Scientific frontiers in faecal microbiota transplantation: joint document of Asia-Pacific Association of Gastroenterology (APAGE) and Asia-Pacific Society for Digestive Endoscopy (APSDE)

Siew C Ng, Michael A Kamm, Yun Kit Yeoh, Paul K S Chan, Tao Zuo, Whitney Tang, Ajit Sood, Akira Andoh, Naoki Ohmiya, Yongjian Zhou, Choon Jin Ooi, Varocha Mahachai, Chun-Ying Wu, Faming Zhang, Kentaro Sugano, Francis K L Chan, Siew C Ng, Michael A Kamm, Yun Kit Yeoh, Paul K S Chan, Tao Zuo, Whitney Tang, Ajit Sood, Akira Andoh, Naoki Ohmiya, Yongjian Zhou, Choon Jin Ooi, Varocha Mahachai, Chun-Ying Wu, Faming Zhang, Kentaro Sugano, Francis K L Chan

Abstract

Objective: The underlying microbial basis, predictors of therapeutic outcome and active constituent(s) of faecal microbiota transplantation (FMT) mediating benefit remain unknown. An international panel of experts presented key elements that will shape forthcoming FMT research and practice.

Design: Systematic search was performed, FMT literature was critically appraised and a 1-day round-table discussion was conducted to derive expert consensus on key issues in FMT research.

Results: 16 experts convened and discussed five questions regarding (1) the role of donor and recipient microbial (bacteria, viruses, fungi) parameters in FMT; (2) methods to assess microbiota alterations; (3) concept of keystone species and microbial predictors of FMT, (4) influence of recipient profile and antibiotics pretreatment on FMT engraftment and maintenance and (5) new developments in FMT formulations and delivery. The panel considered that variable outcomes of FMT relate to compositional and functional differences in recipient's microbiota, and likely donor-associated and recipient-associated physiological and genetic factors. Taxonomic composition of donor intestinal microbiota may influence the efficacy of FMT in recurrent Clostridioides difficile infections and UC. FMT not only alters bacteria composition but also establishes trans-kingdom equilibrium between gut fungi, viruses and bacteria to promote the recovery of microbial homeostasis. FMT is not a one size fits all and studies are required to identify microbial components that have specific effects in patients with different diseases.

Conclusion: FMT requires optimisation before their therapeutic promise can be evaluated for different diseases. This summary will guide future directions and priorities in advancement of the science and practice of FMT.

Keywords: bacteria; donor; faecal microbiota transplantation; fungi; recipient; virus.

Conflict of interest statement

Competing interests: FZ invented the concept of GenFMTer and TET and devices related to it.

© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Flow diagram of studies included after systematic search.
Figure 2
Figure 2
Potential mechanisms of faecal microbiota transplantation (FMT). Potential mechanisms of FMT include direct interaction or competition between donor and recipient gut microbiota to achieve homeostasis, effect of donor microbiota on the host immunity and effect of microbiota on modulating host metabolism and physiology. Competitive niche exclusion is a plausible mechanism behind the therapeutic effects of FMT in the treatment of Clostridioides difficile infections (CDI) by introducing competitors and/or modifying the diseased gut environment to the detriment of Clostridioidesdifficile. For example, introduction of non-toxigenic C. difficile strains can reduce the recurrence of CDI in subjects. Another competition-based strategy harnessed by gut micro-organisms is the production of bacteriocins. Quorum sensing systems have also been detected in C. difficile and are known to affect toxin expression profiles in other coexisting micro-organisms. FMT can alter host bile acid metabolism concomitant with alterations to gut microbiota composition. In CDI, a consistent metabolic signature with reduced primary bile acids and increased secondary bile acid production capacity has been reported after FMT. FMT restores Firmicutes phylum bacteria and secondary bile acid metabolism, providing a prime example of FMT re-establishing normal gut microbiota and host metabolism. FMT also has the ability to restore gut microbiota and mucosal immunity and systemic immunity of the host. In mice models of colitis, FMT reduced colonic inflammation and initiated a simultaneous activation of different immune pathways, leading to interleukin (IL-10) production by innate and adaptive immune cells including CD4+ T cells, invariant natural killer T (iNKT) cells and antigen-presenting cells (APC), and reduced the ability of macrophages, monocytes and dendritic cells to present MHCII-dependent bacterial antigens to colonic T cells. These results demonstrate the immunomodulating capability of FMT to therapeutically control intestinal homeostasis and highlight FMT as a valuable therapeutic option in immune-related pathologies. For instance, a reconstitution of the gut microbiome and a relative increase in the proportion of Foxp3+ regulatory T cells within the colonic mucosa were potential mechanisms through which FMT could abrogate immune check point inhibitors associated toxicity. online supplementary file 1 Please refer to Appendix 3 for references. iNKT, invariant natural killer T; MHCII, major histocompatibility complex class II.
Figure 3
Figure 3
For faecal microbiota transplantation (FMT) administration, lower GI delivery can be achieved through enema or colonoscopy, whereas upper GI delivery is through infusion via gastroscopy or via nasoenteric tubes. Oral capsules are non-invasive and is an ideal delivery method for FMT. Formulation to improve delivery, ensuring organism survival and ensure colonisation along the GI tract are needed.6 An encapsulation technique based on a water-in-oil emulsion can provide better insulation of FMT material from GI environment.7 One potential solution is the use of bile absorbent resins. The use of bile acid binding agents such as cholestyramine, when combined with Vcaps HPMC capsules, provided a 1700-fold increase in freeze-dried and rehydrated Lactobacillus casei in a simulated intestinal fluid containing 1% bile.8 Alternatively, a layer-by-layer encapsulation approach using mucoadhesive polysaccharides, chitosan and alginate has been shown to improve cytoprotection, bioavailability and engraftment of Bacillus coagulans in porcine and human intestines.9 Capsule design content release is dependent on intestinal stimuli.10 Other approaches include genetic engineering, for example, Escherichia coli engineered to bind to colorectal cancer cell receptors and catalyse production of molecules with anticancer activity.11 This novel approach can be considered for FMT if constituent strains that contribute to clinical efficacy are identified. They can then be engineered to improve binding and colonisation in the gut of FMT recipients. Please refer to Appendix 3 for references.(online supplementary file 1)

