Research article Special Issues

Microbiome predictors of dysbiosis and VRE decolonization in patients with recurrent C. difficile infections in a multi-center retrospective study

  • Received: 01 November 2018 Accepted: 23 December 2018 Published: 17 January 2019
  • The gastrointestinal microbiome is intrinsically linked to the spread of antibiotic resistance. Antibiotic treatment puts patients at risk for colonization by opportunistic pathogens like vancomycin resistant Enterococcus and Clostridioides difficile by destroying the colonization resistance provided by the commensal microbiota. Once colonized, the host is at a much higher risk for infection by that pathogen. Furthermore, we know that microbiome community differences are associated with disease states, but we do not have a good understanding of how we can use these changes to classify different patient populations. To that end, we have performed a multicenter retrospective analysis on patients who received fecal microbiota transplants to treat recurrent Clostridioides difficile infection. We performed 16S rRNA gene sequencing on fecal samples collected as part of this study and used these data to develop a microbiome disruption index. Our microbiome disruption index is a simple index that is predictive across cohorts, indications, and batch effects. We are able to classify pre-fecal transplant vs post-fecal transplant samples in patients with recurrent C. difficile infection, and we are able to predict, using previously-published data from a cohort of patients receiving hematopoietic stem cell transplants, which patients would go on to develop bloodstream infections. Finally, we also identified patients in this cohort that were initially colonized with vancomycin resistant Enterococcus and that 92% (11/12) were decolonized after the transplant, but the microbiome disruption index was unable to predict such decolonization. We, however, were able to compare the relative abundance of different taxa between the two groups, and we found that increased abundance of Enterobacteriaceae predicts whether patients were colonized with vancomycin resistant Enterococcus. This work is an early step towards a better understanding of how microbiome predictors can be used to help improve patient care and patient outcomes.

    Citation: Marina Santiago, Lindsay Eysenbach, Jessica Allegretti, Olga Aroniadis, Lawrence J. Brandt, Monika Fischer, Ari Grinspan, Colleen Kelly, Casey Morrow, Martin Rodriguez, Majdi Osman, Zain Kassam, Mark B. Smith, Sonia Timberlake. Microbiome predictors of dysbiosis and VRE decolonization in patients with recurrent C. difficile infections in a multi-center retrospective study[J]. AIMS Microbiology, 2019, 5(1): 1-18. doi: 10.3934/microbiol.2019.1.1

    Related Papers:

  • The gastrointestinal microbiome is intrinsically linked to the spread of antibiotic resistance. Antibiotic treatment puts patients at risk for colonization by opportunistic pathogens like vancomycin resistant Enterococcus and Clostridioides difficile by destroying the colonization resistance provided by the commensal microbiota. Once colonized, the host is at a much higher risk for infection by that pathogen. Furthermore, we know that microbiome community differences are associated with disease states, but we do not have a good understanding of how we can use these changes to classify different patient populations. To that end, we have performed a multicenter retrospective analysis on patients who received fecal microbiota transplants to treat recurrent Clostridioides difficile infection. We performed 16S rRNA gene sequencing on fecal samples collected as part of this study and used these data to develop a microbiome disruption index. Our microbiome disruption index is a simple index that is predictive across cohorts, indications, and batch effects. We are able to classify pre-fecal transplant vs post-fecal transplant samples in patients with recurrent C. difficile infection, and we are able to predict, using previously-published data from a cohort of patients receiving hematopoietic stem cell transplants, which patients would go on to develop bloodstream infections. Finally, we also identified patients in this cohort that were initially colonized with vancomycin resistant Enterococcus and that 92% (11/12) were decolonized after the transplant, but the microbiome disruption index was unable to predict such decolonization. We, however, were able to compare the relative abundance of different taxa between the two groups, and we found that increased abundance of Enterobacteriaceae predicts whether patients were colonized with vancomycin resistant Enterococcus. This work is an early step towards a better understanding of how microbiome predictors can be used to help improve patient care and patient outcomes.


    加载中

    Acknowledgments



    The microbiome disruption index was developed by OpenBiome with the assistance of Finch Therapeutics and the support of CDC (contract numbers 200-2016-91946 and 200-2016-91948).

    Conflict of interest



    MS, ZK, ST, and MBS are employees of Finch Therapeutics Group Inc. JRA consults for and had research support from Finch Therapeutics Group. AG is a consultant for Merck. The authors declare no conflicts of interest in this paper.

