Bose P, Estrov Z, et al. A phase 2 study of ruxolitinib in combination with azacitidine in patients with myelofibrosis. Blood. 2018;132(16):1664–74.CrossRefPubMedPubMedCentral
56.
go back to reference Mascarenhas J, Ch AP, Td FPRR, et al. CPI-0610, a bromodomain and extraterminal domain protein (BET) inhibitor, in combination with ruxolitinib. In JAK inhibitor treatment naïve myelofibrosis patients: update from manifest phase 2 study. EHA25; Virtual2020. Mascarenhas J, Ch AP, Td FPRR, et al. CPI-0610, a bromodomain and extraterminal domain protein (BET) inhibitor, in combination with ruxolitinib. In JAK inhibitor treatment naïve myelofibrosis patients: update from manifest phase 2 study. EHA25; Virtual2020.
57.
go back to reference Verstovsek S, JM, MK, RH, RR, VG, et al. CPI-0610, Bromodomain and extraterminal domain protein (BET) inhibitor, as 'add-on' to ruxolitinib (RUX). In: Advanced myelofibrosis patients with suboptimal response: update of manifest phase 2 study. EHA25; Virtual2020. Verstovsek S, JM, MK, RH, RR, VG, et al. CPI-0610, Bromodomain and extraterminal domain protein (BET) inhibitor, as 'add-on' to ruxolitinib (RUX). In: Advanced myelofibrosis patients with suboptimal response: update of manifest phase 2 study. EHA25; Virtual2020.
58.
go back to reference Moyo T, JP, YH, OO, SM, RC, et al. Resurrecting response to ruxolitinib: a phase I study testing the combination of ruxolitinib and the PI3K delta inhibitor umbralisib in ruxolitinib-experienced myelofibrosis. EHA2018. Moyo T, JP, YH, OO, SM, RC, et al. Resurrecting response to ruxolitinib: a phase I study testing the combination of ruxolitinib and the PI3K delta inhibitor umbralisib in ruxolitinib-experienced myelofibrosis. EHA2018.
59.
go back to reference Yacoub A, ESW, RKR, UB, MK, HA, et al. Addition of parsaclisib, a PI3KDELTA inhibitor. In: Patients (PTS) with suboptimal response to ruxolitinib (RUX): a phase 2 study in PTS with myelofibrosis (MF). EHA 6/12/202020. Yacoub A, ESW, RKR, UB, MK, HA, et al. Addition of parsaclisib, a PI3KDELTA inhibitor. In: Patients (PTS) with suboptimal response to ruxolitinib (RUX): a phase 2 study in PTS with myelofibrosis (MF). EHA 6/12/202020.
60.
go back to reference Harrison CN, Garcia JS, Mesa RA, Somervaille TC, Komrokji RS, Pemmaraju N, et al. Results from a phase 2 study of navitoclax in combination with ruxolitinib in patients with primary or secondary myelofibrosis. Blood. 2019;134(Supplement_1):671.CrossRef Harrison CN, Garcia JS, Mesa RA, Somervaille TC, Komrokji RS, Pemmaraju N, et al. Results from a phase 2 study of navitoclax in combination with ruxolitinib in patients with primary or secondary myelofibrosis. Blood. 2019;134(Supplement_1):671.CrossRef
61.
go back to reference Kiladjian J-J, Soret-Dulphy J, Resche-Rigon M, Boyer-Perrard F, Barraco F, Rolland-Neyret V, et al. Ruxopeg, a multi-center bayesian phase 1/2 adaptive randomized trial of the combination of ruxolitinib and pegylated interferon alpha 2a in patients with myeloproliferative neoplasm (MPN)-associated myelofibrosis. Blood. 2018;132(Supplement_1):581.CrossRef Kiladjian J-J, Soret-Dulphy J, Resche-Rigon M, Boyer-Perrard F, Barraco F, Rolland-Neyret V, et al. Ruxopeg, a multi-center bayesian phase 1/2 adaptive randomized trial of the combination of ruxolitinib and pegylated interferon alpha 2a in patients with myeloproliferative neoplasm (MPN)-associated myelofibrosis. Blood. 2018;132(Supplement_1):581.CrossRef
62.
