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

      Targeted inhibition of gut bacterial β-glucuronidase activity enhances anticancer drug efficacy

      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.

          Significance

          Cancer chemotherapy often causes side effects that require modulations in dosing, which then reduce anticancer efficacy. Here, we show that targeted inhibition of gut bacterial enzymes alleviates key stages of gut epithelial damage caused by the cancer drug irinotecan, blunts stark gut microbial compositional shifts caused by irinotecan, and enhances irinotecan’s antitumor effectiveness by reducing its gastrointestinal toxicity.

          Abstract

          Irinotecan treats a range of solid tumors, but its effectiveness is severely limited by gastrointestinal (GI) tract toxicity caused by gut bacterial β-glucuronidase (GUS) enzymes. Targeted bacterial GUS inhibitors have been shown to partially alleviate irinotecan-induced GI tract damage and resultant diarrhea in mice. Here, we unravel the mechanistic basis for GI protection by gut microbial GUS inhibitors using in vivo models. We use in vitro, in fimo, and in vivo models to determine whether GUS inhibition alters the anticancer efficacy of irinotecan. We demonstrate that a single dose of irinotecan increases GI bacterial GUS activity in 1 d and reduces intestinal epithelial cell proliferation in 5 d, both blocked by a single dose of a GUS inhibitor. In a tumor xenograft model, GUS inhibition prevents intestinal toxicity and maintains the antitumor efficacy of irinotecan. Remarkably, GUS inhibitor also effectively blocks the striking irinotecan-induced bloom of Enterobacteriaceae in immune-deficient mice. In a genetically engineered mouse model of cancer, GUS inhibition alleviates gut damage, improves survival, and does not alter gut microbial composition; however, by allowing dose intensification, it dramatically improves irinotecan’s effectiveness, reducing tumors to a fraction of that achieved by irinotecan alone, while simultaneously promoting epithelial regeneration. These results indicate that targeted gut microbial enzyme inhibitors can improve cancer chemotherapeutic outcomes by protecting the gut epithelium from microbial dysbiosis and proliferative crypt damage.

          Related collections

          Most cited references26

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

          Chemotherapy-induced diarrhea: pathophysiology, frequency and guideline-based management.

          Diarrhea is one of the main drawbacks for cancer patients. Possible etiologies could be radiotherapy, chemotherapeutic agents, decreased physical performance, graft versus host disease and infections. Chemotherapy-induced diarrhea (CID) is a common problem, especially in patients with advanced cancer. The incidence of CID has been reported to be as high as 50-80% of treated patients (≥30% CTC grade 3-5), especially with 5-fluorouracil bolus or some combination therapies of irinotecan and fluoropyrimidines (IFL, XELIRI). Regardless of the molecular targeted approach of tyrosine kinase inhibitors and antibodies, diarrhea is a common side effect in up to 60% of patients with up to 10% having severe diarrhea. Furthermore, the underlying pathophysiology is still under investigation. Despite the number of clinical trials evaluating therapeutic or prophylactic measures in CID, there are just three drugs recommended in current guidelines: loperamide, deodorized tincture of opium and octreotide. Newer strategies and more effective agents are being developed to reduce the morbidity and mortality associated with CID. Recent research focusing on the prophylactic use of antibiotics, budesonide, probiotics or activated charcoal still have to define the role of these drugs in the routine clinical setting. Whereas therapeutic management and clinical work-up of patients presenting with diarrhea after chemotherapy are rather well defined, prediction and prevention of CID is an evolving field. Current research focuses on establishing predictive factors for CID like uridine diphosphate glucuronosyltransferase-1A1 polymorphisms for irinotecan or dihydropyrimidine-dehydrogenase insufficiency for fluoropyrimidines.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            The four-herb Chinese medicine PHY906 reduces chemotherapy-induced gastrointestinal toxicity.

            PHY906, a four-herb Chinese medicine formula first described 1800 years ago, decreases gastrointestinal toxicity induced by the chemotherapeutic drug CPT-11 (irinotecan), as shown in a phase I/II clinical study. Similarly, in a murine colon 38 allograft model, PHY906 increased the antitumor activity of CPT-11 while decreasing animal weight loss caused by CPT-11. Here, we have further examined the effect of PHY906 on the intestinal toxicity caused by CPT-11 in mice. PHY906 did not protect against the initial DNA damage and apoptosis triggered by CPT-11 in the intestine, but by 4 days after CPT-11 treatment, PHY906 had restored the intestinal epithelium by promoting the regeneration of intestinal progenitor or stem cells and several Wnt signaling components. PHY906 also potentiated Wnt3a activity in human embryonic kidney-293 cells. Furthermore, PHY906 exhibited anti-inflammatory effects in mice by decreasing the infiltration of neutrophils or macrophages, tumor necrosis factor-alpha expression in the intestine, and proinflammatory cytokine concentrations in plasma. Chemical constituents of PHY906 potently inhibited nuclear factor kappaB, cyclooxygenase-2, and inducible nitric oxide synthase. Our results show that the herbal medicine PHY906 can counteract the toxicity of CPT-11 via several mechanisms that act simultaneously.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              An Atlas of β-Glucuronidases in the Human Intestinal Microbiome.

