Integrated genomic analysis illustrates the central role of JAK-STAT pathway activation in myeloproliferative neoplasm pathogenesis.

Genomic studies have identified somatic alterations in the majority of myeloproliferative neoplasms (MPN) patients, including JAK2 mutations in the majority of MPN patients and CALR mutations in JAK2-negative MPN patients. However, the role of JAK-STAT pathway activation in different MPNs, and in patients without JAK2 mutations, has not been definitively delineated. We used expression profiling, single nucleotide polymorphism arrays, and mutational profiling to investigate a well-characterized cohort of MPN patients. MPN patients with homozygous JAK2V617F mutations were characterized by a distinctive transcriptional profile. Notably, a transcriptional signature consistent with activated JAK2 signaling is seen in all MPN patients regardless of clinical phenotype or mutational status. In addition, the activated JAK2 signature was present in patients with somatic CALR mutations. Conversely, we identified a gene expression signature of CALR mutations; this signature was significantly enriched in JAK2-mutant MPN patients consistent with a shared mechanism of transformation by JAK2 and CALR mutations. We also identified a transcriptional signature of TET2 mutations in MPN patent samples. Our data indicate that MPN patients, regardless of diagnosis or JAK2 mutational status, are characterized by a distinct gene expression signature with upregulation of JAK-STAT target genes, demonstrating the central importance of the JAK-STAT pathway in MPN pathogenesis.

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

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

[3]  Niccolò Bartalucci,et al.  Co-targeting the PI3K/mTOR and JAK2 signalling pathways produces synergistic activity against myeloproliferative neoplasms , 2013, Journal of cellular and molecular medicine.

[4]  Angela G. Fleischman,et al.  Oncogenic CSF3R mutations in chronic neutrophilic leukemia and atypical CML. , 2013, The New England journal of medicine.

[5]  K. Bhalla,et al.  Dual PI3K/AKT/mTOR Inhibitor BEZ235 Synergistically Enhances the Activity of JAK2 Inhibitor against Cultured and Primary Human Myeloproliferative Neoplasm Cells , 2013, Molecular Cancer Therapeutics.

[6]  Mads Thomassen,et al.  Gene expression profiling with principal component analysis depicts the biological continuum from essential thrombocythemia over polycythemia vera to myelofibrosis. , 2012, Experimental hematology.

[7]  O. Abdel-Wahab,et al.  The role of mutations in epigenetic regulators in myeloid malignancies , 2012, Nature Reviews Cancer.

[8]  M. Gönen,et al.  Genetic analysis of patients with leukemic transformation of myeloproliferative neoplasms shows recurrent SRSF2 mutations that are associated with adverse outcome. , 2012, Blood.

[9]  D. Yan,et al.  Critical requirement for Stat5 in a mouse model of polycythemia vera. , 2012, Blood.

[10]  N. Socci,et al.  Prognostic relevance of integrated genetic profiling in acute myeloid leukemia. , 2012, The New England journal of medicine.

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

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

[13]  S. González,et al.  Ectopic expression of the histone methyltransferase Ezh2 in haematopoietic stem cells causes myeloproliferative disease , 2012, Nature Communications.

[14]  L. Hennighausen,et al.  Essential role for Stat5a/b in myeloproliferative neoplasms induced by BCR-ABL1 and JAK2(V617F) in mice. , 2009, Blood.

[15]  D. Gilliland,et al.  Analysis of genomic aberrations and gene expression profiling identifies novel lesions and pathways in myeloproliferative neoplasms , 2011, Blood cancer journal.

[16]  Feng-Chun Yang,et al.  Deletion of Tet2 in mice leads to dysregulated hematopoietic stem cells and subsequent development of myeloid malignancies. , 2011, Blood.

[17]  E. Pronier,et al.  Inhibition of TET2-mediated conversion of 5-methylcytosine to 5-hydroxymethylcytosine disturbs erythroid and granulomonocytic differentiation of human hematopoietic progenitors. , 2011, Blood.