go back to reference Verstovsek S, Mesa RA, Gotlib J, Gupta V, DiPersio JF, Catalano JV, 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.CrossRefPubMedPubMedCentral Verstovsek S, Mesa RA, Gotlib J, Gupta V, DiPersio JF, Catalano JV, 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.CrossRefPubMedPubMedCentral
63.
go back to reference Tefferi A, Cervantes F, Mesa R, Passamonti F, Verstovsek S, Vannucchi AM, et al. Revised response criteria for myelofibrosis: International Working Group-Myeloproliferative Neoplasms Research and Treatment (IWG-MRT) and European LeukemiaNet (ELN) consensus report. Blood. 2013;122(8):1395–8.CrossRefPubMedPubMedCentral Tefferi A, Cervantes F, Mesa R, Passamonti F, Verstovsek S, Vannucchi AM, et al. Revised response criteria for myelofibrosis: International Working Group-Myeloproliferative Neoplasms Research and Treatment (IWG-MRT) and European LeukemiaNet (ELN) consensus report. Blood. 2013;122(8):1395–8.CrossRefPubMedPubMedCentral
64.
go back to reference Mascarenhas J, Mehra M, He J, Potluri R, Loefgren C. Patient characteristics and outcomes after ruxolitinib discontinuation in patients with myelofibrosis. J Med Econ. 2020;23(7):721–7.CrossRefPubMed Mascarenhas J, Mehra M, He J, Potluri R, Loefgren C. Patient characteristics and outcomes after ruxolitinib discontinuation in patients with myelofibrosis. J Med Econ. 2020;23(7):721–7.CrossRefPubMed
65.
go back to reference Harrison CN, Schaap N, Vannucchi AM, Kiladjian J-J, Jourdan E, Silver RT, 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.CrossRefPubMedPubMedCentral Harrison CN, Schaap N, Vannucchi AM, Kiladjian J-J, Jourdan E, Silver RT, 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.CrossRefPubMedPubMedCentral
66.
go back to reference Asshoff M, Petzer V, Warr MR, Haschka D, Tymoszuk P, Demetz E, et al. Momelotinib inhibits ACVR1/ALK2, decreases hepcidin production, and ameliorates anemia of chronic disease in rodents. Blood. 2017;129(13):1823–30.CrossRefPubMedPubMedCentral Asshoff M, Petzer V, Warr MR, Haschka D, Tymoszuk P, Demetz E, et al. Momelotinib inhibits ACVR1/ALK2, decreases hepcidin production, and ameliorates anemia of chronic disease in rodents. Blood. 2017;129(13):1823–30.CrossRefPubMedPubMedCentral
67.
go back to reference Mesa RA, Kiladjian JJ, Catalano JV, Devos T, Egyed M, Hellmann A, et al. SIMPLIFY-1: a phase III randomized trial of momelotinib versus ruxolitinib in Janus kinase inhibitor-Naïve patients with myelofibrosis. J Clin Oncol. 2017;35(34):3844–50.CrossRefPubMedPubMedCentral Mesa RA, Kiladjian JJ, Catalano JV, Devos T, Egyed M, Hellmann A, et al. SIMPLIFY-1: a phase III randomized trial of momelotinib versus ruxolitinib in Janus kinase inhibitor-Naïve patients with myelofibrosis. J Clin Oncol. 2017;35(34):3844–50.CrossRefPubMedPubMedCentral
68.
go back to reference Harrison CN, Vannucchi AM, Platzbecker U, Cervantes F, Gupta V, Lavie D, 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.CrossRefPubMed Harrison CN, Vannucchi AM, Platzbecker U, Cervantes F, Gupta V, Lavie D, 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.CrossRefPubMed
69.
go back to reference Oh ST, Talpaz M, Gerds AT, Gupta V, Verstovsek S, Mesa RA, et al. Hepcidin suppression by momelotinib is associated with increased iron availability and erythropoiesis in transfusion-dependent myelofibrosis patients. Blood. 2018;132(Supplement 1):4282.CrossRef Oh ST, Talpaz M, Gerds AT, Gupta V, Verstovsek S, Mesa RA, et al. Hepcidin suppression by momelotinib is associated with increased iron availability and erythropoiesis in transfusion-dependent myelofibrosis patients. Blood. 2018;132(Supplement 1):4282.CrossRef
70.
