Abstract

Agonistic monoclonal antibodies (mAb) targeting the T-cell receptor coregulatory molecule GITR exert potent therapeutic activities in preclinical tumor models. Although anti-GITR mAb are thought to act by depleting and destabilizing the intratumoral T regulatory cell (Treg) population, the precise mechanism of action is obscure. Here, we addressed this issue using a Treg fate-mapping approach, which revealed that Treg loss was primarily due to cell depletion, with minimal evidence of Treg conversion to a non-Foxp3-expressing population. Further characterization of persisting Tregs following anti-GITR mAb treatment showed that a highly activated subpopulation of CD44hiICOShi intratumoral Tregs were preferentially targeted for elimination, with the remaining Tregs exhibiting a less suppressive phenotype. With these changes in the Treg population, intratumoral CD8+ T cells acquired a more functional phenotype characterized by downregulation of the exhaustion markers PD-1 and LAG-3. This reversal of CD8+ T-cell exhaustion was dependent on both agonistic GITR signaling and Treg depletion, as neither mechanism by itself could fully rescue the exhaustion phenotype. Tests of anti-human GITR antibody MK-4166 in a humanized mouse model of cancer mimicked many of the effects of anti-mouse GITR mAb in syngeneic tumor models, decreasing both Treg numbers and immune suppressor phenotype while enhancing effector responsiveness. Overall, our results show how anti-GITR mAb shifts Treg populations to enable immune attack on tumors, with clinical implications for molecular markers to modify emerging treatments. Cancer Res; 77(5); 1108-18. ©2016 AACR.

Highlights

  • Glucocorticoid-induced TNFR family-related protein (GITR, TNFRSF18, CD357) is a costimulatory immune modulating receptor expressed on various immune cell subsets, including T cells, natural killer (NK) cells, and B cells, with high expression on regulatory T cells (Tregs; refs. 1–3)

  • We have demonstrated that mouse DTA-1 IgG2a (mDTA-1) treatment in the MC38 tumor model leads to a tumor-specific reduction in T regulatory cell (Treg) numbers and in the functional status of those Tregs that persist

  • Upon examination of intratumoral CD8þ T cells following mDTA-1 treatment, we observed a reversal of an exhaustion phenotype that could not be reproduced with either Treg depletion or agonistic GITR signaling alone

Read more

Summary

Introduction

Glucocorticoid-induced TNFR family-related protein (GITR, TNFRSF18, CD357) is a costimulatory immune modulating receptor expressed on various immune cell subsets, including T cells, natural killer (NK) cells, and B cells, with high expression on regulatory T cells (Tregs; refs. 1–3). A widely studied anti-GITR agonistic monoclonal antibody (mAb), clone DTA-1, has been shown to have costimulatory effects on T cells in vitro and in vivo [4] and potent antitumor efficacy in multiple mouse syngeneic tumor models [5, 6]. Previous studies investigating the mechanism of action behind the antitumor efficacy of DTA-1 have focused heavily on the impact on Tregs, a key population involved in the inhibition of antitumor immune responses in a range of solid tumor types

Methods
Results
Conclusion
Full Text
Published version (Free)

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