11
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Akkermansia Muciniphila Potentiates the Antitumor Efficacy of FOLFOX in Colon Cancer

      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

          FOLFOX (oxaliplatin, fluorouracil and calcium folinate) is the first-line chemotherapy regimen for colon cancer therapy in the clinic. It provides superior efficacy than oxaliplatin alone, but the underlying mechanism remains unclear. In the present study, pharmacomicrobiomics integrated with metabolomics was conducted to uncover the role of the gut microbiome behind this. First, in vivo study demonstrated that FOLFOX exhibited better efficacy than oxaliplatin alone in colon cancer animal models. Second, 16S rDNA gene sequencing analysis showed that the abundance of Akkermansia muciniphila ( A. muciniphila) remarkably increased in the FOLFOX treated individuals and positively correlated with the therapeutic effect. Third, further exploration confirmed A. muciniphila colonization significantly enhanced the anti-cancer efficacy of FOLFOX. Last, metabolomics analysis suggested dipeptides containing branched-chain amino acid (BCAA) might be responsible for gut bacteria mediated FOLFOX efficacy. In conclusion, our study revealed the key role of A. muciniphila in mediating FOLFOX efficacy, and manipulating A. muciniphila might serve as a novel strategy for colon cancer therapy.

          Related collections

          Most cited references52

          • 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

            Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity.

            Obesity and type 2 diabetes are characterized by altered gut microbiota, inflammation, and gut barrier disruption. Microbial composition and the mechanisms of interaction with the host that affect gut barrier function during obesity and type 2 diabetes have not been elucidated. We recently isolated Akkermansia muciniphila, which is a mucin-degrading bacterium that resides in the mucus layer. The presence of this bacterium inversely correlates with body weight in rodents and humans. However, the precise physiological roles played by this bacterium during obesity and metabolic disorders are unknown. This study demonstrated that the abundance of A. muciniphila decreased in obese and type 2 diabetic mice. We also observed that prebiotic feeding normalized A. muciniphila abundance, which correlated with an improved metabolic profile. In addition, we demonstrated that A. muciniphila treatment reversed high-fat diet-induced metabolic disorders, including fat-mass gain, metabolic endotoxemia, adipose tissue inflammation, and insulin resistance. A. muciniphila administration increased the intestinal levels of endocannabinoids that control inflammation, the gut barrier, and gut peptide secretion. Finally, we demonstrated that all these effects required viable A. muciniphila because treatment with heat-killed cells did not improve the metabolic profile or the mucus layer thickness. In summary, this study provides substantial insight into the intricate mechanisms of bacterial (i.e., A. muciniphila) regulation of the cross-talk between the host and gut microbiota. These results also provide a rationale for the development of a treatment that uses this human mucus colonizer for the prevention or treatment of obesity and its associated metabolic disorders.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Bile acid–microbiota crosstalk in gastrointestinal inflammation and carcinogenesis

              Emerging evidence points to a strong association between the gut microbiota and the risk, development and progression of gastrointestinal cancers such as colorectal cancer (CRC) and hepatocellular carcinoma (HCC). Bile acids, produced in the liver, are metabolized by enzymes derived from intestinal bacteria and are critically important for maintaining a healthy gut microbiota, balanced lipid and carbohydrate metabolism, insulin sensitivity and innate immunity. Given the complexity of bile acid signalling and the direct biochemical interactions between the gut microbiota and the host, a systems biology perspective is required to understand the liver-bile acid-microbiota axis and its role in gastrointestinal carcinogenesis to reverse the microbiota-mediated alterations in bile acid metabolism that occur in disease states. An examination of recent research progress in this area is urgently needed. In this Review, we discuss the mechanistic links between bile acids and gastrointestinal carcinogenesis in CRC and HCC, which involve two major bile acid-sensing receptors, farnesoid X receptor (FXR) and G protein-coupled bile acid receptor 1 (TGR5). We also highlight the strategies and cutting-edge technologies to target gut-microbiota-dependent alterations in bile acid metabolism in the context of cancer therapy.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Pharmacol
                Front Pharmacol
                Front. Pharmacol.
                Frontiers in Pharmacology
                Frontiers Media S.A.
                1663-9812
                17 September 2021
                2021
                : 12
                : 725583
                Affiliations
                [ 1 ]Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing, China
                [ 2 ]School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
                [ 3 ]School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
                Author notes

                Edited by: Ester Pagano, University of Naples Federico II, Italy

                Reviewed by: Wenjing Zhao, Sun Yat-sen University, China

                Hui Yang, The Second Affiliated Hospital of Guangzhou Medical University, China

                *Correspondence: Zunjian Zhang, zunjianzhangcpu@ 123456hotmail.com ; Fengguo Xu, fengguoxu@ 123456cpu.edu.cn
                [ † ]

                These authors have contributed equally to this work

                This article was submitted to Gastrointestinal and Hepatic Pharmacology, a section of the journal Frontiers in Pharmacology

                Article
                725583
                10.3389/fphar.2021.725583
                8484791
                34603035
                efa6bf3f-1fa5-4ca1-b73c-9ba67e390610
                Copyright © 2021 Hou, Zhang, Du, Chu, Sun, Qin, Tian, Zhang and Xu.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 15 June 2021
                : 24 August 2021
                Funding
                Funded by: Foundation for Innovative Research Groups of the National Natural Science Foundation of China , doi 10.13039/501100012659;
                Award ID: 82073812 81773682 81773861
                Funded by: Outstanding Youth Foundation of Jiangsu Province of China , doi 10.13039/501100010035;
                Award ID: BK20180027
                Funded by: National Major Science and Technology Projects of China , doi 10.13039/501100013076;
                Award ID: 2017ZX09101001
                Funded by: Graduate Research and Innovation Projects of Jiangsu Province , doi 10.13039/501100012154;
                Award ID: KYCX20_0665
                Funded by: Double First Class University Plan , doi 10.13039/501100012172;
                Funded by: Priority Academic Program Development of Jiangsu Higher Education Institutions , doi 10.13039/501100012246;
                Categories
                Pharmacology
                Original Research

                Pharmacology & Pharmaceutical medicine
                folfox,oxaliplatin,colon cancer,akkermansia muciniphila,pharmacomicrobiomics,metabolomics

                Comments

                Comment on this article