go back to reference Verstovsek S, Manshouri T, Pilling D, Bueso-Ramos CE, Newberry KJ, Prijic S, et al. Role of neoplastic monocyte-derived fibrocytes in primary myelofibrosis. J Exp Med. 2016;213(9):1723–40.CrossRefPubMedPubMedCentral Verstovsek S, Manshouri T, Pilling D, Bueso-Ramos CE, Newberry KJ, Prijic S, et al. Role of neoplastic monocyte-derived fibrocytes in primary myelofibrosis. J Exp Med. 2016;213(9):1723–40.CrossRefPubMedPubMedCentral
71.
go back to reference Verstovsek S, Hasserjian RP, Pozdnyakova O, Veletic I, Mesa RA, Foltz L, et al. PRM-151 in myelofibrosis: efficacy and safety in an open label extension study. Blood. 2018;132(Supplement 1):686.CrossRef Verstovsek S, Hasserjian RP, Pozdnyakova O, Veletic I, Mesa RA, Foltz L, et al. PRM-151 in myelofibrosis: efficacy and safety in an open label extension study. Blood. 2018;132(Supplement 1):686.CrossRef
72.
go back to reference Verstovsek S, MT, MW, JP, AI, PtB, 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. EHA242019. Verstovsek S, MT, MW, JP, AI, PtB, 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. EHA242019.
73.
go back to reference Pettit K, Gerds AT, Yacoub A, Watts JM, Tartaczuch M, Bradley TJ, et al. A phase 2a study of the LSD1 inhibitor img-7289 (bomedemstat) for the treatment of myelofibrosis. Blood. 2019;134(Supplement_1):556.CrossRef Pettit K, Gerds AT, Yacoub A, Watts JM, Tartaczuch M, Bradley TJ, et al. A phase 2a study of the LSD1 inhibitor img-7289 (bomedemstat) for the treatment of myelofibrosis. Blood. 2019;134(Supplement_1):556.CrossRef
74.
go back to reference Garcia-Delgado R, McLornan DP, Rejtő L, Jourdan E, Al-Ali HK, Pluta A, et al. An open-label, phase 2 study of KRT-232, a first-in-class, oral small molecule inhibitor of MDM2, for the treatment of patients with myelofibrosis (mf) who have previously received treatment with a JAK inhibitor. Blood. 2019;134(Supplement_1):2945.CrossRef Garcia-Delgado R, McLornan DP, Rejtő L, Jourdan E, Al-Ali HK, Pluta A, et al. An open-label, phase 2 study of KRT-232, a first-in-class, oral small molecule inhibitor of MDM2, for the treatment of patients with myelofibrosis (mf) who have previously received treatment with a JAK inhibitor. Blood. 2019;134(Supplement_1):2945.CrossRef
75.
go back to reference Al-Ali HK, RGD, AL, AP, DPM, PV, et al. KRT‑232, A first-in-class, murine double minute 2 inhibitor (MDM2I), for myelofibrosis (MF) relapsed or refractory (R/R) to Janus-associated kinase inhibitor (JAKI) Treatment (TX); 2020. Al-Ali HK, RGD, AL, AP, DPM, PV, et al. KRT‑232, A first-in-class, murine double minute 2 inhibitor (MDM2I), for myelofibrosis (MF) relapsed or refractory (R/R) to Janus-associated kinase inhibitor (JAKI) Treatment (TX); 2020.
