To enable TCR activation-dependent cytokine signaling and improve therapeutic T cell functionality without systemic toxicity, Kim et al. modified NY-ESO-1 TCR-transgenic T cells with CRISPR/Cas9 for site-specific insertion of IL-12, replacing PD-1, in the PDCD1 locus. The engineered cells demonstrated antigen-specific IL-12 secretion, loss of PD-1, and tighter control of gene expression than with an NFAT promoter. The IL-12 knock-in improved in vitro T cell cytotoxicity and proliferative capacity after repetitive antigen stimulation, as well as in vivo tumor infiltration and efficacy, over NY-ESO-1 T cell controls or with those with PD-1 knockout alone.

Contributed by Alex Najibi

INTRODUCTION: Although the engineering of T cells to co-express immunostimulatory cytokines has been shown to enhance the therapeutic efficacy of adoptive T cell therapy, the uncontrolled systemic release of potent cytokines can lead to severe adverse effects. To address this, we site-specifically inserted the interleukin-12 (IL-12) gene into the PDCD1 locus in T cells using clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9)-based genome editing to achieve T-cell activation-dependent expression of IL-12 while ablating the expression of inhibitory PD-1. METHODS: New York esophageal squamous cell carcinoma 1(NY-ESO-1)-specific TCR-T cells was investigated as a model system. We generated _PD-1-IL-12 -edited NY-ESO-1 TCR-T cells by sequential lentiviral transduction and CRISPR knock-in into activated human primary T cells. RESULTS: We showed that the endogenous PDCD1 regulatory elements can tightly control the secretion of recombinant IL-12 in a target cell-dependent manner, at an expression level that is more moderate than that obtained using a synthetic NFAT-responsive promoter. The inducible expression of IL-12 from the PDCD1 locus was sufficient to enhance the effector function of NY-ESO-1 TCR-T cells, as determined by upregulation of effector molecules, increased cytotoxic activity, and enhanced expansion upon repeated antigen stimulation in vitro. Mouse xenograft studies also revealed that PD-1-edited IL-12-secreting NY-ESO-1 TCR-T cells could eliminate established tumors and showed significantly greater in vivo expansion capacity than control TCR-T cells. DISCUSSION: Our approach may provide a way to safely harness the therapeutic potential of potent immunostimulatory cytokines for the development of effective adoptive T cell therapies against solid tumors.

Author Info: (1) Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea. (2) Department of Biological Sciences, Korea Advanced Institu

Author Info: (1) Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea. (2) Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea. (3) Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea. (4) Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea. (5) Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea.