Reconstitution of the gut microbiota of antibiotic-treated patients by autologous fecal microbiota transplant

Ying Taur, Katharine Coyte, Jonas Schluter, Elizabeth Robilotti, Cesar Figueroa, Mergim Gjonbalaj, Eric R Littmann, Lilan Ling, Liza Miller, Yangtsho Gyaltshen, Emily Fontana, Sejal Morjaria, Boglarka Gyurkocza, Miguel-Angel Perales, Hugo Castro-Malaspina, Roni Tamari, Doris Ponce, Guenther Koehne, Juliet Barker, Ann Jakubowski, Esperanza Papadopoulos, Parastoo Dahi, Craig Sauter, Brian Shaffer, James W Young, Jonathan Peled, Richard C Meagher, Robert R Jenq, Marcel R M van den Brink, Sergio A Giralt, Eric G Pamer, Joao B Xavier, Ying Taur, Katharine Coyte, Jonas Schluter, Elizabeth Robilotti, Cesar Figueroa, Mergim Gjonbalaj, Eric R Littmann, Lilan Ling, Liza Miller, Yangtsho Gyaltshen, Emily Fontana, Sejal Morjaria, Boglarka Gyurkocza, Miguel-Angel Perales, Hugo Castro-Malaspina, Roni Tamari, Doris Ponce, Guenther Koehne, Juliet Barker, Ann Jakubowski, Esperanza Papadopoulos, Parastoo Dahi, Craig Sauter, Brian Shaffer, James W Young, Jonathan Peled, Richard C Meagher, Robert R Jenq, Marcel R M van den Brink, Sergio A Giralt, Eric G Pamer, Joao B Xavier

Abstract

Antibiotic treatment can deplete the commensal bacteria of a patient's gut microbiota and, paradoxically, increase their risk of subsequent infections. In allogeneic hematopoietic stem cell transplantation (allo-HSCT), antibiotic administration is essential for optimal clinical outcomes but significantly disrupts intestinal microbiota diversity, leading to loss of many beneficial microbes. Although gut microbiota diversity loss during allo-HSCT is associated with increased mortality, approaches to reestablish depleted commensal bacteria have yet to be developed. We have initiated a randomized, controlled clinical trial of autologous fecal microbiota transplantation (auto-FMT) versus no intervention and have analyzed the intestinal microbiota profiles of 25 allo-HSCT patients (14 who received auto-FMT treatment and 11 control patients who did not). Changes in gut microbiota diversity and composition revealed that the auto-FMT intervention boosted microbial diversity and reestablished the intestinal microbiota composition that the patient had before antibiotic treatment and allo-HSCT. These results demonstrate the potential for fecal sample banking and posttreatment remediation of a patient's gut microbiota after microbiota-depleting antibiotic treatment during allo-HSCT.

Copyright © 2018 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.

