Skip to main content
Top
Published in: Current Hematologic Malignancy Reports 4/2013

01-12-2013 | Myeloproliferative Disorders (JJ Kiladjian, Section Editor)

Preclinical Models for Drug Selection in Myeloproliferative Neoplasms

Authors: Niccolò Bartalucci, Costanza Bogani, Alessandro M. Vannucchi

Published in: Current Hematologic Malignancy Reports | Issue 4/2013

Login to get access

Abstract

The discovery that an abnormally activated JAK-STAT signaling pathway is central to the pathogenesis of myeloproliferative neoplasms has promoted the clinical development of small-molecule JAK2 inhibitors. These agents have shown remarkable efficacy in disease control, but do not induce molecular remission; on the other hand, interferon holds the promise to target the putative hematopoietic progenitor cell initiating the disease. The presence of additional molecular abnormalities indicates a high molecular complexity of myeloproliferative neoplasms, and the need for simultaneously targeting different targets. Several drugs are currently under study as single agents and in combination. This review briefly describes the several in vitro and in vivo models of myeloproliferative neoplasms that are being used as preclinical models for drug development.
Literature
1.
go back to reference Tefferi A, Thiele J, Orazi A, et al. Proposals and rationale for revision of the World Health Organization diagnostic criteria for polycythemia vera, essential thrombocythemia, and primary myelofibrosis: recommendations from an ad hoc international expert panel. Blood. 2007;110:1092–7.PubMedCrossRef Tefferi A, Thiele J, Orazi A, et al. Proposals and rationale for revision of the World Health Organization diagnostic criteria for polycythemia vera, essential thrombocythemia, and primary myelofibrosis: recommendations from an ad hoc international expert panel. Blood. 2007;110:1092–7.PubMedCrossRef
2.
go back to reference Swerdlow SH, Campo E, Harris NL, et al. WHO classification of tumors of haematopoietic and lymphoid tissues. Lyon: IARC; 2008. Swerdlow SH, Campo E, Harris NL, et al. WHO classification of tumors of haematopoietic and lymphoid tissues. Lyon: IARC; 2008.
3.
go back to reference Dameshek W. Some speculations on the myeloproliferative syndromes. Blood. 1951;6:372–5.PubMed Dameshek W. Some speculations on the myeloproliferative syndromes. Blood. 1951;6:372–5.PubMed
4.
go back to reference James C, Ugo V, Le Couedic JP, et al. A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. Nature. 2005;434:1144–8.PubMedCrossRef James C, Ugo V, Le Couedic JP, et al. A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. Nature. 2005;434:1144–8.PubMedCrossRef
5.
go back to reference Baxter EJ, Scott LM, Campbell PJ, et al. Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet. 2005;365:1054–61.PubMed Baxter EJ, Scott LM, Campbell PJ, et al. Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet. 2005;365:1054–61.PubMed
6.
go back to reference Levine RL, Wadleigh M, Cools J, et al. Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. Cancer Cell. 2005;7:387–97.PubMedCrossRef Levine RL, Wadleigh M, Cools J, et al. Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. Cancer Cell. 2005;7:387–97.PubMedCrossRef
7.
go back to reference Kralovics R, Passamonti F, Buser AS, et al. A gain-of-function mutation of JAK2 in myeloproliferative disorders. N Engl J Med. 2005;352:1779–90.PubMedCrossRef Kralovics R, Passamonti F, Buser AS, et al. A gain-of-function mutation of JAK2 in myeloproliferative disorders. N Engl J Med. 2005;352:1779–90.PubMedCrossRef
9.
go back to reference •• Vainchenker W, Delhommeau F, Constantinescu SN, Bernard OA. New mutations and pathogenesis of myeloproliferative neoplasms. Blood. 2011;118:1723–35. An excellent review highligting the mutationla complexity of MPN.PubMedCrossRef •• Vainchenker W, Delhommeau F, Constantinescu SN, Bernard OA. New mutations and pathogenesis of myeloproliferative neoplasms. Blood. 2011;118:1723–35. An excellent review highligting the mutationla complexity of MPN.PubMedCrossRef
10.
go back to reference •• Verstovsek S, Mesa RA, Gotlib J, et al. A double-blind, placebo-controlled trial of ruxolitinib for myelofibrosis. New Engl J Med. 2012;366:799–807. One of two phase III studies demonstrating the clinical efficacy of the anti-JAK1 and JAK2 inhibitor ruxolitinib in MF.PubMedCrossRef •• Verstovsek S, Mesa RA, Gotlib J, et al. A double-blind, placebo-controlled trial of ruxolitinib for myelofibrosis. New Engl J Med. 2012;366:799–807. One of two phase III studies demonstrating the clinical efficacy of the anti-JAK1 and JAK2 inhibitor ruxolitinib in MF.PubMedCrossRef
11.
go back to reference •• Harrison C, Kiladjian JJ, Al-Ali HK, et al. JAK inhibition with ruxolitinib versus best available therapy for myelofibrosis. N Engl J Med. 2012;366:787–98. One of two phase III studies demonstrating the clinical efficacy of the anti-JAK1 and JAK2 inhibitor ruxolitinib in MF.PubMedCrossRef •• Harrison C, Kiladjian JJ, Al-Ali HK, et al. JAK inhibition with ruxolitinib versus best available therapy for myelofibrosis. N Engl J Med. 2012;366:787–98. One of two phase III studies demonstrating the clinical efficacy of the anti-JAK1 and JAK2 inhibitor ruxolitinib in MF.PubMedCrossRef
12.
go back to reference Quintás-Cardama A, Kantarjian H, Cortes J, Verstovsek S. Janus kinase inhibitors for the treatment of myeloproliferative neoplasias and beyond. Nat Rev Drug Discov. 2011;10:127–40.PubMedCrossRef Quintás-Cardama A, Kantarjian H, Cortes J, Verstovsek S. Janus kinase inhibitors for the treatment of myeloproliferative neoplasias and beyond. Nat Rev Drug Discov. 2011;10:127–40.PubMedCrossRef
13.
go back to reference Vannucchi AM. From palliation to targeted therapy in myelofibrosis. N Engl J Med. 2010;363:1180–2.PubMedCrossRef Vannucchi AM. From palliation to targeted therapy in myelofibrosis. N Engl J Med. 2010;363:1180–2.PubMedCrossRef
14.
go back to reference •• Bogani C, Bartalucci N, Martinelli S, et al. mTOR inhibitors alone and in combination with JAK2 inhibitors effectively inhibit cells of myeloproliferative neoplasms. PLoS ONE. 2013;8:e54826. Evidence using in vitro models of the activity of anti-mTOR compounds in MPN.PubMedCrossRef •• Bogani C, Bartalucci N, Martinelli S, et al. mTOR inhibitors alone and in combination with JAK2 inhibitors effectively inhibit cells of myeloproliferative neoplasms. PLoS ONE. 2013;8:e54826. Evidence using in vitro models of the activity of anti-mTOR compounds in MPN.PubMedCrossRef
15.
go back to reference Barrio S, Gallardo M, Arenas A, et al. Inhibition of related JAK/STAT pathways with molecular targeted drugs shows strong synergy with ruxolitinib in chronic myeloproliferative neoplasm. Br J Haematol. 2013;161:667–76.PubMedCrossRef Barrio S, Gallardo M, Arenas A, et al. Inhibition of related JAK/STAT pathways with molecular targeted drugs shows strong synergy with ruxolitinib in chronic myeloproliferative neoplasm. Br J Haematol. 2013;161:667–76.PubMedCrossRef
16.
go back to reference Amaru Calzada A, Pedrini O, Finazzi G, et al. Givinostat and hydroxyurea synergize in vitro to induce apoptosis of cells from JAK2(V617F) myeloproliferative neoplasm patients. Exp Hematol. 2013;41:253.e2–60.e2.CrossRef Amaru Calzada A, Pedrini O, Finazzi G, et al. Givinostat and hydroxyurea synergize in vitro to induce apoptosis of cells from JAK2(V617F) myeloproliferative neoplasm patients. Exp Hematol. 2013;41:253.e2–60.e2.CrossRef
17.
go back to reference Hart S, Goh KC, Novotny-Diermayr V, et al. SB1518, a novel macrocyclic pyrimidine-based JAK2 inhibitor for the treatment of myeloid and lymphoid malignancies. Leukemia. 2011;25:1751–9.PubMedCrossRef Hart S, Goh KC, Novotny-Diermayr V, et al. SB1518, a novel macrocyclic pyrimidine-based JAK2 inhibitor for the treatment of myeloid and lymphoid malignancies. Leukemia. 2011;25:1751–9.PubMedCrossRef
18.
go back to reference Fleischman AG, Aichberger KJ, Luty SB, et al. TNFalpha facilitates clonal expansion of JAK2V617F positive cells in myeloproliferative neoplasms. Blood. 2011;118:6392–8.PubMedCrossRef Fleischman AG, Aichberger KJ, Luty SB, et al. TNFalpha facilitates clonal expansion of JAK2V617F positive cells in myeloproliferative neoplasms. Blood. 2011;118:6392–8.PubMedCrossRef