    [1] Antibiotic Resistance Threats in the United States, 2013. US Department of Health and Human Services, Centers for Disease Control and Prevention, 2013.
    [2] Knight GM, Costelloe C, Deeny SR, et al. (2018) Quantifying where human acquisition of antibiotic resistance occurs: a mathematical modelling study. BMC Med 16: 137. doi: 10.1186/s12916-018-1121-8
    [3] Sassone-Corsi M, Raffatellu M (2015) No vacancy: how beneficial microbes cooperate with immunity to provide colonization resistance to pathogens. J Immunol 194: 4081–4087. doi: 10.4049/jimmunol.1403169
    [4] Olsan EE, Byndloss MX, Faber F, et al. (2017) Colonization resistance: the deconvolution of a complex trait. J Biol Chem 292: 8577–8581. doi: 10.1074/jbc.R116.752295
    [5] McKenney ES, Kendall MM (2016) Microbiota and pathogen 'pas de deux': setting up and breaking down barriers to intestinal infection. Pathog Dis 74: ftw051. doi: 10.1093/femspd/ftw051
    [6] Sassone-Corsi M, Nuccio S, Liu H, et al. (2016) Microcins mediate competition among Enterobacteriaceae in the inflamed gut. Nature 540: 280–283. doi: 10.1038/nature20557
    [7] Brandl K, Plitas G, Mihu C, et al. (2008) Vancomycin-resistant enterococci exploit antibiotic-induced innate immune deficits. Nature 455: 804–807. doi: 10.1038/nature07250
    [8] Theriot CM, Young VB (2015) Interactions between the gastrointestinal microbiome and Clostridium difficile. Annu Rev Microbiol 69: 445–461. doi: 10.1146/annurev-micro-091014-104115
    [9] Freedman A (2014) Use of stool transplant to clear fecal colonization with carbapenem-resistant Enterobacteraciae (CRE): proof of concept. IDWeek.
    [10] Singh R, Nood E, Nieuwdorp, et al. (2014) Donor feces infusion for eradication of Extended Spectrum beta-Lactamase producing Escherichia coli in a patient with end stage renal disease. Clin Microbiol Infec 20: O977–O978. doi: 10.1111/1469-0691.12683
    [11] Stripling J, Kumar R, Baddley JW, et al. (2015) Loss of vancomycin-resistant Enterococcus fecal dominance in an organ transplant patient with Clostridium difficile colitis after fecal microbiota transplant. Open Forum Infect Dis 2: 1–4.
    [12] Crun-Cianfione N, Sullivan E, Gonzalo B (2015) Fecal microbiota transplantation and successful resolution of multidrug-resistant-organism colonization. J Clin Microbiol 53: 1986–1989. doi: 10.1128/JCM.00820-15
    [13] Jang M, An J, Jung S, et al. (2014) Refractory Clostridium difficile infection cured with fecal microbiota transplantation in vancomycin-resistant Enterococcus colonized patient. Intest Res 13: 80–84.
    [14] Lombardo M (2015) Vancomycin-resistant enterococcal (VRE) titers diminish among patients with recurrent Clostridium difficile infection after administration of SER-109, a novel microbiome agent. IDWeek.
    [15] Biliński J, Grzesiowski P, Muszynski J, et al. (2016) Fecal microbiota transplantation inhibits multidrug-resistant gut pathogens: preliminary report performed in an immunocompromised host. Arch Immunol Ther Ex 64: 255–258. doi: 10.1007/s00005-016-0387-9
    [16] Lagier J, Million M, Fournier P, et al. (2015) Faecal microbiota transplantation for stool decolonization of OXA-48 carbapenemase-producing Klebsiella pneumoniae. J Hosp Infect 90: 173–174. doi: 10.1016/j.jhin.2015.02.013
    [17] Wei Y, Gong J, Zhu W, et al. (2015) Fecal microbiota transplantation restores dysbiosis in patients with methicillin resistant Staphylococcus aureus enterocolitis. BMC Infect Dis 15: 265. doi: 10.1186/s12879-015-0973-1
    [18] Eysenbach L, Allegretti JR, Aroniadis O, et al. (2016) Clearance of vancomycin-resistant Enterococcus colonization with fecal microbiota transplantation among patients with recurrent Clostridium difficile infection. IDWeek.
    [19] Dubberke ER, Mullane KM, Gerding DN, et al. (2016) Clearance of vancomycin-resistant Enterococcus concomitant with administration of a microbiota-based drug targeted at recurrent Clostridium difficile infection. Open Forum Infect Dis 3: ofw133. doi: 10.1093/ofid/ofw133
