Abstract

Adoptive T cell therapy (ACT) has been established as an efficacious methodology for the treatment of cancer. Identifying targets to enhance the antigen recognition, functional capacity, and longevity of T cells has the potential to broaden the applicability of these approaches in the clinic. We previously reported that targeting expression of phosphotyrosine phosphatase, nonreceptor type 22 (PTPN22) in effector CD8+ T cells enhances the efficacy of ACT for tumor clearance in mice. In the current work, we demonstrate that, upon ACT, PTPN22-deficient effector CD8+ T cells afforded greater protection against tumors expressing very low-affinity antigen but did not survive long term in vivo. Persistence of CD8+ T cells following tumor clearance was improved by ACT of memory phenotype cells that have a distinct metabolic phenotype, as compared with effector T cells. Importantly, PTPN22-deficient T cells have comparable capacity to form long-lived memory cells in vivo but enhanced antitumor activity in vivo and effector responses ex vivo. These findings provide key insights into the regulation of effector and memory T cell responses in vivo and indicate that PTPN22 is a rational target to improve ACT for cancer.

Highlights

  • T cell–directed immunotherapy has delivered remarkable therapeutic benefits in cancer patients

  • We sought to determine the effect of PTPN22 deficiency on the capacity of CTLs to directly mediate target cell killing in response to strong and weak tumor-associated antigens (TAAs)

  • Previous studies have shown that T cell receptors (TCR) triggering influences expression of inhibitory phosphatases [15]; it was of interest to determine the levels of PTPN22 following stimulation of OT-1 T cells with weak and strong agonist peptides

Read more

Summary

Introduction

T cell–directed immunotherapy has delivered remarkable therapeutic benefits in cancer patients. A key consideration for ACT approaches to cancer therapy is to determine which T cell phenotype provides optimal long-lived protection. Upon transfer to naive recipient mice, very low numbers of long-lived Ptpn22–/– memory T cells provided enhanced protection from tumor challenge, as compared with control cells. Ptpn22–/–, but not control, memory phenotype T cell ACT could completely protect mice from low-affinity antigen-bearing tumors when transferred to hosts 2–4 weeks prior to tumor implantation. Together, these experiments have determined that deletion of PTPN22 represents a rational approach to enhance the functional capacity of both short-lived effector and long-lived memory T cells in antitumor immunity

Results
Discussion
Methods
Full Text
Paper version not known

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call