19.
go back to reference Nakaya Y, Shide K, Niwa T, et al. Efficacy of NS-018, a potent and selective JAK2/Src inhibitor, in primary cells and mouse models of myeloproliferative neoplasms. Blood Cancer J. 2011;1:e29.PubMedCrossRef Nakaya Y, Shide K, Niwa T, et al. Efficacy of NS-018, a potent and selective JAK2/Src inhibitor, in primary cells and mouse models of myeloproliferative neoplasms. Blood Cancer J. 2011;1:e29.PubMedCrossRef
20.
go back to reference Anand S, Stedham F, Gudgin E, et al. Increased basal intracellular signaling patterns do not correlate with JAK2 genotype in human myeloproliferative neoplasms. Blood. 2011;118:1610–21.PubMedCrossRef Anand S, Stedham F, Gudgin E, et al. Increased basal intracellular signaling patterns do not correlate with JAK2 genotype in human myeloproliferative neoplasms. Blood. 2011;118:1610–21.PubMedCrossRef
21.
go back to reference Vicari L, Martinetti D, Buccheri S, et al. Increased phospho-mTOR expression in megakaryocytic cells derived from CD34+ progenitors of essential thrombocythaemia and myelofibrosis patients. Br J Haematol. 2012;159:237–40.PubMedCrossRef Vicari L, Martinetti D, Buccheri S, et al. Increased phospho-mTOR expression in megakaryocytic cells derived from CD34+ progenitors of essential thrombocythaemia and myelofibrosis patients. Br J Haematol. 2012;159:237–40.PubMedCrossRef
22.
go back to reference Chou T-C. Drug combination studies and their synergy quantification using the Chou-Talalay method. Cancer Res. 2010;70:440–6.PubMedCrossRef Chou T-C. Drug combination studies and their synergy quantification using the Chou-Talalay method. Cancer Res. 2010;70:440–6.PubMedCrossRef
23.
go back to reference Vannucchi AM, Lasho TL, Guglielmelli P, et al. Mutations and prognosis in primary myelofibrosis. Leukemia. 2013;27:1861–9.PubMedCrossRef Vannucchi AM, Lasho TL, Guglielmelli P, et al. Mutations and prognosis in primary myelofibrosis. Leukemia. 2013;27:1861–9.PubMedCrossRef
24.
go back to reference Wernig G, Mercher T, Okabe R, et al. Expression of Jak2V617F causes a polycythemia vera-like disease with associated myelofibrosis in a murine bone marrow transplant model. Blood. 2006;107:4274–81.PubMedCrossRef Wernig G, Mercher T, Okabe R, et al. Expression of Jak2V617F causes a polycythemia vera-like disease with associated myelofibrosis in a murine bone marrow transplant model. Blood. 2006;107:4274–81.PubMedCrossRef
25.
go back to reference Lacout C, Pisani DF, Tulliez M, et al. JAK2V617F expression in murine hematopoietic cells leads to MPD mimicking human PV with secondary myelofibrosis. Blood. 2006;108:1652–60.PubMedCrossRef Lacout C, Pisani DF, Tulliez M, et al. JAK2V617F expression in murine hematopoietic cells leads to MPD mimicking human PV with secondary myelofibrosis. Blood. 2006;108:1652–60.PubMedCrossRef
26.
go back to reference Zaleskas VM, Krause DS, Lazarides K, et al. Molecular pathogenesis and therapy of polycythemia induced in mice by JAK2 V617F. PLoS ONE. 2006;1:e18.PubMedCrossRef Zaleskas VM, Krause DS, Lazarides K, et al. Molecular pathogenesis and therapy of polycythemia induced in mice by JAK2 V617F. PLoS ONE. 2006;1:e18.PubMedCrossRef
27.
go back to reference Bumm TG, Elsea C, Corbin AS, et al. Characterization of murine JAK2V617F-positive myeloproliferative disease. Cancer Res. 2006;66:11156–65.PubMedCrossRef Bumm TG, Elsea C, Corbin AS, et al. Characterization of murine JAK2V617F-positive myeloproliferative disease. Cancer Res. 2006;66:11156–65.PubMedCrossRef
28.
go back to reference Wernig G, Kharas MG, Okabe R, et al. Efficacy of TG101348, a selective JAK2 inhibitor, in treatment of a murine model of JAK2V617F-induced polycythemia vera. Cancer Cell. 2008;13:311–20.PubMedCrossRef Wernig G, Kharas MG, Okabe R, et al. Efficacy of TG101348, a selective JAK2 inhibitor, in treatment of a murine model of JAK2V617F-induced polycythemia vera. Cancer Cell. 2008;13:311–20.PubMedCrossRef
