Your browser doesn't support javascript.
loading
Spatial and functional targeting of intratumoral Tregs reverses CD8+ T cell exhaustion and promotes cancer immunotherapy.
Zhou, Lei; Velegraki, Maria; Wang, Yi; Mandula, J K; Chang, Yuzhou; Liu, Weiwei; Song, No-Joon; Kwon, Hyunwoo; Xiao, Tong; Bolyard, Chelsea; Hong, Feng; Xin, Gang; Ma, Qin; Rubinstein, Mark P; Wen, Haitao; Li, Zihai.
Affiliation
  • Zhou L; Pelotonia Institute for Immuno-Oncology (PIIO), The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute (OSUCCC), Columbus, Ohio, USA.
  • Velegraki M; Department of Anesthesiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
  • Wang Y; Pelotonia Institute for Immuno-Oncology (PIIO), The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute (OSUCCC), Columbus, Ohio, USA.
  • Mandula JK; Pelotonia Institute for Immuno-Oncology (PIIO), The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute (OSUCCC), Columbus, Ohio, USA.
  • Chang Y; Molecular, Cellular and Developmental Biology Graduate Program, Ohio State University, Columbus, Ohio, USA.
  • Liu W; Pelotonia Institute for Immuno-Oncology (PIIO), The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute (OSUCCC), Columbus, Ohio, USA.
  • Song NJ; Pelotonia Institute for Immuno-Oncology (PIIO), The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute (OSUCCC), Columbus, Ohio, USA.
  • Kwon H; Department of Biomedical Informatics.
  • Xiao T; Pelotonia Institute for Immuno-Oncology (PIIO), The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute (OSUCCC), Columbus, Ohio, USA.
  • Bolyard C; Molecular, Cellular and Developmental Biology Graduate Program, Ohio State University, Columbus, Ohio, USA.
  • Hong F; Pelotonia Institute for Immuno-Oncology (PIIO), The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute (OSUCCC), Columbus, Ohio, USA.
  • Xin G; Pelotonia Institute for Immuno-Oncology (PIIO), The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute (OSUCCC), Columbus, Ohio, USA.
  • Ma Q; Department of Internal Medicine, Ohio State University College of Medicine, Columbus, USA.
  • Rubinstein MP; Pelotonia Institute for Immuno-Oncology (PIIO), The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute (OSUCCC), Columbus, Ohio, USA.
  • Wen H; Molecular, Cellular and Developmental Biology Graduate Program, Ohio State University, Columbus, Ohio, USA.
  • Li Z; Pelotonia Institute for Immuno-Oncology (PIIO), The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute (OSUCCC), Columbus, Ohio, USA.
J Clin Invest ; 134(14)2024 May 23.
Article de En | MEDLINE | ID: mdl-38787791
ABSTRACT
Intratumoral Tregs are key mediators of cancer immunotherapy resistance, including anti-programmed cell death (ligand) 1 [anti-PD-(L)1] immune checkpoint blockade (ICB). The mechanisms driving Treg infiltration into the tumor microenvironment (TME) and the consequence on CD8+ T cell exhaustion remain elusive. Here, we report that heat shock protein gp96 (also known as GRP94) was indispensable for Treg tumor infiltration, primarily through the roles of gp96 in chaperoning integrins. Among various gp96-dependent integrins, we found that only LFA-1 (αL integrin), and not αV, CD103 (αE), or ß7 integrin, was required for Treg tumor homing. Loss of Treg infiltration into the TME by genetic deletion of gp96/LFA-1 potently induced rejection of tumors in multiple ICB-resistant murine cancer models in a CD8+ T cell-dependent manner, without loss of self-tolerance. Moreover, gp96 deletion impeded Treg activation primarily by suppressing IL-2/STAT5 signaling, which also contributed to tumor regression. By competing for intratumoral IL-2, Tregs prevented the activation of CD8+ tumor-infiltrating lymphocytes, drove thymocyte selection-associated high mobility group box protein (TOX) induction, and induced bona fide CD8+ T cell exhaustion. By contrast, Treg ablation led to striking CD8+ T cell activation without TOX induction, demonstrating clear uncoupling of the 2 processes. Our study reveals that the gp96/LFA-1 axis plays a fundamental role in Treg biology and suggests that Treg-specific gp96/LFA-1 targeting represents a valuable strategy for cancer immunotherapy without inflicting autoinflammatory conditions.
Sujet(s)
Mots clés

Texte intégral: 1 Collection: 01-internacional Base de données: MEDLINE Sujet principal: Lymphocytes T régulateurs / Lymphocytes T CD8/ / Microenvironnement tumoral / Immunothérapie Limites: Animals / Humans Langue: En Journal: J Clin Invest Année: 2024 Type de document: Article Pays d'affiliation: États-Unis d'Amérique

Texte intégral: 1 Collection: 01-internacional Base de données: MEDLINE Sujet principal: Lymphocytes T régulateurs / Lymphocytes T CD8/ / Microenvironnement tumoral / Immunothérapie Limites: Animals / Humans Langue: En Journal: J Clin Invest Année: 2024 Type de document: Article Pays d'affiliation: États-Unis d'Amérique