Microbiota restoration reduces antibiotic-resistant bacteria gut colonization in patients with recurrent Clostridioides difficile infection from the open-label PUNCH CD study

Amy Langdon, Drew J Schwartz, Christopher Bulow, Xiaoqing Sun, Tiffany Hink, Kimberly A Reske, Courtney Jones, Carey-Ann D Burnham, Erik R Dubberke, Gautam Dantas, CDC Prevention Epicenter Program, Amy Langdon, Drew J Schwartz, Christopher Bulow, Xiaoqing Sun, Tiffany Hink, Kimberly A Reske, Courtney Jones, Carey-Ann D Burnham, Erik R Dubberke, Gautam Dantas, CDC Prevention Epicenter Program

Abstract

Background: Once antibiotic-resistant bacteria become established within the gut microbiota, they can cause infections in the host and be transmitted to other people and the environment. Currently, there are no effective modalities for decreasing or preventing colonization by antibiotic-resistant bacteria. Intestinal microbiota restoration can prevent Clostridioides difficile infection (CDI) recurrences. Another potential application of microbiota restoration is suppression of non-C. difficile multidrug-resistant bacteria and overall decrease in the abundance of antibiotic resistance genes (the resistome) within the gut microbiota. This study characterizes the effects of RBX2660, a microbiota-based investigational therapeutic, on the composition and abundance of the gut microbiota and resistome, as well as multidrug-resistant organism carriage, after delivery to patients suffering from recurrent CDI.

Methods: An open-label, multi-center clinical trial in 11 centers in the USA for the safety and efficacy of RBX2660 on recurrent CDI was conducted. Fecal specimens from 29 of these subjects with recurrent CDI who received either one (N = 16) or two doses of RBX2660 (N = 13) were analyzed secondarily. Stool samples were collected prior to and at intervals up to 6 months post-therapy and analyzed in three ways: (1) 16S rRNA gene sequencing for microbiota taxonomic composition, (2) whole metagenome shotgun sequencing for functional pathways and antibiotic resistome content, and (3) selective and differential bacterial culturing followed by isolate genome sequencing to longitudinally track multidrug-resistant organisms.

Results: Successful prevention of CDI recurrence with RBX2660 correlated with taxonomic convergence of patient microbiota to the donor microbiota as measured by weighted UniFrac distance. RBX2660 dramatically reduced the abundance of antibiotic-resistant Enterobacteriaceae in the 2 months after administration. Fecal antibiotic resistance gene carriage decreased in direct relationship to the degree to which donor microbiota engrafted.

Conclusions: Microbiota-based therapeutics reduce resistance gene abundance and resistant organisms in the recipient gut microbiome. This approach could potentially reduce the risk of infections caused by resistant organisms within the patient and the transfer of resistance genes or pathogens to others.

Trial registration: ClinicalTrials.gov, NCT01925417 ; registered on August 19, 2013.

Keywords: Antibiotic resistance; Clostridioides difficile; Fecal microbiota transplantation; Metagenomics; Microbiome; Multidrug resistance.

Conflict of interest statement

Rebiotix, Inc. provided access to study specimens and data and reviewed the manuscript prior to submission, but was not involved in this study’s design, specimen processing, data analysis, or interpretation. Washington University investigators had the final say on published content. Erik R. Dubberke is a consultant for Sanofi, Pfizer, Synthetic Biologics, BioK+, and Rebiotix with grants from Pfizer. The remaining authors declare no other competing interests.

Figures

Fig. 1
Fig. 1
Sampling schematic. Patients were given RBX2660 (green square) after vancomycin oral therapy (left panel). Stools (labeled as maroon circles) provided were sequenced and used for subsequent analyses. If a patient had CDI recurrence (red triangle), they were offered a second dose of RBX2660 (green square) with subsequent stools provided after the second study drug (right panel). Any antibiotic treatment during the trial is labeled as yellow diamonds. Patient IDs colored red failed first treatment and received antibiotics or second dose and constitute the RI group (n = 17). Patients who had no recurrence of symptoms or received antibiotics were considered successes (SI group, n = 12). All subsequent figures utilize data after the first dose. Data after second RBX2660 is used only for Figs. 5a–e and 6d. Three stool samples that failed sequencing were excluded from this figure and downstream analyses
Fig. 2
Fig. 2
Microbiota composition similarity to the donor at 7 days is predictive of treatment outcome. The donor and recipient microbiota compositions were assessed via 16S rRNA gene sequencing followed by DADA2, and their similarity to the donor product was quantified by weighted UniFrac at each timepoint. a Gray lines represent individuals successfully treated with one administration (SI group) while red lines are patients who needed further treatment (RI group, a second product or antibiotics). N = 28 patients and 130 samples. b Plot demonstrating average distance from the donor at timepoints 0 and 7 days after treatment. N = 28 total patients and 44 samples. Box subsumes 75% of the data with a horizontal bar at the median. *p < 0.05, Mann-Whitney U test
Fig. 3
Fig. 3
Taxonomy and microbial functional pathways converge after therapy receipt. a, b Principal component analysis (PCA) of patient microbiome taxonomic composition from 16S data (a) and of functional pathway abundances from whole metagenomic sequencing (b) in the SI group. Each colored dot represents an individual fecal sample after the first intervention with the circle representing 95% confidence interval with non-intersecting circles therefore statistically significant. Panel a shows 96 samples from all twelve patients with successful treatment and all four donors, while panel b shows 52 samples from eight successful patients and four donors (all of those who passed shotgun sequencing quality filters). c PCA from timepoint 7 samples after first study treatment only, colored by the SI or RI group (46 samples from all donors and all patients with day 7 samples; patient N = 25; donor N = 4). Each sample is connected to the centroid of its outcome group by a segment of the same color. d Taxonomy biplot shows the vectors of influence from taxa in distinguishing day 7 samples. The input samples, axes, and origin are the same as in c
Fig. 4
Fig. 4
Taxa significantly associated with distance from the donor and successful response to RBX2660. A heatmap demonstrating the relative abundance over time after first RBX2660 is shown for donors and the SI group. These taxa are the top 11 identified by the PCA in Fig. 3d as significantly associated with successful treatment. Dark blue corresponds to 0.001% relative abundance with lighter blue 0.1% relative abundance. Each column represents a sample from a patient over time from left to right with donor samples at the right. N = 109 samples from 12 subjects from the SI group and 4 donors
Fig. 5
Fig. 5
Antibiotic-resistant organisms cultured from patient and donor stools and the corresponding ASVs from species were tracked over time. ae Antibiotic susceptibility profiles for each cultured organism from any sample from donor and patient with the corresponding phylogenetic tree. All breakpoints in antibiotic concentration were determined by CLSI 2016 criteria. Taxonomic labels are derived from DADA2 ASV assignments, with Enterobacter being further specified from family level based on metaphlan2 and MALDI-TOF taxonomy assignments. The designation A indicates recipient and D indicates donor. The following number indicates the study ID number followed by timepoint of isolation. A and E connote single colonies on separate plates. fj Each of the ASVs corresponding to the species in ae are shown in relative abundance over time in 131 fecal samples from 28 patients and 4 donors after the first study treatment. *p < 0.05, **p < 0.01 for relative abundance differences 7 days after therapy between SI and RI groups using Mann-Whitney U test. TMP-SMX, trimethoprim-sulfamethoxazole
Fig. 6
Fig. 6
Antibiotic resistance gene abundance correlates with distance from the donor. a ARGs were quantified in metagenomic sequences (N = 21 patients and 4 donors) and summarized by mechanism. All ARG counts were transformed by log (ARG + 1) for visibility. b Two gene families within the β-lactamase class show opposite trajectories (N = 21 patients and 4 donors). c Patient-origin ARGs shown over time after RBX2660. d ARG abundance is plotted versus 1-(distance from donor) using weighted UniFrac. A generalized mixed effects log normal regression model of the formula ARGs ~ DFD + (1| PatientID) is shown, where DFD was significantly predictive of and correlated with ARG count compared to the null model (Chisq = 72.28, d.f.(full) = 1, p < 2.2 × 10−16). For c and d, all patients of both outcome groups were included for 153 total samples with patient N = 25 and donor N = 4. ac Significance was determined by pairwise Wilcoxon with Benjamini-Hochberg correction. *p < 0.05; **p < 0.001

References

    1. O'Neill J. Tackling drug-resistant infections globally: final report and recommendations. 2016.
    1. Klein EY, Van Boeckel TP, Martinez EM, Pant S, Gandra S, Levin SA, et al. Global increase and geographic convergence in antibiotic consumption between 2000 and 2015. Proc Natl Acad Sci U S A. 2018;115(15):E3463–E3E70.
    1. Pehrsson EC, Tsukayama P, Patel S, Mejía-Bautista M, Sosa-Soto G, Navarrete KM, et al. Interconnected microbiomes and resistomes in low-income human habitats. Nature. 2016;533(7602):212.
    1. Costelloe C, Metcalfe C, Lovering A, Mant D, Hay AD. Effect of antibiotic prescribing in primary care on antimicrobial resistance in individual patients: systematic review and meta-analysis. BMJ. 2010;340:c2096.
    1. Wang J, Foxman B, Mody L, Snitkin ES. Network of microbial and antibiotic interactions drive colonization and infection with multidrug-resistant organisms. Proc Natl Acad Sci U S A. 2017;114(39):10467–10472.
    1. Langdon A, Crook N, Dantas G. The effects of antibiotics on the microbiome throughout development and alternative approaches for therapeutic modulation. Genome Med. 2016;8(1):39.
    1. Chatterjee A, Modarai M, Naylor NR, Boyd SE, Atun R, Barlow J, et al. Quantifying drivers of antibiotic resistance in humans: a systematic review. Lancet Infect Dis. 2018;18(12):e368–ee78.
    1. Buffie CG, Pamer EG. Microbiota-mediated colonization resistance against intestinal pathogens. Nat Rev Immunol. 2013;13(11):790–801.
    1. Chilton CH, Pickering DS, Freeman J. Microbiologic factors affecting Clostridium difficile recurrence. Clin Microbiol Infect. 2018;24(5):476–482.
    1. Lessa FC, Winston LG, McDonald LC, Emerging Infections Program CST. Burden of Clostridium difficile infection in the United States. N Engl J Med. 2015;372(24):2369–2370.
    1. Shah D, Dang MD, Hasbun R, Koo HL, Jiang ZD, DuPont HL, et al. Clostridium difficile infection: update on emerging antibiotic treatment options and antibiotic resistance. Expert Rev Anti-Infect Ther. 2010;8(5):555–564.
    1. Weingarden A, Gonzalez A, Vazquez-Baeza Y, Weiss S, Humphry G, Berg-Lyons D, et al. Dynamic changes in short- and long-term bacterial composition following fecal microbiota transplantation for recurrent Clostridium difficile infection. Microbiome. 2015;3:10.
    1. Youngster I, Sauk J, Pindar C, Wilson RG, Kaplan JL, Smith MB, et al. Fecal microbiota transplant for relapsing Clostridium difficile infection using a frozen inoculum from unrelated donors: a randomized, open-label, controlled pilot study. Clin Infect Dis. 2014;58(11):1515–1522.
    1. Al-Nassir WN, Sethi AK, Li Y, Pultz MJ, Riggs MM, Donskey CJ. Both oral metronidazole and oral vancomycin promote persistent overgrowth of vancomycin-resistant enterococci during treatment of Clostridium difficile-associated disease. Antimicrob Agents Chemother. 2008;52(7):2403–2406.
    1. Roghmann MC, McCarter RJ, Jr, Brewrink J, Cross AS, Morris JG., Jr Clostridium difficile infection is a risk factor for bacteremia due to vancomycin-resistant enterococci (VRE) in VRE-colonized patients with acute leukemia. Clin Infect Dis. 1997;25(5):1056–1059.
    1. Prematunge C, MacDougall C, Johnstone J, Adomako K, Lam F, Robertson J, et al. VRE and VSE bacteremia outcomes in the era of effective VRE therapy: a systematic review and meta-analysis. Infect Control Hosp Epidemiol. 2016;37(1):26–35.
    1. Cammarota G, Ianiro G, Gasbarrini A. Fecal microbiota transplantation for the treatment of Clostridium difficile infection: a systematic review. J Clin Gastroenterol. 2014;48(8):693–702.
    1. Iqbal U, Anwar H, Karim MA. Safety and efficacy of encapsulated fecal microbiota transplantation for recurrent Clostridium difficile infection: a systematic review. Eur J Gastroenterol Hepatol. 2018;30(7):730–734.
    1. Dubberke ER, Lee CH, Orenstein R, Khanna S, Hecht G, Gerding DN. Results from a randomized, placebo-controlled clinical trial of a RBX2660-a microbiota-based drug for the prevention of recurrent Clostridium difficile infection. Clin Infect Dis. 2018;67(8):1198–1204.
    1. Khanna S, Pardi DS, Kelly CR, Kraft CS, Dhere T, Henn MR, et al. A novel microbiome therapeutic increases gut microbial diversity and prevents recurrent Clostridium difficile infection. J Infect Dis. 2016;214(2):173–181.
    1. Suez J, Zmora N, Zilberman-Schapira G, Mor U, Dori-Bachash M, Bashiardes S, et al. Post-antibiotic gut mucosal microbiome reconstitution is impaired by probiotics and improved by autologous FMT. Cell. 2018;174(6):1406–1423.
    1. Smillie CS, Sauk J, Gevers D, Friedman J, Sung J, Youngster I, et al. Strain tracking reveals the determinants of bacterial engraftment in the human gut following fecal microbiota transplantation. Cell Host Microbe. 2018;23(2):229–40.e5–40.e5.
    1. Millan B, Park H, Hotte N, Mathieu O, Burguiere P, Tompkins TA, et al. Fecal microbial transplants reduce antibiotic-resistant genes in patients with recurrent Clostridium difficile infection. Clin Infect Dis. 2016;62(12):1479–1486.
    1. Singh R, de Groot PF, Geerlings SE, Hodiamont CJ, Belzer C, Berge I, et al. Fecal microbiota transplantation against intestinal colonization by extended spectrum beta-lactamase producing Enterobacteriaceae: a proof of principle study. BMC Res Notes. 2018;11(1):190.
    1. Orenstein R, Dubberke E, Hardi R, Ray A, Mullane K, Pardi DS, et al. Safety and durability of RBX2660 (microbiota suspension) for recurrent Clostridium difficile infection: results of the PUNCH CD study. Clin Infect Dis. 2016;62(5):596–602.
    1. Ray A, Jones C. Does the donor matter? Donor vs patient effects in the outcome of a next-generation microbiota-based drug trial for recurrent Clostridium difficile infection. Future Microbiol. 2016;11:611–616.
    1. Langdon A, Schwartz DJ, Bulow C, Sun X, Hink T, Reske KA, et al. Dataset for microbiota restoration reduces antibiotic resistant bacteria gut colonization in patients with recurrent Clostridioides difficile infection from the open-label PUNCH CD study. NCBI Biorepository 2020.
    1. CLSI, editor. Performance standards for antimicrobial susceptibility testing. 26th ed. ed: Clinical and Laboratory Standards Institute; 2016.
    1. Gurevich A, Saveliev V, Vyahhi N, Tesler G. QUAST: quality assessment tool for genome assemblies. Bioinformatics. 2013;29(8):1072–1075.
    1. Zankari E, Hasman H, Cosentino S, Vestergaard M, Rasmussen S, Lund O, et al. Identification of acquired antimicrobial resistance genes. J Antimicrob Chemother. 2012;67(11):2640.
    1. Seemann T. Prokka: rapid prokaryotic genome annotation. Bioinformatics. 2014;30(14):2068–2069.
    1. Page AJ, Cummins CA, Hunt M, Wong VK, Reuter S, Holden MT, et al. Roary: rapid large-scale prokaryote pan genome analysis. Bioinformatics. 2015;31(22):3691–3693.
    1. Stamatakis A. RAxML version 8: a tool for phylogenetic analysis and post-analysis of large phylogenies. Bioinformatics. 2014;30(9):1312–1313.
    1. Seah BKB, Schwaha T, Volland JM, Huettel B, Dubilier N, Gruber-Vodicka HR. Specificity in diversity: single origin of a widespread ciliate-bacteria symbiosis. Proc Biol Sci. 2017;284(1858):1–9.
    1. Langdon A, Schwartz DJ, Bulow C, Sun X, Hink T, Reske KA, et al. Dataset for bacterial isolate assemblies for microbiota restoration reduces antibiotic resistant bacteria gut colonization in patients with recurrent Clostridioides difficile infection from the open-label PUNCH CD study. NCBI Biorepository 2020.
    1. Schloss PD, Westcott SL, Ryabin T, Hall JR, Hartmann M, Hollister EB, et al. Introducing mothur: open-source, platform-independent, community-supported software for describing and comparing microbial communities. Appl Environ Microbiol. 2009;75(23):7537–7541.
    1. Kaminski J, Gibson MK, Franzosa EA, Segata N, Dantas G, Huttenhower C. High-specificity targeted functional profiling in microbial communities with ShortBRED. PLoS Comput Biol. 2015;11(12):e1004557.
    1. Callahan BJ, McMurdie PJ, Rosen MJ, Han AW, Johnson AJA, Holmes SP. DADA2: high-resolution sample inference from Illumina amplicon data. Nature Methods. 2016;13(7):nmeth.3869.
    1. Wright ES. DECIPHER: harnessing local sequence context to improve protein multiple sequence alignment. BMC Bioinformatics. 2015;16:322.
    1. McMurdie PJ, Holmes S. phyloseq: an R package for reproducible interactive analysis and graphics of microbiome census data. Plos One. 2013;8(4):e61217.
    1. Bolger AM, Lohse M, Usadel B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics. 2014;30(15):2114–2120.
    1. Schmieder R, Edwards R. Fast identification and removal of sequence contamination from genomic and metagenomic datasets. Plos One. 2011;6(3):e17288.
    1. Truong DT, Franzosa EA, Tickle TL, Scholz M, Weingart G, Pasolli E, et al. MetaPhlAn2 for enhanced metagenomic taxonomic profiling. Nat Methods. 2015;12(10):902.
    1. Franzosa EA, McIver LJ, Rahnavard G, Thompson LR, Schirmer M, Weingart G, et al. Species-level functional profiling of metagenomes and metatranscriptomes. Nat Methods. 2018;15(11):962.
    1. Le Roy T, Debedat J, Marquet F, Da-Cunha C, Ichou F, Guerre-Millo M, et al. Comparative evaluation of microbiota engraftment following fecal microbiota transfer in mice models: age, kinetic and microbial status matter. Front Microbiol. 2018;9:3289.
    1. Blount KF, Shannon WD, Deych E, Jones C. Restoration of bacterial microbiome composition and diversity among treatment responders in a phase 2 trial of Rbx2660—an investigational microbiome restoration therapeutic. Open Forum Infect Dis. 2019:6(4)ofz095:1–10.
    1. Consortium THMP, Huttenhower C, Gevers D, Knight R, Abubucker S, Badger JH, et al. Structure, function and diversity of the healthy human microbiome. Nature. 2012;486(7402):207.
    1. Citron DM, Tyrrell KL, Dale SE, Chesnel L, Goldstein EJ. Impact of surotomycin on the gut microbiota of healthy volunteers in a phase 1 clinical trial. Antimicrob Agents Chemother. 2016;60(4):2069–2074.
    1. Reuland EA, Sonder GJ, Stolte I, Al Naiemi N, Koek A, Linde GB, et al. Travel to Asia and traveller’s diarrhoea with antibiotic treatment are independent risk factors for acquiring ciprofloxacin-resistant and extended spectrum beta-lactamase-producing Enterobacteriaceae-a prospective cohort study. Clin Microbiol Infect. 2016;22(8):731 e1–731 e7.
    1. Segata N, Izard J, Waldron L, Gevers D, Miropolsky L, Garrett WS, et al. Metagenomic biomarker discovery and explanation. Genome Biol. 2011;12(6):R60.
    1. Callahan BJ, McMurdie PJ, Holmes SP. Exact sequence variants should replace operational taxonomic units in marker-gene data analysis. ISME J. 2017;11(12):2639–2643.
    1. Mukherjee C, Beall CJ, Griffen AL, Leys EJ. High-resolution ISR amplicon sequencing reveals personalized oral microbiome. Microbiome. 2018;6(1):153.
    1. DeFilipp Z, Bloom PP, Torres Soto M, Mansour MK, Sater MRA, Huntley MH, et al. Drug-resistant E. coli bacteremia transmitted by fecal microbiota transplant. N Engl J Med. 2019;381(21):2043–2050.
    1. Babady NE. Hospital-associated infections. Microbiol Spectr. 2016;4(3):1–22.
    1. Naiemi NA, Duim B, Savelkoul PH, Spanjaard L, de Jonge E, Bart A, et al. Widespread transfer of resistance genes between bacterial species in an intensive care unit: implications for hospital epidemiology. J Clin Microbiol. 2005;43(9):4862–4864.
    1. Forsberg KJ, Reyes A, Wang B, Selleck EM, Sommer MO, Dantas G. The shared antibiotic resistome of soil bacteria and human pathogens. Science. 2012;337(6098):1107–1111.
    1. Andersen H, Connolly N, Bangar H, Staat M, Mortensen J, Deburger B, et al. Use of shotgun metagenome sequencing to detect fecal colonization with multidrug-resistant bacteria in children. J Clin Microbiol. 2016;54(7):1804–1813.
    1. Administration FaD. Information pertaining to additional safety protections regarding use of fecal microbiota for transplantation – screening and testing of stool donors for multi-drug resistant organisms FDA.gov2019 [updated 06/18/2019.
    1. Palleja A, Mikkelsen KH, Forslund SK, Kashani A, Allin KH, Nielsen T, et al. Recovery of gut microbiota of healthy adults following antibiotic exposure. Nat Microbiol. 2018;3(11):1255.
    1. Buelow E, Bello Gonzalez TDJ, Fuentes S, de Steenhuijsen Piters WAA, Lahti L, Bayjanov JR, et al. Comparative gut microbiota and resistome profiling of intensive care patients receiving selective digestive tract decontamination and healthy subjects. Microbiome. 2017;5(1):88.
    1. Feng J, Li B, Jiang X, Yang Y, Wells GF, Zhang T, et al. Antibiotic resistome in a large-scale healthy human gut microbiota deciphered by metagenomic and network analyses. Environ Microbiol. 2018;20(1):355–368.
    1. Tariq R, Pardi DS, Bartlett MG, Khanna S. Low cure rates in controlled trials of fecal microbiota transplantation for recurrent Clostridium difficile infection: a systematic review and meta-analysis. Clin Infect Dis. 2019;68(8):1351–1358.
    1. Khan MY, Dirweesh A, Khurshid T, Siddiqui WJ. Comparing fecal microbiota transplantation to standard-of-care treatment for recurrent Clostridium difficile infection: a systematic review and meta-analysis. Eur J Gastroenterol Hepatol. 2018;30(11):1309–1317.
    1. Kwak S, Choi J, Hink T, Reske KA, Blount K, Jones C, et al. Impact of investigational microbiota therapeutic RBX2660 on the gut microbiome and resistome revealed by a placebo-controlled clinical trial. Microbiome. 2020;8(1):125.
    1. Mullish BH, McDonald JAK, Pechlivanis A, Allegretti JR, Kao D, Barker GF, et al. Microbial bile salt hydrolases mediate the efficacy of faecal microbiota transplant in the treatment of recurrent Clostridioides difficile infection. Gut. 2019;68(10):1791–800.
    1. Png CW, Linden SK, Gilshenan KS, Zoetendal EG, McSweeney CS, Sly LI, et al. Mucolytic bacteria with increased prevalence in IBD mucosa augment in vitro utilization of mucin by other bacteria. Am J Gastroenterol. 2010;105(11):2420–2428.
    1. Dao MC, Everard A, Aron-Wisnewsky J, Sokolovska N, Prifti E, Verger EO, et al. Akkermansia muciniphila and improved metabolic health during a dietary intervention in obesity: relationship with gut microbiome richness and ecology. Gut. 2016;65(3):426–436.
    1. Derrien M, Belzer C, de Vos WM. Akkermansia muciniphila and its role in regulating host functions. Microb Pathog. 2017;106:171–181.
    1. Ford C HM, Bryant J, Diao L, Wortman J, Tomlinson A, Litcofsky K, Bernardo P, McGovern B, Aunins JG, Cook DN, Trucksis M. 1641. Treatment of recurrent Clostridium difficile infection with SER-109 reduces gastrointestinal carriage of antimicrobial resistance genes. Open Forum Infect Dis. 2018;5:S44-S47.
    1. Browne HP, Forster SC, Anonye BO, Kumar N, Neville BA, Stares MD, et al. Culturing of ‘unculturable’ human microbiota reveals novel taxa and extensive sporulation. Nature. 2016;533(7604):543–546.
    1. Lawley TD, Clare S, Walker AW, Goulding D, Stabler RA, Croucher N, et al. Antibiotic treatment of clostridium difficile carrier mice triggers a supershedder state, spore-mediated transmission, and severe disease in immunocompromised hosts. Infect Immun. 2009;77(9):3661–3669.
    1. Saha S, Tariq R, Tosh PK, Pardi DS, Khanna S. Faecal microbiota transplantation for eradicating carriage of multidrug-resistant organisms: a systematic review. Clin Microbiol Infect. 2019;25(8):958–963.
    1. McKain N, Genc B, Snelling TJ, Wallace RJ. Differential recovery of bacterial and archaeal 16S rRNA genes from ruminal digesta in response to glycerol as cryoprotectant. J Microbiol Methods. 2013;95(3):381–383.
    1. Ma GK, Brensinger CM, Wu Q, Lewis JD. Increasing incidence of multiply recurrent Clostridium difficile infection in the United States: a cohort study. Ann Intern Med. 2017;167(3):152–158.
    1. Lloyd-Price J, Arze C, Ananthakrishnan AN, Schirmer M, Avila-Pacheco J, Poon TW, et al. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature. 2019;569(7758):655–662.
    1. Lim ES, Zhou Y, Zhao G, Bauer IK, Droit L, Ndao IM, et al. Early life dynamics of the human gut virome and bacterial microbiome in infants. Nat Med. 2015;21(10):1228–1234.

Source: PubMed

3
Se inscrever