Targeting megakaryocytic-induced fibrosis in myeloproliferative neoplasms by AURKA inhibition

Primary myelofibrosis (PMF) is characterized by bone marrow fibrosis, myeloproliferation, extramedullary hematopoiesis, splenomegaly and leukemic progression. Moreover, the bone marrow and spleens of individuals with PMF contain large numbers of atypical megakaryocytes that are postulated to contribute to fibrosis through the release of cytokines, including transforming growth factor (TGF)-β. Although the Janus kinase inhibitor ruxolitinib provides symptomatic relief, it does not reduce the mutant allele burden or substantially reverse fibrosis. Here we show through pharmacologic and genetic studies that aurora kinase A (AURKA) represents a new therapeutic target in PMF. Treatment with MLN8237, a selective AURKA inhibitor, promoted polyploidization and differentiation of megakaryocytes with PMF-associated mutations and had potent antifibrotic and antitumor activity in vivo in mouse models of PMF. Moreover, heterozygous deletion of Aurka was sufficient to ameliorate fibrosis and other PMF features in vivo. Our data suggest that megakaryocytes drive fibrosis in PMF and that targeting them with AURKA inhibitors has the potential to provide therapeutic benefit.

[1]  Sandra A. Moore,et al.  Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. , 2005, Cancer cell.

[2]  R. Mesa,et al.  Current Outlook on Molecular Pathogenesis and Treatment of Myeloproliferative Neoplasms , 2012, Molecular Diagnosis & Therapy.

[3]  H. Castro-Malaspina Pathogenesis of myelofibrosis: role of ineffective megakaryopoiesis and megakaryocyte components. , 1984, Progress in clinical and biological research.

[4]  I. Weissman,et al.  A clonogenic common myeloid progenitor that gives rise to all myeloid lineages , 2000, Nature.

[5]  Jason Gotlib,et al.  A double-blind, placebo-controlled trial of ruxolitinib for myelofibrosis. , 2012, The New England journal of medicine.

[6]  K. Uozumi,et al.  Establishment and characterization of a new human megakaryoblastic cell line (SET-2) that spontaneously matures to megakaryocytes and produces platelet-like particles , 2000, Leukemia.

[7]  N. Mahmud,et al.  Pivotal contributions of megakaryocytes to the biology of idiopathic myelofibrosis. , 2007, Blood.

[8]  M. Le Bousse-Kerdilès,et al.  Involvement of the fibrogenic cytokines, TGF-beta and bFGF, in the pathogenesis of idiopathic myelofibrosis. , 2001, Pathologie-biologie.

[9]  S. Verstovsek,et al.  Investigational Janus kinase inhibitors , 2013, Expert opinion on investigational drugs.

[10]  P. Guglielmelli,et al.  Abnormalities of GATA-1 in megakaryocytes from patients with idiopathic myelofibrosis. , 2005, The American journal of pathology.

[11]  A. Mead,et al.  Practical management of patients with myelofibrosis receiving ruxolitinib , 2013, Expert review of hematology.

[12]  A. Tefferi JAK inhibitors for myeloproliferative neoplasms: clarifying facts from myths. , 2012, Blood.

[13]  Francisco Cervantes,et al.  JAK inhibition with ruxolitinib versus best available therapy for myelofibrosis. , 2012, The New England journal of medicine.

[14]  A. Bergman,et al.  Megakaryocytes regulate hematopoietic stem cell quiescence via Cxcl4 secretion , 2013, Nature Medicine.

[15]  O. Abdel-Wahab,et al.  Efficacy of the JAK2 inhibitor INCB16562 in a murine model of MPLW515L-induced thrombocytosis and myelofibrosis. , 2010, Blood.

[16]  M. Weiss,et al.  Early block to erythromegakaryocytic development conferred by loss of transcription factor GATA-1. , 2005, Blood.

[17]  W. Vainchenker,et al.  High thrombopoietin production by hematopoietic cells induces a fatal myeloproliferative syndrome in mice. , 1997, Blood.

[18]  R. Hoffman,et al.  A comprehensive review and analysis of the effect of ruxolitinib therapy on the survival of patients with myelofibrosis. , 2013, Blood.

[19]  G. Superti-Furga,et al.  Somatic mutations of calreticulin in myeloproliferative neoplasms. , 2013, The New England journal of medicine.

[20]  M. Cazzola,et al.  Mutations and prognosis in primary myelofibrosis , 2013, Leukemia.

