关键词: ADU-S100 CD8(+) T cell ImmunoOncology STING abscopal immunity anti-tumor immunity checkpoint inhibitor cyclic dinucleotide intratumoral type I interferon

Mesh : Animals CD8-Positive T-Lymphocytes / immunology CTLA-4 Antigen / metabolism Cell Line, Tumor Cytokines / metabolism Dose-Response Relationship, Immunologic Drug Resistance, Neoplasm Hematopoiesis Immunity Membrane Proteins / metabolism Mice, Inbred BALB C Mice, Inbred C57BL Neoplasms / immunology pathology Programmed Cell Death 1 Receptor / metabolism S100 Proteins / administration & dosage immunology

来  源:   DOI:10.1016/j.celrep.2018.11.047   PDF(Sci-hub)

Abstract:
Intratumoral (IT) STING activation results in tumor regression in preclinical models, yet factors dictating the balance between innate and adaptive anti-tumor immunity are unclear. Here, clinical candidate STING agonist ADU-S100 (S100) is used in an IT dosing regimen optimized for adaptive immunity to uncover requirements for a T cell-driven response compatible with checkpoint inhibitors (CPIs). In contrast to high-dose tumor ablative regimens that result in systemic S100 distribution, low-dose immunogenic regimens induce local activation of tumor-specific CD8+ effector T cells that are responsible for durable anti-tumor immunity and can be enhanced with CPIs. Both hematopoietic cell STING expression and signaling through IFNAR are required for tumor-specific T cell activation, and in the context of optimized T cell responses, TNFα is dispensable for tumor control. In a poorly immunogenic model, S100 combined with CPIs generates a survival benefit and durable protection. These results provide fundamental mechanistic insights into STING-induced anti-tumor immunity.
摘要:
暂无翻译
公众号