Login / Signup

An oncolytic virus-delivered TGFβ inhibitor overcomes the immunosuppressive tumor microenvironment.

Kristin DePeauxDayana B RivadeneiraKonstantinos LontosVictoria G DeanWilliam G GunnMcLane J WatsonTianhong YaoDrew WilfahrtCynthia S HinckLukasz WieteskaStephen H ThorneAndrew P HinckGreg M Delgoffe
Published in: The Journal of experimental medicine (2023)
While checkpoint blockade immunotherapies have widespread success, they rely on a responsive immune infiltrate; as such, treatments enhancing immune infiltration and preventing immunosuppression are of critical need. We previously generated αPD-1 resistant variants of the murine HNSCC model MEER. While entirely αPD-1 resistant, these tumors regress after single dose of oncolytic vaccinia virus (VV). We then generated a VV-resistant MEER line to dissect the immunologic features of sensitive and resistant tumors. While treatment of both tumor types induced immune infiltration and IFNγ, we found a defining feature of resistance was elevation of immunosuppressive cytokines like TGFβ, which blunted IFNγ signaling, especially in regulatory T cells. We engineered VV to express a genetically encoded TGFβRII inhibitor. Inhibitor-expressing VV produced regressions in resistant tumor models and showed impressive synergy with checkpoint blockade. Importantly, tumor-specific, viral delivery of TGFβ inhibition had no toxicities associated with systemic TGFβ/TGFβR inhibition. Our data suggest that aside from stimulating immune infiltration, oncolytic viruses are attractive means to deliver agents to limit immunosuppression in cancer.
Keyphrases