BackgroundThe suppressive nature of immune cells in the tumor microenvironment plays a major role in regulating anti-tumor immune responses. Our previous work demonstrated that a soluble factor from tumor cells is able to induce a suppressor phenotype (SP) in human CD8+ T cells typified by loss of CD27/CD28 expression and acquisition of a potent suppressor function. The present study hypothesized that the soluble mechanism that is inducing the SP in CD8+ T cells are tumor-derived exosomes (TDEs).MethodsMembrane vesicles and TDEs from multiple head and neck cancer cell line’s conditioned growth media were isolated by ultracentrifugation and precipitation, respectively. Human purified CD3+CD8+ T cells were assessed for their induction of the T cell SP by flow cytometry identifying loss of CD27/CD28 expression and in vitro suppression assays. Furthermore, the T cell SP was characterized for the attenuation of IFN-γ production. To delineate exosomal proteins contributing to T cell SP, mass spectrometry was used to identify unique proteins that were present in TDEs. CRISPR/Cas9 knockout constructs were used to examine the role of one of these proteins, galectin-1. To assess the role of exosomal RNA, RNA purified from TDEs was nucleofected into CD8+ T cells followed by suppression analysis.ResultsUsing fractionated conditioned growth media, factors >200 kDa induced CD8+ T cell SP, which was determined to be an exosome by mass spectrometry analysis. Multiple head and neck cancer-derived cell lines were found to secrete T cell SP-inducing exosomes. Mass spectrometry analysis revealed that an immunoregulatory protein, galectin-1 (Gal-1), was expressed in those exosomes, but not in TDEs unable to induce T cell SP. Galectin-1 knockout cells were found to be less able to induce T cell SP. Furthermore, RNA purified from the T cell SP-inducing exosomes were found to partially induce the SP when transfected into normal CD8+ T cells.ConclusionsFor the first-time, TDEs have been identified to induce a SP in CD8+ T cells and their mode of action may be synergistic effects from exosomal proteins and RNA. One protein in particular, galectin-1, appears to play a significant role in inducing T cell SP. Therefore, tumor-derived immunosuppressive exosomes are a potential therapeutic target to prevent T cell dysfunction and enhance anti-tumor immune responses.
Introduction Triple negative breast cancer (TNBC) is a heterogeneous breast cancer subtype which continues to portend a particularly poor prognosis compared to other breast cancer subtypes. Immune checkpoint inhibitor (ICI) therapies have emerged as promising options for locally advanced and metastatic TNBC. However, clinical trials have demonstrated mixed results with respect to ICI response. Given varied outcomes and the potential for immune-related adverse events associated with ICIs, there is critical need for identification of accurate biomarkers of response and improved strategies to counteract ICI resistance and/or toxicity. Methods Advanced immunoprofiling of peripheral blood mononuclear cells (PBMCs) and plasma from 7 metastatic TNBC patients with variable responses to anti-PD1 or anti-PDL1 therapy was performed. Samples were analyzed from blood draws obtained: (i) prior to first administration, (ii) while receiving ICI treatment, and (iii) at the time of conformed clinical progression or response to therapy. Response was determined by standard radiological assessment. Immunoprofiling included high parameter flow cytometry, single cell transcriptomics (10x genomics) and secretome analysis (Isoplexis). Single cell RNA profiles from 63,984 cells were analyzed. Results High parameter flow cytometry identified higher circulating levels of a subpopulation of activated CD4+ T cells with a phenotype of CXCR3low CD62Llow CD45RAhigh and CD57high expression in responders versus non-responders. Higher effector function of CD4+ T cells was corroborated by significantly elevated plasma concentrations of IL-2 (p=0.02) and IL-5 (p< 0.0001) among responders. Single cell transcriptomic analysis revealed clusters of B and T cells with distinct activation patterns that were associated with radiographic response. Strikingly, genes involved in B cell activation and T cell-B cell conjugation such as CD81 were found to be highly upregulated among CD4+ T cells from responders. Conclusions Our results are consistent with previous reports describing an association of increased B cell activity in TNBC with improved overall survival. We identified a subpopulation of CD4+ T cells with effector functions consistent with type 2 helper T cells that may not only target cancer cells by direct cytotoxic function, but also promote increased B cell anti-tumor activity. Our study provides insight into specific mechanisms of immune cell interplay that may drive response to ICI therapy. These cell populations and their associated pathways may represent potential biomarkers of response and/or targets for resistance reversal. Citation Format: Avia D. Wilkerson, Patricia A. Rayman, Paul G. Pavicic Jr, Hana Husic, Vladimir Makarov, Ivan Juric, Timothy Chan, Alberto J. Montero, Marcela Diaz-Montero. A multiomic approach to the identification of immune signatures of anti-PD1/PDL1 therapy responders in metastatic triple negative breast cancer: new implications in the role of helper T-cell and B-cell interplay [abstract]. In: Proceedings of the 2022 San Antonio Breast Cancer Symposium; 2022 Dec 6-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2023;83(5 Suppl):Abstract nr P1-04-04.
Glioblastoma (GBM) creates an immunosuppressive environment that presents a challenge to efficacy of immunotherapeutic approaches. Results from the CheckMate-143 trial demonstrated responses in 8% of patients with nivolumab, underscoring the need for further insight into the mechanisms and markers of immune suppression and response. Given a limited set of biomarkers predictive of immunotherapy response in GBM, we explored the changes in immune cell populations in nivolumab and bevacizumab-treated GBM patients pre and post-treatment in order to help predict response. In these studies, we utilized traditional and newly developed approaches, including mass cytometry time-of-flight (CyTOF), single-cell RNA sequencing, and 10X Genomics simultaneous cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq). We analyzed patients’ samples in a randomized, phase 2 study of nivolumab and bevacizumab at GBM first recurrence (NCT03452579). Nine patients were identified as responders or non-responders at 8 weeks after therapy initiation. Utilizing peripheral blood samples, we observed a 6.4-fold decrease in immunosuppressive myeloid-derived suppressor cells (MDSCs) between baseline and first imaging follow-up in responders compared to non-responders, with a 4.9-fold decrease in the granulocytic MDSC (G-MDSC) subtype in responders over non-responders. While no significant changes in overall T-cell numbers were noted, expression of PD-1 on CD4+ T cells was significantly elevated at baseline and follow-up in responders as compared to non-responders – signatures which were confirmed by CyTOF. Given these immunophenotypic changes, preliminary results of a detailed investigation of this cohort by CITE-seq indicate that responders had increased IL7R-positive T cells post-treatment, which was not observed in non-responders. These results are currently being validated in an additional 40 patients that have been enrolled. Altogether, differences in immunophenotypes that were specific to responders and non-responders were observed, and characterization of these immune populations may be helpful in identifying GBM patients likely to benefit from immunotherapy.
scite is a Brooklyn-based organization that helps researchers better discover and understand research articles through Smart Citations–citations that display the context of the citation and describe whether the article provides supporting or contrasting evidence. scite is used by students and researchers from around the world and is funded in part by the National Science Foundation and the National Institute on Drug Abuse of the National Institutes of Health.
customersupport@researchsolutions.com
10624 S. Eastern Ave., Ste. A-614
Henderson, NV 89052, USA
This site is protected by reCAPTCHA and the Google Privacy Policy and Terms of Service apply.
Copyright © 2024 scite LLC. All rights reserved.
Made with 💙 for researchers
Part of the Research Solutions Family.