76.
go back to reference Pemmaraju N, Carter BZ, Kantarjian HM, Cortes JE, Bose P, Kadia TM, et al. Final results of phase 2 clinical trial of LCL161, a novel oral SMAC mimetic/IAP antagonist, for patients with intermediate to high risk myelofibrosis. Blood. 2019;134(Supplement_1):555.CrossRef Pemmaraju N, Carter BZ, Kantarjian HM, Cortes JE, Bose P, Kadia TM, et al. Final results of phase 2 clinical trial of LCL161, a novel oral SMAC mimetic/IAP antagonist, for patients with intermediate to high risk myelofibrosis. Blood. 2019;134(Supplement_1):555.CrossRef
77.
go back to reference Gangat N, Marinaccio C, Swords R, Watts JM, Gurbuxani S, Rademaker A, Fought AJ, Frankfurt O, Altman JK, Wen QJ, Farnoud N, Famulare CA, Patel A, Tapia R, Vallapureddy RR, Barath S, Graf A, Handlogten A, Zblewski D, Patnaik MM, Al-kali A, Dinh YT, Prahl KE, Patel S, Nobrega JC, Tejera D, Thomassen A, Gao J, Ji P, Rampal RK, Giles FJ, Tefferi A, Stein B, Crispino JD. Aurora kinase a inhibition provides clinical benefit, normalizes megakaryocytes, and reduces bone marrow fibrosis in patients with myelofibrosis: a phase I trial. Clin Cancer Res. 2019;25(16):4898–906.CrossRefPubMed Gangat N, Marinaccio C, Swords R, Watts JM, Gurbuxani S, Rademaker A, Fought AJ, Frankfurt O, Altman JK, Wen QJ, Farnoud N, Famulare CA, Patel A, Tapia R, Vallapureddy RR, Barath S, Graf A, Handlogten A, Zblewski D, Patnaik MM, Al-kali A, Dinh YT, Prahl KE, Patel S, Nobrega JC, Tejera D, Thomassen A, Gao J, Ji P, Rampal RK, Giles FJ, Tefferi A, Stein B, Crispino JD. Aurora kinase a inhibition provides clinical benefit, normalizes megakaryocytes, and reduces bone marrow fibrosis in patients with myelofibrosis: a phase I trial. Clin Cancer Res. 2019;25(16):4898–906.CrossRefPubMed
78.
go back to reference Pemmaraju N, Lane AA, Sweet KL, Stein AS, Vasu S, Blum W, Rizzieri DA, Wang ES, Madeleine Duvic J, Sloan M, Spence S, Shemesh S, Brooks CL, Balser J, Bergstein I, Lancet JE, Kantarjian HM, Konopleva M. Tagraxofusp in blastic plasmacytoid dendritic-cell neoplasm. N Engl J Med. 2019;380(17):1628–37.CrossRefPubMed Pemmaraju N, Lane AA, Sweet KL, Stein AS, Vasu S, Blum W, Rizzieri DA, Wang ES, Madeleine Duvic J, Sloan M, Spence S, Shemesh S, Brooks CL, Balser J, Bergstein I, Lancet JE, Kantarjian HM, Konopleva M. Tagraxofusp in blastic plasmacytoid dendritic-cell neoplasm. N Engl J Med. 2019;380(17):1628–37.CrossRefPubMed
79.
go back to reference Pemmaraju N, Gupta V, Ali H, Yacoub A, Wang ES, Lee S, et al. Results from a phase 1/2 clinical trial of tagraxofusp (SL-401) in patients with intermediate, or high risk, relapsed/refractory myelofibrosis. Blood. 2019;134(Supplement_1):558.CrossRef Pemmaraju N, Gupta V, Ali H, Yacoub A, Wang ES, Lee S, et al. Results from a phase 1/2 clinical trial of tagraxofusp (SL-401) in patients with intermediate, or high risk, relapsed/refractory myelofibrosis. Blood. 2019;134(Supplement_1):558.CrossRef
80.
go back to reference 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:e366.CrossRefPubMedPubMedCentral 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:e366.CrossRefPubMedPubMedCentral
81.
go back to reference Mascarenhas J, Komrokji RS, Cavo M, Martino B, Niederwieser D, Reiter A, 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.CrossRef Mascarenhas J, Komrokji RS, Cavo M, Martino B, Niederwieser D, Reiter A, 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.CrossRef
82.
go back to reference Mascarenhas J, RSK, BM, DN, AR, BLS, et al. Telomerase activity, telomere length and htert expression correlate with clinical outcomes in higher-risk myelofibrosis (MF) relapsed/refractory (R/R) to Janus kinase inhibitor treated with Imetelstat; 2020. Mascarenhas J, RSK, BM, DN, AR, BLS, et al. Telomerase activity, telomere length and htert expression correlate with clinical outcomes in higher-risk myelofibrosis (MF) relapsed/refractory (R/R) to Janus kinase inhibitor treated with Imetelstat; 2020.
