Your browser doesn't support javascript.
loading
EZH2 Cooperates with BRD4-NUT to Drive NUT Carcinoma Growth by Silencing Key Tumor Suppressor Genes.
Huang, Yeying; Durall, R Taylor; Luong, Nhi M; Hertzler, Hans J; Huang, Julianna; Gokhale, Prafulla C; Leeper, Brittaney A; Persky, Nicole S; Root, David E; Anekal, Praju V; Montero Llopis, Paula D L M; David, Clement N; Kutok, Jeffery L; Raimondi, Alejandra; Saluja, Karan; Luo, Jia; Zahnow, Cynthia A; Adane, Biniam; Stegmaier, Kimberly; Hawkins, Catherine E; Ponne, Christopher; Le, Quan; Shapiro, Geoffrey I; Lemieux, Madeleine E; Eagen, Kyle P; French, Christopher A.
Afiliación
  • Huang Y; Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
  • Durall RT; Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
  • Luong NM; Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
  • Hertzler HJ; Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
  • Huang J; Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
  • Gokhale PC; Experimental Therapeutics Core and Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Leeper BA; Experimental Therapeutics Core and Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Persky NS; Broad Institute of MIT and Harvard, Cambridge, Massachusetts.
  • Root DE; Broad Institute of MIT and Harvard, Cambridge, Massachusetts.
  • Anekal PV; MicRoN, Department of Microbiology, Harvard Medical School, Boston, Massachusetts.
  • Montero Llopis PDLM; MicRoN, Department of Microbiology, Harvard Medical School, Boston, Massachusetts.
  • David CN; NanoString Technologies, Inc., Seattle, Washington.
  • Kutok JL; Epizyme Inc., Cambridge, Massachusetts.
  • Raimondi A; Epizyme Inc., Cambridge, Massachusetts.
  • Saluja K; Department of Pathology and Laboratory Medicine, University of Texas Health Science Center, Houston, Texas.
  • Luo J; Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
  • Zahnow CA; Department of Oncology, The Johns Hopkins School of Medicine, Baltimore, Maryland.
  • Adane B; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Stegmaier K; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
  • Hawkins CE; Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts.
  • Ponne C; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas.
  • Le Q; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas.
  • Shapiro GI; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas.
  • Lemieux ME; Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
  • Eagen KP; Bioinfo, Plantagenet, Ontario, Canada.
  • French CA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas.
Cancer Res ; 83(23): 3956-3973, 2023 12 01.
Article en En | MEDLINE | ID: mdl-37747726
ABSTRACT
NUT carcinoma is an aggressive carcinoma driven by the BRD4-NUT fusion oncoprotein, which activates chromatin to promote expression of progrowth genes. BET bromodomain inhibitors (BETi) are a promising treatment for NUT carcinoma that can impede BRD4-NUT's ability to activate genes, but the efficacy of BETi as monotherapy is limited. Here, we demonstrated that enhancer of zeste homolog 2 (EZH2), which silences genes through establishment of repressive chromatin, is a dependency in NUT carcinoma. Inhibition of EZH2 with the clinical compound tazemetostat potently blocked growth of NUT carcinoma cells. Epigenetic and transcriptomic analysis revealed that tazemetostat reversed the EZH2-specific H3K27me3 silencing mark and restored expression of multiple tumor suppressor genes while having no effect on key oncogenic BRD4-NUT-regulated genes. Indeed, H3K27me3 and H3K27ac domains were found to be mutually exclusive in NUT carcinoma cells. CDKN2A was identified as the only gene among all tazemetostat-derepressed genes to confer resistance to tazemetostat in a CRISPR-Cas9 screen. Combined inhibition of EZH2 and BET synergized to downregulate cell proliferation genes, resulting in more pronounced growth arrest and differentiation than either inhibitor alone. In preclinical models, combined tazemetostat and BETi synergistically blocked tumor growth and prolonged survival of NUT carcinoma-xenografted mice, with complete remission without relapse in one cohort. Identification of EZH2 as a dependency in NUT carcinoma substantiates the reliance of NUT carcinoma tumor cells on epigenetic dysregulation of functionally opposite, yet highly complementary, chromatin regulatory pathways to maintain NUT carcinoma growth.

SIGNIFICANCE:

Repression of tumor suppressor genes, including CDKN2A, by EZH2 provides a mechanistic rationale for combining EZH2 and BET inhibitors for the clinical treatment of NUT carcinoma. See related commentary by Kazansky and Kentsis, p. 3827.
Asunto(s)

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Proteínas Nucleares / Carcinoma Límite: Animals / Humans Idioma: En Revista: Cancer Res Año: 2023 Tipo del documento: Article

Texto completo: 1 Colección: 01-internacional Base de datos: MEDLINE Asunto principal: Proteínas Nucleares / Carcinoma Límite: Animals / Humans Idioma: En Revista: Cancer Res Año: 2023 Tipo del documento: Article
...