Your browser doesn't support javascript.
loading
EGFR-Driven Lung Adenocarcinomas Co-opt Alveolar Macrophage Metabolism and Function to Support EGFR Signaling and Growth.
Kuhlmann-Hogan, Alexandra; Cordes, Thekla; Xu, Ziyan; Kuna, Ramya S; Traina, Kacie A; Robles-Oteíza, Camila; Ayeni, Deborah; Kwong, Elizabeth M; Levy, Stellar; Globig, Anna-Maria; Nobari, Matthew M; Cheng, George Z; Leibel, Sandra L; Homer, Robert J; Shaw, Reuben J; Metallo, Christian M; Politi, Katerina; Kaech, Susan M.
Afiliação
  • Kuhlmann-Hogan A; Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut.
  • Cordes T; NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, California.
  • Xu Z; Molecular and Cellular Biology Laboratory, Salk Institute for Biological Studies, La Jolla, California.
  • Kuna RS; Department of Bioinformatics and Biochemistry, Braunshweig Integrated Centre of Systems Biology (BRICS), Technishe Universität Braunschweig, Germany.
  • Traina KA; Research Group Cellular Metabolism in Infection, Helmholtz Centre for Infection Research, Braunschweig, Germany.
  • Robles-Oteíza C; NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, California.
  • Ayeni D; Division of Biological Sciences, University of California San Diego, La Jolla, California.
  • Kwong EM; Molecular and Cellular Biology Laboratory, Salk Institute for Biological Studies, La Jolla, California.
  • Levy S; NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, California.
  • Globig AM; Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut.
  • Nobari MM; Departments of Pathology and Internal Medicine (Section of Medical Oncology), Yale School of Medicine, New Haven, Connecticut.
  • Cheng GZ; Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California.
  • Leibel SL; Sanford Consortium for Regenerative Medicine, La Jolla, California.
  • Homer RJ; Departments of Pathology and Internal Medicine (Section of Medical Oncology), Yale School of Medicine, New Haven, Connecticut.
  • Shaw RJ; NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, California.
  • Metallo CM; Division of Pulmonary and Critical Sleep Medicine, University of California San Diego, Department of Medicine, La Jolla, California.
  • Politi K; Division of Pulmonary and Critical Sleep Medicine, University of California San Diego, Department of Medicine, La Jolla, California.
  • Kaech SM; Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, California.
Cancer Discov ; : OF1-OF22, 2024 Jan 25.
Article em En | MEDLINE | ID: mdl-38270272
ABSTRACT
The limited efficacy of currently approved immunotherapies in EGFR-driven lung adenocarcinoma (LUAD) underscores the need to better understand alternative mechanisms governing local immunosuppression to fuel novel therapies. Elevated surfactant and GM-CSF secretion from the transformed epithelium induces tumor-associated alveolar macrophage (TA-AM) proliferation, which supports tumor growth by rewiring inflammatory functions and lipid metabolism. TA-AM properties are driven by increased GM-CSF-PPARγ signaling and inhibition of airway GM-CSF or PPARγ in TA-AMs suppresses cholesterol efflux to tumor cells, which impairs EGFR phosphorylation and restrains LUAD progression. In the absence of TA-AM metabolic support, LUAD cells compensate by increasing cholesterol synthesis, and blocking PPARγ in TA-AMs simultaneous with statin therapy further suppresses tumor progression and increases proinflammatory immune responses. These results reveal new therapeutic combinations for immunotherapy-resistant EGFR-mutant LUADs and demonstrate how cancer cells can metabolically co-opt TA-AMs through GM-CSF-PPARγ signaling to provide nutrients that promote oncogenic signaling and growth.

SIGNIFICANCE:

Alternate strategies harnessing anticancer innate immunity are required for lung cancers with poor response rates to T cell-based immunotherapies. This study identifies a targetable, mutually supportive, metabolic relationship between macrophages and transformed epithelium, which is exploited by tumors to obtain metabolic and immunologic support to sustain proliferation and oncogenic signaling.

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Idioma: En Revista: Cancer Discov Ano de publicação: 2024 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Idioma: En Revista: Cancer Discov Ano de publicação: 2024 Tipo de documento: Article