Super-Enhancer analysis defines novel epigenomic subtypes of non-APL AML including an RARα dependency targetable by SY-1425, a potent and selective RARα agonist AUTHORS and AFFILITATIONS:

We characterized the enhancer landscape of 66 AML patients, identifying 6 novel subgroups and their associated regulatory loci. These subgroups are defined by their super-enhancer (SE) maps, orthogonal to somatic mutations, and are associated with distinct leukemic cell states. Examination of transcriptional drivers for these epigenomic subtypes uncovers a subset of patients with a particularly strong super-enhancer at the retinoic acid receptor alpha (RARA) gene locus. Presence of a RARA SE and concomitant high levels of RARA mRNA predisposes cell lines and ex vivo models to exquisite sensitivity to a selective agonist of RARα, SY-1425 (tamibarotene). Furthermore, only AML patient-derived xenograft (PDX) models with high RARA mRNA were found to respond to SY-1425. Mechanistically, we show that the response to SY-1425 in RARA-high AML cells is similar to that of APL treated with retinoids, characterized by the induction of known retinoic acid response genes, increased differentiation, and loss of proliferation. Significance We use the super-enhancer landscape of primary human AML to elucidate transcriptional circuitry and identify novel cancer vulnerabilities. A subset of patients were found to have an SE at RARA which is predictive for response to SY-1425, a potent and selective RARα agonist, in preclinical models, forming the rationale for its clinical investigation in biomarker-selected patients.

[1]  A. Yokoyama Transcriptional activation by MLL fusion proteins in leukemogenesis. , 2017, Experimental hematology.

[2]  Jonathan M. Cairns,et al.  Lineage-Specific Genome Architecture Links Enhancers and Non-coding Disease Variants to Target Gene Promoters , 2016, Cell.

[3]  M. Fratelli,et al.  RARα2 and PML-RAR similarities in the control of basal and retinoic acid induced myeloid maturation of acute myeloid leukemia cells , 2016, Oncotarget.

[4]  R. Levine,et al.  Epigenetic regulators and their impact on therapy in acute myeloid leukemia , 2016, Haematologica.

[5]  Bas J. Wouters,et al.  Epigenetics and approaches to targeted epigenetic therapy in acute myeloid leukemia. , 2016, Blood.

[6]  Jun Yu Li,et al.  Tamibarotene Compared to All-Trans Retinoic Acid (ATRA) As Add-on to Arsenic Trioxide (ATO) in Subjects with Relapsed Acute Promyelocytic Leukemia (APL) , 2015 .

[7]  Stephen C. J. Parker,et al.  Super Enhancers in Cancers, Complex Disease, and Developmental Disorders , 2015, Genes.

[8]  V. Lefebvre,et al.  The transcription factors SOX9 and SOX5/SOX6 cooperate genome-wide through super-enhancers to drive chondrogenesis , 2015, Nucleic acids research.

[9]  R. Slany,et al.  Pbx3 and Meis1 cooperate through multiple mechanisms to support Hox-induced murine leukemia , 2015, Haematologica.

[10]  Daniel E. Johnson,et al.  An ATRActive future for differentiation therapy in AML. , 2015, Blood reviews.

[11]  M. Tallman,et al.  Acute promyelocytic leukemia: where did we start, where are we now, and the future , 2015, Blood Cancer Journal.

[12]  Charles Y. Lin,et al.  Convergence of developmental and oncogenic signaling pathways at transcriptional super-enhancers. , 2015, Molecular cell.

[13]  W. L. Nelson,et al.  Induction of CYP26A1 by Metabolites of Retinoic Acid: Evidence That CYP26A1 Is an Important Enzyme in the Elimination of Active Retinoids , 2015, Molecular Pharmacology.

[14]  D. Zheng,et al.  Pioneer factors govern super-enhancer dynamics in stem cell plasticity and lineage choice , 2015, Nature.

[15]  Tyler B. Hughes,et al.  Enhancer sequence variants and transcription-factor deregulation synergize to construct pathogenic regulatory circuits in B-cell lymphoma. , 2015, Immunity.

[16]  J. Stender,et al.  Environment Drives Selection and Function of Enhancers Controlling Tissue-Specific Macrophage Identities , 2015, Cell.

[17]  P. Campbell,et al.  Combining gene mutation with gene expression data improves outcome prediction in myelodysplastic syndromes , 2015, Nature Communications.

[18]  Stephen C. J. Parker,et al.  Stretch-Enhancers Delineate Disease-Associated Regulatory Nodes in T Cells , 2014, Nature.

[19]  T. Naoe,et al.  Tamibarotene as maintenance therapy for acute promyelocytic leukemia: results from a randomized controlled trial. , 2014, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[20]  A. Zelent,et al.  Unlocking the potential of retinoic acid in anticancer therapy , 2014, British Journal of Cancer.

[21]  S. Garattini,et al.  Retinoids and breast cancer: from basic studies to the clinic and back again. , 2014, Cancer treatment reviews.

[22]  Mark K. Adams,et al.  The Retinaldehyde Reductase Activity of DHRS3 Is Reciprocally Activated by Retinol Dehydrogenase 10 to Control Retinoid Homeostasis* , 2014, The Journal of Biological Chemistry.

[23]  Charles Y. Lin,et al.  Discovery and characterization of super-enhancer-associated dependencies in diffuse large B cell lymphoma. , 2013, Cancer cell.

