1,868
Views
5
CrossRef citations to date
0
Altmetric
Editorial

Just another crappy commentary: the future of fecal microbiota transplantation

Pages 987-989 | Received 11 Jul 2019, Accepted 13 Aug 2019, Published online: 08 Oct 2019

The journey through life is laden with ironies.

Table 1. Proposed indications for FMT based on a search of Clinicaltrials.gov

But perhaps the greatest irony of all is that feces, which are disgusting to virtually everyone, are also absolutely vital to life. Fecal microbiota transplant (FMT) has proven to be a lifesaving technique for Clostridia difficile infection [Citation1,Citation2]. And it might have therapeutic value for a host of other incompletely treated conditions. A search of the website, Clinicaltrials.gov, on 27 May 2019 with the keywords, fecal transplant, found listings for 307 trials. Pubmed lists nearly 2000 manuscripts on this topic. Quite a few physicians and scientists apparently now accept that disgusting and lifesaving are not mutually exclusive terms.

Joshua Lederberg coined the term, microbiome, for the microbes living in and on each of us [Citation3]. In addition to bacteria, the microbiome includes viruses, fungi, and sometimes other organisms such as helminths. We now appreciate that the microbiome is essential for health. Our microbes are a source of vitamins and neurotransmitters. Most importantly, our microbes educate our immune system. Mice which are raised in a germ free environment have shrunken lymphoid organs and an impaired immune response [Citation4]. We should not forget that our gut microbiome metabolizes many of the medications that we ingest [Citation5,Citation6]. The microbiome even alters the efficacy of anti-cancer therapy, such as checkpoint inhibitors [Citation7,Citation8]. Anyone who has ever taken an antibiotic knows that drugs alter bugs, but the range of drugs altering bugs is being increasingly explored. And we are just starting to explore how bugs alter drugs. Acetaminophen was one of the first recognized examples [Citation5], but many more have followed [Citation6].

Medical literature includes reports on FMT for C. difficile colitis [Citation1] and other refractory bowel infections [Citation9], ulcerative colitis [Citation10,Citation11] Crohn’s disease [Citation12], irritable bowel syndrome [Citation13,Citation14], sclerosing cholangitis [Citation15], constipation [Citation16], cirrhosis [Citation17], hepatic encephalopathy [Citation18,Citation19], psoriatic arthritis [Citation20], graft versus host disease [Citation21], diabetic neuropathy [Citation22] and metabolic syndrome [Citation23Citation25], although data are sparse and sometimes negative or inconclusive for some of these indications. Additional indications are under study or were under study based on listings with clinicaltrials.gov and as shown in .

While FMT is a movement with tremendous momentum (pun intended), we are lacking many answers. What is the best route (e.g. oral, rectal, jejeunal) to deliver FMT? (For C. difficile infection, delivery by colonoscopy appears to be superior) [Citation2]. How frequently must FMT be repeated? How durable is the effect? Can FMT be used as a strategy for prevention rather than treatment? Does pretreatment with antibiotics help or hinder the engraftment of FMT? Are there risks from future antibiotics? How safe is FMT? Could there be unanticipated long-term side effects? Not all healthy donors are equally effective for specific diseases; more work needs to be done to identify the bacteria or ratio of bacteria that result in benefit. In addition, recently Khoruts and Brandt appropriately contended that intestinal microbiota transplantation might be a more appropriate designation for this therapeutic approach [Citation26].

FMT is invaluable as a proof of concept regarding the microbiome’s role in health and disease, but it is clearly not the ultimate approach to treat microbiome-driven diseases. One obvious problem is the esthetics of fecal transplant. But there are also dangers. FMT can certainly transmit viral diseases [Citation27] and in theory, it could transmit prions or other incompletely detected infectious vectors. On 13 June 2019, the New York Times reported that the Food and Drug Administration was investigating an FMT-related death from E. Coli sepsis in an immunocompromised host in a trial in which guidelines were not followed. (https://www.nytimes.com/2019/06/13/health/fecal-transplant-fda.html). As a consequence, some FMT clinical trials were being halted. It should also be emphasized that some stool banks screen more rigorously than others for disease. Data support the safety of FMT even in immunocompromised hosts [Citation28].

