Randomized clinical trial to evaluate the effect of fecal microbiota transplant for initial Clostridium difficile infection in intestinal microbiome

Adrián Camacho-Ortiz, Eva María Gutiérrez-Delgado, Jose F Garcia-Mazcorro, Soraya Mendoza-Olazarán, Adrián Martínez-Meléndez, Laura Palau-Davila, Simon D Baines, Héctor Maldonado-Garza, Elvira Garza-González, Adrián Camacho-Ortiz, Eva María Gutiérrez-Delgado, Jose F Garcia-Mazcorro, Soraya Mendoza-Olazarán, Adrián Martínez-Meléndez, Laura Palau-Davila, Simon D Baines, Héctor Maldonado-Garza, Elvira Garza-González

Abstract

Objective: The aim of this study was to evaluate the impact of fecal donor-unrelated donor mix (FMT-FURM) transplantation as first-line therapy for C. difficile infection (CDI) in intestinal microbiome.

Methods: We designed an open, two-arm pilot study with oral vancomycin (250mg every 6 h for 10-14 days) or FMT-FURM as treatments for the first CDI episode in hospitalized adult patients in Hospital Universitario "Dr. Jose Eleuterio Gonzalez". Patients were randomized by a closed envelope method in a 1: 1 ratio to either oral vancomycin or FMT-FURM. CDI resolution was considered when there was a reduction on the Bristol scale of at least 2 points, a reduction of at least 50% in the number of bowel movements, absence of fever, and resolution of abdominal pain (at least two criteria). From each patient, a fecal sample was obtained at days 0, 3, and 7 after treatment. Specimens were cultured to isolate C. difficile, and isolates were characterized by PCR. Susceptibility testing of isolates was performed using the agar dilution method. Fecal samples and FMT-FURM were analyzed by 16S rRNA sequencing.

Results: We included 19 patients; 10 in the vancomycin arm and 9 in the FMT-FURM arm. However, one of the patients in the vancomycin arm and two patients in the FMT-FURM arm were eliminated. Symptoms resolved in 8/9 patients (88.9%) in the vancomycin group, while symptoms resolved in 4/7 patients (57.1%) after the first FMT-FURM dose (P = 0.26) and in 5/7 patients (71.4%) after the second dose (P = 0.55). During the study, no adverse effects attributable to FMT-FURM were observed in patients. Twelve isolates were recovered, most isolates carried tcdB, tcdA, cdtA, and cdtB, with an 18-bp deletion in tcdC. All isolates were resistant to ciprofloxacin and moxifloxacin but susceptible to metronidazole, linezolid, fidaxomicin, and tetracycline. In the FMT-FURM group, the bacterial composition was dominated by Firmicutes, Bacteroidetes, and Proteobacteria at all-time points and the microbiota were remarkably stable over time. The vancomycin group showed a very different pattern of the microbial composition when comparing to the FMT-FURM group over time.

Conclusion: The results of this preliminary study showed that FMT-FURM for initial CDI is associated with specific bacterial communities that do not resemble the donors' sample.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Fig 1. Flowchart of the study design.
Fig 1. Flowchart of the study design.
Fig 2. Resolution rates (%) of initial…
Fig 2. Resolution rates (%) of initial C. difficile infection in patients treated with vancomycin and fecal microbiota transplantation (FMT).
Fig 3. Bar charts showing the relative…
Fig 3. Bar charts showing the relative abundance of OTUs at the phylum level.
Distribution of bacterial phyla in the fecal microbiota of transplantation (FMT) group and the vancomycin group (Vanco). The label of each sample (e.g., FMT-2-0) denotes Treatment (FMT or Vancomycin), the assigned number of each patient, and days on treatment (0 = baseline, 3 = 3 days on treatment, 7 = 7 days on treatment). To facilitate comparison and visualization, the distribution of bacterial phyla of the donors’ pool is presented for the FMT group (Donors’ pool). Please note that the border of the three most abundant groups was highlighted for better visualization and the bars were organized based on the abundance of the highest abundant group (i.e., Firmicutes).
Fig 4. Average bacterial composition at the…
Fig 4. Average bacterial composition at the phylum level.
FMT: fecal microbiota transplantation.
Fig 5. Bar charts showing the relative…
Fig 5. Bar charts showing the relative abundance of OTUs at the family level.
Distribution of bacterial families in the fecal microbiota of transplantation (FMT) group and the vancomycin group (Vanco). The label of each sample (e.g., FMT-5-0) denotes Treatment (FMT or Vancomycin), the assigned number of each patient, and days on treatment (0 = baseline, 3 = 3 days on treatment, 7 = 7 days on treatment). To facilitate comparison and visualization, the distribution of bacterial families of the donors’ pool is presented for the FMT group (Donors’ pool). Please note that the border of the three most abundant groups was highlighted for better visualization and the bars were organized based on the abundance of the highest abundant group (i.e., Enterobacteriaceae).
Fig 6. Number of observed species.
Fig 6. Number of observed species.
The figure shows the number of observed species (i.e., operational taxonomic units at 97% similarity, y-axis) and the number of sequences per sample (x-axis) for the FMT group and Vancomycin group across collection days of fecal samples. Error bars represent standard deviations.
Fig 7. PCoA plots of weighted UniFrac…
Fig 7. PCoA plots of weighted UniFrac distances for the FMT group and the vancomycin group.
PCoA plots of weighted UniFrac distances for both groups across the days of the collection of fecal samples. There was no significant clustering of microbial communities according to time point (please note that the analysis of unweighted UniFrac distances did not reveal any significant clustering of microbial communities either).

References

    1. Hamilton MJ, Weingarden AR, Sadowsky MJ, Khoruts A. Standardized frozen preparation for transplantation of fecal microbiota for recurrent Clostridium difficile infection. Am J Gastroenterol. 2012;107(5):761–767. doi:
    1. Rineh A, Kelso MJ, Vatansever F, Tegos GP, Hamblin MR. Clostridium difficile infection: molecular pathogenesis and novel therapeutics. Expert Rev Anti Infect Ther. 2014;12(1):131–150. doi:
    1. Lessa FC, Winston LG, McDonald LC, Team EIPCdS. Burden of Clostridium difficile infection in the United States. N Engl J Med. 2015;372(24):2369–2370. doi:
    1. Oake N, Taljaard M, van Walraven C, Wilson K, Roth V, Forster AJ. The effect of hospital-acquired Clostridium difficile infection on in-hospital mortality. Arch Intern Med. 2010;170(20):1804–1810. doi:
    1. Cohen SH, Gerding DN, Johnson S, Kelly CP, Loo VG, McDonald LC, et al. Clinical practice guidelines for Clostridium difficile infection in adults: 2010 update by the society for healthcare epidemiology of America (SHEA) and the infectious diseases society of America (IDSA). Infect Control Hosp Epidemiol. 2010;31(5):431–455. doi:
    1. Vardakas KZ, Polyzos KA, Patouni K, Rafailidis PI, Samonis G, Falagas ME. Treatment failure and recurrence of Clostridium difficile infection following treatment with vancomycin or metronidazole: a systematic review of the evidence. Int J Antimicrob Agents. 2012;40(1):1–8. doi:
    1. Pépin J, Valiquette L, Gagnon S, Routhier S, Brazeau I. Outcomes of Clostridium difficile-associated disease treated with metronidazole or vancomycin before and after the emergence of NAP1/027. Am J Gastroenterol. 2007;102(12):2781–2788. doi:
    1. Mattila E, Uusitalo-Seppälä R, Wuorela M, Lehtola L, Nurmi H, Ristikankare M, et al. Fecal transplantation, through colonoscopy, is effective therapy for recurrent Clostridium difficile infection. Gastroenterology. 2012;142(3):490–496. doi:
    1. Cammarota G, Masucci L, Ianiro G, Bibbò S, Dinoi G, Costamagna G, et al. Randomised clinical trial: faecal microbiota transplantation by colonoscopy vs. vancomycin for the treatment of recurrent Clostridium difficile infection. Aliment Pharmacol Ther. 2015;41(9):835–843. doi:
    1. van Nood E, Vrieze A, Nieuwdorp M, Fuentes S, Zoetendal EG, de Vos WM, et al. Duodenal infusion of donor feces for recurrent Clostridium difficile. N Engl J Med. 2013;368(5):407–415. doi:
    1. Lemee L, Dhalluin A, Testelin S, Mattrat MA, Maillard K, Lemeland JF, et al. Multiplex PCR targeting tpi (triose phosphate isomerase), tcdA (Toxin A), and tcdB (Toxin B) genes for toxigenic culture of Clostridium difficile. J Clin Microbiol. 2004;42(12):5710–5714. doi:
    1. Persson S, Jensen JN, Olsen KE. Multiplex PCR method for detection of Clostridium difficile tcdA, tcdB, cdtA, and cdtB and internal in-frame deletion of tcdC. J Clin Microbiol. 2011;49(12):4299–4300. doi:
    1. Freeman J, Wilcox MH. Antibiotic activity against genotypically distinct and indistinguishable Clostridium difficile isolates. J Antimicrob Chemother. 2001;47(2):244–246.
    1. Buchler AC, Rampini SK, Stelling S, Ledergerber B, Peter S, Schweiger A, et al. Antibiotic susceptibility of Clostridium difficile is similar worldwide over two decades despite widespread use of broad-spectrum antibiotics: an analysis done at the University Hospital of Zurich. BMC Infect Dis. 2014;14:607 doi:
