Epigenetic reprogramming of tumor cell–intrinsic STING function sculpts antigenicity and T cell recognition of melanoma

R Falahat, A Berglund, RM Putney… - Proceedings of the …, 2021 - National Acad Sciences
R Falahat, A Berglund, RM Putney, P Perez-Villarroel, S Aoyama, S Pilon-Thomas…
Proceedings of the National Academy of Sciences, 2021National Acad Sciences
Lack or loss of tumor antigenicity represents one of the key mechanisms of immune escape
and resistance to T cell–based immunotherapies. Evidence suggests that activation of
stimulator of interferon genes (STING) signaling in tumor cells can augment their antigenicity
by triggering a type I IFN-mediated sequence of autocrine and paracrine events. Although
suppression of this pathway in melanoma and other tumor types has been consistently
reported, the mechanistic basis remains unclear. In this study, we asked whether this …
Lack or loss of tumor antigenicity represents one of the key mechanisms of immune escape and resistance to T cell–based immunotherapies. Evidence suggests that activation of stimulator of interferon genes (STING) signaling in tumor cells can augment their antigenicity by triggering a type I IFN-mediated sequence of autocrine and paracrine events. Although suppression of this pathway in melanoma and other tumor types has been consistently reported, the mechanistic basis remains unclear. In this study, we asked whether this suppression is, in part, epigenetically regulated and whether it is indeed a driver of melanoma resistance to T cell–based immunotherapies. Using genome-wide DNA methylation profiling, we show that promoter hypermethylation of cGAS and STING genes mediates their coordinated transcriptional silencing and contributes to the widespread impairment of the STING signaling function in clinically-relevant human melanomas and melanoma cell lines. This suppression is reversible through pharmacologic inhibition of DNA methylation, which can reinstate functional STING signaling in at least half of the examined cell lines. Using a series of T cell recognition assays with HLA-matched human melanoma tumor-infiltrating lymphocytes (TIL), we further show that demethylation-mediated restoration of STING signaling in STING-defective melanoma cell lines can improve their antigenicity through the up-regulation of MHC class I molecules and thereby enhance their recognition and killing by cytotoxic T cells. These findings not only elucidate the contribution of epigenetic processes and specifically DNA methylation in melanoma-intrinsic STING signaling impairment but also highlight their functional significance in mediating tumor-immune evasion and resistance to T cell–based immunotherapies.
National Acad Sciences