[21]  F. Passamonti,et al.  DIPSS plus: a refined Dynamic International Prognostic Scoring System for primary myelofibrosis that incorporates prognostic information from karyotype, platelet count, and transfusion status. , 2011, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[22]  Michael G. Kharas,et al.  Physiological Jak2V617F expression causes a lethal myeloproliferative neoplasm with differential effects on hematopoietic stem and progenitor cells. , 2010, Cancer cell.

[23]  E. Hsiao,et al.  Myeloproliferative neoplasia remodels the endosteal bone marrow niche into a self-reinforcing leukemic niche. , 2013, Cell stem cell.

[24]  Xi C. He,et al.  Megakaryocytes maintain homeostatic quiescence and promote post-injury regeneration of hematopoietic stem cells , 2014, Nature Medicine.

[25]  M. Bryckaert,et al.  Increased intraplatelet levels of platelet‐derived growth factor and transforming growth factor‐β in patients with myelofibrosis with myeloid metaplasia , 1991, British journal of haematology.

[26]  B. Wiese,et al.  Growth Factors , Cytokines , Cell Cycle Molecules Bone Morphogenetic Proteins Are Overexpressed in the Bone Marrow of Primary Myelofibrosis and Are Apparently Induced by Fibrogenic Cytokines , 2010 .

[27]  M. Cazzola,et al.  Unraveling the genetic underpinnings of myeloproliferative neoplasms and understanding their effect on disease course and response to therapy: Proceedings from the 6th international post‐ASH symposium , 2012, American journal of hematology.

[28]  T. Kasahara,et al.  Aurora kinase A critically contributes to the resistance to anti‐cancer drug cisplatin in JAK2 V617F mutant‐induced transformed cells , 2011, FEBS letters.

[29]  T. Chou,et al.  Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors. , 1984, Advances in enzyme regulation.

[30]  A. Migliaccio,et al.  Development of myelofibrosis in mice genetically impaired for GATA-1 expression (GATA-1(low) mice). , 2002, Blood.

[31]  Q. Wen,et al.  Aurora kinase A is required for hematopoiesis but is dispensable for murine megakaryocyte endomitosis and differentiation. , 2015, Blood.

[32]  R. Kralovics,et al.  Identification of oncostatin M as a JAK2 V617F‐dependent amplifier of cytokine production and bone marrow remodeling in myeloproliferative neoplasms , 2012, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[33]  Sandra A. Moore,et al.  Efficacy of TG101348, a selective JAK2 inhibitor, in treatment of a murine model of JAK2V617F-induced polycythemia vera. , 2008, Cancer cell.

[34]  W. Vainchenker,et al.  Prominent role of TGF-beta 1 in thrombopoietin-induced myelofibrosis in mice. , 2002, Blood.

[35]  Mark Coles,et al.  Transgenic mice with hematopoietic and lymphoid specific expression of Cre , 2003, European journal of immunology.

[36]  M Aguet,et al.  Inducible gene targeting in mice , 1995, Science.

[37]  R. Bayliss,et al.  A Kinetic Test Characterizes Kinase Intramolecular and Intermolecular Autophosphorylation Mechanisms , 2013, Science Signaling.

[38]  W. Vainchenker,et al.  Prominent role of TGF-1 in thrombopoietin-induced myelofibrosis in mice , 2002 .

[39]  J. D. Fitzpatrick,et al.  Somatic CALR mutations in myeloproliferative neoplasms with nonmutated JAK2. , 2013, The New England journal of medicine.

[40]  J. Crispino,et al.  The Aurora Kinases in Cell Cycle and Leukemia , 2014, Oncogene.

[41]  Mark A. Hall,et al.  Aurora kinases A and B are up-regulated by Myc and are essential for maintenance of the malignant state. , 2010, Blood.

[42]  M. Tomasson,et al.  The Jak2V617F oncogene associated with myeloproliferative diseases requires a functional FERM domain for transformation and for expression of the Myc and Pim proto-oncogenes. , 2008, Blood.

[43]  T. Magnuson,et al.  Aurora-A Kinase Is Essential for Bipolar Spindle Formation and Early Development , 2008, Molecular and Cellular Biology.

[44]  S. Orkin,et al.  A lineage‐selective knockout establishes the critical role of transcription factor GATA‐1 in megakaryocyte growth and platelet development , 1997, The EMBO journal.

[45]  Anne E Carpenter,et al.  Identification of Regulators of Polyploidization Presents Therapeutic Targets for Treatment of AMKL , 2012, Cell.