29.
go back to reference Tyner JW, Bumm TG, Deininger J, et al. CYT387, a novel JAK2 inhibitor, induces hematologic responses and normalizes inflammatory cytokines in murine myeloproliferative neoplasms. Blood. 2010;115:5232–40.PubMedCrossRef Tyner JW, Bumm TG, Deininger J, et al. CYT387, a novel JAK2 inhibitor, induces hematologic responses and normalizes inflammatory cytokines in murine myeloproliferative neoplasms. Blood. 2010;115:5232–40.PubMedCrossRef
30.
go back to reference Shide K, Shimoda HK, Kumano T, et al. Development of ET, primary myelofibrosis and PV in mice expressing JAK2 V617F. Leukemia. 2008;22:87–95.PubMedCrossRef Shide K, Shimoda HK, Kumano T, et al. Development of ET, primary myelofibrosis and PV in mice expressing JAK2 V617F. Leukemia. 2008;22:87–95.PubMedCrossRef
31.
go back to reference Xing S, Ho WT, Zhao W, et al. Transgenic expression of JAK2V617F causes myeloproliferative disorders in mice. Blood. 2008;111:5109–17.PubMedCrossRef Xing S, Ho WT, Zhao W, et al. Transgenic expression of JAK2V617F causes myeloproliferative disorders in mice. Blood. 2008;111:5109–17.PubMedCrossRef
32.
go back to reference Tiedt R, Hao-Shen H, Sobas MA, et al. Ratio of mutant JAK2-V617F to wild-type Jak2 determines the MPD phenotypes in transgenic mice. Blood. 2008;111:3931–40.PubMedCrossRef Tiedt R, Hao-Shen H, Sobas MA, et al. Ratio of mutant JAK2-V617F to wild-type Jak2 determines the MPD phenotypes in transgenic mice. Blood. 2008;111:3931–40.PubMedCrossRef
33.
go back to reference •• Kubovcakova L, Lundberg P, Grisouard J, et al. Differential effects of hydroxyurea and INC424 on mutant allele burden and myeloproliferative phenotype in a JAK2-V617F polycythemia vera mouse model. Blood. 2013;121:1188–99. Detailed study of the different mechanisms of action of hydroxyurea and interferon-alpha in a JAK2V617F-mutated animal model of MPN.PubMedCrossRef •• Kubovcakova L, Lundberg P, Grisouard J, et al. Differential effects of hydroxyurea and INC424 on mutant allele burden and myeloproliferative phenotype in a JAK2-V617F polycythemia vera mouse model. Blood. 2013;121:1188–99. Detailed study of the different mechanisms of action of hydroxyurea and interferon-alpha in a JAK2V617F-mutated animal model of MPN.PubMedCrossRef
34.
go back to reference Akada H, Yan D, Zou H, et al. Conditional expression of heterozygous or homozygous Jak2V617F from its endogenous promoter induces a polycythemia vera-like disease. Blood. 2010;115:3589–97.PubMedCrossRef Akada H, Yan D, Zou H, et al. Conditional expression of heterozygous or homozygous Jak2V617F from its endogenous promoter induces a polycythemia vera-like disease. Blood. 2010;115:3589–97.PubMedCrossRef
35.
go back to reference Marty C, Lacout C, Martin A, et al. Myeloproliferative neoplasm induced by constitutive expression of JAK2V617F in knock-in mice. Blood. 2010;116:783–7.PubMedCrossRef Marty C, Lacout C, Martin A, et al. Myeloproliferative neoplasm induced by constitutive expression of JAK2V617F in knock-in mice. Blood. 2010;116:783–7.PubMedCrossRef
36.
go back to reference Mullally A, Lane SW, Ball B, et al. Physiological Jak2V617F expression causes a lethal myeloproliferative neoplasm with differential effects on hematopoietic stem and progenitor cells. Cancer Cell. 2010;17:584–96.PubMedCrossRef Mullally A, Lane SW, Ball B, et al. Physiological Jak2V617F expression causes a lethal myeloproliferative neoplasm with differential effects on hematopoietic stem and progenitor cells. Cancer Cell. 2010;17:584–96.PubMedCrossRef
37.
go back to reference Li J, Spensberger D, Ahn JS, et al. JAK2 V617F impairs hematopoietic stem cell function in a conditional knock-in mouse model of JAK2 V617F-positive essential thrombocythemia. Blood. 2010;116:1528–38.PubMedCrossRef Li J, Spensberger D, Ahn JS, et al. JAK2 V617F impairs hematopoietic stem cell function in a conditional knock-in mouse model of JAK2 V617F-positive essential thrombocythemia. Blood. 2010;116:1528–38.PubMedCrossRef
38.
go back to reference Akada H, Hamada S, Mohi MG. Efficacy of vorinostat in a murine model of polycythemia vera. Blood (ASH Annual Meeting Abstracts). 2010;116 Abstract 629. Akada H, Hamada S, Mohi MG. Efficacy of vorinostat in a murine model of polycythemia vera. Blood (ASH Annual Meeting Abstracts). 2010;116 Abstract 629.
39.
go back to reference •• Mullally A, Bruedigam C, Poveromo L, et al. Depletion of Jak2V617F MPN-propagating stem cells by interferon-alpha in a murine model of polycythemia vera. Blood. 2013;121:3692–702. Evidence using an animal JAK2V617F mutated model that interferon-alpha can target the MPN stem cell.PubMedCrossRef •• Mullally A, Bruedigam C, Poveromo L, et al. Depletion of Jak2V617F MPN-propagating stem cells by interferon-alpha in a murine model of polycythemia vera. Blood. 2013;121:3692–702. Evidence using an animal JAK2V617F mutated model that interferon-alpha can target the MPN stem cell.PubMedCrossRef
40.
go back to reference Ulich TR, del Castillo J, Senaldi G, et al. Systemic hematologic effects of PEG-rHuMGDF-induced megakaryocyte hyperplasia in mice. Blood. 1996;87:5006–15.PubMed Ulich TR, del Castillo J, Senaldi G, et al. Systemic hematologic effects of PEG-rHuMGDF-induced megakaryocyte hyperplasia in mice. Blood. 1996;87:5006–15.PubMed
41.
go back to reference Ohwada A, Rafii S, Moore MA, Crystal RG. In vivo adenovirus vector-mediated transfer of the human thrombopoietin cDNA maintains platelet levels during radiation- and chemotherapy-induced bone marrow suppression. Blood. 1996;88:778–84.PubMed Ohwada A, Rafii S, Moore MA, Crystal RG. In vivo adenovirus vector-mediated transfer of the human thrombopoietin cDNA maintains platelet levels during radiation- and chemotherapy-induced bone marrow suppression. Blood. 1996;88:778–84.PubMed
42.
go back to reference Cannizzo SJ, Frey BM, Raffi S, et al. Augmentation of blood platelet levels by intratracheal administration of an adenovirus vector encoding human thrombopoietin cDNA. Nat Biotechnol. 1997;15:570–3.PubMedCrossRef Cannizzo SJ, Frey BM, Raffi S, et al. Augmentation of blood platelet levels by intratracheal administration of an adenovirus vector encoding human thrombopoietin cDNA. Nat Biotechnol. 1997;15:570–3.PubMedCrossRef
43.
go back to reference Abina MA, Tulliez M, Duffour MT, et al. Thrombopoietin (TPO) knockout phenotype induced by cross-reactive antibodies against TPO following injection of mice with recombinant adenovirus encoding human TPO. J Immunol. 1998;160:4481–9.PubMed Abina MA, Tulliez M, Duffour MT, et al. Thrombopoietin (TPO) knockout phenotype induced by cross-reactive antibodies against TPO following injection of mice with recombinant adenovirus encoding human TPO. J Immunol. 1998;160:4481–9.PubMed
44.
go back to reference Frey BM, Rafii S, Teterson M, et al. Adenovector-mediated expression of human thrombopoietin cDNA in immune-compromised mice: insights into the pathophysiology of osteomyelofibrosis. J Immunol. 1998;160:691–9.PubMed Frey BM, Rafii S, Teterson M, et al. Adenovector-mediated expression of human thrombopoietin cDNA in immune-compromised mice: insights into the pathophysiology of osteomyelofibrosis. J Immunol. 1998;160:691–9.PubMed
45.
go back to reference Yan XQ, Lacey D, Fletcher F, et al. Chronic exposure to retroviral vector encoded MGDF (mpl-ligand) induces lineage-specific growth and differentiation of megakaryocytes in mice. Blood. 1995;86:4025–33.PubMed Yan XQ, Lacey D, Fletcher F, et al. Chronic exposure to retroviral vector encoded MGDF (mpl-ligand) induces lineage-specific growth and differentiation of megakaryocytes in mice. Blood. 1995;86:4025–33.PubMed
46.
go back to reference Villeval JL, Cohen-Solal K, Tulliez M, et al. High thrombopoietin production by hematopoietic cells induces a fatal myeloproliferative syndrome in mice. Blood. 1997;90:4369–83.PubMed Villeval JL, Cohen-Solal K, Tulliez M, et al. High thrombopoietin production by hematopoietic cells induces a fatal myeloproliferative syndrome in mice. Blood. 1997;90:4369–83.PubMed
47.
