Your browser doesn't support javascript.
loading
Myeloid-Derived Suppressor Cell Subsets Drive Glioblastoma Growth in a Sex-Specific Manner.
Bayik, Defne; Zhou, Yadi; Park, Chihyun; Hong, Changjin; Vail, Daniel; Silver, Daniel J; Lauko, Adam; Roversi, Gustavo; Watson, Dionysios C; Lo, Alice; Alban, Tyler J; McGraw, Mary; Sorensen, Mia; Grabowski, Matthew M; Otvos, Balint; Vogelbaum, Michael A; Horbinski, Craig; Kristensen, Bjarne Winther; Khalil, Ahmad M; Hwang, Tae Hyun; Ahluwalia, Manmeet S; Cheng, Feixiong; Lathia, Justin D.
Afiliação
  • Bayik D; Cancer Impact Area and Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.
  • Zhou Y; Case Comprehensive Cancer Center, Cleveland, Ohio.
  • Park C; Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.
  • Hong C; Quantitative Health Science, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.
  • Vail D; Quantitative Health Science, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.
  • Silver DJ; Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, Ohio.
  • Lauko A; Cancer Impact Area and Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.
  • Roversi G; Case Comprehensive Cancer Center, Cleveland, Ohio.
  • Watson DC; Cancer Impact Area and Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.
  • Lo A; Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland, Ohio.
  • Alban TJ; Department of Pathology, Case Western Reserve University, Cleveland, Ohio.
  • McGraw M; Cancer Impact Area and Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.
  • Sorensen M; Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland, Ohio.
  • Grabowski MM; Cancer Impact Area and Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.
  • Otvos B; Case Comprehensive Cancer Center, Cleveland, Ohio.
  • Vogelbaum MA; University Hospitals Cleveland Medical Center, Cleveland, Ohio.
  • Horbinski C; School of Medicine, Case Western Reserve University, Cleveland, Ohio.
  • Kristensen BW; Cancer Impact Area and Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.
  • Khalil AM; Case Western Reserve University, Cleveland, Ohio.
  • Hwang TH; Cancer Impact Area and Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio.
  • Ahluwalia MS; Case Comprehensive Cancer Center, Cleveland, Ohio.
  • Cheng F; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine at Case Western Reserve University, Cleveland, Ohio.
  • Lathia JD; Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, Ohio.
Cancer Discov ; 10(8): 1210-1225, 2020 08.
Article em En | MEDLINE | ID: mdl-32300059
ABSTRACT
Myeloid-derived suppressor cells (MDSC) that block antitumor immunity are elevated in glioblastoma (GBM) patient blood and tumors. However, the distinct contributions of monocytic (mMDSC) versus granulocytic (gMDSC) subsets have yet to be determined. In mouse models of GBM, we observed that mMDSCs were enriched in the male tumors, whereas gMDSCs were elevated in the blood of females. Depletion of gMDSCs extended survival only in female mice. Using gene-expression signatures coupled with network medicine analysis, we demonstrated in preclinical models that mMDSCs could be targeted with antiproliferative agents in males, whereas gMDSC function could be inhibited by IL1ß blockade in females. Analysis of patient data confirmed that proliferating mMDSCs were predominant in male tumors and that a high gMDSC/IL1ß gene signature correlated with poor prognosis in female patients. These findings demonstrate that MDSC subsets differentially drive immune suppression in a sex-specific manner and can be leveraged for therapeutic intervention in GBM.

SIGNIFICANCE:

Sexual dimorphism at the level of MDSC subset prevalence, localization, and gene-expression profile constitutes a therapeutic opportunity. Our results indicate that chemotherapy can be used to target mMDSCs in males, whereas IL1 pathway inhibitors can provide benefit to females via inhibition of gMDSCs.See related commentary by Gabrilovich et al., p. 1100.This article is highlighted in the In This Issue feature, p. 1079.
Assuntos

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Neoplasias Encefálicas / Caracteres Sexuais / Glioblastoma / Células Supressoras Mieloides Tipo de estudo: Prognostic_studies / Risk_factors_studies Limite: Animals / Female / Humans / Male Idioma: En Ano de publicação: 2020 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Neoplasias Encefálicas / Caracteres Sexuais / Glioblastoma / Células Supressoras Mieloides Tipo de estudo: Prognostic_studies / Risk_factors_studies Limite: Animals / Female / Humans / Male Idioma: En Ano de publicação: 2020 Tipo de documento: Article