DOE PAGES title logo U.S. Department of Energy
Office of Scientific and Technical Information

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

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 resultsmore » indicate that targeted gut microbial enzyme inhibitors can improve cancer chemotherapeutic outcomes by protecting the gut epithelium from microbial dysbiosis and proliferative crypt damage.« less

Authors:
ORCiD logo [1]; ORCiD logo [1];  [1];  [1];  [1];  [1];  [2];  [2];  [2];  [1];  [1];  [1];  [1]; ORCiD logo [1]; ORCiD logo [1]; ORCiD logo [1];  [3];  [1]; ORCiD logo [1]
  1. Univ. of North Carolina, Chapel Hill, NC (United States)
  2. Imperial College, London (United Kingdom)
  3. Univ. of Florida, Gainesville, FL (United States)
Publication Date:
Research Org.:
Argonne National Lab. (ANL), Argonne, IL (United States). Advanced Photon Source (APS)
Sponsoring Org.:
USDOE; University of Florida Health Cancer Center; University Cancer Research Fund; Crohn’s and Colitis Foundation; USDA; National Institutes of Health (NIH)
OSTI Identifier:
1644107
Grant/Contract Number:  
055336; T32-DK007737; P30 DK034987; P40 OD010995; CA098468; CA207416
Resource Type:
Accepted Manuscript
Journal Name:
Proceedings of the National Academy of Sciences of the United States of America
Additional Journal Information:
Journal Volume: 117; Journal Issue: 13; Journal ID: ISSN 0027-8424
Publisher:
National Academy of Sciences
Country of Publication:
United States
Language:
ENGLISH
Subject:
60 APPLIED LIFE SCIENCES; microbiome; chemotherapy; cancer; gastrointestinal toxicity

Citation Formats

Bhatt, Aadra P., Pellock, Samuel J., Biernat, Kristen A., Walton, William G., Wallace, Bret D., Creekmore, Benjamin C., Letertre, Marine M., Swann, Jonathan R., Wilson, Ian D., Roques, Jose R., Darr, David B., Bailey, Sean T., Montgomery, Stephanie A., Roach, Jeffrey M., Azcarate-Peril, M. Andrea, Sartor, R. Balfour, Gharaibeh, Raad Z., Bultman, Scott J., and Redinbo, Matthew R. Targeted inhibition of gut bacterial β-glucuronidase activity enhances anticancer drug efficacy. United States: N. p., 2020. Web. doi:10.1073/pnas.1918095117.
Bhatt, Aadra P., Pellock, Samuel J., Biernat, Kristen A., Walton, William G., Wallace, Bret D., Creekmore, Benjamin C., Letertre, Marine M., Swann, Jonathan R., Wilson, Ian D., Roques, Jose R., Darr, David B., Bailey, Sean T., Montgomery, Stephanie A., Roach, Jeffrey M., Azcarate-Peril, M. Andrea, Sartor, R. Balfour, Gharaibeh, Raad Z., Bultman, Scott J., & Redinbo, Matthew R. Targeted inhibition of gut bacterial β-glucuronidase activity enhances anticancer drug efficacy. United States. https://doi.org/10.1073/pnas.1918095117
Bhatt, Aadra P., Pellock, Samuel J., Biernat, Kristen A., Walton, William G., Wallace, Bret D., Creekmore, Benjamin C., Letertre, Marine M., Swann, Jonathan R., Wilson, Ian D., Roques, Jose R., Darr, David B., Bailey, Sean T., Montgomery, Stephanie A., Roach, Jeffrey M., Azcarate-Peril, M. Andrea, Sartor, R. Balfour, Gharaibeh, Raad Z., Bultman, Scott J., and Redinbo, Matthew R. Fri . "Targeted inhibition of gut bacterial β-glucuronidase activity enhances anticancer drug efficacy". United States. https://doi.org/10.1073/pnas.1918095117. https://www.osti.gov/servlets/purl/1644107.
@article{osti_1644107,
title = {Targeted inhibition of gut bacterial β-glucuronidase activity enhances anticancer drug efficacy},
author = {Bhatt, Aadra P. and Pellock, Samuel J. and Biernat, Kristen A. and Walton, William G. and Wallace, Bret D. and Creekmore, Benjamin C. and Letertre, Marine M. and Swann, Jonathan R. and Wilson, Ian D. and Roques, Jose R. and Darr, David B. and Bailey, Sean T. and Montgomery, Stephanie A. and Roach, Jeffrey M. and Azcarate-Peril, M. Andrea and Sartor, R. Balfour and Gharaibeh, Raad Z. and Bultman, Scott J. and Redinbo, Matthew R.},
abstractNote = {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.},
doi = {10.1073/pnas.1918095117},
journal = {Proceedings of the National Academy of Sciences of the United States of America},
number = 13,
volume = 117,
place = {United States},
year = {Fri Mar 13 00:00:00 EDT 2020},
month = {Fri Mar 13 00:00:00 EDT 2020}
}

Journal Article:
Free Publicly Available Full Text
Publisher's Version of Record

Citation Metrics:
Cited by: 77 works
Citation information provided by
Web of Science

Save / Share:

Works referenced in this record:

Structure, function, and inhibition of drug reactivating human gut microbial β-glucuronidases
journal, January 2019


Structure-activity relationships of flavonoids as natural inhibitors against E. coli β-glucuronidase
journal, November 2017


Three structurally and functionally distinct β-glucuronidases from the human gut microbe Bacteroides uniformis
journal, October 2018

  • Pellock, Samuel J.; Walton, William G.; Biernat, Kristen A.
  • Journal of Biological Chemistry, Vol. 293, Issue 48
  • DOI: 10.1074/jbc.RA118.005414

Mouse Gut Microbiome-Encoded β-Glucuronidases Identified Using Metagenome Analysis Guided by Protein Structure
journal, August 2019


Generation of Multimillion-Sequence 16S rRNA Gene Libraries from Complex Microbial Communities by Assembling Paired-End Illumina Reads
journal, April 2011

  • Bartram, Andrea K.; Lynch, Michael D. J.; Stearns, Jennifer C.
  • Applied and Environmental Microbiology, Vol. 77, Issue 11
  • DOI: 10.1128/AEM.02772-10

Sacituzumab Govitecan-hziy in Refractory Metastatic Triple-Negative Breast Cancer
journal, February 2019

  • Bardia, Aditya; Mayer, Ingrid A.; Vahdat, Linda T.
  • New England Journal of Medicine, Vol. 380, Issue 8
  • DOI: 10.1056/NEJMoa1814213

Old formula, new Rx: The journey of PHY906 as cancer adjuvant therapy
journal, April 2012


Microbes, Microbiota, and Colon Cancer
journal, March 2014


Topoisomerase I inhibitors: camptothecins and beyond
journal, October 2006

  • Pommier, Yves
  • Nature Reviews Cancer, Vol. 6, Issue 10
  • DOI: 10.1038/nrc1977

Review: Chemotherapy-induced diarrhea: pathophysiology, frequency and guideline-based management
journal, December 2009

  • Stein, Alexander; Voigt, Wieland; Jordan, Karin
  • Therapeutic Advances in Medical Oncology, Vol. 2, Issue 1
  • DOI: 10.1177/1758834009355164

In Fimo: A Term Proposed for Excrement Examined Experimentally
journal, April 2019


Discovering the Microbial Enzymes Driving Drug Toxicity with Activity-Based Protein Profiling
journal, October 2019


The role of the microbiome in cancer development and therapy: Microbiome and Cancer
journal, May 2017

  • Bhatt, Aadra P.; Redinbo, Matthew R.; Bultman, Scott J.
  • CA: A Cancer Journal for Clinicians, Vol. 67, Issue 4
  • DOI: 10.3322/caac.21398

An Atlas of β-Glucuronidases in the Human Intestinal Microbiome
journal, July 2017


FOLFIRINOX versus Gemcitabine for Metastatic Pancreatic Cancer
journal, May 2011

  • Conroy, Thierry; Desseigne, Françoise; Ychou, Marc
  • New England Journal of Medicine, Vol. 364, Issue 19
  • DOI: 10.1056/NEJMoa1011923

The Four-Herb Chinese Medicine PHY906 Reduces Chemotherapy-Induced Gastrointestinal Toxicity
journal, August 2010


Pharmacologic Targeting of Bacterial β-Glucuronidase Alleviates Nonsteroidal Anti-Inflammatory Drug-Induced Enteropathy in Mice
journal, February 2012

  • LoGuidice, Amanda; Wallace, Bret D.; Bendel, Lauren
  • Journal of Pharmacology and Experimental Therapeutics, Vol. 341, Issue 2
  • DOI: 10.1124/jpet.111.191122

Good Bug, Bad Bug: Breaking through Microbial Stereotypes
journal, January 2018


Microbial Glucuronidase Inhibition Reduces Severity of Diclofenac-Induced Anastomotic Leak in Rats
journal, May 2018