References

    1. Mullish BH, Quraishi MN, Segal JP, et al. . The use of faecal microbiota transplant as treatment for recurrent or refractory Clostridium difficile infection and other potential indications: joint British Society of Gastroenterology (BSG) and Healthcare Infection Society (HIS) guidelines. Gut 2018;67:1920–41. 10.1136/gutjnl-2018-316818
    1. Krajicek E, Fischer M, Allegretti JR, et al. . Nuts and bolts of fecal microbiota transplantation. Clin Gastroenterol Hepatol 2019;17:345–52. 10.1016/j.cgh.2018.09.029
    1. Moayyedi P, Surette MG, Kim PT, et al. . Fecal microbiota transplantation induces remission in patients with active ulcerative colitis in a randomized controlled trial. Gastroenterology 2015;149:102–9. 10.1053/j.gastro.2015.04.001
    1. Rossen NG, Fuentes S, van der Spek MJ, et al. . Findings from a randomized controlled trial of fecal transplantation for patients with ulcerative colitis. Gastroenterology 2015;149:110–8. 10.1053/j.gastro.2015.03.045
    1. Paramsothy S, Kamm MA, Kaakoush NO, et al. . Multidonor intensive faecal microbiota transplantation for active ulcerative colitis: a randomised placebo-controlled trial. The Lancet 2017;389:1218–28. 10.1016/S0140-6736(17)30182-4
    1. Costello SP, Hughes PA, Waters O, et al. . Effect of fecal microbiota transplantation on 8-Week remission in patients with ulcerative colitis. JAMA 2019;321:156–64. 10.1001/jama.2018.20046
    1. Xu D, Chen VL, Steiner CA, et al. . Efficacy of fecal microbiota transplantation in irritable bowel syndrome: a systematic review and meta-analysis. Am J Gastroenterol 2019.
    1. Bajaj JS, Kassam Z, Fagan A, et al. . Fecal microbiota transplant from a rational stool donor improves hepatic encephalopathy: a randomized clinical trial. Hepatology 2017;66:1727–38. 10.1002/hep.29306
    1. Bajaj JS, Fagan A, Gavis EA, et al. . Long-Term outcomes of fecal microbiota transplantation in patients with cirrhosis. Gastroenterology 2019;156:1921–3. 10.1053/j.gastro.2019.01.033
    1. Kang D-W, Adams JB, Coleman DM, et al. . Long-Term benefit of microbiota transfer therapy on autism symptoms and gut microbiota. Sci Rep 2019;9:5821 10.1038/s41598-019-42183-0
    1. Kootte RS, Levin E, Salojärvi J, et al. . Improvement of insulin sensitivity after lean donor feces in metabolic syndrome is driven by baseline intestinal microbiota composition. Cell Metab 2017;26:611–9. 10.1016/j.cmet.2017.09.008
    1. Vrieze A, Van Nood E, Holleman F, et al. . Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Gastroenterology 2012;143:913–6. 10.1053/j.gastro.2012.06.031
    1. Lai CY, Sung J, Cheng F, et al. . Systematic review with meta-analysis: review of donor features, procedures and outcomes in 168 clinical studies of faecal microbiota transplantation. Aliment Pharmacol Ther 2019;49:354–63. 10.1111/apt.15116
    1. . 2019.
    1. Wilson BC, Vatanen T, Cutfield WS, et al. . The Super-Donor phenomenon in fecal microbiota transplantation. Front Cell Infect Microbiol 2019;9:2 10.3389/fcimb.2019.00002
    1. Cammarota G, Ianiro G, Tilg H, et al. . European consensus conference on faecal microbiota transplantation in clinical practice. Gut 2017;66:569–80. 10.1136/gutjnl-2016-313017
    1. Hoffmann D, Palumbo F, Ravel J, et al. . Improving regulation of microbiota transplants. Science 2017;358:1390–1. 10.1126/science.aaq0034
    1. Verbeke F, Janssens Y, Wynendaele E, et al. . Faecal microbiota transplantation: a regulatory hurdle? BMC Gastroenterol 2017;17:128 10.1186/s12876-017-0687-5
    1. Barnes D, Ng K, Smits S, et al. . Competitively selected donor fecal microbiota transplantation: butyrate concentration and diversity as measures of donor quality. J Pediatr Gastroenterol Nutr 2018;67:185–7.
    1. Ott SJ, Waetzig GH, Rehman A, et al. . Efficacy of sterile fecal filtrate transfer for treating patients with Clostridium difficile infection. Gastroenterology 2017;152:799–811. 10.1053/j.gastro.2016.11.010
