Drug-microbiota interactions and treatment response: Relevance to rheumatoid arthritis

Ellie Sayers, Alex MacGregor, Simon R Carding, Ellie Sayers, Alex MacGregor, Simon R Carding

Abstract

Knowledge about associations between changes in the structure and/or function of intestinal microbes (the microbiota) and the pathogenesis of various diseases is expanding. However, interactions between the intestinal microbiota and different pharmaceuticals and the impact of these on responses to treatment are less well studied. Several mechanisms are known by which drug-microbiota interactions can influence drug bioavailability, efficacy, and/or toxicity. This includes direct activation or inactivation of drugs by microbial enzymes which can enhance or reduce drug effectiveness. The extensive metabolic capabilities of the intestinal microbiota make it a hotspot for drug modification. However, drugs can also influence the microbiota profoundly and change the outcome of interactions with the host. Additionally, individual microbiota signatures are unique, leading to substantial variation in host responses to particular drugs. In this review, we describe several known and emerging examples of how drug-microbiota interactions influence the responses of patients to treatment for various diseases, including inflammatory bowel disease, type 2 diabetes and cancer. Focussing on rheumatoid arthritis (RA), a chronic inflammatory disease of the joints which has been linked with microbial dysbiosis, we propose mechanisms by which the intestinal microbiota may affect responses to treatment with methotrexate which are highly variable. Furthering our knowledge of this subject will eventually lead to the adoption of new treatment strategies incorporating microbiota signatures to predict or improve treatment outcomes.

Keywords: drug response; intestinal microbiota; metabolism; methotrexate.

Conflict of interest statement

Conflict of interest: The authors declare no conflicts of interest in this paper.

Figures

Figure 1.. Mechanisms for drug-microbiota interactions. There…
Figure 1.. Mechanisms for drug-microbiota interactions. There are several known mechanisms for drug-microbiota interaction that may affect treatment outcomes. Of course, it is not always the case that there will be interactions. For example, some drugs may bypass the intestinal microbiota altogether (e). However, others will be enzymatically activated (b) or inactivated (a) by certain microbes or be converted into potentially toxic substances (f). More recently, the composition of the microbiota has been linked to treatment outcomes, either in association with taking a drug (c) or in response to the existence of certain microbes prior to its use (d). ICI = immune checkpoint inhibitor; SCFA = short chain fatty acid.
Figure 2.. Transport and metabolism of methotrexate…
Figure 2.. Transport and metabolism of methotrexate in humans. Methotrexate (MTX) is transported into cells via the reduced folate transporter (RFT) or proton-coupled folate transporter (PCFT). On entry, MTX is polyglutamated to MTX-PG by the enzyme folylpolyglutamate synthase (FPGS), which retains the drug inside the cell and increases its affinity for the drug target. MTX-PG may then have glutamate entities removed by glutamate carboxypeptidase (GCP), and the single-glutamate MTX may be transported out of the cell and re-enter circulation.

