[HTML][HTML] Overcoming resistance to STING agonist therapy to incite durable protective antitumor immunity

H Lemos, R Ou, C McCardle, Y Lin… - … for Immunotherapy of …, 2020 - ncbi.nlm.nih.gov
H Lemos, R Ou, C McCardle, Y Lin, J Calver, J Minett, A Chadli, L Huang, AL Mellor
Journal for Immunotherapy of Cancer, 2020ncbi.nlm.nih.gov
Background Activating the Stimulator of Interferon Genes (STING) adaptor incites antitumor
immunity against immunogenic tumors in mice, prompting clinical trials to test STING
activators. However, STING signaling in the tumor microenvironment (TME) during
development of Lewis lung carcinoma (LLC) suppresses antitumor immunity to promote
tumor growth. We hypothesized that local immune balance favoring suppression of
antitumor immunity also attenuates antitumor responses following STING activation. The …
Abstract
Background
Activating the Stimulator of Interferon Genes (STING) adaptor incites antitumor immunity against immunogenic tumors in mice, prompting clinical trials to test STING activators. However, STING signaling in the tumor microenvironment (TME) during development of Lewis lung carcinoma (LLC) suppresses antitumor immunity to promote tumor growth. We hypothesized that local immune balance favoring suppression of antitumor immunity also attenuates antitumor responses following STING activation. The purpose of this study was to evaluate how STING activation impacts antitumor responses in mice bearing LLC tumors.
Methods
Mice bearing established LLC tumors were treated with synthetic cyclic diadenyl monophosphate (CDA) to activate STING. Mice were monitored to assess LLC tumor growth, survival and protective antitumor immunity. Transcriptional and metabolic analyses were used to identify pathways responsive to CDA, and mice were co-treated with CDA and drugs that disrupt these pathways.
Results
CDA slowed LLC tumor growth but most CDA-treated mice (77%) succumbed to tumor growth. No evidence of tumor relapse was found in surviving CDA-treated mice at experimental end points but mice were not immune to LLC challenge. CDA induced rapid increase in immune regulatory pathways involving programmed death-1 (PD-1), indoleamine 2, 3 dioxygenase (IDO) and cyclooxygenase-2 (COX2) in the TME. PD-1 blockade enhanced antitumor responses to CDA and increased mouse survival but mice did not eliminate primary tumor burdens. Two IDO inhibitor drugs had little or no beneficial effects on antitumor responses to CDA. A third IDO inhibitor drug synergized with CDA to enhance tumor control and survival but mice did not eliminate primary tumor burdens. In contrast, co-treatments with CDA and the COX2-selective inhibitor celecoxib controlled tumor growth, leading to uniform survival without relapse, and mice acquired resistance to LLC re-challenge and growth of distal tumors not exposed directly to CDA. Thus, mice co-treated with CDA and celecoxib acquired stable and systemic antitumor immunity.
Conclusions
STING activation incites potent antitumor responses and boosts local immune regulation to attenuate antitumor responses. Blocking STING-responsive regulatory pathways synergizes with CDA to enhance antitumor responses, particularly COX2 inhibition. Thus, therapy-induced resistance to STING may necessitate co-treatments to disrupt regulatory pathways responsive to STING in patients with cancer.
ncbi.nlm.nih.gov