Inhibition of ATM increases interferon signaling and sensitizes pancreatic cancer to immune checkpoint blockade therapy

Q Zhang, MD Green, X Lang, J Lazarus, JD Parsels… - Cancer research, 2019 - AACR
Q Zhang, MD Green, X Lang, J Lazarus, JD Parsels, S Wei, LA Parsels, J Shi, N Ramnath
Cancer research, 2019AACR
Combinatorial strategies are needed to overcome the resistance of pancreatic cancer to
immune checkpoint blockade (ICB). DNA damage activates the innate immune response
and improves ICB efficacy. Because ATM is an apical kinase in the radiation-induced DNA
damage response, we investigated the effects of ATM inhibition and radiation on pancreatic
tumor immunogenicity. ATM was inhibited through pharmacologic and genetic strategies in
human and murine pancreatic cancer models both in vitro and in vivo. Tumor …
Abstract
Combinatorial strategies are needed to overcome the resistance of pancreatic cancer to immune checkpoint blockade (ICB). DNA damage activates the innate immune response and improves ICB efficacy. Because ATM is an apical kinase in the radiation-induced DNA damage response, we investigated the effects of ATM inhibition and radiation on pancreatic tumor immunogenicity. ATM was inhibited through pharmacologic and genetic strategies in human and murine pancreatic cancer models both in vitro and in vivo. Tumor immunogenicity was evaluated after ATM inhibition alone and in combination with radiation by assessing TBK1 and Type I interferon (T1IFN) signaling as well as tumor growth following PD-L1/PD-1 checkpoint inhibition. Inhibition of ATM increased tumoral T1IFN expression in a cGAS/STING-independent, but TBK1- and SRC-dependent, manner. The combination of ATM inhibition with radiation further enhanced TBK1 activity, T1IFN production, and antigen presentation. Furthermore, ATM silencing increased PD-L1 expression and increased the sensitivity of pancreatic tumors to PD-L1–blocking antibody in association with increased tumoral CD8+ T cells and established immune memory. In patient pancreatic tumors, low ATM expression inversely correlated with PD-L1 expression. Taken together, these results demonstrate that the efficacy of ICB in pancreatic cancer is enhanced by ATM inhibition and further potentiated by radiation as a function of increased tumoral immunogenicity, underscoring the potential of ATM inhibition in combination with ICB and radiation as an efficacious treatment strategy for pancreatic cancer.
Significance
This study demonstrates that ATM inhibition induces a T1IFN-mediated innate immune response in pancreatic cancer that is further enhanced by radiation and leads to increased sensitivity to anti–PD-L1 therapy.
See related commentary by Gutiontov and Weichselbaum, p. 3815
AACR