    1. Broecker F, Russo G, Klumpp J, et al. . Stable core virome despite variable microbiome after fecal transfer. Gut Microbes 2017;8:214–20. 10.1080/19490976.2016.1265196
    1. Zuo T, Wong SH, Lam K, et al. . Bacteriophage transfer during faecal microbiota transplantation in Clostridium difficile infection is associated with treatment outcome. Gut 2018;67:634–43.
    1. Park H, Millan BT, Hotte N, et al. . A11 effect of fecal microbial transplant on microbial and phage composition in patients with Clostridium difficile infection. J Can Assoc Gastroenterol 2018;1(suppl_1):20–1. 10.1093/jcag/gwy008.012
    1. Hoffmann C, Dollive S, Grunberg S, et al. . Archaea and fungi of the human gut microbiome: correlations with diet and bacterial residents. PLoS One 2013;8:e66019 10.1371/journal.pone.0066019
    1. Schoch CL, Seifert KA, Huhndorf S, et al. . Nuclear ribosomal internal transcribed spacer (ITS) region as a universal DNA barcode marker for fungi. Proc Natl Acad Sci U S A 2012;109:6241–6. 10.1073/pnas.1117018109
    1. Zuo T, Wong SH, Cheung CP, et al. . Gut fungal dysbiosis correlates with reduced efficacy of fecal microbiota transplantation in Clostridium difficile infection. Nat Commun 2018;9:3663 10.1038/s41467-018-06103-6
    1. Erb Downward JR, Falkowski NR, Mason KL, et al. . Modulation of post-antibiotic bacterial community reassembly and host response by Candida albicans. Sci Rep 2013;3:2191 10.1038/srep02191
    1. Reyes A, Wu M, McNulty NP, et al. . Gnotobiotic mouse model of phage-bacterial host dynamics in the human gut. Proc Natl Acad Sci U S A 2013;110:20236–41. 10.1073/pnas.1319470110
    1. Wheeler ML, Limon JJ, Bar AS, et al. . Immunological consequences of intestinal fungal dysbiosis. Cell Host Microbe 2016;19:865–73. 10.1016/j.chom.2016.05.003
    1. Vermeire S, Joossens M, Verbeke K, et al. . Donor species richness determines faecal microbiota transplantation success in inflammatory bowel disease. ECCOJC 2016;10:387–94. 10.1093/ecco-jcc/jjv203
    1. Kump P, Wurm P, Gröchenig HP, et al. . The taxonomic composition of the donor intestinal microbiota is a major factor influencing the efficacy of faecal microbiota transplantation in therapy refractory ulcerative colitis. Aliment Pharmacol Ther 2018;47:67–77. 10.1111/apt.14387
    1. Paramsothy S, Nielsen S, Kamm MA, et al. . Specific bacteria and metabolites associated with response to fecal microbiota transplantation in patients with ulcerative colitis. Gastroenterology 2019;156:1440–54. 10.1053/j.gastro.2018.12.001
    1. Fuentes S, Rossen NG, van der Spek MJ, et al. . Microbial shifts and signatures of long-term remission in ulcerative colitis after faecal microbiota transplantation. Isme J 2017;11:1877–89. 10.1038/ismej.2017.44
    1. Kazerouni A, Wein LM. Exploring the efficacy of pooled stools in fecal microbiota transplantation for Microbiota-Associated chronic diseases. PLoS One 2017;12:e0163956 10.1371/journal.pone.0163956
    1. Conceição-Neto N, Deboutte W, Dierckx T, et al. . DOP080 low viral richness at baseline in ulcerative ulcerative colitis associated with faecal microbiota transplantation success. Journal of Crohns Colitis 2017;11(suppl_1):S73–S74. 10.1093/ecco-jcc/jjx002.117
    1. Smillie CS, Sauk J, Gevers D, et al. . Strain tracking reveals the determinants of bacterial engraftment in the human gut following fecal microbiota transplantation. Cell Host Microbe 2018;23:229–40. 10.1016/j.chom.2018.01.003
    1. SS L, Zhu A, Benes V, et al. . Durable coexistence of donor and recipient strains after fecal microbiota transplantation. Science 2016;352:586–9.
    1. Liu Q, Yang K, Xu Z, et al. . 625 – donor fungi and bacteria engraftment after successful eradication of carbapenem-resistant Enterobacteriaceae with fecal microbiota transplantation: serial fecal Metagenomics analysis. Gastroenterology 2019;156:S-131 10.1016/S0016-5085(19)37116-1