[24]  R. Young,et al.  Super-Enhancers in the Control of Cell Identity and Disease , 2013, Cell.

[25]  M. Kane,et al.  The retinaldehyde reductase DHRS3 is essential for preventing the formation of excess retinoic acid during embryonic development , 2013, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[26]  David A. Orlando,et al.  Selective Inhibition of Tumor Oncogenes by Disruption of Super-Enhancers , 2013, Cell.

[27]  David A. Orlando,et al.  Master Transcription Factors and Mediator Establish Super-Enhancers at Key Cell Identity Genes , 2013, Cell.

[28]  S. Hooper,et al.  Gene rearrangements in hormone receptor negative breast cancers revealed by mate pair sequencing , 2013, BMC Genomics.

[29]  F. Ferrara,et al.  Acute myeloid leukaemia in adults , 2013, The Lancet.

[30]  A. Ross,et al.  DHRS3, a retinal reductase, is differentially regulated by retinoic acid and lipopolysaccharide-induced inflammation in THP-1 cells and rat liver. , 2012, American journal of physiology. Gastrointestinal and liver physiology.

[31]  P. Hughes,et al.  Retinoid Differentiation Therapy for Common Types of Acute Myeloid Leukemia , 2012, Leukemia research and treatment.

[32]  A. Hero,et al.  Identification and characterization of Hoxa9 binding sites in hematopoietic cells. , 2012, Blood.

[33]  Cory Y. McLean,et al.  GREAT improves functional interpretation of cis-regulatory regions , 2010, Nature Biotechnology.

[34]  John Quackenbush,et al.  Decomposition of Gene Expression State Space Trajectories , 2009, PLoS Comput. Biol..

[35]  P. Pandolfi,et al.  Eradication of acute promyelocytic leukemia-initiating cells through PML-RARA degradation , 2008, Nature Medicine.

[36]  P. Dollé,et al.  Retinoic acid in development: towards an integrated view , 2008, Nature Reviews Genetics.

[37]  Guang-Biao Zhou,et al.  Treatment of acute promyelocytic leukaemia with all-trans retinoic acid and arsenic trioxide: a paradigm of synergistic molecular targeting therapy , 2007, Philosophical Transactions of the Royal Society B: Biological Sciences.

[38]  J. Licht Reconstructing a disease: What essential features of the retinoic acid receptor fusion oncoproteins generate acute promyelocytic leukemia? , 2006, Cancer cell.

[39]  M. Caligiuri,et al.  Overexpression of the ETS-related gene, ERG, predicts a worse outcome in acute myeloid leukemia with normal karyotype: a Cancer and Leukemia Group B study. , 2005, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[40]  Natalia Meani,et al.  Molecular signature of retinoic acid treatment in acute promyelocytic leukemia , 2005, Oncogene.

[41]  M. Osanai,et al.  Expression of the Retinoic Acid-Metabolizing Enzyme CYP26A1 Limits Programmed Cell Death , 2005, Molecular Pharmacology.

[42]  H. de Thé,et al.  PML–RARA-RXR Oligomers Mediate Retinoid and Rexinoid/cAMP Cross-Talk in Acute Promyelocytic Leukemia Cell Differentiation , 2004, The Journal of experimental medicine.

[43]  J. She,et al.  Gene expression profiling during all-trans retinoic acid-induced cell differentiation of acute promyelocytic leukemia cells. , 2003, The Journal of molecular diagnostics : JMD.

[44]  M. Levis,et al.  FLT3: ITDoes matter in leukemia , 2003, Leukemia.

[45]  Z. Wang,et al.  Monitoring all-trans-retinoic acid-induced differentiation of human acute promyelocytic leukemia NB4 cells by Fourier-transform infrared spectroscopy , 2003, Leukemia.

[46]  R. Chandraratna,et al.  Synthesis and biological activity of retinoic acid receptor-α specific amides , 2002 .

[47]  Unnur Thorsteinsdottir,et al.  Defining Roles for HOX and MEIS1 Genes in Induction of Acute Myeloid Leukemia , 2001, Molecular and Cellular Biology.

[48]  P Chambon,et al.  Structural basis for engineering of retinoic acid receptor isotype-selective agonists and antagonists. , 1999, Chemistry & biology.

[49]  H. Gronemeyer,et al.  Regulation of retinoidal actions by diazepinylbenzoic acids. Retinoid synergists which activate the RXR-RAR heterodimers. , 1997, Journal of medicinal chemistry.

[50]  T. Naoe,et al.  Treatment with a new synthetic retinoid, Am80, of acute promyelocytic leukemia relapsed from complete remission induced by all-trans retinoic acid. , 1997, Blood.

[51]  E. Dmitrovsky,et al.  Continuous treatment with all-trans retinoic acid causes a progressive reduction in plasma drug concentrations: implications for relapse and retinoid "resistance" in patients with acute promyelocytic leukemia. , 1992, Blood.

[52]  B. Shroot,et al.  Selective high affinity retinoic acid receptor alpha or beta-gamma ligands. , 1991, Molecular pharmacology.

[53]  O. Yokosuka,et al.  An open-label phase I/II study of tamibarotene in patients with advanced hepatocellular carcinoma , 2013, Hepatology International.

[54]  M. Lanotte,et al.  Granulocytic differentiation of human NB4 promyelocytic leukemia cells induced by all-trans retinoic acid metabolites. , 2001, Cancer research.