Role of melanoma cell-intrinsic RIG-I and STING signaling for checkpoint inhibitor-mediated anticancer immunity.

Authors

null

Simon Heidegger

Klinikum rechts der Isar, Technical University Munich, Munich, Germany

Simon Heidegger , Alexander Wintges , Sarah Bek , Martina Schmickl , Julius Fischer , Paul-Albert Koenig , Tobias Haas , Hendrik Poeck

Organizations

Klinikum rechts der Isar, Technical University Munich, Munich, Germany, Department of Hematology and Oncology, University Hospital rechts der Isar, Technical University of Munich, Munich, Germany

Research Funding

Other Foundation

Background: Strong inter-individual variation in clinical response to immune checkpoint inhibitors (ICB) including anti-CTLA-4 remains a major challenge, but the molecular pathways that modulate ICB efficacy remain ill defined. Methods: Using CRISPR/Cas9 technology to generate melanoma cells lines that lack nucleic acid receptors or downstream signaling molecules together with available genetically deficient mouse models, we addressed the importance of nucleic acid receptor signaling in both tumor and host cells for the efficacy of anti-CTLA-4 immunotherapy. Results: We demonstrate that anti-CTLA-4 immunotherapy relies on melanoma cell-intrinsic activation of the cytosolic RNA receptor RIG-I (DDx58) but not the DNA sensing adaptor protein STING. Mechanistically, RIG-I signaling induced caspase-3-mediated tumor cell death, cross-presentation of tumor-associated antigen by CD103+ dendritic cells, subsequent expansion of tumor antigen-specific CD8+ T cells and the accumulation of CD8+ T cells within the tumor tissue. These processes were independent of tumor-cell derived type I IFN (IFN-I), but additionally required host STING, MAVS and IFN-I signaling. Consistently, therapeutic targeting of RIG-I with 5’-phosphorylated-RNA in both tumor and non-malignant host cells potently augmented the efficacy of CTLA-4 checkpoint blockade. Conclusions: Our data are consistent with the finding that expression of RIG-I in human melanomas has been associated with clinical benefit to CTLA-4 blockade and identify activation of RIG-I/MAVS signaling in tumors and their microenvironment as a crucial component for checkpoint inhibitor-mediated immunotherapy of cancer. These findings not only nominate tumor intrinsic RIG-I activity as potential biomarker for treatment response to checkpoint inhibitors, but predict that targeting this pathway may serve as a basis for the development of new combined modality approaches to increase the response rate of checkpoint inhibitor-based immunotherapy, particularly in individuals that do not have a sufficient spontaneous antitumor T-cell immune response.

Disclaimer

This material on this page is ©2024 American Society of Clinical Oncology, all rights reserved. Licensing available upon request. For more information, please contact licensing@asco.org

Abstract Details

Meeting

2018 ASCO Annual Meeting

Session Type

Poster Session

Session Title

Developmental Therapeutics—Immunotherapy

Track

Developmental Therapeutics and Translational Research

Sub Track

Immune Checkpoint Inhibitors

Citation

J Clin Oncol 36, 2018 (suppl; abstr 3081)

DOI

10.1200/JCO.2018.36.15_suppl.3081

Abstract #

3081

Poster Bd #

295

Abstract Disclosures

Similar Abstracts

Abstract

2023 ASCO Annual Meeting

Identification of RPTOR mutation as a novel predictor of efficacious immunotherapy in melanoma.

First Author: Yongzhao Li

First Author: Alessandra Anna Anna Prete

First Author: Guillermo De Velasco