              Microbiome-encoded β-glucuronidase (GUS) enzymes play important roles in human health by metabolizing drugs in the gastrointestinal (GI) tract. The numbers, types, and diversity of these proteins in the human GI microbiome, however, remain undefined. We present an atlas of GUS enzymes comprehensive for the Human Microbiome Project GI database. We identify 3,013 total and 279 unique microbiome-encoded GUS proteins clustered into six unique structural categories. We assign their taxonomy, assess cellular localization, reveal the inter-individual variability within the 139 individuals sampled, and discover 112 novel microbial GUS enzymes. A representative in vitro panel of the most common GUS proteins by read abundances highlights structural and functional variabilities within the family, including their differential processing of smaller glucuronides and larger carbohydrates. These data provide a sequencing-to-molecular roadmap for examining microbiome-encoded enzymes essential to human health.
                Bookmark

                Author and article information

                Journal
                Proc Natl Acad Sci U S A
                Proc. Natl. Acad. Sci. U.S.A
                pnas
                pnas
                PNAS
                Proceedings of the National Academy of Sciences of the United States of America
                National Academy of Sciences
                0027-8424
                1091-6490
                31 March 2020
                13 March 2020
                13 March 2020
                : 117
                : 13
                : 7374-7381
                Affiliations
                [1] aDepartment of Chemistry, University of North Carolina at Chapel Hill , Chapel Hill, NC 27599-3290;
                [2] bDepartment of Medicine, Division of Gastroenterology and Hepatology, University of North Carolina at Chapel Hill , Chapel Hill, NC 27599-7555;
                [3] cCenter for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill , Chapel Hill, NC 27599-7555;
                [4] dComputational and Systems Medicine, Department of Surgery & Cancer, Imperial College London , SW7 2AZ London, United Kingdom;
                [5] eLineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill , Chapel Hill, NC 27599;
                [6] fDepartment of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill , Chapel Hill, NC 27599-7525;
                [7] gDepartment of Microbiology and Immunology, University of North Carolina at Chapel Hill , Chapel Hill, NC 27599;
                [8] hDepartment of Medicine, Division of Gastroenterology, University of Florida , Gainesville, FL 32610;
                [9] iDepartment of Genetics, University of North Carolina at Chapel Hill , Chapel Hill, NC 27599-7264;
                [10] jDepartment of Biochemistry, Integrated Program for Biological and Genome Science, University of North Carolina at Chapel Hill , Chapel Hill, NC 27599-3290;
                [11] kDepartment of Biophysics, Integrated Program for Biological and Genome Science, University of North Carolina at Chapel Hill , Chapel Hill, NC 27599-3290
                Author notes
                2To whom correspondence may be addressed. Email: redinbo@ 123456unc.edu .

                Edited by Robert J. Fletterick, University of California, San Francisco, CA, and approved February 17, 2020 (received for review October 16, 2019)

                Author contributions: A.P.B., R.B.S., S.J.B., and M.R.R. designed research; A.P.B., S.J.P., K.A.B., W.G.W., B.D.W., B.C.C., M.M.L., J.R.R., and S.A.M. performed research; A.P.B., S.J.P., K.A.B., B.D.W., B.C.C., M.M.L., J.R.S., I.D.W., D.B.D., S.T.B., R.B.S., and R.Z.G. contributed new reagents/analytic tools; A.P.B., S.J.P., K.A.B., W.G.W., B.C.C., M.M.L., J.R.S., I.D.W., J.R.R., S.T.B., S.A.M., J.M.R., M.A.A.-P., S.J.B., and R.Z.G. analyzed data; and A.P.B., M.M.L., J.R.S., I.D.W., S.A.M., J.M.R., R.Z.G., and M.R.R. wrote the paper.

                1Present address: Symberix, Inc., Durham, NC 27703.

                Author information
                http://orcid.org/0000-0002-0134-6543
                http://orcid.org/0000-0002-7557-7985
                http://orcid.org/0000-0001-9817-5877
                http://orcid.org/0000-0003-0325-1289
                http://orcid.org/0000-0002-7820-632X
                http://orcid.org/0000-0003-0814-5346
                Article
                201918095
                10.1073/pnas.1918095117
                7132129
                32170007
                2bd0411e-8edb-4902-a349-e86bd43144eb
                Copyright © 2020 the Author(s). Published by PNAS.

                This open access article is distributed under Creative Commons Attribution-NonCommercial-NoDerivatives License 4.0 (CC BY-NC-ND).

                History
                Page count
                Pages: 8
                Funding
                Funded by: HHS | NIH | National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) 100000062
                Award ID: T32-DK007737
                Award Recipient : Aadra Prashant Bhatt Award Recipient : M. Andrea Azcarate-Peril Award Recipient : R. Balfour Sartor
                Funded by: HHS | NIH | National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) 100000062
                Award ID: P30 DK034987
                Award Recipient : Aadra Prashant Bhatt Award Recipient : M. Andrea Azcarate-Peril Award Recipient : R. Balfour Sartor
                Funded by: HHS | NIH | National Cancer Institute (NCI) 100000054
                Award ID: CA207416
                Award Recipient : Scott J. Bultman Award Recipient : Matthew R. Redinbo
                Funded by: HHS | NIH | National Cancer Institute (NCI) 100000054
                Award ID: CA098468
                Award Recipient : Scott J. Bultman Award Recipient : Matthew R. Redinbo
                Funded by: U.S. Department of Agriculture (USDA) 100000199
                Award ID: 055336
                Award Recipient : Scott J. Bultman
                Funded by: HHS | NIH | National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) 100000062
                Award ID: P40 OD010995
                Award Recipient : Aadra Prashant Bhatt Award Recipient : M. Andrea Azcarate-Peril Award Recipient : R. Balfour Sartor
                Categories
                Biological Sciences
                Microbiology

                microbiome,chemotherapy,cancer,gastrointestinal toxicity

                Comments

                Comment on this article