Short- and long-term effects of oral vancomycin on the human intestinal microbiota

Sandrine Isaac, Jose U Scher, Ana Djukovic, Nuria Jiménez, Dan R Littman, Steven B Abramson, Eric G Pamer, Carles Ubeda, Sandrine Isaac, Jose U Scher, Ana Djukovic, Nuria Jiménez, Dan R Littman, Steven B Abramson, Eric G Pamer, Carles Ubeda

Abstract

Background: Oral vancomycin remains the mainstay of therapy for severe infections produced by Clostridium difficile, the most prevalent cause of healthcare-associated infectious diarrhoea in developed countries. However, its short- and long-term effects on the human intestinal microbiota remain largely unknown.

Methods: We utilized high-throughput sequencing to analyse the effects of vancomycin on the faecal human microbiota up to 22 weeks post-antibiotic cessation. The clinical relevance of the observed microbiota perturbations was studied in mice.

Results: During vancomycin therapy, most intestinal microbiota genera and operational taxonomic units (OTUs) were depleted in all analysed subjects, including all baseline OTUs from the phylum Bacteroidetes. This was accompanied by a vast expansion of genera associated with infections, including Klebsiella and Escherichia/Shigella. Following antibiotic cessation, marked differences in microbiota resilience were observed among subjects. While some individuals recovered a microbiota close to baseline composition, in others, up to 89% of abundant OTUs could no longer be detected. The clinical relevance of the observed microbiota changes was further demonstrated in mice, which developed analogous microbiota alterations. During vancomycin treatment, mice were highly susceptible to intestinal colonization by an antibiotic-resistant pathogen and, upon antibiotic cessation, a less-resilient microbiota allowed higher levels of pathogen colonization.

Conclusions: Oral vancomycin induces drastic and consistent changes in the human intestinal microbiota. Upon vancomycin cessation, the microbiota recovery rate varied considerably among subjects, which could influence, as validated in mice, the level of susceptibility to pathogen intestinal colonization. Our results demonstrate the negative long-term effects of vancomycin, which should be considered as a fundamental aspect of the cost-benefit equation for antibiotic prescription.

© The Author 2016. Published by Oxford University Press on behalf of the British Society for Antimicrobial Chemotherapy.

Figures

Figure 1.
Figure 1.
Vancomycin induces persistent changes in the structure and richness of the human microbiota. Patients received vancomycin treatment for 2 weeks (T, vancomycin treated) or did not receive vancomycin (C, control group). Faecal samples were obtained immediately before treatment (Baseline), the day of antibiotic cessation (Vanco), and 2, 6, 14 and 22 weeks (w) after antibiotic cessation (Recovery). As control, faecal samples from patients that did not receive vancomycin were obtained at similar timepoints. (a) Hierarchical clustering based on microbiota similarity (see the supplementary methods) among the faecal samples analysed from vancomycin-treated and control patients. Samples with a more similar microbiota are clustered within the same branch of the tree. Colours indicate the time frame where the faecal sample was obtained. Numbers indicate the patient ID. Those samples obtained at the last timepoint (22 weeks post-antibiotic withdrawal) are labelled with asterisks. (b) Number of OTUs (a close estimate of bacterial species) and (c) Shannon index of microbial diversity calculated from the microbiota of faecal samples collected at baseline and at different timepoints after vancomycin treatment. For comparison, the same indices were calculated from the faecal samples collected at similar timepoints from patients who did not receive vancomycin (control). Bar graphs represent mean ± SEM. *P < 0.05, **P < 0.01, ***P < 0.001, two-tailed t-test. ns, not significant. n = 5–12 per group and timepoint except for the second timepoint from control patients where n = 3. This figure appears in colour in the online version of JAC and in black and white in the print version of JAC.
Figure 2.
Figure 2.
The abundance of the majority of taxa and OTUs is altered during vancomycin treatment. (a) Relative abundance of different phyla in patients treated with vancomycin at baseline or 2 weeks after treatment initiation (Vanco). Only phyla that are present in at least 50% of the patients at baseline or after vancomycin treatment are shown. **P < 0.01, *P < 0.05, FDR < 0.2, two-tailed Wilcoxon test. ns, not significant. (b) Log2 average fold change (FC) between the genera abundance from samples obtained immediately after vancomycin treatment compared with their respective baseline samples. Only genera that are present in at least 50% of the patients at baseline or after vancomycin treatment are shown. Genera are sorted by phyla, FC difference and then alphabetically. UC, unclassified; vanco, vancomycin. (c) Heatmap representing the relative abundance (%) of OTUs present in at least 50% of the patients at baseline or after vancomycin treatment, showing a depletion of all prevalent Bacteroidetes OTUs and most Firmicutes OTUs. Additional analysis including all detected OTUs confirmed the depletion of all baseline OTUs from the Bacteroidetes phylum (not shown). For both (b) and (c): black squares indicate significant changes (P < 0.05, FDR < 0.1); grey squares indicate close to significance changes (P < 0.073, FDR < 0.1); two-tailed Wilcoxon test. n = 5. This figure appears in colour in the online version of JAC and in black and white in the print version of JAC.
Figure 3.
Figure 3.
Changes in the human microbiota following vancomycin withdrawal. Heatmap representing the average fold change of (a) phyla, (b) genera and (c) OTUs that were significantly increased (red) or decreased (green) (P < 0.05, FDR < 0.2, two-tailed Wilcoxon test) at a given timepoint as compared with the baseline. Time 0 represents the sample obtained the day of vancomycin withdrawal. n = 6–9 per timepoint. UC, unclassified; Enterobact., Enterobacteriaceae. This figure appears in colour in the online version of JAC and in black and white in the print version of JAC.
Figure 4.
Figure 4.
Incomplete and individualized recovery of the human microbiota after vancomycin withdrawal. Relative abundance at baseline (Bs) and at different weeks post-antibiotic cessation of OTUs whose abundance was ≥10 counts in the baseline sample of the analysed patient. Note that each patient baseline OTUs are distinct. The ID of each patient is indicated. As control, samples at similar timepoints were analysed from patients who did not receive vancomycin. Four representative patients from each group are shown (the four control patients from whom we were able to collect and analyse faecal samples at every timepoint and four out of the five vancomycin-treated patients for whom we were able to collect and analyse faecal samples at every timepoint). The rest of the patients are shown in Figure S5. This figure appears in colour in the online version of JAC and in black and white in the print version of JAC.
Figure 5.
Figure 5.
Microbiota recovery rate after vancomycin cessation influences the level of susceptibility to VRE intestinal colonization in mice. (a) Heatmap showing the most prevalent OTUs (≥10 counts per group of mice) found in the faeces of mice treated with vancomycin for 7 days (Vanco), 2 weeks after antibiotic cessation (Recov) or in mice that remained untreated (Untreat.). n = 5 per group. (b) Number of OTUs identified in faecal samples from untreated mice or humans, or mice/humans that were treated with oral vancomycin, the day the treatment was stopped (Vanco) or 2 weeks post-antibiotic cessation (Recov). Boxes extend from the 25th percentile to the 75th percentile. The line in the middle of the box represents the median. Whiskers extend from the minimum value to the maximum value. n = 5–12 per group. (c) VRE cfu/10 mg of faeces 2 days post-VRE inoculation in untreated mice (UT), mice that received vancomycin for 1 week and were inoculated with VRE immediately after vancomycin therapy (Vanco) or were inoculated 2 weeks post-antibiotic cessation (Recov). n = 5 for the UT and Vanco groups and n = 25 for the Recov group. (d) Correlation analysis between the y-axis variable (log2 VRE cfu/10 mg of faeces detected the second day after VRE inoculation in mice that recover for 2 weeks after vancomycin administration) and the x-axis variable, which is either (i) the number of OTUs identified in mice, the day of VRE inoculation, divided by the average number of OTUs identified in untreated mice, or (ii) the microbiota similarity between VRE-colonized mice, the day of VRE inoculation and untreated mice (based on UniFrac distance, see the supplementary methods). n = 25. This figure appears in colour in the online version of JAC and in black and white in the print version of JAC.

References

    1. Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F et al. . Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell 2004; 118: 229–41.
    1. Buffie CG, Pamer EG. Microbiota-mediated colonization resistance against intestinal pathogens. Nat Rev Immunol 2013; 13: 790–801.
    1. Ivanov II, Atarashi K, Manel N et al. . Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell 2009; 139: 485–98.
    1. Heijtz RD, Wang S, Anuar F et al. . Normal gut microbiota modulates brain development and behavior. Proc Natl Acad Sci USA 2011; 108: 3047–52.
    1. Ubeda C, Pamer EG. Antibiotics, microbiota, and immune defense. Trends Immunol 2012; 33: 459–66.
    1. Ubeda C, Taur Y, Jenq RR et al. . Vancomycin-resistant Enterococcus domination of intestinal microbiota is enabled by antibiotic treatment in mice and precedes bloodstream invasion in humans. J Clin Invest 2010; 120: 4332–41.
    1. Million M, Thuny F, Angelakis E et al. . Lactobacillus reuteri and Escherichia coli in the human gut microbiota may predict weight gain associated with vancomycin treatment. Nutr Diabetes 2013; 3: e87.
    1. Zar FA, Bakkanagari SR, Moorthi KMLST et al. . Comparison of vancomycin and metronidazole for the treatment of Clostridium difficile-associated diarrhea, stratified by disease severity. Clin Infect Dis 2007; 45: 302–7.
    1. Johnson S, Gerding DN, Louie TJ et al. . Sustained clinical response as an endpoint in treatment trials of Clostridium difficile-associated diarrhea. Antimicrob Agents Chemother 2012; 56: 4043–5.
    1. Lewis B, Buffie CG, Carter RA et al. . Loss of microbiota-mediated colonization resistance to Clostridium difficile infection is greater following oral vancomycin as compared with metronidazole. J Infect Dis 2015; 212: 1656–65.
    1. Russell SL, Gold MJ, Hartmann M et al. . Early life antibiotic-driven changes in microbiota enhance susceptibility to allergic asthma. EMBO Rep 2012; 13: 440–7.
    1. Candon S, Perez-Arroyo A, Marquet C et al. . Antibiotics in early life alter the gut microbiome and increase disease incidence in a spontaneous mouse model of autoimmune insulin-dependent diabetes. PLoS ONE 2015; 10: e0125448.
    1. Tannock GW, Munro K, Taylor C et al. . A new macrocyclic antibiotic, fidaxomicin (OPT-80), causes less alteration to the bowel microbiota of Clostridium difficile-infected patients than does vancomycin. Microbiology 2010; 156: 3354–9.
    1. Louie TJ, Cannon K, Byrne B et al. . Fidaxomicin preserves the intestinal microbiome during and after treatment of Clostridium difficile infection (CDI) and reduces both toxin reexpression and recurrence of CDI. Clin Infect Dis 2012; 55 Suppl 2: S132–42.
    1. Louie TJ, Byrne B, Emery J et al. . Differences of the fecal microflora with Clostridium difficile therapies. Clin Infect Dis 2015; 60 Suppl 2: S91–7.
    1. Abujamel T, Cadnum JL, Jury LA et al. . Defining the vulnerable period for re-establishment of Clostridium difficile colonization after treatment of C. difficile infection with oral vancomycin or metronidazole. PLoS ONE 2013; 8: e76269.
    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. Magurran AE. Measuring Biological Diversity. African J Aquatic Sci 2004; 29: 285–6.
    1. Benjamini Y, Hockberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc 1995; 57: 289–300.
    1. Vrieze A, Out C, Fuentes S et al. . Impact of oral vancomycin on gut microbiota, bile acid metabolism, and insulin sensitivity. J Hepatol 2014; 60: 824–31.
    1. Wu H-J, Ivanov II, Darce J et al. . Gut-residing segmented filamentous bacteria drive autoimmune arthritis via T helper 17 cells. Immunity 2010; 32: 815–27.
    1. Zhang D, Jia H, Feng Q et al. . The oral and gut microbiomes are perturbed in rheumatoid arthritis and partly normalized after treatment. Nat Med 2015; 21: 895–905.
    1. Rashid MU, Zaura E, Buijs MJ et al. . Determining the long-term effect of antibiotic administration on the human normal intestinal microbiota using culture and pyrosequencing methods. Clin Infect Dis 2015; 60 Suppl 2: S77–84.
    1. Louie TJ, Peppe J, Watt CK et al. . Tolevamer, a novel nonantibiotic polymer, compared with vancomycin in the treatment of mild to moderately severe Clostridium difficile-associated diarrhea. Clin Infect Dis 2006; 43: 411–20.
    1. Pepin J, Valiquette L, Gagnon S et al. . Outcomes of Clostridium difficile-associated disease treated with metronidazole or vancomycin before and after the emergence of NAP1/027. Am J Gastroenterol 2007; 102: 2781–8.
    1. Falcone M, Russo A, Iraci F et al. . Risk factors and outcomes for bloodstream infections secondary to Clostridium difficile infection. Antimicrob Agents Chemother 2015; 60: 252–7.

Source: PubMed

3
Subscribe