    1. Zhang F, Yeoh YK, Cheng F, et al. . Tu1879 – rapid and durable engraftment of donor fungi and bacteria after successful fecal microbiota transplantation in acute graft-versus-host disease: intensive serial Metagenomics study. Gastroenterology 2019;156:S-1157–0. 10.1016/S0016-5085(19)39862-2
    1. Yeoh YK, Chen Z, Hui M, et al. . Impact of inter- and intra-individual variation, sample storage and sampling fraction on human stool microbial community profiles. PeerJ 2019;7:e6172 10.7717/peerj.6172
    1. Seekatz AM, Aas J, Gessert CE, et al. . Recovery of the gut microbiome following fecal microbiota transplantation. MBio 2014;5:e00893–14. 10.1128/mBio.00893-14
    1. Fuentes S, van Nood E, Tims S, et al. . Reset of a critically disturbed microbial ecosystem: faecal transplant in recurrent Clostridium difficile infection. Isme J 2014;8:1621–33. 10.1038/ismej.2014.13
    1. Khanna S, Vazquez-Baeza Y, González A, et al. . Changes in microbial ecology after fecal microbiota transplantation for recurrent C. difficile infection affected by underlying inflammatory bowel disease. Microbiome 2017;5 10.1186/s40168-017-0269-3
    1. Joseph OD, Thompson SV, Bogner A, et al. . Longitudinal study of the human gastrointestinal microbiota following fecal microbiota transplant (FMT) for Clostridium difficile infections. Faseb J 2017;31.
    1. Rothschild D, Weissbrod O, Barkan E, et al. . Environment dominates over host genetics in shaping human gut microbiota. Nature 2018;555:210–5. 10.1038/nature25973
    1. Lozupone CA, Stombaugh JI, Gordon JI, et al. . Diversity, stability and resilience of the human gut microbiota. Nature 2012;489:220–30. 10.1038/nature11550 10.1038/nature11550
    1. Tatusov RL, Galperin MY, Natale DA. The COG database: a tool for genome-scale analysis of protein functions and evolution. Nucleic Acids Res 2000;28:33–6. 10.1093/nar/28.1.33
    1. Kanehisa M, Sato Y, Kawashima M, et al. . Kegg as a reference resource for gene and protein annotation. Nucleic Acids Res 2016;44:D457–D462. 10.1093/nar/gkv1070
    1. Almeida A, Mitchell AL, Boland M, et al. . A new genomic blueprint of the human gut microbiota. Nature 2019;568:499–504. 10.1038/s41586-019-0965-1
    1. Pasolli E, Asnicar F, Manara S, et al. . Extensive unexplored human microbiome diversity revealed by over 150,000 genomes from metagenomes spanning age, geography, and lifestyle. Cell 2019;176:649–62. 10.1016/j.cell.2019.01.001
    1. Ianiro G, Valerio L, Masucci L, et al. . Predictors of failure after single faecal microbiota transplantation in patients with recurrent Clostridium difficile infection: results from a 3-year, single-centre cohort study. Clinical Microbiology and Infection 2017;23:337.e1–337.e3. 10.1016/j.cmi.2016.12.025
    1. Fischer M, Kao D, Mehta SR, et al. . Predictors of early failure after fecal microbiota transplantation for the therapy of Clostridium difficile infection: a multicenter study. Am J Gastroenterol 2016;111:1024–31. 10.1038/ajg.2016.180
    1. Duvallet C, Gibbons SM, Gurry T, et al. . Meta-Analysis of gut microbiome studies identifies disease-specific and shared responses. Nat Commun 2017;8 10.1038/s41467-017-01973-8
    1. Narula N, Kassam Z, Yuan Y, et al. . Systematic review and meta-analysis: fecal microbiota transplantation for treatment of active ulcerative colitis. Inflamm Bowel Dis 2017;23:1702–9. 10.1097/MIB.0000000000001228
    1. Petrof EO, Gloor GB, Vanner SJ, et al. . Stool substitute transplant therapy for the eradication of Clostridium difficile infection: ‘RePOOPulating’ the gut. Microbiome 2013;1 10.1186/2049-2618-1-3
    1. Emanuelsson F, Claesson BEB, Ljungström L, et al. . Faecal microbiota transplantation and bacteriotherapy for recurrent Clostridium difficile infection: a retrospective evaluation of 31 patients. Scand J Infect Dis 2014;46:89–97. 10.3109/00365548.2013.858181