go back to reference Zhou W, Toombs CF, Zou T, et al. Transgenic mice overexpressing human c-mpl ligand exhibit chronic thrombocytosis and display enhanced recovery from 5-fluorouracil or antiplatelet serum treatment. Blood. 1997;89:1551–9.PubMed Zhou W, Toombs CF, Zou T, et al. Transgenic mice overexpressing human c-mpl ligand exhibit chronic thrombocytosis and display enhanced recovery from 5-fluorouracil or antiplatelet serum treatment. Blood. 1997;89:1551–9.PubMed
48.
go back to reference Kakumitsu H, Kamezaki K, Shimoda K, et al. Transgenic mice overexpressing murine thrombopoietin develop myelofibrosis and osteosclerosis. Leuk Res. 2005;29:761–9.PubMedCrossRef Kakumitsu H, Kamezaki K, Shimoda K, et al. Transgenic mice overexpressing murine thrombopoietin develop myelofibrosis and osteosclerosis. Leuk Res. 2005;29:761–9.PubMedCrossRef
49.
go back to reference Yanagida M, Ide Y, Imai A, et al. The role of transforming growth factor-beta in PEG-rHuMGDF-induced reversible myelofibrosis in rats. Br J Haematol. 1997;99:739–45.PubMedCrossRef Yanagida M, Ide Y, Imai A, et al. The role of transforming growth factor-beta in PEG-rHuMGDF-induced reversible myelofibrosis in rats. Br J Haematol. 1997;99:739–45.PubMedCrossRef
50.
go back to reference Chagraoui H, Komura E, Tulliez M, et al. Prominent role of TGF-beta 1 in thrombopoietin-induced myelofibrosis in mice. Blood. 2002;100:3495–503.PubMedCrossRef Chagraoui H, Komura E, Tulliez M, et al. Prominent role of TGF-beta 1 in thrombopoietin-induced myelofibrosis in mice. Blood. 2002;100:3495–503.PubMedCrossRef
51.
go back to reference Chagraoui H, Tulliez M, Smayra T, et al. Stimulation of osteoprotegerin production is responsible for osteosclerosis in mice overexpressing TPO. Blood. 2003;101:2983–9.PubMedCrossRef Chagraoui H, Tulliez M, Smayra T, et al. Stimulation of osteoprotegerin production is responsible for osteosclerosis in mice overexpressing TPO. Blood. 2003;101:2983–9.PubMedCrossRef
52.
go back to reference Wagner-Ballon O, Pisani DF, Gastinne T, et al. Proteasome inhibitor bortezomib impairs both myelofibrosis and osteosclerosis induced by high thrombopoietin levels in mice. Blood. 2007;110:345–53.PubMedCrossRef Wagner-Ballon O, Pisani DF, Gastinne T, et al. Proteasome inhibitor bortezomib impairs both myelofibrosis and osteosclerosis induced by high thrombopoietin levels in mice. Blood. 2007;110:345–53.PubMedCrossRef
53.
go back to reference Barosi G, Gattoni E, Guglielmelli P, et al. Phase I/II study of single-agent bortezomib for the treatment of patients with myelofibrosis. Clinical and biological effects of proteasome inhibition. Am J Hematol. 2010;85:616–9.PubMedCrossRef Barosi G, Gattoni E, Guglielmelli P, et al. Phase I/II study of single-agent bortezomib for the treatment of patients with myelofibrosis. Clinical and biological effects of proteasome inhibition. Am J Hematol. 2010;85:616–9.PubMedCrossRef
54.
go back to reference McDevitt MA, Fujiwara Y, Shivdasani RA, Orkin SH. An upstream, DNase I hypersensitive region of the hematopoietic-expressed transcription factor GATA-1 gene confers developmental specificity in transgenic mice. Proc Natl Acad Sci U S A. 1997;94:7976–81.PubMedCrossRef McDevitt MA, Fujiwara Y, Shivdasani RA, Orkin SH. An upstream, DNase I hypersensitive region of the hematopoietic-expressed transcription factor GATA-1 gene confers developmental specificity in transgenic mice. Proc Natl Acad Sci U S A. 1997;94:7976–81.PubMedCrossRef
55.
go back to reference McDevitt MA, Shivdasani RA, Fujiwara Y, et al. A "knockdown" mutation created by cis-element gene targeting reveals the dependence of erythroid cell maturation on the level of transcription factor GATA-1. Proc Natl Acad Sci U S A. 1997;94:6781–5.PubMedCrossRef McDevitt MA, Shivdasani RA, Fujiwara Y, et al. A "knockdown" mutation created by cis-element gene targeting reveals the dependence of erythroid cell maturation on the level of transcription factor GATA-1. Proc Natl Acad Sci U S A. 1997;94:6781–5.PubMedCrossRef
56.