References

    1. Carding S, Verbeke K, Vipond DT, et al. Dysbiosis of the gut microbiota in disease. Microb Ecol Heal Dis. 2015;26:26191–26199.
    1. Scher JU, Sczesnak A, Longman RS, et al. Expansion of intestinal Prevotella copri correlates with enhanced susceptibility to arthritis. Elife. 2013;2:e01202.
    1. Liu X, Zou Q, Zeng B, et al. Analysis of fecal Lactobacillus community structure in patients with early rheumatoid arthritis. Curr Microbiol. 2013;67:170–176.
    1. Zhang X, Zhang D, Jia H, et al. The oral and gut microbiotas are perturbed in rheumatoid arthritis and partly normalized after treatment. Nat Med. 2015;21:895–905.
    1. Chen J, Wright K, Davis JM, et al. An expansion of rare lineage intestinal microbes characterizes rheumatoid arthritis. Genome Med. 2016;8:43–51.
    1. Sender R, Fuchs S, Milo R. Revised estimates for the number of human and bacteria cells in the body. PLoS Biol. 2016;14:e1002533.
    1. Falony G, Joossens M, Vieira-Silva S, et al. Population-level analysis of gut microbiota variation. Science. 2016;352:560–564.
    1. Koppel N, Rekdal VM, Balskus EP. Chemical transformation of xenobiotics by the human gut microbiota. Science. 2017;356:1246–1257.
    1. Klatt NR, Cheu R, Birse K, et al. Vaginal bacteria modify HIV tenofovir microbicide efficacy in African women. Science. 2017;356:938–945.
    1. Koh A, De Vadder F, Kovatcheva-Datchary P, et al. From dietary fiber to host physiology: Short-chain fatty acids as key bacterial metabolites. Cell. 2016;165:1332–1345.
    1. Takasuna K, Hagiwara T, Hirohashi M, et al. Involvement of β-glucuronidase in intestinal microflora in the intestinal toxicity of the antitumor camptothecin derivative irinotecan hydrochloride (CPT-11) in rats. Cancer Res. 1996;56:3752–3757.
    1. Lindenbaum J, Rund DG, Butler VP, et al. Inactivation of digoxin by the gut flora: Reversal by antibiotic therapy. N Engl J Med. 1981;305:789–794.
    1. Dobkin J, Saha J, Butler V, et al. Digoxin-inactivating bacteria: Identification in human gut flora. Science. 1982;220:325–327.
    1. Haiser HJ, Gootenberg DB, Chatman K, et al. Predicting and manipulating cardiac drug inactivation by the human gut bacterium Eggerthella lenta. Science. 2013;341:295–298.
    1. Rafii F, Franklin W, Cerniglia CE. Azoreductase activity of anaerobic bacteria isolated from human intestinal microflora. Appl Environ Microbiol. 1990;7:2146–2151.
    1. Rafii F, Cerniglia CE. Reduction of azo dyes and nitroaromatic compounds by bacterial enzymes from the human intestinal tract. Environ Health Persp. 1995;103:17–19.
    1. Azadkhan A, Truelove S, Aronson J. The disposition and metabolism of sulphasalazine (salicylazosulphapyridine) in man. Brit J Clin Pharmaco. 1982;13:523–528.
    1. Bishop JB, Witt KL, Gulati DK, et al. Evaluation of the mutagenicity of the anti-inflammatory drug salicylazosulfapyridine (SASP) Mutagenesis. 1990;5:549–554.
    1. Das KM, Eastwood MA, McManus JPA, et al. Adverse reactions during salicylazosulfapyridine therapy and the relation with drug metabolism and acetylator phenotype. N Engl J Med. 1973;289:491–495.
    1. Dingsdag SA, Hunter N. Metronidazole: an update on metabolism, structure-cytotoxicity and resistance mechanisms. J Antimicrob Chemoth. 2017;73:265–279.
    1. Bendesky A, Menéndez D, Ostrosky-Wegman P. Is metronidazole carcinogenic? Mutat Res-Rev Mutat. 2002;511:133–144.
    1. Chrystal EJT, Koch RL, McLafferty MA, et al. Relationship between metronidazole metabolism and bactericidal activity. Antimicrob Agents Ch. 1980;18:566–573.
    1. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12:252–264.
    1. Dubin K, Callahan MK, Ren B, et al. Intestinal microbiota analyses identify melanoma patients at risk for checkpoint-blockade-induced colitis. Nat Commun. 2016;7:10391–10397.
    1. Sivan A, Corrales L, Hubert N, et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science. 2015;350:1084–1089.
    1. Routy B, Le Chatelier E, Derosa L, et al. Gut microbiota influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science. 2018;359:91–97.
