4
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Helicobacter pylori infection has a detrimental impact on the efficacy of cancer immunotherapies

      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

          In this study, we determined whether Helicobacter pylori ( H. pylori) infection dampens the efficacy of cancer immunotherapies.

          Design

          Using mouse models, we evaluated whether immune checkpoint inhibitors or vaccine-based immunotherapies are effective in reducing tumour volumes of H. pylori-infected mice. In humans, we evaluated the correlation between H. pylori seropositivity and the efficacy of the programmed cell death protein 1 (PD-1) blockade therapy in patients with non-small-cell lung cancer (NSCLC).

          Results

          In mice engrafted with MC38 colon adenocarcinoma or B16-OVA melanoma cells, the tumour volumes of non-infected mice undergoing anticytotoxic T-lymphocyte-associated protein 4 and/or programmed death ligand 1 or anti-cancer vaccine treatments were significantly smaller than those of infected mice. We observed a decreased number and activation status of tumour-specific CD8 + T cells in the tumours of infected mice treated with cancer immunotherapies independent of the gut microbiome composition. Additionally, by performing an in vitro co-culture assay, we observed that dendritic cells of infected mice promote lower tumour-specific CD8 + T cell proliferation. We performed retrospective human clinical studies in two independent cohorts. In the Dijon cohort, H. pylori seropositivity was found to be associated with a decreased NSCLC patient survival on anti-PD-1 therapy. The survival median for H. pylori seropositive patients was 6.7 months compared with 15.4 months for seronegative patients (p=0.001). Additionally, in the Montreal cohort, H. pylori seropositivity was found to be associated with an apparent decrease of NSCLC patient progression-free survival on anti-PD-1 therapy.

          Conclusion

          Our study unveils for the first time that the stomach microbiota affects the response to cancer immunotherapies and that H. pylori serology would be a powerful tool to personalize cancer immunotherapy treatment.

          Related collections

          Most cited references49

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

              Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy.

              T cell infiltration of solid tumors is associated with favorable patient outcomes, yet the mechanisms underlying variable immune responses between individuals are not well understood. One possible modulator could be the intestinal microbiota. We compared melanoma growth in mice harboring distinct commensal microbiota and observed differences in spontaneous antitumor immunity, which were eliminated upon cohousing or after fecal transfer. Sequencing of the 16S ribosomal RNA identified Bifidobacterium as associated with the antitumor effects. Oral administration of Bifidobacterium alone improved tumor control to the same degree as programmed cell death protein 1 ligand 1 (PD-L1)-specific antibody therapy (checkpoint blockade), and combination treatment nearly abolished tumor outgrowth. Augmented dendritic cell function leading to enhanced CD8(+) T cell priming and accumulation in the tumor microenvironment mediated the effect. Our data suggest that manipulating the microbiota may modulate cancer immunotherapy.
                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
                March 2022
                12 July 2021
                : 71
                : 3
                : 457-466
                Affiliations
                [1 ] departmentService of Gastroenterology and Hepatology , Centre Hospitalier Universitaire Vaudois, University of Lausanne , Lausanne, Switzerland
                [2 ] departmentDepartment of Medical Oncology , Centre Georges François Leclerc , Dijon, France
                [3 ] departmentResearch Centre for the University of Montréal (CRCHUM), Hematology-Oncology Division, Department of Medicine , University of Montreal Healthcare Centre (CHUM) , Montreal, Quebec, Canada
                [4 ] departmentDepartment of Oncology , Centre Hospitalier Universitaire Vaudois, University of Lausanne , Lausanne, Switzerland
                Author notes
                [Correspondence to ] Dr Dominique Velin, Service of Gastroenterology and Hepatology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, 1011 Lausanne, Switzerland; dominique.velin@ 123456chuv.ch
                Author information
                http://orcid.org/0000-0002-5465-8305
                http://orcid.org/0000-0001-6033-5871
                Article
                gutjnl-2020-323392
                10.1136/gutjnl-2020-323392
                8862014
                34253574
                bebfaf94-67e9-4b70-bd60-5c68482d352d
                © Author(s) (or their employer(s)) 2022. 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
                : 17 October 2020
                : 24 June 2021
                Funding
                Funded by: Foundation Emma Muschamp;
                Award ID: NA
                Funded by: Faculté de Biologie et de Médecine de Lausanne;
                Award ID: NA
                Funded by: Foundation Max Cloëtta;
                Award ID: NA
                Funded by: FundRef http://dx.doi.org/10.13039/501100013362, Swiss Cancer Research Foundation;
                Award ID: KFS-4452-02-2018; KFS-4840-08-2019
                Funded by: Foundation Porphyrogenis;
                Award ID: NA
                Categories
                Helicobacter Pylori
                1506
                2312
                Original research
                Custom metadata
                unlocked

                Gastroenterology & Hepatology
                helicobacter pylori,cancer,immunotherapy,cancer immunobiology
                Gastroenterology & Hepatology
                helicobacter pylori, cancer, immunotherapy, cancer immunobiology

                Comments

                Comment on this article