Your browser doesn't support javascript.
loading
IL12 integrated into the CAR exodomain converts CD8+ T cells to poly-functional NK-like cells with superior killing of antigen-loss tumors.
Hombach, Andreas; Barden, Markus; Hannappel, Lisa; Chmielewski, Markus; Rappl, Gunter; Sachinidis, Agapios; Abken, Hinrich.
Afiliação
  • Hombach A; Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany; Department I Internal Medicine, University Hospital Cologne, 50931 Cologne, Germany.
  • Barden M; RCI, Regensburg Center for Interventional Immunology, Department Genetic Immunotherapy, and University Hospital Regensburg, 93053 Regensburg, Germany.
  • Hannappel L; Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany.
  • Chmielewski M; Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany; Department I Internal Medicine, University Hospital Cologne, 50931 Cologne, Germany.
  • Rappl G; Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany.
  • Sachinidis A; Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany; University of Cologne, Faculty of Medicine and Center for Physiology, University Hospital Cologne, 50931 Cologne, Germany.
  • Abken H; RCI, Regensburg Center for Interventional Immunology, Department Genetic Immunotherapy, and University Hospital Regensburg, 93053 Regensburg, Germany. Electronic address: hinrich.abken@ukr.de.
Mol Ther ; 30(2): 593-605, 2022 02 02.
Article em En | MEDLINE | ID: mdl-34678512
ABSTRACT
Chimeric antigen receptor (CAR)-redirected T cell therapy often fails to control tumors in the long term due to selecting cancer cells that downregulated or lost CAR targeted antigen. To reprogram the functional capacities specifically of engineered CARcells, we inserted IL12 into the extracellular moiety of a CD28-ζ CAR; both the CAR endodomain and IL12 were functionally active, as indicated by antigen-redirected effector functions and STAT4 phosphorylation, respectively. The IL12-CAR reprogrammed CD8+ T cells toward a so far not recognized natural killer (NK) cell-like signature and a CD94+CD56+CD62Lhigh phenotype closely similar, but not identical, to NK and cytokine induced killer (CIK) cells. In contrast to conventional CARcells, IL12-CARcells acquired antigen-independent, human leukocyte antigen E (HLA-E) restricted cytotoxic capacities eliminating antigen-negative cancer cells in addition to eliminating cancer cells with CAR cognate antigen. Simultaneous signaling through both the CAR endodomain and IL12 were required for inducing maximal NK-like cytotoxicity; adding IL12 to conventional CARcells was not sufficient. Antigen-negative tumors were attacked by IL12-CARcells, but not by conventional CARcells. Overall, we present a prototype of a new family of CARs that augments tumor recognition and elimination through expanded functional capacities by an appropriate cytokine integrated into the CAR exodomain.
Assuntos
Palavras-chave

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Imunoterapia Adotiva / Interleucina-12 / Linfócitos T CD8-Positivos / Neoplasias Limite: Humans Idioma: En Ano de publicação: 2022 Tipo de documento: Article

Texto completo: 1 Base de dados: MEDLINE Assunto principal: Imunoterapia Adotiva / Interleucina-12 / Linfócitos T CD8-Positivos / Neoplasias Limite: Humans Idioma: En Ano de publicação: 2022 Tipo de documento: Article