    1. Staley C, Vaughn BP, Graiziger CT, et al. . Community dynamics drive punctuated engraftment of the fecal microbiome following transplantation using freeze-dried, encapsulated fecal microbiota. Gut Microbes 2017;8:276–88. 10.1080/19490976.2017.1299310
    1. Keshteli AH, Millan B, Madsen KL. Pretreatment with antibiotics may enhance the efficacy of fecal microbiota transplantation in ulcerative colitis: a meta-analysis. Mucosal Immunol 2017;10:565–6. 10.1038/mi.2016.123
    1. Ji SK, Yan H, Jiang T, et al. . Preparing the gut with antibiotics enhances gut microbiota reprogramming efficiency by promoting Xenomicrobiota colonization. Front Microbiol 2017;8:1208 10.3389/fmicb.2017.01208
    1. Millan B, Park H, Hotte N, et al. . Antibiotics and bowel preparation enhance the ability of fecal microbial transplantation to reshape the gut microbiota in IL-10<sup>-/-</sup> mice. Canadian Journal of Gastroenterology and Hepatology Conference 2016.
    1. Ishikawa D, Takahashi M, Haga K, et al. . P074 microbial composition is effectively improved by combination therapy with fecal microbial transplantation and multiple antibiotics for ulcerative colitis. Inflamm Bowel Dis 2018;24(suppl_1):S27–S. 10.1093/ibd/izy019.082
    1. Bharat M, Curran J, Herfarth HH, et al. . 85 - SER-287, an Investigational Microbiome Therapeutic, Induces Remission and Endoscopic Improvement in a Placebo-Controlled, Double-Blind Randomized Trial in Patients with Active Mild-to-Moderate Ulcerative Colitis. Gastroenterology 2018;154:S-25–-20. 10.1016/S0016-5085(18)30561-4
    1. Simmons S, Diao L, O'Brien E, et al. . Tu2019 - Engraftment of Ser-287, an Investigational Microbiome Therapeutic, is Related to Clinical Remission in a Placebo-Controlled, Double-Blind Randomized Trial (Seres-101) in Patients with Active Mild to Moderate Ulcerative Colitis (UC). Gastroenterology 2018;154:S-1371–0. 10.1016/S0016-5085(18)34478-0
    1. Peng Z, Xiang J, He Z, et al. . Colonic transendoscopic enteral tubing: a novel way of transplanting fecal microbiota. Endoscopy International Open 2016;04:E610–E613. 10.1055/s-0042-105205
    1. Ding X, Li Q, Li P, et al. . Long-Term safety and efficacy of fecal microbiota transplant in active ulcerative colitis. Drug Saf 2019;42:869–80. 10.1007/s40264-019-00809-2
    1. Wang JW, Wang YK, Zhang F, et al. . Initial experience of fecal microbiota transplantation in gastrointestinal disease: a case series. Kaohsiung J Med Sci 2019.
    1. Kao D, Roach B, Silva M, et al. . Effect of Oral Capsule– vs Colonoscopy-Delivered Fecal Microbiota Transplantation on Recurrent Clostridium difficile Infection. JAMA 2017;318:1985–93. 10.1001/jama.2017.17077
    1. Youngster I, Mahabamunuge J, Systrom HK, et al. . Oral, frozen fecal microbiota transplant (FMT) capsules for recurrent Clostridium difficile infection. BMC Med 2016;14:134 10.1186/s12916-016-0680-9
    1. Staley C, Hamilton MJ, Vaughn BP, et al. . Successful resolution of recurrent Clostridium difficile infection using freeze-dried, encapsulated fecal microbiota; pragmatic cohort study. American Journal of Gastroenterology 2017;112:940–7. 10.1038/ajg.2017.6
    1. Lee CH, Steiner T, Petrof EO, et al. . Frozen vs Fresh Fecal Microbiota Transplantation and Clinical Resolution of Diarrhea in Patients With Recurrent Clostridium difficile Infection. JAMA 2016;315:142–9. 10.1001/jama.2015.18098
    1. Ramai D, Zakhia K, Ofosu A, et al. . Fecal microbiota transplantation: donor relation, fresh or frozen, delivery methods, cost-effectiveness. Ann Gastroenterol 2019;32:30–8. 10.20524/aog.2018.0328
    1. Jiang Z-D, Alexander A, Ke S, et al. . Stability and efficacy of frozen and lyophilized fecal microbiota transplant (FMT) product in a mouse model of Clostridium difficile infection (CDI). Anaerobe 2017;48:110–4. 10.1016/j.anaerobe.2017.08.003
    1. Jiang ZD, Ajami NJ, Petrosino JF, et al. . Randomised clinical trial: faecal microbiota transplantation for recurrent Clostridum difficile infection - fresh, or frozen, or lyophilised microbiota from a small pool of healthy donors delivered by colonoscopy. Aliment Pharmacol Ther 2017;45:899–908. 10.1111/apt.13969
