5
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Lactobacillus gallinarum-derived metabolites boost anti-PD1 efficacy in colorectal cancer by inhibiting regulatory T cells through modulating IDO1/Kyn/AHR axis

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Objective

          Gut microbiota is a key player in dictating immunotherapy response. We aimed to explore the immunomodulatory effect of probiotic Lactobacillus gallinarum and its role in improving anti-programmed cell death protein 1 (PD1) efficacy against colorectal cancer (CRC).

          Design

          The effects of L. gallinarum in anti-PD1 response were assessed in syngeneic mouse models and azoxymethane/dextran sulfate sodium-induced CRC model. The change of immune landscape was identified by multicolour flow cytometry and validated by immunohistochemistry staining and in vitro functional assays. Liquid chromatography-mass spectrometry was performed to identify the functional metabolites.

          Results

          L. gallinarum significantly improved anti-PD1 efficacy in two syngeneic mouse models with different microsatellite instability (MSI) statuses (MSI-high for MC38, MSI-low for CT26). Such effect was confirmed in CRC tumourigenesis model. L. gallinarum synergised with anti-PD1 therapy by reducing Foxp3 + CD25 + regulatory T cell (Treg) intratumoural infiltration, and enhancing effector function of CD8 + T cells. L. gallinarum-derived indole-3-carboxylic acid (ICA) was identified as the functional metabolite. Mechanistically, ICA inhibited indoleamine 2,3-dioxygenase (IDO1) expression, therefore suppressing kynurenine (Kyn) production in tumours. ICA also competed with Kyn for binding site on aryl hydrocarbon receptor (AHR) and antagonised Kyn binding on CD4 + T cells, thereby inhibiting Treg differentiation in vitro. ICA phenocopied L. gallinarum effect and significantly improved anti-PD1 efficacy in vivo, which could be reversed by Kyn supplementation.

          Conclusion

          L. gallinarum-derived ICA improved anti-PD1 efficacy in CRC through suppressing CD4+Treg differentiation and enhancing CD8+T cell function by modulating the IDO1/Kyn/AHR axis. L. gallinarum is a potential adjuvant to augment anti-PD1 efficacy against CRC.

          Related collections

          Most cited references45

          • Record: found
          • Abstract: found
          • Article: not found

          Cancer immunotherapy using checkpoint blockade

          The release of negative regulators of immune activation (immune checkpoints) that limit antitumor responses has resulted in unprecedented rates of long-lasting tumor responses in patients with a variety of cancers. This can be achieved by antibodies blocking the cytotoxic T lymphocyte antigen-4 (CTLA-4) or the programmed death-1 (PD-1) pathway, either alone or in combination. The main premise for inducing an immune response is the pre-existence of antitumor T cells that were limited by specific immune checkpoints. Most patients who have tumor responses maintain long lasting disease control, yet one third of patients relapse. Mechanisms of acquired resistance are currently poorly understood, but evidence points to alterations that converge on the antigen presentation and interferon gamma signaling pathways. New generation combinatorial therapies may overcome resistance mechanisms to immune checkpoint therapy.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Gut microbiome influences efficacy of PD-1–based immunotherapy against epithelial tumors

            Immune checkpoint inhibitors (ICI) targeting the PD-1/PD-L1 axis induce sustained clinical responses in a sizeable minority of cancer patients. Here, we show that primary resistance to ICI can be due to abnormal gut microbiome composition. Antibiotics (ATB) inhibited the clinical benefit of ICI in patients with advanced cancer. Fecal microbiota transplantation (FMT) from cancer patients who responded to ICI (but not from non-responding patients) into germ-free or ATB-treated mice ameliorated the antitumor effects of PD-1 blockade. Metagenomics of patient stools at diagnosis revealed correlations between clinical responses to ICI and the relative abundance of Akkermansia muciniphila. Oral supplementation with A. muciniphila post-FMT with non-responder feces restored the efficacy of PD-1 blockade in an IL-12-dependent manner, by increasing the recruitment of CCR9+CXCR3+CD4+ T lymphocytes into tumor beds.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Gut microbiome modulates response to anti–PD-1 immunotherapy in melanoma patients

              Pre-clinical mouse models suggest that the gut microbiome modulates tumor response to checkpoint blockade immunotherapy; however, this has not been well-characterized in human cancer patients. Here we examined the oral and gut microbiome of melanoma patients undergoing anti-PD-1 immunotherapy (n=112). Significant differences were observed in the diversity and composition of the patient gut microbiome of responders (R) versus non-responders (NR). Analysis of patient fecal microbiome samples (n=43, 30R, 13NR) showed significantly higher alpha diversity (p<0.01) and relative abundance of Ruminococcaceae bacteria (p<0.01) in responding patients. Metagenomic studies revealed functional differences in gut bacteria in R including enrichment of anabolic pathways. Immune profiling suggested enhanced systemic and anti-tumor immunity in responding patients with a favorable gut microbiome, as well as in germ-free mice receiving fecal transplants from responding patients. Together, these data have important implications for the treatment of melanoma patients with immune checkpoint inhibitors.
                Bookmark

                Author and article information

                Journal
                Gut
                Gut
                gutjnl
                gut
                Gut
                BMJ Publishing Group (BMA House, Tavistock Square, London, WC1H 9JR )
                0017-5749
                1468-3288
                December 2023
                28 September 2023
                : 72
                : 12
                : 2272-2285
                Affiliations
                [1 ] departmentInstitute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences , Ringgold_26451The Chinese University of Hong Kong , Hong Kong, China
                [2 ] departmentDepartment of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute , The Chinese University of Hong Kong , Hong Kong, China
                [3 ] departmentInstitute of Precision Medicine , Ringgold_71068The First Affiliated Hospital of Sun Yat-Sen University , Guangzhou, China
                [4 ] departmentSchool of Pharmacy , Ringgold_26451The Chinese University of Hong Kong , Hong Kong, China
                [5 ] departmentLee Kong Chian School of Medicine , Ringgold_54761Nanyang Technological University , Singapore
                Author notes
                [Correspondence to ] Professor Jun Yu, Institute of Digestive Disease, Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China; junyu@ 123456cuhk.edu.hk
                Author information
                http://orcid.org/0000-0003-3581-2909
                http://orcid.org/0000-0001-8791-7505
                http://orcid.org/0000-0003-3125-5199
                http://orcid.org/0000-0001-5008-2153
                Article
                gutjnl-2023-329543
                10.1136/gutjnl-2023-329543
                10715476
                37770127
                12aa38d7-a881-4f00-ae1b-2b65dc90cb65
                © Author(s) (or their employer(s)) 2023. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

                This is an open access article distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited, appropriate credit is given, any changes made indicated, and the use is non-commercial. See:  http://creativecommons.org/licenses/by-nc/4.0/.

                History
                : 19 January 2023
                : 16 August 2023
                Funding
                Funded by: Shenzhen-Hong Kong-Macao Science and Technology Program (Category C) Shenzhen;
                Award ID: SGDX20210823103535016
                Funded by: Research Talent Hub-Innovation and Technology Fund Hong Kong;
                Award ID: ITS/177/21FP
                Funded by: RGC Research Impact Fund Hong Kong;
                Award ID: R4032-21F
                Categories
                Gut Microbiota
                1506
                2312
                Original research
                Custom metadata
                unlocked

                Gastroenterology & Hepatology
                probiotics,immunotherapy,colorectal cancer
                Gastroenterology & Hepatology
                probiotics, immunotherapy, colorectal cancer

                Comments

                Comment on this article