Your browser doesn't support javascript.
loading
Immunogenetic Metabolomics Reveals Key Enzymes That Modulate CAR T-cell Metabolism and Function.
Renauer, Paul; Park, Jonathan J; Bai, Meizhu; Acosta, Arianny; Lee, Won-Ho; Lin, Guang Han; Zhang, Yueqi; Dai, Xiaoyun; Wang, Guangchuan; Errami, Youssef; Wu, Terence; Clark, Paul; Ye, Lupeng; Yang, Quanjun; Chen, Sidi.
Afiliação
  • Renauer P; Department of Genetics, Yale University School of Medicine, New Haven, Connecticut.
  • Park JJ; System Biology Institute, Yale University, West Haven, Connecticut.
  • Bai M; Center for Cancer Systems Biology, Yale University, West Haven, Connecticut.
  • Acosta A; Combined Program in the Biological and Biomedical Sciences, Yale University, New Haven, Connecticut.
  • Lee WH; Molecular Cell Biology, Genetics, and Development Program, Yale University, New Haven, Connecticut.
  • Lin GH; Department of Genetics, Yale University School of Medicine, New Haven, Connecticut.
  • Zhang Y; System Biology Institute, Yale University, West Haven, Connecticut.
  • Dai X; Center for Cancer Systems Biology, Yale University, West Haven, Connecticut.
  • Wang G; Molecular Cell Biology, Genetics, and Development Program, Yale University, New Haven, Connecticut.
  • Errami Y; M.D.-Ph.D. Program, Yale University, West Haven, Connecticut.
  • Wu T; Department of Genetics, Yale University School of Medicine, New Haven, Connecticut.
  • Clark P; System Biology Institute, Yale University, West Haven, Connecticut.
  • Ye L; Center for Cancer Systems Biology, Yale University, West Haven, Connecticut.
  • Yang Q; Department of Genetics, Yale University School of Medicine, New Haven, Connecticut.
  • Chen S; System Biology Institute, Yale University, West Haven, Connecticut.
Cancer Immunol Res ; 11(8): 1068-1084, 2023 08 03.
Article em En | MEDLINE | ID: mdl-37253111
ABSTRACT
Immune evasion is a critical step of cancer progression that remains a major obstacle for current T cell-based immunotherapies. Hence, we investigated whether it is possible to genetically reprogram T cells to exploit a common tumor-intrinsic evasion mechanism whereby cancer cells suppress T-cell function by generating a metabolically unfavorable tumor microenvironment (TME). In an in silico screen, we identified ADA and PDK1 as metabolic regulators. We then showed that overexpression (OE) of these genes enhanced the cytolysis of CD19-specific chimeric antigen receptor (CAR) T cells against cognate leukemia cells, and conversely, ADA or PDK1 deficiency dampened this effect. ADA-OE in CAR T cells improved cancer cytolysis under high concentrations of adenosine, the ADA substrate, and an immunosuppressive metabolite in the TME. High-throughput transcriptomics and metabolomics analysis of these CAR T cells revealed alterations of global gene expression and metabolic signatures in both ADA- and PDK1-engineered CAR T cells. Functional and immunologic analyses demonstrated that ADA-OE increased proliferation and decreased exhaustion in CD19-specific and HER2-specific CAR T cells. ADA-OE improved tumor infiltration and clearance by HER2-specific CAR T cells in an in vivo colorectal cancer model. Collectively, these data unveil systematic knowledge of metabolic reprogramming directly in CAR T cells and reveal potential targets for improving CAR T-cell therapy.
Assuntos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Linfócitos T / Neoplasias Limite: Humans Idioma: En Ano de publicação: 2023 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Linfócitos T / Neoplasias Limite: Humans Idioma: En Ano de publicação: 2023 Tipo de documento: Article