Your browser doesn't support javascript.
loading
CD38-RyR2 axis-mediated signaling impedes CD8+ T cell response to anti-PD1 therapy in cancer.
Kar, Anwesha; Ghosh, Puspendu; Gautam, Anupam; Chowdhury, Snehanshu; Basak, Debashree; Sarkar, Ishita; Bhoumik, Arpita; Barman, Shubhrajit; Chakraborty, Paramita; Mukhopadhyay, Asima; Mehrotra, Shikhar; Ganesan, Senthil Kumar; Paul, Sandip; Chatterjee, Shilpak.
Afiliação
  • Kar A; Division of Cancer Biology and Inflammatory Disorder, Translational Research Unit of Excellence, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata 700032, West Bengal, India.
  • Ghosh P; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India.
  • Gautam A; Division of Cancer Biology and Inflammatory Disorder, Translational Research Unit of Excellence, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata 700032, West Bengal, India.
  • Chowdhury S; Algorithms in Bioinformatics, Institute for Bioinformatics and Medical Informatics, University of Tübingen, Sand 14 72076, Tübingen, Baden-Württemberg, Germany.
  • Basak D; International Max Planck Research School "From Molecules to Organisms", Max Planck Institute for Biology Tübingen 72076, Tübingen, Baden-Württemberg, Germany.
  • Sarkar I; Division of Cancer Biology and Inflammatory Disorder, Translational Research Unit of Excellence, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata 700032, West Bengal, India.
  • Bhoumik A; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India.
  • Barman S; Division of Cancer Biology and Inflammatory Disorder, Translational Research Unit of Excellence, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata 700032, West Bengal, India.
  • Chakraborty P; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India.
  • Mukhopadhyay A; Division of Cancer Biology and Inflammatory Disorder, Translational Research Unit of Excellence, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata 700032, West Bengal, India.
  • Mehrotra S; Division of Cancer Biology and Inflammatory Disorder, Translational Research Unit of Excellence, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata 700032, West Bengal, India.
  • Ganesan SK; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India.
  • Paul S; Division of Structural Biology & Bioinformatics, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Kolkata 700032, West Bengal, India.
  • Chatterjee S; Department of Surgery, Medical University of South Carolina, Charleston, South Carolina SC- 29425.
Proc Natl Acad Sci U S A ; 121(11): e2315989121, 2024 Mar 12.
Article em En | MEDLINE | ID: mdl-38451948
ABSTRACT
PD1 blockade therapy, harnessing the cytotoxic potential of CD8+ T cells, has yielded clinical success in treating malignancies. However, its efficacy is often limited due to the progressive differentiation of intratumoral CD8+ T cells into a hypofunctional state known as terminal exhaustion. Despite identifying CD8+ T cell subsets associated with immunotherapy resistance, the molecular pathway triggering the resistance remains elusive. Given the clear association of CD38 with CD8+ T cell subsets resistant to anti-PD1 therapy, we investigated its role in inducing resistance. Phenotypic and functional characterization, along with single-cell RNA sequencing analysis of both in vitro chronically stimulated and intratumoral CD8+ T cells, revealed that CD38-expressing CD8+ T cells are terminally exhausted. Exploring the molecular mechanism, we found that CD38 expression was crucial in promoting terminal differentiation of CD8+ T cells by suppressing TCF1 expression, thereby rendering them unresponsive to anti-PD1 therapy. Genetic ablation of CD38 in tumor-reactive CD8+ T cells restored TCF1 levels and improved the responsiveness to anti-PD1 therapy in mice. Mechanistically, CD38 expression on exhausted CD8+ T cells elevated intracellular Ca2+ levels through RyR2 calcium channel activation. This, in turn, promoted chronic AKT activation, leading to TCF1 loss. Knockdown of RyR2 or inhibition of AKT in CD8+ T cells maintained TCF1 levels, induced a sustained anti-tumor response, and enhanced responsiveness to anti-PD1 therapy. Thus, targeting CD38 represents a potential strategy to improve the efficacy of anti-PD1 treatment in cancer.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Linfócitos T CD8-Positivos / Neoplasias Limite: Animals Idioma: En Ano de publicação: 2024 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Linfócitos T CD8-Positivos / Neoplasias Limite: Animals Idioma: En Ano de publicação: 2024 Tipo de documento: Article