83.
go back to reference Kuykendall A, Wan Y, Mascarenhas J, Kiladjian J-J, Vannucchi A, Wang J, et al. PS1456 favorable overall survival of imetelstat-treated relapsed/refractory myelofibrosis patients compared with closely matched real world data. HemaSphere. 2019;3(S1):669–70.CrossRef Kuykendall A, Wan Y, Mascarenhas J, Kiladjian J-J, Vannucchi A, Wang J, et al. PS1456 favorable overall survival of imetelstat-treated relapsed/refractory myelofibrosis patients compared with closely matched real world data. HemaSphere. 2019;3(S1):669–70.CrossRef
85.
go back to reference Stegelmann F, Koschmieder S, Isfort S, Hochhaus A, Heidel F, Hebart H, et al. S1608 Ruxolitinib plus pomalidomide in myelofibrosis with Anemia: results from the MPNSG-0212 combination trial (NCT01644110). HemaSphere. 2019;3(S1):740–1.CrossRef Stegelmann F, Koschmieder S, Isfort S, Hochhaus A, Heidel F, Hebart H, et al. S1608 Ruxolitinib plus pomalidomide in myelofibrosis with Anemia: results from the MPNSG-0212 combination trial (NCT01644110). HemaSphere. 2019;3(S1):740–1.CrossRef
86.
go back to reference Iancu-Rubin C, Mosoyan G, Wang J, Kraus T, Sung V, Hoffman R. Stromal cell-mediated inhibition of erythropoiesis can be attenuated by Sotatercept (ACE-011), an activin receptor type II ligand trap. Exp Hematol. 2013;41(2):155–66.CrossRefPubMed Iancu-Rubin C, Mosoyan G, Wang J, Kraus T, Sung V, Hoffman R. Stromal cell-mediated inhibition of erythropoiesis can be attenuated by Sotatercept (ACE-011), an activin receptor type II ligand trap. Exp Hematol. 2013;41(2):155–66.CrossRefPubMed
87.
go back to reference Carrancio S, Markovics J, Wong P, Leisten J, Castiglioni P, Groza MC, et al. An activin receptor II A ligand trap promotes erythropoiesis resulting in a rapid induction of red blood cells and haemoglobin. Br J Haematol. 2014;165(6):870–82.CrossRefPubMedPubMedCentral Carrancio S, Markovics J, Wong P, Leisten J, Castiglioni P, Groza MC, et al. An activin receptor II A ligand trap promotes erythropoiesis resulting in a rapid induction of red blood cells and haemoglobin. Br J Haematol. 2014;165(6):870–82.CrossRefPubMedPubMedCentral
88.
go back to reference Fenaux P, Platzbecker U, Mufti GJ, Garcia-Manero G, Buckstein R, Santini V, et al. Luspatercept in patients with lower-risk myelodysplastic syndromes. N Engl J Med. 2020;382(2):140–51.CrossRefPubMed Fenaux P, Platzbecker U, Mufti GJ, Garcia-Manero G, Buckstein R, Santini V, et al. Luspatercept in patients with lower-risk myelodysplastic syndromes. N Engl J Med. 2020;382(2):140–51.CrossRefPubMed
89.
go back to reference Bose P, Daver N, Pemmaraju N, Jabbour EJ, Estrov Z, Pike A, et al. Sotatercept (ACE-011) alone and in combination with ruxolitinib in patients (pts) with myeloproliferative neoplasm (MPN)-associated myelofibrosis (MF) and anemia. Blood. 2017;130(Supplement_1):255. Bose P, Daver N, Pemmaraju N, Jabbour EJ, Estrov Z, Pike A, et al. Sotatercept (ACE-011) alone and in combination with ruxolitinib in patients (pts) with myeloproliferative neoplasm (MPN)-associated myelofibrosis (MF) and anemia. Blood. 2017;130(Supplement_1):255.