The mechanisms by which FMT could restore health are multiple. In the case of C. difficile, FMT might simply restore a healthy gut ecosystem such that the pathogen no longer has a major competitive advantage. In obesity, gut bacteria affect the metabolism of ingested food and therefore have a profound effect on the availability of food derived calories. But bacteria could also affect neurotransmitters and thus have a fundamental effect on appetite. Bacterial mimicry of self-antigens, enhanced bowel permeability, changes in immune homeostasis such as a reduction in regulatory T cells or an expansion of TH17 T cells, and activation of lymphocytes or other immune cells from the lamina propria to remote organs are additional explanations as to how the intestinal microbiome contributes to health or disease [Citation29].

If the gut microbiome is vital to health, but FMT is not likely to be the ultimate solution, what alternatives exist? Rather than extracts of feces, we could deliver a defined consortium of bacteria. Such an approach would provide for greater uniformity, a critical component for judging efficacy, and could easily avoid unknown pathogens. We can also alter the microbiome through diet [Citation30]. The ecosystem represented by gut bacteria is greatly influenced by the substrates provided by what we ingest, although diet is complex and the impact of dietary changes is incompletely defined. If a disease is driven by a single microbe, vaccination against that microbe could have beneficial effects. Such an approach has proven successful in a mouse model of systemic lupus [Citation31]. The mechanisms by which the gut microbiome could cause disease are multiple. One frequently invoked mechanism is an increase in bowel permeability leading to dispersion of bacterial products. If this mechanism does indeed contribute, then a strategy that repairs the leakiness of the bowel could be effective without any knowledge of specific bacteria. Finally, we might not need to know which bacteria are responsible for disease. If we understand metabolically how the bacteria are contributing to disease, blocking of a specific bacterial enzyme could be salutary. Such a paradigm has been described in laboratory animals to prevent bacteria from generating Trimethylamine-N-oxides (TMAOs) which are atherogenic [Citation32,Citation33].

Considering that the efforts to catalog our microbiome through the Human Microbiome Project in the US [Citation34] and the Meta Hit Project in Europe [Citation35] are arguably still in their infancy, it is remarkable how rapidly interest in fecal transplantation has grown.

Having a crappy day truly has found a new meaning because crappy can be life-saving.

Declaration of interest

JT Rosenbaum is a collaborator with Viome. He also consults for Abbvie, Santen, Gilead, Roche, Novartis, Janssen, Corvus, Celldex, and Horizon on topics not related to this commentary. The author receives royalties from UpToDate. OHSU receives clinical trial support funds from Pfizer for a trial conducted by the author and not related to this submission. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Reviewer disclosures

One of the peer reviewers has declared research support from Finch/Crestovo for research in Intestinal Microbiota Transplantation, other research support from Roche, Takeda, Celgene and Diasorin, and speaking/consulting honoraria from Abbvie. Peer reviewers on this manuscript have no other relevant financial relationships or otherwise to disclose.

Additional information

Funding

This work was supported by NIH Grant EY029266, the Grandmaison Fund for Autoimmunity Research, the William and Mary Bauman Foundation, Research to Prevent Blindness, the Rheumatology Research Foundation and the Stan and Madelle Rosenfeld Family Trust.

References

  • Juul FE, Garborg K, Bretthauer M, et al. Fecal microbiota transplantation for primary clostridium difficile infection. N Engl J Med. 2018;378(26):2535–2536. Epub 2018/06/05. PubMed PMID: 29860912.
  • 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(8):1232–1244. Epub 2018/ 10/06. PubMed PMID: 30288286; PMCID: PMC6169051.
  • Lederberg J. ‘Ome Sweet’ Omics: A geneological treasury of words. The Scientist. 2001.
  • Bauer H, Horowitz RE, Levenson SM, et al. The response of the lymphatic tissue to the microbial flora. Studies on germfree mice. Am J Pathol. 1963;42:471–483. Epub 1963/04/01. PubMed PMID: 13966929; PMCID: PMC1949649.
  • Clayton TA, Baker D, Lindon JC, et al. Pharmacometabonomic identification of a significant host-microbiome metabolic interaction affecting human drug metabolism. Proc Natl Acad Sci U S A. 2009;106(34):14728–14733. Epub 2009/08/12. PubMed PMID: 19667173; PMCID: PMC2731842.
  • Zimmermann M, Zimmermann-Kogadeeva M, Wegmann R, et al. Separating host and microbiome contributions to drug pharmacokinetics and toxicity. Science. 2019;363(6427). Epub 2019/02/09. PubMed PMID: 30733391; PMCID: PMC6533120. DOI:10.1126/science.aat9931
  • Wang Y, Wiesnoski DH, Helmink BA, et al. Fecal microbiota transplantation for refractory immune checkpoint inhibitor-associated colitis. Nat Med. 2018; 12(24):1804–1808. PubMed PMID: 30420754; PMCID: PMC6322556. Epub 2018/11/14.
  • Routy B, Le Chatelier E, Derosa L, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science. 2018;359(6371):91–97. Epub 2017/ 11/04. PubMed PMID: 29097494.
  • Dinh A, Fessi H, Duran C, et al. Clearance of carbapenem-resistant Enterobacteriaceae vs vancomycin-resistant enterococci carriage after faecal microbiota transplant: a prospective comparative study. J Hosp Infect. 2018;99(4):481–486. Epub 2018/ 02/27. PubMed PMID: 29477634.
  • Costello SP, Soo W, Bryant RV, et al. Systematic review with meta-analysis: faecal microbiota transplantation for the induction of remission for active ulcerative colitis. Aliment Pharmacol Ther. 2017;46(3):213–224. Epub 2017/ 06/15. PubMed PMID: 28612983.
  • Ding X, Li Q, Li P, et al. Long-term safety and efficacy of fecal microbiota transplant in active ulcerative colitis. Drug Saf 2019. Epub 2019/04/12. PubMed PMID: 30972640. DOI: 10.1007/s40264-019-00809-2.
  • Li P, Zhang T, Xiao Y, et al. Timing for the second fecal microbiota transplantation to maintain the long-term benefit from the first treatment for Crohn’s disease. Appl Microbiol Biotechnol. 2019;103(1):349–360. Epub 2018/ 10/26. PubMed PMID: 30357440; PMCID: PMC6311185. DOI: 10.1007/s00253-018-9447-x.
  • Huang HL, Chen HT, Luo QL, et al. Relief of irritable bowel syndrome by fecal microbiota transplantation is associated with changes in diversity and composition of the gut microbiota. J Dig Dis. 2019. Epub 2019/05/10. PubMed PMID: 31070838. DOI:10.1111/1751-2980.12756.
  • Ianiro G, Eusebi LH, Black CJ, et al. Systematic review with meta-analysis: efficacy of faecal microbiota transplantation for the treatment of irritable bowel syndrome. Aliment Pharmacol Ther. 2019;50(3):240–248. Epub 2019/ 05/29. PubMed PMID: 31136009.
  • Allegretti JR, Kassam Z, Carrellas M, et al. Fecal microbiota transplantation in patients with primary sclerosing cholangitis: a pilot clinical trial. Am J Gastroenterol. 2019. Epub 2019/ 02/08. PubMed PMID: 30730351. DOI:10.14309/ajg.0000000000000115.
  • Ohara T. Identification of the microbial diversity after fecal microbiota transplantation therapy for chronic intractable constipation using 16s rRNA amplicon sequencing. PLoS One. 2019;14(3):e0214085. Epub 2019/03/20. PubMed PMID: 30889205; PMCID: PMC6424423.
  • Bajaj JS, Kakiyama G, Savidge T, et al. Antibiotic-associated disruption of microbiota composition and function in cirrhosis is restored by fecal transplant. Hepatology. 2018;68(4):1549–1558. Epub 2018/04/18. PubMed PMID: 29665102.
  • Bajaj JS, Salzman NH, Acharya C, et al. Fecal microbial transplant capsules are safe in hepatic encephalopathy: a phase 1, randomized, placebo-controlled trial. Hepatology. 2019 PubMed PMID: 31038755. Epub 2019/ 05/01. DOI:10.1002/hep.30690.
  • Mehta R, Kabrawala M, Nandwani S, et al. Preliminary experience with single fecal microbiota transplant for treatment of recurrent overt hepatic encephalopathy-A case series. Indian J Gastroenterol. 2018;37(6):559–562. Epub 2018/11/27. PubMed PMID: 30474827.
  • Kragsnaes MS, Kjeldsen J, Horn HC, et al. Efficacy and safety of faecal microbiota transplantation in patients with psoriatic arthritis: protocol for a 6-month, double-blind, randomised, placebo-controlled trial. BMJ Open. 2018;8(4):e019231. Epub 2018/04/29. PubMed PMID: 29703851; PMCID: PMC5922473.
  • Kakihana K, Fujioka Y, Suda W, et al. Fecal microbiota transplantation for patients with steroid-resistant acute graft-versus-host disease of the gut. Blood. 2016;128(16):2083–2088. Epub 2016/07/28. PubMed PMID: 27461930; PMCID: PMC5085256.
  • Cai TT, Ye XL, Yong HJ, et al. Fecal microbiota transplantation relieve painful diabetic neuropathy: A case report. Medicine (Baltimore). 2018;97(50):e13543. Epub 2018/ 12/19. PubMed PMID: 30558014; PMCID: PMC6320208.
  • Vrieze A, Van Nood E, Holleman F, et al. Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Gastro. 2012;143(4):913–6 e7. Epub 2012/06/26. PubMed PMID: 22728514.
  • Kootte RS, Levin E, Salojarvi 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(4):611–9 e6. Epub 2017/10/06. PubMed PMID: 28978426.
  • Smits LP, Kootte RS, Levin E, et al. Effect of vegan fecal microbiota transplantation on carnitine- and choline-derived trimethylamine-n-oxide production and vascular inflammation in patients with metabolic syndrome. J Am Heart Assoc. 2018;7(7). Epub 2018/03/28. PubMed PMID: 29581220; PMCID: PMC5907601. DOI:10.1161/JAHA.117.008342
  • Khoruts A, Brandt LJ. Fecal microbiota transplant: a rose by any other name. Am J Gastroenterol. 2019;114(7):1176. Epub 2019/ 06/18. PubMed PMID: 31205129.
  • Dailey FE, Turse EP, Daglilar E, et al. The dirty aspects of fecal microbiota transplantation: a review of its adverse effects and complications. Curr Opin Pharmacol. 2019;49:29–33. Epub 2019/05/20. PubMed PMID: 31103793.
  • Kelly CR, Ihunnah C, Fischer M, et al. Fecal microbiota transplant for treatment of Clostridium difficile infection in immunocompromised patients. Am J Gastroenterol. 2014;109(7):1065–1071. Epub 2014/06/04. PubMed PMID: 24890442; PMCID: PMC5537742.
  • Rosenbaum JT, Asquith M, et al. The microbiome and HLA-B27-associated acute anterior uveitis. Nat Rev Rheumatol. 2018;14:704–713. Epub 2018/10/12. PubMed PMID: 30301938.
  • Bibbo S, Ianiro G, Giorgio V, et al.. The role of diet on gut microbiota composition. Eur Rev Med Pharmacol Sci. 2016;20(22): 4742–4749. Epub 2016/12/03. . PubMed PMID: 27906427
  • Manfredo Vieira S, Hiltensperger M, Kumar V, et al. Translocation of a gut pathobiont drives autoimmunity in mice and humans. Science. 2018;359(6380):1156–1161. Epub 2018/03/29. PubMed PMID: 29590047. DOI: 10.1126/science.aar7201
  • Roberts AB, Gu X, Buffa JA, et al. Development of a gut microbe-targeted nonlethal therapeutic to inhibit thrombosis potential. Nat Med. 2018;24(9):1407–1417. Epub 2018/08/08. PubMed PMID: 30082863; PMCID: PMC6129214.
  • Zhuang R, Ge X, Han L, et al. Gut microbe-generated metabolite trimethylamine N-oxide and the risk of diabetes: A systematic review and dose-response meta-analysis. Obes Rev. 2019;20(6):883–894. Epub 2019/ 03/15. PubMed PMID: 30868721.
  • Turnbaugh PJ, Ley RE, Hamady M, et al. The human microbiome project. Nature. 2007;449(7164):804–810. Epub 2007/ 10/19. PubMed PMID: 17943116; PMCID: 3709439.
  • Qin J, Li R, Raes J, et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010;464(7285):59–65. PubMed PMID: 20203603; PMCID: 3779803. .

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.