Figures

Fig. 1.. The gut microbiota is disrupted…
Fig. 1.. The gut microbiota is disrupted during allo-HSCT.
A total of 3237 fecal samples from 753 patients who underwent allo-HSCT were obtained, and the microbiota composition was analyzed using high-throughput 16S rRNA amplicon sequencing. (A) Antibiotics with a varying spectrum of activity were given over the course of allo-HSCT, either as prophylaxis or for treatment of infections. Antibiotics with anti-anaerobic activity included β-lactams, carbapenems, metronidazole, clindamycin, and vancomycin (oral only); these antibiotics were defined as microbiota perturbing in the study protocol (see the Supplementary Materials). (B) The α-diversity of each microbiota sample was measured using the IS index. Intestinal microbiota diversity dropped sharply as patients progressed through allo-HSCT treatment, reaching very low levels (IS < 4 or lower). (C) The average diversity trend showed that the drop in average microbiota diversity began several days before hematopoietic stem cell infusion, during the conditioning regimen and antibiotic prophylaxis treatment.
Fig. 2.. Timeline for a study patient…
Fig. 2.. Timeline for a study patient undergoing allo-HSCT and randomized to receive auto-FMT.
(A) Allo-HSCT for study patient T5 was initiated with pretransplant conditioning [chemotherapy and total body irradiation (TBI)], followed by allogeneic hematopoietic stem cell infusion (day 0). Various antibiotics were given throughout this period for prophylactic and treatment purposes. (B) Neutrophil count reached a nadir but later recovered after hematopoietic stem cell engraftment. (C) The subject’s intestinal microbiota composition was altered as shown by sequencing of longitudinally collected fecal samples obtained before allo-HSCT and up to day 91 after transplant. After stem cell engraftment, randomization assigned patient T5 to the treatment arm and the patient received an auto-FMT on day 49 using the patient’s initial pretreatment feces, which had been collected and stored before allo-HSCT (initial feces collected at day −21). The intestinal microbiota was restored to that before transplant in terms of (D) its diversity (α-diversity quantified by the IS index) and (E) its percent similarity to the initial fecal microbiota.
Fig. 3.. Auto-FMT improves gut microbiota diversity…
Fig. 3.. Auto-FMT improves gut microbiota diversity and composition in allo-HSCT patients.
(A) t-SNE plot provides visualization of gut microbiota composition during allo-HSCT. t-SNE plots were constructed from 3237 fecal samples collected from allo-HSCT patients since 2010; the patients were randomized in the auto-FMT trial. The plot shows a large region of high diversity (IS > 4) and clusters of low diversity corresponding to domination by specific bacterial species, for example, Enterococcus and Klebsiella. (B) Each patient followed a unique trajectory in the t-SNE space. The control patients (C) and those receiving auto-FMT treatment (T) were ranked by post-randomization gut microbiota recovery. The patient’s number and the percentage similarity of the gut microbiota to the initial fecal sample are shown. Patients started with a diverse gut microbiota composition, but then many progressed to domination states with loss of diversity due to the impact of antibiotic and chemotherapy administration. The microbiota composition in patients after auto-FMT (patients T1 to T14) moved closer to the initial composition than it did for control patients (patients C1 to C11). (C) Effect sizes of clinical parameters quantified by a mixed-effects model (along with a 95% CI) show that auto-FMT for allo-HSCT patients improved gut microbiota diversity (left-hand plot; P = 5 × 10−6) and recovery of the original gut microbiota composition (right-hand plot; P = 5 × 10−6).
Fig. 4.. Auto-FMT restores commensal members of…
Fig. 4.. Auto-FMT restores commensal members of the gut microbiota in allo-HSCT patients.
(A) Commensal species (ranked by their abundance) in the initial fecal sample from allo-HSCT patients disappeared after allo-HSCT before randomization to the auto-FMT or control arm (Pre-Rand); they reappeared after auto-FMT (Post-Rand) compared to the control group, who did not receive auto-FMT. Sixty percent of the patients enrolled in this trial (15 of 25) had a gut microbial diversity of IS < 4 before randomization; after randomization, 3 of 11 (27%) control patients but only 1 of 14 (7%) patients receiving auto-FMT retained this depleted diversity. The percent recovery also showed that auto-FMT helped to reconstitute the original gut microbial composition. Only 3 of 11 (27%) of the control patients had a composition 75% or more similar to their initial fecal sample, whereas 11 of 14 (79%) of the auto-FMT–treated patients showed 75% or more recovery. The bacteria in the left-hand grayscale heat map represent the top 50 bacterial strains present in the patient’s initial fecal sample; the right-hand grayscale heat map highlights seven bacterial species that increased after allo-HSCT (colors underlying the heat maps are defined in the key in Fig. 2). (B) Bacterial taxa from the patient’s initial fecal sample were classified on the basis of their presence in pre- and post-randomization fecal samples from the same patient. This is shown for all taxonomic groups (bottom panel) and for three important taxonomic groups (top three panels). A taxon was considered lost (red) if it was not detected in both pre- and post-randomization samples, restored (blue) if it was lost in the pre-randomization sample but was present in the post-randomization sample, and never lost (gray) if it remained detectable throughout the study (note that the initial sample of patient T4 did not have detectable Bacteroidetes).