    [20] Jouhten H, Mattila E, Arkkila P, et al. (2016) Reduction of antibiotic resistance genes in intestinal microbiota of patients with recurrent Clostridium difficile infection after fecal microbiota transplantation. Clin Infect Dis 63: 710–711. doi: 10.1093/cid/ciw390
    [21] Millan B, Park H, Hotte N, et al. (2016) Fecal microbial transplants reduce antibiotic-resistant genes in patients with recurrent Clostridium difficile infection. Clin Infect Dis 62: 1479–1486. doi: 10.1093/cid/ciw185
    [22] García-Fernández S, Morosini M, Cobo M, et al. (2016) Gut eradication of VIM-1 producing ST9 Klebsiella oxytoca after fecal microbiota transplantation for diarrhea caused by a Clostridium difficile hypervirulent R027 strain. Diagn Micr Infec Dis 86: 470–471. doi: 10.1016/j.diagmicrobio.2016.09.004
    [23] Sohn KM, Cheon S, Kim YS (2016) Can fecal microbiota transplantation (FMT) eradicate fecal colonization with vancomycin-resistant Enterococci (VRE)? Infect Cont Hosp Ep 37: 1519–1521.
    [24] Davido B, Batista R, Michelon H, et al. (2017) Is faecal microbiota transplantation an option to eradicate highly drug-resistant enteric bacteria carriage? J Hosp Infect 95: 433–437. doi: 10.1016/j.jhin.2017.02.001
    [25] Ponte A, Pinho R, Mota M (2017) Fecal microbiota transplantation: is there a role in the eradication of carbapenem-resistant Klebsiella pneumoniae intestinal carriage? Rev Esp Enferm Dig 109: 392.
    [26] Bilinski J, Grzesiowski P, Sorensen N, et al. (2017) Fecal microbiota transplantation in patients with blood disorders inhibits gut colonization with antibiotic-resistant bacteria: results of a prospective, single-center study. Clin Infect Dis 65: 364–370. doi: 10.1093/cid/cix252
    [27] Dias C, Pipa S, Duarte-Ribeiro F, et al. (2018) Fecal microbiota transplantation as a potential way to eradicate multiresistant microorganisms. IDCases 13: e00432. doi: 10.1016/j.idcr.2018.e00432
    [28] Safdar N, Sengupta S, Musuuza JS, et al. (2017) Status of the prevention of multidrug-resistant organisms in international settings: a survey of the society for healthcare epidemiology of america research network. Infect Cont Hosp Ep 38: 53–60. doi: 10.1017/ice.2016.242
    [29] Kelly CR, Khoruts A, Staley C, et al. (2016) Effect of fecal microbiota transplantation on recurrence in multiply recurrent Clostridium difficile infection: a randomized trial. Ann Intern Med 165: 609–616. doi: 10.7326/M16-0271
    [30] Kozich JJ, Westcott SL, Baxter NT, et al. (2013) Development of a dual-index sequencing strategy and curation pipeline for analyzing amplicon sequence data on the MiSeq Illumina sequencing platform. Appl Environ Microb 79: 5112–5120. doi: 10.1128/AEM.01043-13
    [31] Caporaso JG, Kucznyski J, Stombaugh J, et al. (2010) QIIME allows analysis of high-throughput community sequencing data. Nat Methods 7: 335–336. doi: 10.1038/nmeth.f.303
    [32] Taur Y, Xavier J, Lipuma L, et al. (2012) Intestinal domination and the risk of bacteremia in patients undergoing allogeneic hematopoietic stem cell transplantation. Clin Infect Dis 5: 905–914.
    [33] Moayyedi P, Yuan Y, Baharith H, et al. (2017) Faecal microbiota transplantation for Clostridium difficile-associated diarrhoea: a systematic review of randomised controlled trials. Med J Aust 207: 166–172. doi: 10.5694/mja17.00295
    [34] Burns LJ, Dubois N, Smith MB, et al. (2015) 499 donor recruitment and eligibility for fecal microbiota transplantation: results from an international public stool bank. Gastroenterology 48: 96–97.
    [35] Halpin AL, de Man TJB, Kraft CS, et al. (2016) Intestinal microbiome disruption in patients in a long-term acute care hospital: A case for development of microbiome disruption indices to improve infection prevention. Am J Infect Control 44: 830–836. doi: 10.1016/j.ajic.2016.01.003
    [36] Staley C, Kaiser T, Vaughn BP, et al. (2018) Predicting recurrence of Clostridium difficile infection following encapsulated fecal microbiota transplantation. Microbiome 6: 166. doi: 10.1186/s40168-018-0549-6
    [37] Chang JY, Antonopoulos DA, Kaira A, et al. (2008) Decreased diversity of the fecal microbiome in recurrent Clostridium difficile-associated diarrhea. J Infect Dis 197: 435–438. doi: 10.1086/525047