[18]  T. Barbui,et al.  Safety and efficacy of everolimus, a mTOR inhibitor, as single agent in a phase 1/2 study in patients with myelofibrosis. , 2011, Blood.

[19]  K. Rajewsky,et al.  Ten-Eleven-Translocation 2 (TET2) negatively regulates homeostasis and differentiation of hematopoietic stem cells in mice , 2011, Proceedings of the National Academy of Sciences.

[20]  W. Vainchenker,et al.  New mutations and pathogenesis of myeloproliferative neoplasms. , 2011, Blood.

[21]  P. Opolon,et al.  TET2 inactivation results in pleiotropic hematopoietic abnormalities in mouse and is a recurrent event during human lymphomagenesis. , 2011, Cancer cell.

[22]  O. Abdel-Wahab,et al.  Tet2 loss leads to increased hematopoietic stem cell self-renewal and myeloid transformation. , 2011, Cancer cell.

[23]  M. Cazzola,et al.  Molecular and clinical features of the myeloproliferative neoplasm associated with JAK2 exon 12 mutations. , 2011, Blood.

[24]  E. Dermitzakis,et al.  Distinct clinical phenotypes associated with JAK2V617F reflect differential STAT1 signaling. , 2010, Cancer cell.

[25]  L. Aravind,et al.  Impaired hydroxylation of 5-methylcytosine in myeloid cancers with mutant TET2 , 2010, Nature.

[26]  A. Tefferi,et al.  LNK mutations in JAK2 mutation-negative erythrocytosis. , 2010, The New England journal of medicine.

[27]  Martin Dugas,et al.  Next-generation sequencing technology reveals a characteristic pattern of molecular mutations in 72.8% of chronic myelomonocytic leukemia by detecting frequent alterations in TET2, CBL, RAS, and RUNX1. , 2010, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[28]  A. Tefferi,et al.  LNK mutation studies in blast-phase myeloproliferative neoplasms, and in chronic-phase disease with TET2, IDH, JAK2 or MPL mutations , 2010, Leukemia.

[29]  Erin F. Simonds,et al.  Novel mutations in the inhibitory adaptor protein LNK drive JAK-STAT signaling in patients with myeloproliferative neoplasms. , 2010, Blood.

[30]  D. Gilliland,et al.  Transcriptional Profiling of Polycythemia Vera Identifies Gene Expression Patterns Both Dependent and Independent from the Action of JAK2V617F , 2010, Clinical Cancer Research.

[31]  R. Kusec,et al.  Two routes to leukemic transformation after a JAK2 mutation-positive myeloproliferative neoplasm. , 2010, Blood.

[32]  A. Tichelli,et al.  Clonal analysis of TET2 and JAK2 mutations suggests that TET2 can be a late event in the progression of myeloproliferative neoplasms. , 2010, Blood.

[33]  O. Abdel-Wahab,et al.  Genetic analysis of transforming events that convert chronic myeloproliferative neoplasms to leukemias. , 2010, Cancer research.

[34]  S. Fiering,et al.  Conditional expression of heterozygous or homozygous Jak2V617F from its endogenous promoter induces a polycythemia vera-like disease. , 2009, Blood.

[35]  J. Soulier,et al.  Mutation in TET2 in myeloid cancers. , 2009, The New England journal of medicine.

[36]  Keith L. Ligon,et al.  Profiling Critical Cancer Gene Mutations in Clinical Tumor Samples , 2009, PloS one.

[37]  D. Birnbaum,et al.  Mutations of ASXL1 gene in myeloproliferative neoplasms , 2009, Leukemia.

[38]  D. Gilliland,et al.  Genetic characterization of TET1, TET2, and TET3 alterations in myeloid malignancies. , 2009, Blood.

[39]  L. Arenillas,et al.  Gene expression profiling distinguishes JAK2V617F-negative from JAK2V617F-positive patients in essential thrombocythemia , 2008, Leukemia.

[40]  L. Arenillas,et al.  Gene expression profiling distinguishes JAK2V617F-negative from JAK2V617F-positive patients in essential thrombocythemia , 2008, Leukemia.

[41]  H. Pahl,et al.  JAK2V617F-negative ET patients do not display constitutively active JAK/STAT signaling. , 2007, Experimental hematology.

[42]  M. Stratton,et al.  JAK2 exon 12 mutations in polycythemia vera and idiopathic erythrocytosis. , 2007, The New England journal of medicine.

[43]  D. Gilliland,et al.  MPL515 mutations in myeloproliferative and other myeloid disorders: a study of 1182 patients. , 2006, Blood.

[44]  Jill P. Mesirov,et al.  Comparative gene marker selection suite , 2006, Bioinform..

[45]  Sandra A. Moore,et al.  MPLW515L Is a Novel Somatic Activating Mutation in Myelofibrosis with Myeloid Metaplasia , 2006, PLoS medicine.

[46]  J. Mesirov,et al.  GenePattern 2.0 , 2006, Nature Genetics.

[47]  I. Weissman,et al.  The JAK2 V617F mutation occurs in hematopoietic stem cells in polycythemia vera and predisposes toward erythroid differentiation. , 2006, Proceedings of the National Academy of Sciences of the United States of America.

[48]  Anne E Carpenter,et al.  A Lentiviral RNAi Library for Human and Mouse Genes Applied to an Arrayed Viral High-Content Screen , 2006, Cell.

[49]  M. Wadleigh,et al.  The clinical phenotype of wild‐type, heterozygous, and homozygous JAK2V617F in polycythemia vera , 2006, Cancer.

[50]  R. Levine,et al.  X-inactivation-based clonality analysis and quantitative JAK2V617F assessment reveal a strong association between clonality and JAK2V617F in PV but not ET/MMM, and identifies a subset of JAK2V617F-negative ET and MMM patients with clonal hematopoiesis. , 2005, Blood.

[51]  M. Cazzola,et al.  Altered gene expression in myeloproliferative disorders correlates with activation of signaling by the V617F mutation of Jak2. , 2005, Blood.

[52]  W. Vainchenker,et al.  Role of tyrosine kinases and phosphatases in polycythemia vera. , 2005, Seminars in hematology.

[53]  Pablo Tamayo,et al.  Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[54]  Stefan N. Constantinescu,et al.  A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera , 2005, Nature.

[55]  Mario Cazzola,et al.  A gain-of-function mutation of JAK2 in myeloproliferative disorders. , 2005, The New England journal of medicine.

[56]  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.

[57]  T. Maiwald,et al.  Gene expression profiling in polycythaemia vera: overexpression of transcription factor NF‐E2 , 2005, British journal of haematology.

[58]  P. Campbell,et al.  Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders , 2005, The Lancet.

[59]  G. Morrone,et al.  Enforced Activation of STAT5A Facilitates the Generation of Embryonic Stem–Derived Hematopoietic Stem Cells That Contribute to Hematopoiesis In Vivo , 2004, Stem cells.

[60]  H. Heimpel,et al.  Quantification of PRV-1 mRNA distinguishes polycythemia vera from secondary erythrocytosis. , 2003, Blood.

[61]  C. Langford,et al.  Gene expression profiling in polycythemia vera using cDNA microarray technology. , 2003, Cancer research.

[62]  Terence P. Speed,et al.  A comparison of normalization methods for high density oligonucleotide array data based on variance and bias , 2003, Bioinform..

[63]  R. Kralovics,et al.  Acquired uniparental disomy of chromosome 9p is a frequent stem cell defect in polycythemia vera. , 2002, Experimental hematology.

[64]  E. Lander,et al.  Classification of human lung carcinomas by mRNA expression profiling reveals distinct adenocarcinoma subclasses. , 2001, Proceedings of the National Academy of Sciences of the United States of America.

[65]  J. Mesirov,et al.  Molecular classification of cancer: class discovery and class prediction by gene expression monitoring. , 1999, Science.

[66]  Y. Benjamini,et al.  More powerful procedures for multiple significance testing. , 1990, Statistics in medicine.