90.
go back to reference Gerds AT, Vannucchi AM, Passamonti F, Kremyanskaya M, Gotlib JR, Palmer JM, et al. A phase 2 study of luspatercept in patients with myelofibrosis-associated anemia. Blood. 2019;134(Supplement_1):557.CrossRef Gerds AT, Vannucchi AM, Passamonti F, Kremyanskaya M, Gotlib JR, Palmer JM, et al. A phase 2 study of luspatercept in patients with myelofibrosis-associated anemia. Blood. 2019;134(Supplement_1):557.CrossRef
91.
go back to reference Mesa RA, Steensma DP, Pardanani A, Li CY, Elliott M, Kaufmann SH, et al. A phase 2 trial of combination low-dose thalidomide and prednisone for the treatment of myelofibrosis with myeloid metaplasia. Blood. 2003;101(7):2534–41.CrossRefPubMed Mesa RA, Steensma DP, Pardanani A, Li CY, Elliott M, Kaufmann SH, et al. A phase 2 trial of combination low-dose thalidomide and prednisone for the treatment of myelofibrosis with myeloid metaplasia. Blood. 2003;101(7):2534–41.CrossRefPubMed
92.
go back to reference Rampal RK, Verstovsek S, Devlin SM, King AC, Stein EM, Pemmaraju N, et al. Safety and efficacy of combined ruxolitinib and thalidomide in patients with myelofibrosis: a phase II study. Blood. 2019;134(Supplement_1):4163.CrossRef Rampal RK, Verstovsek S, Devlin SM, King AC, Stein EM, Pemmaraju N, et al. Safety and efficacy of combined ruxolitinib and thalidomide in patients with myelofibrosis: a phase II study. Blood. 2019;134(Supplement_1):4163.CrossRef
93.
go back to reference Rampal RK, Maria P-O, Amritha Varshini HS, Levine RL, Cao A. Synergistic therapeutic efficacy of combined JAK1/2, Pan-PIM, and CDK4/6 inhibition in myeloproliferative neoplasms. Blood. 2016;128(22):634.CrossRef Rampal RK, Maria P-O, Amritha Varshini HS, Levine RL, Cao A. Synergistic therapeutic efficacy of combined JAK1/2, Pan-PIM, and CDK4/6 inhibition in myeloproliferative neoplasms. Blood. 2016;128(22):634.CrossRef
94.
go back to reference McKenney AS, Lau AN, Somasundara AVH, Spitzer B, Intlekofer AM, Ahn J, et al. JAK2/IDH-mutant-driven myeloproliferative neoplasm is sensitive to combined targeted inhibition. J Clin Invest. 2018;128(2):789–804.CrossRefPubMedPubMedCentral McKenney AS, Lau AN, Somasundara AVH, Spitzer B, Intlekofer AM, Ahn J, et al. JAK2/IDH-mutant-driven myeloproliferative neoplasm is sensitive to combined targeted inhibition. J Clin Invest. 2018;128(2):789–804.CrossRefPubMedPubMedCentral
95.
go back to reference Pemmaraju N, Gundabolu K, Pettit K, Talpaz M, Podoltsev NA, Schiller GJ, et al. Phase 1b study of the epichaperome inhibitor PU-H71 administered orally with ruxolitinib continuation for the treatment of patients with myelofibrosis. Blood. 2019;134(Supplement_1):4178.CrossRef Pemmaraju N, Gundabolu K, Pettit K, Talpaz M, Podoltsev NA, Schiller GJ, et al. Phase 1b study of the epichaperome inhibitor PU-H71 administered orally with ruxolitinib continuation for the treatment of patients with myelofibrosis. Blood. 2019;134(Supplement_1):4178.CrossRef
96.
go back to reference Wu X, Stenson M, Abeykoon J, Nowakowski K, Zhang L, Lawson J, et al. Targeting glycogen synthase kinase 3 for therapeutic benefit in lymphoma. Blood. 2019;134(4):363–73.CrossRefPubMedPubMedCentral Wu X, Stenson M, Abeykoon J, Nowakowski K, Zhang L, Lawson J, et al. Targeting glycogen synthase kinase 3 for therapeutic benefit in lymphoma. Blood. 2019;134(4):363–73.CrossRefPubMedPubMedCentral
97.