    [38] Fuentes S, van Nood E, Tims S, et al. (2014) Reset of a critically disturbed microbial ecosystem: faecal transplant in recurrent Clostridium difficile infection. ISME J 8: 1621–1633. doi: 10.1038/ismej.2014.13
    [39] Ubeda C, Taur Y, Jenq R, et al. (2010) Vancomycin-resistant Enterococcus domination of intestinal microbiota is enabled by antibiotic treatment in mice and precedes bloodstream invasion in humans. J Clin Invest 120: 4332–4341. doi: 10.1172/JCI43918
    [40] Lebreton F, Willems RJL, Gilmore MS (2014) Enterococcus Diversity, Origins in Nature, and Gut Colonization, In: Enterococci: From Commensals to Leading Causes of Drug Resistant Infection, 4 Eds., Boston: Massachusetts Eye and Ear Infirmary.
    [41] Patel R, Piper KE, Rouse MS, et al. (1998) Determination of 16S rRNA sequences of Enterococci and application to species identification of nonmotile Enterococcus gallinarum isolates. J Clin Microbiol 36: 3399–3407.
    [42] Huttenhower C, Gevers D, Knight R, et al. (2012) Structure, function and diversity of the healthy human microbiome. Nature 486: 207–214. doi: 10.1038/nature11234
    [43] Kotlowski R, Bernstein CN, Sepehri S, et al. (2007) High prevalence of Escherichia coli belonging to the B2+D phylogenetic group in inflammatory bowel disease. Gut 56: 669–675. doi: 10.1136/gut.2006.099796
    [44] Buffie CG, Jarchum I, Equinda M, et al. (2012) Profound alterations of intestinal microbiota following a single dose of clindamycin results in sustained susceptibility to Clostridium difficile-induced colitis. Infect Immun 80: 62–73. doi: 10.1128/IAI.05496-11
    [45] Desai MS, Seekatz AM, Koropatkin NM, et al. (2016) A dietary fiber-deprived gut microbiota degrades the colonic mucus barrier and enhances pathogen susceptibility. Cell 167: 1339–1353. doi: 10.1016/j.cell.2016.10.043
    [46] Wu W, Sun M, Chen F, et al. (2016) Microbiota metabolite short-chain fatty acid acetate promotes intestinal IgA response to microbiota which is mediated by GPR43. Mucosal Immunol 10: 946–956.
    [47] Goverse G, Molenaar R, Maci L, et al. (2017) Diet-derived short chain fatty acids stimulate intestinal epithelial cells to induce mucosal tolerogenic dendritic cells. J Immunol 198: 2172–2181. doi: 10.4049/jimmunol.1600165
    [48] Faber F, Bäumler AJ (2014) The impact of intestinal inflammation on the nutritional environment of the gut microbiota. Immunol Lett 162: 48–53. doi: 10.1016/j.imlet.2014.04.014
    [49] Lopez CA, Winter SE, Rivera-Chavez F, et al. (2012) Phage-mediated acquisition of a type III secreted effector protein boosts growth of Salmonella by nitrate respiration. Mbio 3: e00143-12.
    [50] Winter SE, Winter MG, Xavier MN, et al. (2013) Host-derived nitrate boosts growth of E. coli in the inflamed gut. Science 339: 708–711.
    [51] Thiennimitr P, Winter SE, Winter MG, et al. (2011) Intestinal inflammation allows Salmonella to use ethanolamine to compete with the microbiota. P Natl Acad Sci USA 108: 17480–17485. doi: 10.1073/pnas.1107857108
    [52] Fujitani S, George WL, Morgan MA, et al. (2011) Implications for vancomycin-resistant Enterococcus colonization associated with Clostridium difficile infections. Am J Infect Control 39: 188–193. doi: 10.1016/j.ajic.2010.10.024
    [53] Sangster W, Hegarty JP, Schieffer KM, et al. (2016) Bacterial and fungal microbiota changes distinguish C. difficile infection from other forms of diarrhea: results of a prospective inpatient study. Front Microbiol 7: 789.
    [54] Horvat S, Mahnic A, Breskvar M, et al. (2017) Evaluating the effect of Clostridium difficile conditioned medium on fecal microbiota community structure. Sci Rep 7: 16448. doi: 10.1038/s41598-017-15434-1
    [55] Papa E, Docktor M, Smillie C, et al. (2012) Non-invasive mapping of the gastrointestinal microbiota identifies children with inflammatory bowel disease. PLoS One 7: e39242. doi: 10.1371/journal.pone.0039242
  • Reader Comments
  • © 2019 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(4511) PDF downloads(884) Cited by(9)

Article outline

Figures and Tables

Figures(5)  /  Tables(2)

/

DownLoad:  Full-Size Img  PowerPoint
Return
Return

Catalog