go back to reference Vyas P, Ault K, Jackson CW, et al. Consequences of GATA-1 deficiency in megakaryocytes and platelets. Blood. 1999;93:2867–75.PubMed Vyas P, Ault K, Jackson CW, et al. Consequences of GATA-1 deficiency in megakaryocytes and platelets. Blood. 1999;93:2867–75.PubMed
57.
go back to reference Vannucchi AM, Bianchi L, Cellai C, et al. Development of myelofibrosis in mice genetically impaired for GATA-1 expression (GATA-1(low) mice). Blood. 2002;100:1123–32.PubMedCrossRef Vannucchi AM, Bianchi L, Cellai C, et al. Development of myelofibrosis in mice genetically impaired for GATA-1 expression (GATA-1(low) mice). Blood. 2002;100:1123–32.PubMedCrossRef
58.
go back to reference Vannucchi AM, Migliaccio AR, Paoletti F, et al. Pathogenesis of myelofibrosis with myeloid metaplasia: lessons from mouse models of the disease. Semin Oncol. 2005;32:365–72.PubMedCrossRef Vannucchi AM, Migliaccio AR, Paoletti F, et al. Pathogenesis of myelofibrosis with myeloid metaplasia: lessons from mouse models of the disease. Semin Oncol. 2005;32:365–72.PubMedCrossRef
59.
go back to reference Vannucchi AM, Bianchi L, Paoletti F, et al. A pathobiologic pathway linking thrombopoietin, GATA-1, and TGF-beta1 in the development of myelofibrosis. Blood. 2005;105:3493–501.PubMedCrossRef Vannucchi AM, Bianchi L, Paoletti F, et al. A pathobiologic pathway linking thrombopoietin, GATA-1, and TGF-beta1 in the development of myelofibrosis. Blood. 2005;105:3493–501.PubMedCrossRef
60.
go back to reference Martelli F, Ghinassi B, Panetta B, et al. Variegation of the phenotype induced by the Gata1low mutation in mice of different genetic backgrounds. Blood. 2005;106:4102–13.PubMedCrossRef Martelli F, Ghinassi B, Panetta B, et al. Variegation of the phenotype induced by the Gata1low mutation in mice of different genetic backgrounds. Blood. 2005;106:4102–13.PubMedCrossRef
61.
go back to reference Vannucchi AM, Pancrazzi A, Guglielmelli P, et al. Abnormalities of GATA-1 in megakaryocytes from patients with idiopathic myelofibrosis. Am J Pathol. 2005;167:849–58.PubMedCrossRef Vannucchi AM, Pancrazzi A, Guglielmelli P, et al. Abnormalities of GATA-1 in megakaryocytes from patients with idiopathic myelofibrosis. Am J Pathol. 2005;167:849–58.PubMedCrossRef
62.
go back to reference Migliaccio AR, Martelli F, Verrucci M, et al. Altered SDF-1/CXCR4 axis in patients with primary myelofibrosis and in the Gata1(low) mouse model of the disease. Exp Hematol. 2008;36:158–71.PubMedCrossRef Migliaccio AR, Martelli F, Verrucci M, et al. Altered SDF-1/CXCR4 axis in patients with primary myelofibrosis and in the Gata1(low) mouse model of the disease. Exp Hematol. 2008;36:158–71.PubMedCrossRef
63.
go back to reference Rosti V, Massa M, Vannucchi AM, et al. The expression of CXCR4 is down-regulated on the CD34+ cells of patients with myelofibrosis with myeloid metaplasia. Blood Cells Mol Dis. 2007;38:280–6.PubMedCrossRef Rosti V, Massa M, Vannucchi AM, et al. The expression of CXCR4 is down-regulated on the CD34+ cells of patients with myelofibrosis with myeloid metaplasia. Blood Cells Mol Dis. 2007;38:280–6.PubMedCrossRef
64.
go back to reference Bogani C, Ponziani V, Guglielmelli P, et al. Hypermethylation of CXCR4 promoter in CD34+ cells from patients with primary myelofibrosis. Stem Cells. 2008;26:1920–30.PubMedCrossRef Bogani C, Ponziani V, Guglielmelli P, et al. Hypermethylation of CXCR4 promoter in CD34+ cells from patients with primary myelofibrosis. Stem Cells. 2008;26:1920–30.PubMedCrossRef
65.
go back to reference Verrucci M, Pancrazzi A, Aracil M, et al. CXCR4-independent rescue of the myeloproliferative defect of the Gata1(low) myelofibrosis mouse model by Aplidin(R). J Cell Physiol. 2010;225:490–9.PubMedCrossRef Verrucci M, Pancrazzi A, Aracil M, et al. CXCR4-independent rescue of the myeloproliferative defect of the Gata1(low) myelofibrosis mouse model by Aplidin(R). J Cell Physiol. 2010;225:490–9.PubMedCrossRef
66.
go back to reference •• Zingariello M, Martelli F, Ciaffoni F, et al. Characterization of the TGF-beta1 signaling abnormalities in the Gata1low mouse model of myelofibrosis. Blood. 2013;121:3345–63. Strong experimental support of the prominent role of TGF-beta 1 signaling in the GATA-1low animal model of MF.PubMedCrossRef •• Zingariello M, Martelli F, Ciaffoni F, et al. Characterization of the TGF-beta1 signaling abnormalities in the Gata1low mouse model of myelofibrosis. Blood. 2013;121:3345–63. Strong experimental support of the prominent role of TGF-beta 1 signaling in the GATA-1low animal model of MF.PubMedCrossRef
67.
go back to reference Baffert F, Regnier C, De Pover A, et al. Potent and selective inhibition of polycythemia by the quinoxaline JAK2 inhibitor NVP-BSK805. Mol Cancer Ther. 2010;9:1945–55.PubMedCrossRef Baffert F, Regnier C, De Pover A, et al. Potent and selective inhibition of polycythemia by the quinoxaline JAK2 inhibitor NVP-BSK805. Mol Cancer Ther. 2010;9:1945–55.PubMedCrossRef
68.
go back to reference Quintas-Cardama A, Vaddi K, Liu P, et al. Preclinical characterization of the selective JAK1/2 inhibitor INCB018424: therapeutic implications for the treatment of myeloproliferative neoplasms. Blood. 2010;115:3109–17.PubMedCrossRef Quintas-Cardama A, Vaddi K, Liu P, et al. Preclinical characterization of the selective JAK1/2 inhibitor INCB018424: therapeutic implications for the treatment of myeloproliferative neoplasms. Blood. 2010;115:3109–17.PubMedCrossRef
69.
go back to reference Pikman Y, Lee BH, Mercher T, et al. MPLW515L is a novel somatic activating mutation in myelofibrosis with myeloid metaplasia. PLoS Med. 2006;3:e270.PubMedCrossRef Pikman Y, Lee BH, Mercher T, et al. MPLW515L is a novel somatic activating mutation in myelofibrosis with myeloid metaplasia. PLoS Med. 2006;3:e270.PubMedCrossRef
70.
go back to reference Pardanani AD, Levine RL, Lasho T, et al. MPL515 mutations in myeloproliferative and other myeloid disorders: a study of 1182 patients. Blood. 2006;108:3472–6.PubMedCrossRef Pardanani AD, Levine RL, Lasho T, et al. MPL515 mutations in myeloproliferative and other myeloid disorders: a study of 1182 patients. Blood. 2006;108:3472–6.PubMedCrossRef
71.
go back to reference Koppikar P, Abdel-Wahab O, Hedvat C, et al. Efficacy of the JAK2 inhibitor INCB16562 in a murine model of MPLW515L-induced thrombocytosis and myelofibrosis. Blood. 2010;115:2919–27.PubMedCrossRef Koppikar P, Abdel-Wahab O, Hedvat C, et al. Efficacy of the JAK2 inhibitor INCB16562 in a murine model of MPLW515L-induced thrombocytosis and myelofibrosis. Blood. 2010;115:2919–27.PubMedCrossRef
72.
go back to reference Wernig G, Kharas MG, Mullally A, et al. EXEL-8232, a small-molecule JAK2 inhibitor, effectively treats thrombocytosis and extramedullary hematopoiesis in a murine model of myeloproliferative neoplasm induced by MPLW515L. Leukemia. 2012;26:720–7.PubMedCrossRef Wernig G, Kharas MG, Mullally A, et al. EXEL-8232, a small-molecule JAK2 inhibitor, effectively treats thrombocytosis and extramedullary hematopoiesis in a murine model of myeloproliferative neoplasm induced by MPLW515L. Leukemia. 2012;26:720–7.PubMedCrossRef
Metadata
Title
Preclinical Models for Drug Selection in Myeloproliferative Neoplasms
Authors
Niccolò Bartalucci
Costanza Bogani
Alessandro M. Vannucchi
Publication date
01-12-2013
Publisher
Springer US
Published in
Current Hematologic Malignancy Reports / Issue 4/2013
Print ISSN: 1558-8211
Electronic ISSN: 1558-822X
DOI
https://doi.org/10.1007/s11899-013-0182-1

Other articles of this Issue 4/2013

Current Hematologic Malignancy Reports 4/2013 Go to the issue

Myelodysplastic Syndromes (M Sekeres, Section Editor)

Standardizing the Initial Evaluation for Myelodysplastic Syndromes

Myeloproliferative Disorders (JJ Kiladjian, Section Editor)

The New Landscape of Therapy for Myelofibrosis

Myeloproliferative Disorders (JJ Kiladjian, Section Editor)

Are MPNs Vascular Diseases?

Multiple Myeloma (R Niesvizky, Section Editor)

New Insights and Modern Treatment of AL Amyloidosis

Myeloproliferative Disorders (JJ Kiladjian, Section Editor)

WHO Classification of Myeloproliferative Neoplasms (MPN): A Critical Update

Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.