    1. Matson V, Fessler J, Bao R, et al. The commensal microbiota is associated with anti- PD-1 efficacy in metastatic melanoma patients. Science. 2018;359:104–108.
    1. Vétizou M, Pitt JM, Daillère R, et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gutmicrobiota. Science. 2015;350:1079–1084.
    1. Shin NR, Lee JC, Lee HY, et al. An increase in the Akkermansia spp. population induced by metformin treatment improves glucose homeostasis in diet-induced obese mice. Gut. 2014;63:727–735.
    1. Bonora E, Cigolini M, Bosello O, et al. Lack of effect of intravenous metformin on plasma concentrations of glucose, insulin, C-peptide, glucagon and growth hormone in non-diabetic subjects. Curr Med Res Opin. 1984;9:47–51.
    1. Foretz M, Guigas B, Bertrand L, et al. Metformin: From mechanisms of action to therapies. Cell Metab. 2014;20:953–966.
    1. Qin J, Li Y, Cai K, et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature. 2012;490:55–60.
    1. Forslund K, Hildebrand F, Nielsen T, et al. Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Nature. 2015;528:262–266.
    1. De La Cuesta-Zuluaga J, Mueller NT, Corrales-Agudelo V, et al. Metformin is associated with higher relative abundance of mucin-degrading akkermansia muciniphila and several short-chain fatty acid-producing microbiota in the gut. Diabetes Care. 2017;40:54–62.
    1. Lee H. Effect of metformin on metabolic improvement and gut microbiota. Appl Environ Microbiol. 2014;80:5935–5943.
    1. Everard A, Belzer C, Geurts L, et al. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. P Natl Acad Sci USA. 2013;110:9066–9071.
    1. Lie E, Uhlig T, van der Heijde D, et al. Effectiveness of sulfasalazine and methotrexate in 1102 DMARD-nave patients with early RA. Rheumatology. 201;51:670–678.
    1. Wijbrandts CA, Tak PP. Prediction of response to targeted treatment in rheumatoid arthritis. Mayo Clin Proc. 2017;92:1129–1143.
    1. Cutulo M, Sulli A, Pizzorni C, et al. Anti-inflammatory mechanisms of methotrexate in rheumatoid arthritis. Ann Rheum Dis. 2001;60:729–735.
    1. Takahashi C, Kaneko Y, Okano Y, et al. Association of erythrocyte methotrexate- polyglutamate levels with the efficacy and hepatotoxicity of methotrexate in patients with rheumatoid arthritis: a 76-week prospective study. RMD Open. 2017;3:e000363.
    1. Tian H, Cronstein BN. Understanding the mechanisms of action of methotrexate—Implications for the treatment of rheumatoid arthritis. Bull NYU Hosp Jt Dis. 2007;65:168–173.
    1. Levy CC, Goldman P. The enzymatic hydrolysis of methotrexate. J Biol Chem. 1967;242:2933–2938.
    1. Webb M. Inactivation of analogues of folic acid by certain non-exacting bacteria. Biochem Biophys Acta. 1955;17:212–225.
    1. Larimer CM, Slavnic D, Pitstick LD, et al. Comparison of substrate specificity of Escherichia coli p-Aminobenzoyl-glutamate hydrolase with Pseudomonas carboxypeptidase G. Adv Enzym Res. 2014;2:39–48.
    1. Nayak R, O'Loughlin C, Fischbach M, et al. Methotrexate is an antibacterial drug metabolized by human gut bacteria—ACR Meeting Abstracts. Arthritis Rheumatol. 2016;68:10.
    1. Dervieux T, Zablocki R, Kremer J. Red blood cell methotrexate polyglutamates emerge as a function of dosage intensity and route of administration during pulse methotrexate therapy in rheumatoid arthritis. Rheumatology. 2010;49:2337–2345.
    1. Busquets D, Mas-de-Xaxars T, López-Siles M, et al. Anti-tumour necrosis factor treatment with adalimumab induces changes in the microbiota of Crohn's Disease. J Crohns Colitis. 2015;9:899–906.
    1. Bazin T, Hooks KB, Barnetche T, et al. Microbiota composition may predict anti-TNF alpha response in spondyloarthritis patients: An exploratory study. Sci Rep. 2018;8:5446–5449.
    1. Andersen V, Holmskov U, Sørensen SB, et al. Proposal for a study on treatment selection and lifestyle recommendations in chronic inflammatory diseases: A Danish multidisciplinary collaboration on prognostic factors and personalised medicine. Nutrients. 2017;9:499–508.

Source: PubMed

3
Abonner