    1. Goldstein EJ, Citron DM, Merriam CV, Warren YA, Tyrrell KL, Fernandez HT. In vitro activities of daptomycin, vancomycin, quinupristin- dalfopristin, linezolid, and five other antimicrobials against 307 gram-positive anaerobic and 31 Corynebacterium clinical isolates. Antimicrob Agents Chemother. 2003;47(1):337–341. doi:
    1. O'Connor JR, Galang MA, Sambol SP, Hecht DW, Vedantam G, Gerding DN, et al. Rifampin and rifaximin resistance in clinical isolates of Clostridium difficile. Antimicrob Agents Chemother. 2008;52(8):2813–2817. doi:
    1. CLSI. M100-S26. Performance Standards for Antimicrobial Susceptibility Testing; Twenty-Second Informational Supplement. Wayne, PA: Clinical and Laboratory Standars Institute. 2016.
    1. Caporaso JG, Lauber CL, Walters WA, Berg-Lyons D, Huntley J, Fierer N, et al. Ultra-high-throughput microbial community analysis on the Illumina HiSeq and MiSeq platforms. ISME J. 2012;6(8):1621–1624. doi:
    1. Rideout JR, He Y, Navas-Molina JA, Walters WA, Ursell LK, Gibbons SM, et al. Subsampled open-reference clustering creates consistent, comprehensive OTU definitions and scales to billions of sequences. PeerJ. 2014;2:e545 doi:
    1. DeSantis TZ, Hugenholtz P, Larsen N, Rojas M, Brodie EL, Keller K, et al. Greengenes, a chimera-checked 16S rRNA gene database and workbench compatible with ARB. Appl Environ Microbiol. 2006;72(7):5069–5072. doi:
    1. Navas-Molina JA, Peralta-Sánchez JM, González A, McMurdie PJ, Vázquez-Baeza Y, Xu Z, et al. Advancing our understanding of the human microbiome using QIIME. Methods Enzymol. 2013;531:371–444. doi:
    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 doi:
    1. Ø H. PAST: paleontological statistics software package for education and data analysis. In: DAT H, editor. Paleontol Electron2001. p. 1–9.
    1. Bent SJ, Forney LJ. The tragedy of the uncommon: understanding limitations in the analysis of microbial diversity. ISME J. 2008;2(7):689–695. doi:
    1. Lozupone C, Hamady M, Knight R. UniFrac—an online tool for comparing microbial community diversity in a phylogenetic context. BMC Bioinformatics. 2006;7:371 doi:
    1. Lozupone CA, Hamady M, Kelley ST, Knight R. Quantitative and qualitative beta diversity measures lead to different insights into factors that structure microbial communities. Appl Environ Microbiol. 2007;73(5):1576–1585. doi:
    1. Brandt LJ, Aroniadis OC, Mellow M, Kanatzar A, Kelly C, Park T, et al. Long-term follow-up of colonoscopic fecal microbiota transplant for recurrent Clostridium difficile infection. Am J Gastroenterol. 2012;107(7):1079–1087. doi:
    1. Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, Contreras M, et al. Human gut microbiome viewed across age and geography. Nature. 2012;486(7402):222–227. doi:
    1. Dethlefsen L, Huse S, Sogin ML, Relman DA. The pervasive effects of an antibiotic on the human gut microbiota, as revealed by deep 16S rRNA sequencing. PLoS Biol. 2008;6(11):e280 doi:
    1. Quraishi MN, Widlak M, Bhala N, Moore D, Price M, Sharma N, et al. Systematic review with meta-analysis: the efficacy of faecal microbiota transplantation for the treatment of recurrent and refractory Clostridium difficile infection. Aliment Pharmacol Ther. 2017;46(5):479–493. doi:

Source: PubMed

3
Abonnere