Review Special Issues

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

  • Received: 20 August 2018 Accepted: 24 October 2018 Published: 26 October 2018
  • 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.

    Citation: Ellie Sayers, Alex MacGregor, Simon R. Carding. Drug-microbiota interactions and treatment response: Relevance to rheumatoid arthritis[J]. AIMS Microbiology, 2018, 4(4): 642-654. doi: 10.3934/microbiol.2018.4.642

    Related Papers:

  • 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.


    加载中
    [1] Carding S, Verbeke K, Vipond DT, et al. (2015) Dysbiosis of the gut microbiota in disease. Microb Ecol Heal Dis 26: 26191–26199.
    [2] Scher JU, Sczesnak A, Longman RS, et al. (2013) Expansion of intestinal Prevotella copri correlates with enhanced susceptibility to arthritis. Elife 2: e01202. doi: 10.7554/eLife.01202
    [3] Liu X, Zou Q, Zeng B, et al. (2013) Analysis of fecal Lactobacillus community structure in patients with early rheumatoid arthritis. Curr Microbiol 67: 170–176. doi: 10.1007/s00284-013-0338-1
    [4] Zhang X, Zhang D, Jia H, et al. (2015) The oral and gut microbiotas are perturbed in rheumatoid arthritis and partly normalized after treatment. Nat Med 21: 895–905. doi: 10.1038/nm.3914
    [5] Chen J, Wright K, Davis JM, et al. (2016) An expansion of rare lineage intestinal microbes characterizes rheumatoid arthritis. Genome Med 8: 43–51. doi: 10.1186/s13073-016-0299-7
    [6] Sender R, Fuchs S, Milo R (2016) Revised estimates for the number of human and bacteria cells in the body. PLoS Biol 14: e1002533. doi: 10.1371/journal.pbio.1002533
    [7] Falony G, Joossens M, Vieira-Silva S, et al. (2016) Population-level analysis of gut microbiota variation. Science 352: 560–564. doi: 10.1126/science.aad3503
    [8] Koppel N, Rekdal VM, Balskus EP (2017) Chemical transformation of xenobiotics by the human gut microbiota. Science 356: 1246–1257.
    [9] Klatt NR, Cheu R, Birse K, et al. (2017) Vaginal bacteria modify HIV tenofovir microbicide efficacy in African women. Science 356: 938–945. doi: 10.1126/science.aai9383
    [10] Koh A, De Vadder F, Kovatcheva-Datchary P, et al. (2016) From dietary fiber to host physiology: Short-chain fatty acids as key bacterial metabolites. Cell 165: 1332–1345. doi: 10.1016/j.cell.2016.05.041
    [11] Takasuna K, Hagiwara T, Hirohashi M, et al. (1996) Involvement of b-glucuronidase in intestinal microflora in the intestinal toxicity of the antitumor camptothecin derivative irinotecan hydrochloride (CPT-11 ) in rats. Cancer Res 56: 3752–3757.
    [12] Lindenbaum J, Rund DG, Butler VP, et al. (1981) Inactivation of digoxin by the gut flora: Reversal by antibiotic therapy. N Engl J Med 305: 789–794. doi: 10.1056/NEJM198110013051403
    [13] Dobkin J, Saha J, Butler V, et al. (1982) Digoxin-inactivating bacteria: Identification in human gut flora. Science 220: 325–327.
    [14] Haiser HJ, Gootenberg DB, Chatman K, et al. (2013) Predicting and manipulating cardiac drug inactivation by the human gut bacterium Eggerthella lenta. Science 341: 295–298. doi: 10.1126/science.1235872
    [15] Rafii F, Franklin W, Cerniglia CE (1990) Azoreductase activity of anaerobic bacteria isolated from human intestinal microflora. Appl Environ Microbiol 7: 2146–2151.
    [16] Rafii F, Cerniglia CE (1995) Reduction of azo dyes and nitroaromatic compounds by bacterial enzymes from the human intestinal tract. Environ Health Persp 103: 17–19.
    [17] Azadkhan A, Truelove S, Aronson J (1982) The disposition and metabolism of sulphasalazine (salicylazosulphapyridine) in man. Brit J Clin Pharmaco 13: 523–528. doi: 10.1111/j.1365-2125.1982.tb01415.x
    [18] Bishop JB, Witt KL, Gulati DK, et al. (1990) Evaluation of the mutagenicity of the anti-inflammatory drug salicylazosulfapyridine (SASP). Mutagenesis 5: 549–554. doi: 10.1093/mutage/5.6.549
    [19] Das KM, Eastwood MA, McManus JPA, et al. (1973) Adverse reactions during salicylazosulfapyridine therapy and the relation with drug metabolism and acetylator phenotype. N Engl J Med 289: 491–495. doi: 10.1056/NEJM197309062891001
    [20] Dingsdag SA, Hunter N (2017) Metronidazole: an update on metabolism, structure-cytotoxicity and resistance mechanisms. J Antimicrob Chemoth 73: 265–279.
    [21] Bendesky A, Menéndez D, Ostrosky-Wegman P (2002) Is metronidazole carcinogenic? Mutat Res-Rev Mutat 511: 133–144. doi: 10.1016/S1383-5742(02)00007-8
    [22] Chrystal EJT, Koch RL, McLafferty MA, et al. (1980) Relationship between metronidazole metabolism and bactericidal activity. Antimicrob Agents Ch 18: 566–573. doi: 10.1128/AAC.18.4.566
    [23] Pardoll DM (2012) The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 12: 252–264. doi: 10.1038/nrc3239
    [24] Dubin K, Callahan MK, Ren B, et al. (2016) Intestinal microbiota analyses identify melanoma patients at risk for checkpoint-blockade-induced colitis. Nat Commun 7: 10391–10397. doi: 10.1038/ncomms10391