  • Yauw, Simon T. K.; Arron, Melissa; Lomme, Roger M. L. M.
  • Surgical Infections, Vol. 19, Issue 4
  • DOI: 10.1089/sur.2017.245

Irinotecan-induced mucositis manifesting as diarrhoea corresponds with an amended intestinal flora and mucin profile
journal, October 2009

  • Stringer, Andrea M.; Gibson, Rachel J.; Bowen, Joanne M.
  • International Journal of Experimental Pathology, Vol. 90, Issue 5
  • DOI: 10.1111/j.1365-2613.2009.00671.x

Discovery and Characterization of FMN-Binding β-Glucuronidases in the Human Gut Microbiome
journal, March 2019

  • Pellock, Samuel J.; Walton, William G.; Ervin, Samantha M.
  • Journal of Molecular Biology, Vol. 431, Issue 5
  • DOI: 10.1016/j.jmb.2019.01.013

Gefitinib Enhances the Antitumor Activity and Oral Bioavailability of Irinotecan in Mice
journal, October 2004


The C3(1)/SV40 T-antigen transgenic mouse model of mammary cancer: ductal epithelial cell targeting with multistage progression to carcinoma
journal, February 2000

  • Green, Jeffrey E.; Shibata, Masa-Aki; Yoshidome, Katsuhide
  • Oncogene, Vol. 19, Issue 8
  • DOI: 10.1038/sj.onc.1203280

Molecular Insights into Microbial β -Glucuronidase Inhibition to Abrogate CPT-11 Toxicity
journal, May 2013

  • Roberts, Adam B.; Wallace, Bret D.; Venkatesh, Madhu Kumar
  • Molecular Pharmacology, Vol. 84, Issue 2
  • DOI: 10.1124/mol.113.085852

Vancomycin relieves mycophenolate mofetil–induced gastrointestinal toxicity by eliminating gut bacterial β-glucuronidase activity
journal, August 2019

  • Taylor, Michael R.; Flannigan, Kyle L.; Rahim, Hannah
  • Science Advances, Vol. 5, Issue 8
  • DOI: 10.1126/sciadv.aax2358

Dynamic Reprogramming of the Kinome in Response to Targeted MEK Inhibition in Triple-Negative Breast Cancer
journal, April 2012


Structural basis for the regulation of β-glucuronidase expression by human gut Enterobacteriaceae
journal, December 2017

  • Little, Michael S.; Pellock, Samuel J.; Walton, William G.
  • Proceedings of the National Academy of Sciences, Vol. 115, Issue 2
  • DOI: 10.1073/pnas.1716241115

Structure and Inhibition of Microbiome β-Glucuronidases Essential to the Alleviation of Cancer Drug Toxicity
journal, September 2015


Irinotecan (CPT-11) Chemotherapy Alters Intestinal Microbiota in Tumour Bearing Rats
journal, July 2012


Akkermansia muciniphila gen. nov., sp. nov., a human intestinal mucin-degrading bacterium
journal, September 2004

  • Derrien, M.
  • INTERNATIONAL JOURNAL OF SYSTEMATIC AND EVOLUTIONARY MICROBIOLOGY, Vol. 54, Issue 5
  • DOI: 10.1099/ijs.0.02873-0

Alleviating Cancer Drug Toxicity by Inhibiting a Bacterial Enzyme
journal, November 2010


Selecting a Single Stereocenter: The Molecular Nuances That Differentiate β-Hexuronidases in the Human Gut Microbiome
journal, February 2019


A High Throughput Assay for Discovery of Bacterial β-Glucuronidase Inhibitors
journal, April 2011


Gut Microbial β-Glucuronidase Inhibition via Catalytic Cycle Interception
journal, July 2018

  • Pellock, Samuel J.; Creekmore, Benjamin C.; Walton, William G.
  • ACS Central Science, Vol. 4, Issue 7
  • DOI: 10.1021/acscentsci.8b00239

Multiple NSAID-Induced Hits Injure the Small Intestine: Underlying Mechanisms and Novel Strategies
journal, October 2012

  • Boelsterli, Urs A.; Redinbo, Matthew R.; Saitta, Kyle S.
  • Toxicological Sciences, Vol. 131, Issue 2
  • DOI: 10.1093/toxsci/kfs310

Pharmacogenetics of irinotecan metabolism and transport: An update
journal, March 2006