Your browser doesn't support javascript.
loading
Metabolic Reprogramming of Tumor-Associated Macrophages Using Glutamine Antagonist JHU083 Drives Tumor Immunity in Myeloid-Rich Prostate and Bladder Cancers.
Praharaj, Monali; Shen, Fan; Lee, Alex J; Zhao, Liang; Nirschl, Thomas R; Theodros, Debebe; Singh, Alok K; Wang, Xiaoxu; Adusei, Kenneth M; Lombardo, Kara A; Williams, Raekwon A; Sena, Laura A; Thompson, Elizabeth A; Tam, Ada; Yegnasubramanian, Srinivasan; Pearce, Edward J; Leone, Robert D; Alt, Jesse; Rais, Rana; Slusher, Barbara S; Pardoll, Drew M; Powell, Jonathan D; Zarif, Jelani C.
Afiliação
  • Praharaj M; Pathobiology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland.
  • Shen F; Bloomberg∼Kimmel Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.
  • Lee AJ; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
  • Zhao L; The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
  • Nirschl TR; Bloomberg∼Kimmel Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.
  • Theodros D; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
  • Singh AK; The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
  • Wang X; Bloomberg∼Kimmel Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.
  • Adusei KM; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
  • Lombardo KA; The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
  • Williams RA; Graduate Program in Immunology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
  • Sena LA; Bloomberg∼Kimmel Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.
  • Thompson EA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
  • Tam A; The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
  • Yegnasubramanian S; Pathobiology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, Maryland.
  • Pearce EJ; Bloomberg∼Kimmel Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.
  • Leone RD; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
  • Alt J; The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
  • Rais R; Bloomberg∼Kimmel Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland.
  • Slusher BS; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
  • Pardoll DM; The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
  • Powell JD; Graduate Program in Immunology, Johns Hopkins University School of Medicine, Baltimore, Maryland.
  • Zarif JC; Medical Scientist Training Program, Johns Hopkins University School of Medicine, Baltimore, Maryland.
Cancer Immunol Res ; 12(7): 854-875, 2024 Jul 02.
Article em En | MEDLINE | ID: mdl-38701369
ABSTRACT
Glutamine metabolism in tumor microenvironments critically regulates antitumor immunity. Using the glutamine-antagonist prodrug JHU083, we report potent tumor growth inhibition in urologic tumors by JHU083-reprogrammed tumor-associated macrophages (TAMs) and tumor-infiltrating monocytes. We show JHU083-mediated glutamine antagonism in tumor microenvironments induced by TNF, proinflammatory, and mTORC1 signaling in intratumoral TAM clusters. JHU083-reprogrammed TAMs also exhibited increased tumor cell phagocytosis and diminished proangiogenic capacities. In vivo inhibition of TAM glutamine consumption resulted in increased glycolysis, a broken tricarboxylic acid (TCA) cycle, and purine metabolism disruption. Although the antitumor effect of glutamine antagonism on tumor-infiltrating T cells was moderate, JHU083 promoted a stem cell-like phenotype in CD8+ T cells and decreased the abundance of regulatory T cells. Finally, JHU083 caused a global shutdown in glutamine-utilizing metabolic pathways in tumor cells, leading to reduced HIF-1α, c-MYC phosphorylation, and induction of tumor cell apoptosis, all key antitumor features. Altogether, our findings demonstrate that targeting glutamine with JHU083 led to suppressed tumor growth as well as reprogramming of immunosuppressive TAMs within prostate and bladder tumors that promoted antitumor immune responses. JHU083 can offer an effective therapeutic benefit for tumor types that are enriched in immunosuppressive TAMs.
Assuntos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Neoplasias da Próstata / Neoplasias da Bexiga Urinária / Microambiente Tumoral / Macrófagos Associados a Tumor / Glutamina Limite: Animals / Humans / Male Idioma: En Revista: Cancer Immunol Res Ano de publicação: 2024 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Neoplasias da Próstata / Neoplasias da Bexiga Urinária / Microambiente Tumoral / Macrófagos Associados a Tumor / Glutamina Limite: Animals / Humans / Male Idioma: En Revista: Cancer Immunol Res Ano de publicação: 2024 Tipo de documento: Article