go back to reference Yan D, Pomicter AD, Tantravahi S, Mason CC, Senina AV, Ahmann JM, et al. Nuclear-cytoplasmic transport is a therapeutic target in myelofibrosis. Clin Cancer Res. 2019;25(7):2323–35.CrossRefPubMed Yan D, Pomicter AD, Tantravahi S, Mason CC, Senina AV, Ahmann JM, et al. Nuclear-cytoplasmic transport is a therapeutic target in myelofibrosis. Clin Cancer Res. 2019;25(7):2323–35.CrossRefPubMed
98.
go back to reference Fisher DAC, Miner CA, Engle EK, Hu H, Collins TB, Zhou A, et al. Cytokine production in myelofibrosis exhibits differential responsiveness to JAK-STAT, MAP kinase, and NFκB signaling. Leukemia. 2019;33(8):1978–95.CrossRefPubMedPubMedCentral Fisher DAC, Miner CA, Engle EK, Hu H, Collins TB, Zhou A, et al. Cytokine production in myelofibrosis exhibits differential responsiveness to JAK-STAT, MAP kinase, and NFκB signaling. Leukemia. 2019;33(8):1978–95.CrossRefPubMedPubMedCentral
99.
go back to reference Prestipino A, Emhardt AJ, Aumann K, O’Sullivan D, Gorantla SP, Duquesne S, et al. Oncogenic JAK2V617F causes PD-L1 expression, mediating immune escape in myeloproliferative neoplasms. Sci Transl Med. 2018;10(429):eaam7729.CrossRefPubMedPubMedCentral Prestipino A, Emhardt AJ, Aumann K, O’Sullivan D, Gorantla SP, Duquesne S, et al. Oncogenic JAK2V617F causes PD-L1 expression, mediating immune escape in myeloproliferative neoplasms. Sci Transl Med. 2018;10(429):eaam7729.CrossRefPubMedPubMedCentral
100.
go back to reference Varricchio L, Mascarenhas J, Migliaccio AR, O’Connor-McCourt M, Tremblay G, Denis J-F, et al. AVID200, a potent trap for TGF-β ligands inhibits TGF-β1 signaling in human myelofibrosis. Blood. 2018;132(Supplement 1):1791.CrossRef Varricchio L, Mascarenhas J, Migliaccio AR, O’Connor-McCourt M, Tremblay G, Denis J-F, et al. AVID200, a potent trap for TGF-β ligands inhibits TGF-β1 signaling in human myelofibrosis. Blood. 2018;132(Supplement 1):1791.CrossRef
101.
go back to reference Lu M, Xia L, Hoffman R. A novel combination of drugs which target both the intrinsic and extrinsic apoptotic pathways to eliminate myelofibrosis CD34+ cells. Blood. 2019;134(Supplement_1):4201.CrossRef Lu M, Xia L, Hoffman R. A novel combination of drugs which target both the intrinsic and extrinsic apoptotic pathways to eliminate myelofibrosis CD34+ cells. Blood. 2019;134(Supplement_1):4201.CrossRef
102.
go back to reference Dutta A, Nath D, Yang Y, Mohi G. Abstract 1874: The PIM kinase inhibitor TP-3654 demonstrates efficacy in a murine model of myelofibrosis. Cancer Res. 2018;78(13 Supplement):1874. Dutta A, Nath D, Yang Y, Mohi G. Abstract 1874: The PIM kinase inhibitor TP-3654 demonstrates efficacy in a murine model of myelofibrosis. Cancer Res. 2018;78(13 Supplement):1874.
Metadata
Title
Novel therapeutics in myeloproliferative neoplasms
Authors
Sangeetha Venugopal
John Mascarenhas
Publication date
01-12-2020
Publisher
BioMed Central
Published in
Journal of Hematology & Oncology / Issue 1/2020
Electronic ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-020-00995-y

Other articles of this Issue 1/2020

Journal of Hematology & Oncology 1/2020 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine