» Articles » PMID: 36302895

Anti-CTLA-4 Antibodies Drive Myeloid Activation and Reprogram the Tumor Microenvironment Through FcγR Engagement and Type I Interferon Signaling

Overview
Journal Nat Cancer
Specialty Oncology
Date 2022 Oct 27
PMID 36302895
Authors
Affiliations
Soon will be listed here.
Abstract

Despite the clinical success of checkpoint inhibitors, a substantial gap still exists in our understanding of their mechanism of action. While antibodies to cytotoxic T lymphocyte-associated protein-4 (CTLA-4) were developed to block inhibitory signals in T cells, several recent studies have demonstrated that Fcγ receptor (FcγR)-dependent depletion of regulatory T cells (T) is critical for antitumor activity. Here, using single-cell RNA sequencing, we dissect the impact of anti-CTLA-4-blocking, T cell-depleting and FcR-engaging activity on the immune response within tumors. We observed a rapid remodeling of the innate immune landscape as early as 24 h after treatment. Using genetic T cell ablation models, we show that immune remodeling was not driven solely by T cell depletion or CTLA-4 blockade but mainly through FcγR engagement, downstream activation of type I interferon signaling and reduction of suppressive macrophages. Our findings indicate that FcγR engagement and innate immune remodeling are involved in successful anti-CTLA-4 treatment, supporting the development of optimized immunotherapy agents bearing these features.

Citing Articles

Nivolumab plus chemotherapy or ipilimumab in gastroesophageal cancer: exploratory biomarker analyses of a randomized phase 3 trial.

Shitara K, Janjigian Y, Ajani J, Moehler M, Yao J, Wang X Nat Med. 2025; .

PMID: 40055521 DOI: 10.1038/s41591-025-03575-0.


pH-dependent dissociation from CTLA-4 in early endosomes improves both safety and antitumor activity of anti-CTLA-4 antibodies.

Zhang M, Li J, Yan K, Zhou H, Mei S, Wang B Proc Natl Acad Sci U S A. 2025; 122(8):e2422731122.

PMID: 39964714 PMC: 11874271. DOI: 10.1073/pnas.2422731122.


Arginase-1-specific T cells target and modulate tumor-associated macrophages.

Martinenaite E, Lecoq I, Aaboe-Jorgensen M, Ahmad S, Perez-Penco M, Glockner H J Immunother Cancer. 2025; 13(1).

PMID: 39880485 PMC: 11781113. DOI: 10.1136/jitc-2024-009930.


FAT1 knockdown enhances the CSC properties of HNSCC through p-CaMKII-mediated inactivation of the IFN pathway.

Wang J, Chen Y, Sun Y, Liu H, Du R, Wang X Int J Biol Sci. 2025; 21(2):671-684.

PMID: 39781458 PMC: 11705627. DOI: 10.7150/ijbs.95723.


Spatial multiplex analysis of lung cancer reveals that regulatory T cells attenuate KRAS-G12C inhibitor-induced immune responses.

Cole M, Anastasiou P, Lee C, Yu X, de Castro A, Roelink J Sci Adv. 2024; 10(44):eadl6464.

PMID: 39485838 PMC: 11529713. DOI: 10.1126/sciadv.adl6464.


References
1.
Sharma P, Siddiqui B, Anandhan S, Yadav S, Subudhi S, Gao J . The Next Decade of Immune Checkpoint Therapy. Cancer Discov. 2021; 11(4):838-857. DOI: 10.1158/2159-8290.CD-20-1680. View

2.
Weiss S, Wolchok J, Sznol M . Immunotherapy of Melanoma: Facts and Hopes. Clin Cancer Res. 2019; 25(17):5191-5201. PMC: 6726509. DOI: 10.1158/1078-0432.CCR-18-1550. View

3.
Hegde P, Chen D . Top 10 Challenges in Cancer Immunotherapy. Immunity. 2020; 52(1):17-35. DOI: 10.1016/j.immuni.2019.12.011. View

4.
Walker L, Sansom D . Confusing signals: recent progress in CTLA-4 biology. Trends Immunol. 2015; 36(2):63-70. PMC: 4323153. DOI: 10.1016/j.it.2014.12.001. View

5.
Walker L . Treg and CTLA-4: two intertwining pathways to immune tolerance. J Autoimmun. 2013; 45:49-57. PMC: 3989116. DOI: 10.1016/j.jaut.2013.06.006. View