Stereotactic radiotherapy increases functionally suppressive regulatory T cells in the tumor microenvironment

Y Muroyama, TR Nirschl, CM Kochel… - Cancer immunology …, 2017 - AACR
Y Muroyama, TR Nirschl, CM Kochel, Z Lopez-Bujanda, D Theodros, W Mao…
Cancer immunology research, 2017AACR
Radiotherapy (RT) enhances innate and adaptive antitumor immunity; however, the effects
of radiation on suppressive immune cells, such as regulatory T cells (Treg), in the tumor
microenvironment (TME) are not fully elucidated. Although previous reports suggest an
increased Treg infiltration after radiation, whether these Tregs are functionally suppressive
remains undetermined. To test the hypothesis that RT enhances the suppressive function of
Treg in the TME, we selectively irradiated implanted tumors using the small animal radiation …
Abstract
Radiotherapy (RT) enhances innate and adaptive antitumor immunity; however, the effects of radiation on suppressive immune cells, such as regulatory T cells (Treg), in the tumor microenvironment (TME) are not fully elucidated. Although previous reports suggest an increased Treg infiltration after radiation, whether these Tregs are functionally suppressive remains undetermined. To test the hypothesis that RT enhances the suppressive function of Treg in the TME, we selectively irradiated implanted tumors using the small animal radiation research platform (SARRP), which models stereotactic radiotherapy in human patients. We then analyzed tumor-infiltrating lymphocytes (TIL) with flow-cytometry and functional assays. Our data showed that RT significantly increased tumor-infiltrating Tregs (TIL-Treg), which had higher expression of CTLA-4, 4-1BB, and Helios compared with Tregs in nonirradiated tumors. This observation held true across several tumor models (B16/F10, RENCA, and MC38). We found that TIL-Tregs from irradiated tumors had equal or improved suppressive capacity compared with nonirradiated tumors. Our data also indicated that after RT, Tregs proliferated more robustly than other T-cell subsets in the TME. In addition, after RT, expansion of Tregs occurred when T-cell migration was inhibited using Fingolimod, suggesting that the increased Treg frequency was likely due to preferential proliferation of intratumoral Treg after radiation. Our data also suggested that Treg expansion after irradiation was independent of TGFβ and IL33 signaling. These data demonstrate that RT increased phenotypically and functionally suppressive Tregs in the TME. Our results suggest that RT might be combined effectively with Treg-targeting agents to maximize antitumor efficacy. Cancer Immunol Res; 5(11); 992–1004. ©2017 AACR.
AACR