References

    1. Pamer EG, Resurrecting the intestinal microbiota to combat antibiotic-resistant pathogens. Science 352, 535–538 (2016).
    1. Taur Y, Xavier JB, Lipuma L, Ubeda C, Goldberg J, Gobourne A, Lee YJ, Dubin KA, Socci ND, Viale A, Perales M-A, Jenq RR, van den Brink MRM, Pamer EG, Intestinal domination and the risk of bacteremia in patients undergoing allogeneic hematopoietic stem cell transplantation. Clin. Infect. Dis 55, 905–914 (2012).
    1. Holler E, Butzhammer P, Schmid K, Hundsrucker C, Koestler J, Peter K, Zhu W, Sporrer D, Hehlgans T, Kreutz M, Holler B, Wolff D, Edinger M, Andreesen R, Levine JE, Ferrara JL, Gessner A, Spang R, Oefner PJ, Metagenomic analysis of the stool microbiome in patients receiving allogeneic stem cell transplantation: Loss of diversity is associated with use of systemic antibiotics and more pronounced in gastrointestinal graft-versus-host disease. Biol. Blood Marrow Transplant 20, 640–645 (2014).
    1. Jenq RR, Taur Y, Devlin SM, Ponce DM, Goldberg JD, Ahr KF, Littmann ER, Ling L, Gobourne AC, Miller LC, Docampo MD, Peled JU, Arpaia N, Cross JR, Peets TK, Lumish MA, Shono Y, Dudakov JA, Poeck H, Hanash AM, Barker JN, Perales M-A, Giralt SA, Pamer EG, van den Brink MRM, Intestinal Blautia is associated with reduced death from graft-versus-host disease. Biol. Blood Marrow Transplant 21, 1373–1383 (2015).
    1. Jenq RR, Ubeda C, Taur Y, Menezes CC, Khanin R, Dudakov JA, Liu C, West ML, Singer NV, Equinda MJ, Gobourne A, Lipuma L, Young LF, Smith OM, Ghosh A, Hanash AM, Goldberg JD, Aoyama K, Blazar BR, Pamer EG, van den Brink MRM, Regulation of intestinal inflammation by microbiota following allogeneic bone marrow transplantation. J. Exp. Med 209, 903–911 (2012).
    1. Shono Y, Docampo MD, Peled JU, Perobelli SM, Velardi E, Tsai JJ, Slingerland AE, Smith OM, Young LF, Gupta J, Lieberman SR, Jay HV, Ahr KF, Porosnicu Rodriguez KA, Xu K, Calarfiore M, Poeck H, Caballero S, Devlin SM, Rapaport F, Dudakov JA, Hanash AM, Gyurkocza B, Murphy GF, Gomes C, Liu C, Moss EL, Falconer SB, Bhatt AS, Taur Y, Pamer EG, van den Brink MRM, Jenq RR, Increased GVHD-related mortality with broad-spectrum antibiotic use after allogeneic hematopoietic stem cell transplantation in human patients and mice. Sci. Transl. Med 8, 339ra71 (2016).
    1. Pamer EG, Taur Y, Jenq R, van den Brink MRM, Impact of the intestinal microbiota on infections and survival following hematopoietic stem cell transplantation. Blood 124, SCI–48 (2014).
    1. Marchesi JR, Adams DH, Fava F, Hermes GDA, Hirschfield GM, Hold G, Quraishi MN, Kinross J, Smidt H, Tuohy KM, Thomas LV, Zoetendal EG, Hart A, The gut microbiota and host health: A new clinical frontier. Gut 65, 330–339 (2016).
    1. David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, Ling AV, Devlin AS, Varma Y, Fischbach MA, Biddinger SB, Dutton RJ, Turnbaugh PJ, Diet rapidly and reproducibly alters the human gut microbiome. Nature 505, 559–563 (2014).
    1. Kakihana K, Fujioka Y, Suda W, Najima Y, Kuwata G, Sasajima S, Mimura I, Morita H, Sugiyama D, Nishikawa H, Hattori M, Hino Y, Ikegawa S, Yamamoto K, Toya T, Doki N, Koizumi K, Honda K, Ohashi K, Fecal microbiota transplantation for patients with steroid-resistant acute graft-versus-host disease of the gut. Blood 128, 2083–2088 (2016).
    1. Kelly CR, Ihunnah C, Fischer M, Khoruts A, Surawicz C, Afzali A, Aroniadis O, Barto A, Borody T, Giovanelli A, Gordon S, Gluck M, Hohmann EL, Kao D, Kao JY, McQuillen DP, Mellow M, Rank KM, Rao K, Ray A, Schwartz MA, Singh N,Stollman N, Suskind DL, Vindigni SM, Youngster I, Brandt L, Fecal microbiota transplant for treatment of Clostridium difficile infection in immunocompromised patients. Am. J. Gastroenterol 109, 1065–1071 (2014).
    1. van der Maaten L, Hinton G, Visualizing data using t-SNE. J. Mach. Learn. Res 9, 2579–2605 (2008).