    1. Cui B, Feng Q, Wang H, et al. . Fecal microbiota transplantation through mid-gut for refractory Crohn's disease: safety, feasibility, and efficacy trial results. J Gastroenterol Hepatol 2015;30:51–8. 10.1111/jgh.12727
    1. Fang H, Fu L, Wang J. Protocol for fecal microbiota transplantation in inflammatory bowel disease: a systematic review and meta-analysis. Biomed Res Int 2018;2018:8941340–11. 10.1155/2018/8941340
    1. Kao DH, Roach B, Walter J, et al. . A51 effect of lyophilized sterile fecal filtrate vs lyophilized donor stool on recurrent Clostridium difficile infection (RCDI): preliminary results from a randomized, double-blind pilot study. J Can Assoc Gastroenterol 2019;2(Supplement_2):101–2. 10.1093/jcag/gwz006.050
    1. Khanna S. Microbiota replacement therapies: innovation in gastrointestinal care. Clin Pharmacol Ther 2018;103:102–11. 10.1002/cpt.923
    1. Kelly CR, Khoruts A, Staley C, et al. . Effect of Fecal Microbiota Transplantation on Recurrence in Multiply Recurrent Clostridium difficile Infection. Ann Intern Med 2016;165:609–16. 10.7326/M16-0271
    1. Chu ND, Smith MB, Perrotta AR, et al. . Profiling living bacteria informs preparation of fecal microbiota transplantations. PLoS One 2017;12:e0170922 10.1371/journal.pone.0170922
    1. Gough E, Shaikh H, Manges AR. Systematic review of intestinal microbiota transplantation (fecal bacteriotherapy) for recurrent Clostridium difficile infection. Clin Infect Dis 2011;53:994–1002. 10.1093/cid/cir632
    1. Ianiro G, Maida M, Burisch J, et al. . Efficacy of different faecal microbiota transplantation protocols for Clostridium difficile infection: A systematic review and meta-analysis. United European Gastroenterol J 2018;6:1232–44. 10.1177/2050640618780762
    1. Allegretti JR, Fischer M, Sagi SV, et al. . Fecal microbiota transplantation capsules with targeted colonic versus gastric delivery in recurrent Clostridium difficile infection: a comparative cohort analysis of high and lose dose. Dig Dis Sci 2019;64:1672–8. 10.1007/s10620-018-5396-6
    1. Jørgensen SMD, Hansen MM, Erikstrup C, et al. . Faecal microbiota transplantation: establishment of a clinical application framework. Eur J Gastroenterol Hepatol 2017;29:e36–45. 10.1097/MEG.0000000000000958
    1. Zhang F, Zhang T, Zhu H, et al. . Evolution of fecal microbiota transplantation in methodology and ethical issues. Curr Opin Pharmacol 2019;49:11–16. 10.1016/j.coph.2019.04.004 10.1016/j.coph.2019.04.004
    1. Khanna S, Pardi DS, Kelly CR, et al. . A Novel Microbiome Therapeutic Increases Gut Microbial Diversity and Prevents Recurrent Clostridium difficile Infection. J Infect Dis. 2016;214:173–81. 10.1093/infdis/jiv766
    1. Jiang Z-D, Jenq RR, Ajami NJ, et al. . Safety and preliminary efficacy of orally administered lyophilized fecal microbiota product compared with frozen product given by enema for recurrent Clostridium difficile infection: a randomized clinical trial. PLoS One 2018;13:e0205064 10.1371/journal.pone.0205064
    1. Blount K, Gerding DN, Khanna S, et al. . 624 – comparison of two open-label trials demonstrates similar 6-month outcomes for Rbx2660 and Rbx7455—Investigational microbiota restoration therapeutics administered by enema or oral capsules for preventing recurrent Clostridium difficile infections. Gastroenterology 2019;156:S-130–S-131. 10.1016/S0016-5085(19)37115-X
    1. Diao L, Nnamani MC, OBrien E, et al. . 623 – Ser-287, an investigational microbiome therapeutic, induces widespread transcriptional changes related to clinical remission in a placebo-controlled, double-blind randomized trial (Seres-101) in patients with active mild-to-moderate ulcerative colitis. Gastroenterology 2019;156:S-130 10.1016/S0016-5085(19)37114-8

Source: PubMed

3
Předplatit