Therapeutically active RIG-I agonist induces immunogenic tumor cell killing in breast cancers

DL Elion, ME Jacobson, DJ Hicks, B Rahman… - Cancer Research, 2018 - AACR
DL Elion, ME Jacobson, DJ Hicks, B Rahman, V Sanchez, PI Gonzales-Ericsson…
Cancer Research, 2018AACR
Cancer immunotherapies that remove checkpoint restraints on adaptive immunity are
gaining clinical momentum but have not achieved widespread success in breast cancers, a
tumor type considered poorly immunogenic and which harbors a decreased presence of
tumor-infiltrating lymphocytes. Approaches that activate innate immunity in breast cancer
cells and the tumor microenvironment are of increasing interest, based on their ability to
induce immunogenic tumor cell death, type I IFNs, and lymphocyte-recruiting chemokines. In …
Abstract
Cancer immunotherapies that remove checkpoint restraints on adaptive immunity are gaining clinical momentum but have not achieved widespread success in breast cancers, a tumor type considered poorly immunogenic and which harbors a decreased presence of tumor-infiltrating lymphocytes. Approaches that activate innate immunity in breast cancer cells and the tumor microenvironment are of increasing interest, based on their ability to induce immunogenic tumor cell death, type I IFNs, and lymphocyte-recruiting chemokines. In agreement with reports in other cancers, we observe loss, downregulation, or mutation of the innate viral nucleotide sensor retinoic acid-inducible gene I (RIG-I/DDX58) in only 1% of clinical breast cancers, suggesting potentially widespread applicability for therapeutic RIG-I agonists that activate innate immunity. This was tested using an engineered RIG-I agonist in a breast cancer cell panel representing each of three major clinical breast cancer subtypes. Treatment with RIG-I agonist resulted in upregulation and mitochondrial localization of RIG-I and activation of proinflammatory transcription factors STAT1 and NF-κB. RIG-I agonist triggered the extrinsic apoptosis pathway and pyroptosis, a highly immunogenic form of cell death in breast cancer cells. RIG-I agonist also induced expression of lymphocyte-recruiting chemokines and type I IFN, confirming that cell death and cytokine modulation occur in a tumor cell–intrinsic manner. Importantly, RIG-I activation in breast tumors increased tumor lymphocytes and decreased tumor growth and metastasis. Overall, these findings demonstrate successful therapeutic delivery of a synthetic RIG-I agonist to induce tumor cell killing and to modulate the tumor microenvironment in vivo.
Significance: These findings describe the first in vivo delivery of RIG-I mimetics to tumors, demonstrating a potent immunogenic and therapeutic effect in the context of otherwise poorly immunogenic breast cancers. Cancer Res; 78(21); 6183–95. ©2018 AACR.
AACR