Your browser doesn't support javascript.
loading
The creatine kinase pathway is a metabolic vulnerability in EVI1-positive acute myeloid leukemia.
Fenouille, Nina; Bassil, Christopher F; Ben-Sahra, Issam; Benajiba, Lina; Alexe, Gabriela; Ramos, Azucena; Pikman, Yana; Conway, Amy S; Burgess, Michael R; Li, Qing; Luciano, Frédéric; Auberger, Patrick; Galinsky, Ilene; DeAngelo, Daniel J; Stone, Richard M; Zhang, Yi; Perkins, Archibald S; Shannon, Kevin; Hemann, Michael T; Puissant, Alexandre; Stegmaier, Kimberly.
Affiliation
  • Fenouille N; Koch Institute for Integrative Cancer Research at Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.
  • Bassil CF; Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA.
  • Ben-Sahra I; Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, Massachusetts, USA.
  • Benajiba L; Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA.
  • Alexe G; Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA.
  • Ramos A; Broad Institute of Harvard University and Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.
  • Pikman Y; Bioinformatics Graduate Program, Boston University, Boston, Massachusetts, USA.
  • Conway AS; Koch Institute for Integrative Cancer Research at Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.
  • Burgess MR; Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA.
  • Li Q; Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA.
  • Luciano F; Department of Medicine, University of California San Francisco, San Francisco, California, USA.
  • Auberger P; Internal Medicine Hematology-Oncology, University of Michigan, Ann Arbor, Michigan, USA.
  • Galinsky I; Université Côte d'Azur, UMR INSERM U1065, C3M, Nice, France.
  • DeAngelo DJ; Université Côte d'Azur, UMR INSERM U1065, C3M, Nice, France.
  • Stone RM; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA.
  • Zhang Y; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA.
  • Perkins AS; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA.
  • Shannon K; Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York, New York, USA.
  • Hemann MT; Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York, New York, USA.
  • Puissant A; Department of Pediatrics and Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California, USA.
  • Stegmaier K; Koch Institute for Integrative Cancer Research at Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.
Nat Med ; 23(3): 301-313, 2017 Mar.
Article in En | MEDLINE | ID: mdl-28191887
ABSTRACT
Expression of the MECOM (also known as EVI1) proto-oncogene is deregulated by chromosomal translocations in some cases of acute myeloid leukemia (AML) and is associated with poor clinical outcome. Here, through transcriptomic and metabolomic profiling of hematopoietic cells, we reveal that EVI1 overexpression alters cellular metabolism. A screen using pooled short hairpin RNAs (shRNAs) identified the ATP-buffering, mitochondrial creatine kinase CKMT1 as necessary for survival of EVI1-expressing cells in subjects with EVI1-positive AML. EVI1 promotes CKMT1 expression by repressing the myeloid differentiation regulator RUNX1. Suppression of arginine-creatine metabolism by CKMT1-directed shRNAs or by the small molecule cyclocreatine selectively decreased the viability, promoted the cell cycle arrest and apoptosis of human EVI1-positive cell lines, and prolonged survival in both orthotopic xenograft models and mouse models of primary AML. CKMT1 inhibition altered mitochondrial respiration and ATP production, an effect that was abrogated by phosphocreatine-mediated reactivation of the arginine-creatine pathway. Targeting CKMT1 is thus a promising therapeutic strategy for this EVI1-driven AML subtype that is highly resistant to current treatment regimens.
Subject(s)

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Transcription Factors / Proto-Oncogenes / Leukemia, Myeloid, Acute / Gene Expression Regulation, Neoplastic / Creatine Kinase / DNA-Binding Proteins / Core Binding Factor Alpha 2 Subunit Type of study: Prognostic_studies Limits: Adult / Aged / Aged80 / Female / Humans / Male / Middle aged Language: En Journal: Nat Med Journal subject: BIOLOGIA MOLECULAR / MEDICINA Year: 2017 Document type: Article Affiliation country: United States

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: Transcription Factors / Proto-Oncogenes / Leukemia, Myeloid, Acute / Gene Expression Regulation, Neoplastic / Creatine Kinase / DNA-Binding Proteins / Core Binding Factor Alpha 2 Subunit Type of study: Prognostic_studies Limits: Adult / Aged / Aged80 / Female / Humans / Male / Middle aged Language: En Journal: Nat Med Journal subject: BIOLOGIA MOLECULAR / MEDICINA Year: 2017 Document type: Article Affiliation country: United States
...