    1. Smits SA, Leach J, Sonnenburg ED, Gonzalez CG, Lichtman JS, Reid G, Knight R, Manjurano A, Changalucha J, Elias JE, Dominguez-Bello MG, Sonnenburg JL, Seasonal cycling in the gut microbiome of the Hadza hunter-gatherers of Tanzania. Science 357, 802–806 (2017).
    1. Deshmukh HS, Liu Y, Menkiti OR, Mei J, Dai N, O’Leary CE, Oliver PM, Kolls JK, Weiser JN, Worthen GS, The microbiota regulates neutrophil homeostasis and host resistance to Escherichia coli K1 sepsis in neonatal mice. Nat. Med 20, 524–530 (2014).
    1. Lee YJ, Arguello ES, Jenq RR, Littmann E, Kim GJ, Miller LC, Ling L, Figueroa C, Robilotti E, Perales M-A, Barker JN, Giralt S, van den Brink MRM, Pamer EG, Taur Y, Protectivefactors in the intestinal microbiome against Clostridium difficile infection in recipients of allogeneic hematopoietic stem cell transplantation. J. Infect. Dis 215, 1117–1123 (2017).
    1. Lee SM, Donaldson GP, Mikulski Z, Boyajian S, Ley K,Mazmanian SK, Bacterial colonization factors control specificity and stability of the gut microbiota. Nature 501, 426–429 (2013).
    1. Reeves AE, Koenigsknecht MJ, Bergin IL, Young VB, Suppression of Clostridium difficile in the gastrointestinal tracts of germfree mice inoculated with a murine isolate from the family Lachnospiraceae. Infect. Immun 80, 3786–3794 (2012).
    1. Weber D, Jenq RR, Peled JU, Taur Y, Hiergeist A, Koestler J, Dettmer K, Weber M, Wolff D, Hahn J, Pamer EG, Herr W, Gessner A, Oefner PJ, van den Brink MRM,Holler E, Microbiota disruption induced by early use of broad-spectrum antibiotics is an independent risk factor of outcome after allogeneic stem cell transplantation. Biol. Blood Marrow Transplant 23, 845–852 (2017).
    1. Weber D, Oefner PJ, Hiergeist A, Koestler J, Gessner A, Weber M, Hahn J, Wolff D, Stammler F, Spang R, Herr W, Dettmer K, Holler E, Low urinary indoxyl sulfate levels early after transplantation reflect a disrupted microbiome and are associated with poor outcome. Blood 126, 1723–1728 (2015).
    1. Kelly CR, Khoruts A, Staley C, Sadowsky MJ, Abd M, Alani M, Bakow B, Curran P, McKenney J, Tisch A, Reinert SE, Machan JT, Brandt LJ, Effect of fecal microbiota transplantation on recurrence in multiply recurrent Clostridium difficile infection: A randomized trial. Ann. Intern. Med 165, 609–616 (2016).
    1. Buffie CG, Bucci V, Stein RR, McKenney PT, Ling L, Gobourne A, No D, Liu H, Kinnebrew M, Viale A, Littmann E, van den Brink MRM, Jenq RR, Taur Y, Sander C, Cross JR, Toussaint NC, Xavier JB, Pamer EG, Precision microbiome reconstitution restores bile acid mediated resistance to Clostridium difficile. Nature 517, 205–208 (2015).
    1. Caporaso JG, Lauber CL, Walters WA, Berg-Lyons D, Huntley J, Fierer N, Owens SM, Betley J, Fraser L, Bauer M, Gormley N, Gilbert JA, Smith G, Knight R, Ultra-high-throughput microbial community analysis on the Illumina HiSeq and MiSeq platforms. ISME J. 6, 1621–1624 (2012).
    1. Edgar RC, UPARSE: Highly accurate OTU sequences from microbial amplicon reads. Nat. Methods 10, 996–998 (2013).
    1. Edgar RC, Flyvbjerg H, Error filtering, pair assembly and error correction for next-generation sequencing reads. Bioinformatics 31, 3476–3482 (2015).
    1. Altschul SF, Gish W, Miller W, Myers EW, Lipman DJ, Basic local alignment search tool. J. Mol. Biol 215, 403–410 (1990).
    1. Tatusova T, Ciufo S, Fedorov B, O’Neill K, Tolstoy I, RefSeq microbial genomes database: New representation and annotation strategy. Nucleic Acids Res. 42, D553–D559 (2014).
    1. Amir E.-a. D., Davis KL, Tadmor MD, Simonds EF, Levine JH, Bendall SC, Shenfeld DK, Krishnaswamy S, Nolan GP, Pe’er D, viSNE enables visualization of high dimensional single-cell data and reveals phenotypic heterogeneity of leukemia. Nat. Biotechnol 31, 545–552 (2013).
    1. Xiong Q, Mukherjee S, Furey TS, GSAASeqSP: A toolset for gene set association analysis of RNA-Seq data. Sci. Rep 4, 6347 (2014).
    1. Irizarry RA, Wang C, Zhou Y, Speed TP, Gene set enrichment analysis made simple. Stat. Methods Med. Res 18, 565–575 (2009).

Source: PubMed

3
S'abonner