    [25] Sivan A, Corrales L, Hubert N, et al. (2015) Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science 350: 1084–1089. doi: 10.1126/science.aac4255
    [26] Routy B, Le Chatelier E, Derosa L, et al. (2018) Gut microbiota influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 359: 91–97. doi: 10.1126/science.aan3706
    [27] Matson V, Fessler J, Bao R, et al. (2018) The commensal microbiota is associated with anti- PD-1 efficacy in metastatic melanoma patients. Science 359: 104–108. doi: 10.1126/science.aao3290
    [28] Vétizou M, Pitt JM, Daillère R, et al. (2015) Anticancer immunotherapy by CTLA-4 blockade relies on the gutmicrobiota. Science 350: 1079–1084. doi: 10.1126/science.aad1329
    [29] Shin NR, Lee JC, Lee HY, et al. (2014) An increase in the Akkermansia spp. population induced by metformin treatment improves glucose homeostasis in diet-induced obese mice. Gut 63: 727–735.
    [30] Bonora E, Cigolini M, Bosello O, et al. (1984) 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 9: 47–51. doi: 10.1185/03007998409109558
    [31] Foretz M, Guigas B, Bertrand L, et al. (2014) Metformin: From mechanisms of action to therapies. Cell Metab 20: 953–966. doi: 10.1016/j.cmet.2014.09.018
    [32] Qin J, Li Y, Cai K, et al. (2012) A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 490: 55–60. doi: 10.1038/nature11450
    [33] Forslund K, Hildebrand F, Nielsen T, et al. (2015) Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Nature 528: 262–266. doi: 10.1038/nature15766
    [34] De La Cuesta-Zuluaga J, Mueller NT, Corrales-Agudelo V, et al. (2017) 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 40: 54–62. doi: 10.2337/dc16-1324
    [35] Lee H (2014) Effect of metformin on metabolic improvement and gut microbiota. Appl Environ Microbiol 80: 5935–5943. doi: 10.1128/AEM.01357-14
    [36] Everard A, Belzer C, Geurts L, et al. (2013) Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. P Natl Acad Sci USA 110: 9066–9071. doi: 10.1073/pnas.1219451110
    [37] Lie E, Uhlig T, van der Heijde D, et al. (201) Effectiveness of sulfasalazine and methotrexate in 1102 DMARD-nave patients with early RA. Rheumatology 51: 670–678.
    [38] Wijbrandts CA, Tak PP (2017) Prediction of response to targeted treatment in rheumatoid arthritis. Mayo Clin Proc 92: 1129–1143. doi: 10.1016/j.mayocp.2017.05.009
    [39] Cutulo M, Sulli A, Pizzorni C, et al. (2001) Anti-inflammatory mechanisms of methotrexate in rheumatoid arthritis. Ann Rheum Dis 60: 729–735. doi: 10.1136/ard.60.8.729
    [40] Takahashi C, Kaneko Y, Okano Y, et al. (2017) 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 3: e000363. doi: 10.1136/rmdopen-2016-000363
    [41] Tian H, Cronstein BN (2007) Understanding the mechanisms of action of methotrexate-Implications for the treatment of rheumatoid arthritis. Bull NYU Hosp Jt Dis 65: 168–173.
    [42] Levy CC, Goldman P (1967) The enzymatic hydrolysis of methotrexate. J Biol Chem 242: 2933–2938.
    [43] Webb M (1955) Inactivation of analogues of folic acid by certain non-exacting bacteria. Biochem Biophys Acta 17: 212–225. doi: 10.1016/0006-3002(55)90352-8
    [44] Larimer CM, Slavnic D, Pitstick LD, et al. (2014) Comparison of substrate specificity of Escherichia coli p-Aminobenzoyl-glutamate hydrolase with Pseudomonas carboxypeptidase G. Adv Enzym Res 2: 39–48. doi: 10.4236/aer.2014.21004
    [45] Nayak R, O'Loughlin C, Fischbach M, et al. (2016) Methotrexate is an antibacterial drug metabolized by human gut bacteria-ACR Meeting Abstracts. Arthritis Rheumatol 68: 10.
    [46] Dervieux T, Zablocki R, Kremer J (2010) Red blood cell methotrexate polyglutamates emerge as a function of dosage intensity and route of administration during pulse methotrexate therapy in rheumatoid arthritis. Rheumatology 49: 2337–2345. doi: 10.1093/rheumatology/keq216
    [47] Busquets D, Mas-de-Xaxars T, López-Siles M, et al. (2015) Anti-tumour necrosis factor treatment with adalimumab induces changes in the microbiota of Crohn's Disease. J Crohns Colitis 9: 899–906. doi: 10.1093/ecco-jcc/jjv119
    [48] Bazin T, Hooks KB, Barnetche T, et al. (2018) Microbiota composition may predict anti-TNF alpha response in spondyloarthritis patients: An exploratory study. Sci Rep 8: 5446–5449. doi: 10.1038/s41598-018-23571-4
    [49] Andersen V, Holmskov U, Sørensen SB, et al. (2017) 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 9: 499–508. doi: 10.3390/nu9050499
  • Reader Comments
  • © 2018 the Author(s), licensee AIMS Press. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0)
通讯作者: 陈斌, bchen63@163.com
  • 1. 

    沈阳化工大学材料科学与工程学院 沈阳 110142

  1. 本站搜索
  2. 百度学术搜索
  3. 万方数据库搜索
  4. CNKI搜索

Metrics

Article views(4841) PDF downloads(835) Cited by(24)

Article outline

Figures and Tables

Figures(2)

Other Articles By Authors

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog