Register      Login
Microbiology Australia Microbiology Australia Society
Microbiology Australia, bringing Microbiologists together
RESEARCH ARTICLE

Faecal microbiota transplantation: a review

Holly A Sinclair A C and Paul Chapman A B
+ Author Affiliations
- Author Affiliations

A Infectious Diseases Department, Royal Brisbane and Women’s Hospital, Herston, Qld, Australia

B QIMR Berghofer Medical Research Institute, Herston, Qld, Australia

C Tel.: +61 7 3646 8111, Email: holly.sinclair@health.qld.gov.au

Microbiology Australia 41(2) 65-69 https://doi.org/10.1071/MA20019
Published: 21 May 2020

Abstract

Faecal microbiota transplantation (FMT) is the transfer of human faeces from a healthy donor to a recipient with a disease associated with gut dysbiosis. Here we review faecal microbiota transplantation as a treatment for Clostridioides difficile infection (CDI) and other conditions including decolonisation of multiresistant organisms. Donor selection and screening, adverse events, processing, administration and regulation of FMT are discussed.

Introduction

Faecal microbiota transplantation (FMT) is not a new concept, being first described in traditional Chinese medicine over 1000 years ago1. FMT delivered by faecal enema was successfully used in the treatment of pseudomembranous enterocolitis in 19582. A timeline for FMT over the years is shown in Figure 1. FMT is now accepted to be the most effective treatment for recurrent or refractory Clostridioides difficile infection (CDI). Clinical trials have also been conducted using FMT in primary sclerosing cholangitis, non-alcoholic steatohepatitis, type II diabetes mellitus, irritable bowel syndrome, inflammatory bowel disease, hepatic encephalopathy, and eradication of multiresistant organisms3.

Perturbations in the composition of intestinal microbiota occur after administration of antibiotics, other medications, dietary changes and travel. Antibiotic exposure decreases the alpha diversity with reduction in Firmicutes and Bacteroidetes phyla and proliferation of Proteobacteria including Enterobacteriaceae4. Following FMT there is reduction in Proteobacteria and expansion of Firmucutes, Ruminococcaceae, Lachnospiraceae, Clostridiaceae and Bacteroidetes4. Recipient microbiota engraftment has been demonstrated by day three after FMT5. This microbial community correlates with that of the donor’s microbial community and has been observed to be stable for 4 months and up to one year46. Complete donor engraftment may not be necessary if functionally effective taxa are present and bacteria associate with secondary bile acid metabolism to provide resistance to recurrent infection4.


C. difficile infection (CDI) and FMT

C. difficile is a Gram-positive anaerobic, spore forming and toxin-producing bacillus1. Spores are transmitted via the faecal–oral route and are an important cause of hospital-acquired infection. Between 15–70% of infants and 5% of adults are colonised, being more frequent in hospital and nursing home residents1.

Antibiotic exposure, older age and hospitalisation are major risk factors for CDI1. Clinical spectrum spans diarrhoea, ileus and toxic megacolon, with severe CDI presenting with fever, haemodynamic instability and peritonitis. Recurrent CDI is classified as recurrence of CDI within 8 weeks of successful treatment and refractory CDI is defined as absent clinical improvement after 3–4 days of appropriate treatment7.

FMT has been shown to be the most effective treatment for recurrent CDI and has repeatedly demonstrated superiority to comparators since the first randomised trial in 20138,9. In a meta-analysis of seven randomised controlled trials and 30 case series, FMT was more effective than vancomycin (RR: 0.23) for recurrent and refractory CDI with clinical resolution rates of 92%10.

The Australasian Society for Infectious Diseases published guidelines for management of CDI that includes FMT7. Australian therapeutic guidelines recommend FMT as preferred treatment for second and subsequent recurrences or ongoing refractory disease11. This is similar to American, European and British guidelines1214.


Adverse events

In general, FMT is considered a safe procedure with rare adverse events. Some of the common adverse effects include fever, abdominal pain, bloating and alteration to bowel habits15,16. Procedural complications include bowel perforation and mucosal tears15,16. Infectious complications including transmission of norovirus, Gram-negative bacteraemia and transmission of multiresistant organisms have been reported15. Deaths have been due to polymicrobial bacteremia in the setting of toxic megacolon, aspiration pneumonia as a complication of anaesthesia during colonoscopic FMT and regurgitation of faeculant material during endoscopic FMT1518. Donor stool screening for multiresistant organisms is now mandatory following two cases of donor derived Escherichia coli Extended Spectrum Beta-Lactamase bacteraemia, resulting in the death of one patient19. The United States Food and Drug Administration (FDA) have recently issued a safety alert regarding FMT after cases of enteropathogenic E. coli (EPEC) and Shigatoxin-producing E. coli (STEC) infection in recipients possibly linked to a stool bank (www.fda.gov).

Food allergy with anaphylaxis is a contraindication to FMT12. FMT should be offered with caution in patients with decompensated chronic liver disease or immunosuppression and special consideration to donor screening (for CMV, EBV and Strongyloides) should be given for immunosuppressed recipients12. Elderly and debilitated patients have been treated with FMT for CDI with success, however they may have a lower primary cure rate and higher recurrence rate compared to a younger cohort18,20. Adverse events in the elderly population have included aspiration; therefore the colonoscopy route has been suggested as the preferred route of administration18.

Limited data exists on long-term adverse effects. Jalanka et al.21 found no difference in incidence of severe diseases or weight gain after 3.8 years of FMT and improved bowel habits and mental health were reported.


Donors

Traditionally, donors known to the patient were selected, however this could result in ethical and confidentiality issues if identifying a disease in the donor or a transmission event to recipient22. Alternatively, FMT is best sourced from a centralised stool bank from healthy unrelated donors12. Donors should be between 18 and 60 years of age and BMI between 18 and 30 kg/m3 12,23. Donors are screened with a questionnaire followed by blood and stool testing with recommendations in Table 1. Woodworth et al.3 recommend screening for carbapenem resistant Enterobacteriaceae, vancomycin resistant Enterococci and those with frequent contact with health care should be excluded. The risk of transmission of non-communicable diseases remains unknown; therefore, donors with cardiovascular disease, stroke, diabetes mellitus, obesity, metabolic syndrome and malnutrition are excluded3.


Table 1.  Example of donor questionnaire and donor blood and stool testing.
Click to zoom


Processing and preparation: impact on efficiency

Stool should be processed within 6 hours of defaecation. FMT material prepared in aerobic conditions has been effective for the treatment of recurrent C. difficile associated diarrhea8. However, ambient air exposure impacts on viable bacterial composition particularly for oxygen sensitive species24. Processing stool in an anaerobic chamber allows preservation of commensal species24. Freezing reduces the overall viability but the microbiota composition is not significantly different to fresh specimens24, with viable bacteria remaining after 6 months of frozen storage in 10% glycerol22,25 and no difference in FMT efficacy observed when used for CDI10.

There are a number of preparations for FMT including fresh, frozen and encapsulated faecal suspensions. Encapsulated freeze-dried preparations had 88% clinical success (49 patients) with no recurrence over two months26. In a randomised study of 72 patients with recurrent CDI, cure rates were highest for fresh faeces (100%), lowest for lyophilized product (78%; P = 0.022 vs fresh) and intermediate for frozen product (83%; P = 0.233 vs fresh)27. CDI recurrence was prevented in 84% receiving oral lyophilized microbiota capsules compared to 88% with FMT by enema (P = 0.74)28. In a non-inferiority randomised trial there was no difference after single treatment with capsule or colonoscopy delivery (both 96.2% without recurrent CDI at 12 weeks)29.


Administration procedure: impact on efficiency

Bowel lavage is administered prior to FMT particularly for the lower gastrointestinal route. There should be minimum 24 hours free from antibiotics before FMT and at least 72 hours after FMT12. FMT can be delivered to upper (nasogastric, nasoduodenal or nasojejunal tube or upper endoscopy) or lower gastrointestinal tract (colonoscopic administration to caecum or terminal ileum or enema if not possible). Ianiro et al.30 conducted a systematic review and meta-analysis of fifteen studies on different protocols of FMT for CDI. Multiple infusions increased efficacy compared to single infusion (93% vs 76%)30. Duodenal delivery had lower efficacy (P = 0.039) and colonoscopy had higher efficacy rates (P = 0.006). Lower faecal amount (≤50g) and enema had lower efficacy rates after single infusion30. Another meta-analysis also demonstrated administration by lower gastrointestinal route was more effective (95%) compared to upper gastrointestinal delivery (85%) with no difference between fresh or frozen FMT10. Consecutive courses after failure of first FMT showed incremental effect10.


FMT services, stool banking and regulation

Historically, FMT has been performed with varying levels of sophistication across Australia, ranging from the ad hoc and infrequent preparation of fresh FMT material for recurrent CDI to specialised centres operating stool banks, such as the Biomebank (Adelaide, SA) and the Centre for Digestive Diseases (Sydney, NSW). In September 2019, the Australian Minister for Health determined that supply of faecal microbiota transplant products be regulated by the Therapeutic Goods Administration (TGA). The new regulatory model classifies most FMT products as class 1 or 2 biologicals depending on the extent of manipulation and whether manufactured in a hospital and used onsite. A Draft Standards for Faecal Microbiota Transplant Products is available with finalised FMT regulatory requirements expected in early 2020 (www.tga.gov.au). The American Gastroenterological Association (AGA) has proposed an FMT National Registry to collect outcomes to assess short- and long-term safety and effectiveness and current practices31. An international consensus on stool banking for FMT in clinical practice is available32. There are now Australian consensus statements for the regulation, production and use of FMT in clinical practice23.


FMT for decolonisation of multiresistant organisms and treatment of other conditions

Small sample studies have shown that FMT was effective in reducing the number of antibiotic resistance genes in patients’ resistome33. Huttner et al.34 hypothesised that decolonisation could be achieved with oral antibiotics (colistin and neomycin) followed by recolonisation to restore intestinal microbiota. The results were only slightly in favour of the intervention group (OR 1.7). Nine uncontrolled studies with heterogeneity have evaluated the use of FMT for multidrug resistant Gram-negative bacteria decolonisation. However, the European guidelines suggest there is insufficient evidence for or against FMT in this context35. Similarly, UK guidelines do not recommend FMT as treatment for inflammatory bowel disease or other gastrointestinal or non-gastrointestinal disease12. Australian guidelines suggest FMT has been shown to be successful in induction therapy for mild to moderate ulcerative colitis however more studies are required before it can be implemented into standard care23. This is a developing research field and future treatment of conditions with FMT will be seen in the future.


Conflicts of interest

The authors declare no conflicts of interest.



Figure 1.  Timeline for faecal microbiota transplantation.
Click to zoom



Acknowledgements

This research did not receive any specific funding.


References

[1]  Czepiel, J. et al. (2019) Clostridium difficile infection Eur. J. Clin. Microbiol. Infect. Dis. 38, 1211–1221.
Clostridium difficile infectionCrossref | GoogleScholarGoogle Scholar | 30945014PubMed |

[2]  Eiseman, B. et al. (1958) Fecal enema as an adjunct in the treatment of pseudomembranous enterocolitis. Surgery 44, 854–859.
| 13592638PubMed |

[3]  Woodworth, M.H. et al. (2017) Challenges in fecal donor selection and screening for fecal microbiota transplantation: a review. Gut Microbes 8, 225–237.
Challenges in fecal donor selection and screening for fecal microbiota transplantation: a review.Crossref | GoogleScholarGoogle Scholar | 28129018PubMed |

[4]  Staley, C. et al. (2016) Complete microbiota engraftment is not essential for recovery from recurrent Clostridium difficile infection following fecal microbiota transplantation. MBio 7, e01965-16.
Complete microbiota engraftment is not essential for recovery from recurrent Clostridium difficile infection following fecal microbiota transplantation.Crossref | GoogleScholarGoogle Scholar | 27999162PubMed |

[5]  Shankar, V. et al. (2014) Species and genus level resolution analysis of gut microbiota in Clostridium difficile patients following fecal microbiota transplantation. Microbiome 2, 13.
Species and genus level resolution analysis of gut microbiota in Clostridium difficile patients following fecal microbiota transplantation.Crossref | GoogleScholarGoogle Scholar | 24855561PubMed |

[6]  Jalanka, J. et al. (2016) Long-term effects on luminal and mucosal microbiota and commonly acquired taxa in faecal microbiota transplantation for recurrent Clostridium difficile infection. BMC Med. 14, 155.
Long-term effects on luminal and mucosal microbiota and commonly acquired taxa in faecal microbiota transplantation for recurrent Clostridium difficile infection.Crossref | GoogleScholarGoogle Scholar | 27724956PubMed |

[7]  Trubiano, J.A. et al. (2016) Australasian Society of Infectious Diseases updated guidelines for the management of Clostridium difficile infection in adults and children in Australia and New Zealand. Intern. Med. J. 46, 479–493.
Australasian Society of Infectious Diseases updated guidelines for the management of Clostridium difficile infection in adults and children in Australia and New Zealand.Crossref | GoogleScholarGoogle Scholar | 27062204PubMed |

[8]  van Nood, E. et al. (2013) Duodenal infusion of donor feces for recurrent Clostridium difficile. N. Engl. J. Med. 368, 407–415.
Duodenal infusion of donor feces for recurrent Clostridium difficile.Crossref | GoogleScholarGoogle Scholar | 23323867PubMed |

[9]  Cammarota, G. et al. (2015) Randomised clinical trial: faecal microbiota transplantation by colonoscopy vs. vancomycin for the treatment of recurrent Clostridium difficile infection. Aliment. Pharmacol. Ther. 41, 835–843.
Randomised clinical trial: faecal microbiota transplantation by colonoscopy vs. vancomycin for the treatment of recurrent Clostridium difficile infection.Crossref | GoogleScholarGoogle Scholar | 25728808PubMed |

[10]  Quraishi, M.N. et al. (2017) Systematic review with meta-analysis: the efficacy of faecal microbiota transplantation for the treatment of recurrent and refractory Clostridium difficile infection. Aliment. Pharmacol. Ther. 46, 479–493.
Systematic review with meta-analysis: the efficacy of faecal microbiota transplantation for the treatment of recurrent and refractory Clostridium difficile infection.Crossref | GoogleScholarGoogle Scholar | 28707337PubMed |

[11]  (2019) eTG complete. www.tg.org.au

[12]  Mullish, B.H. et al. (2018) 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. J. Hosp. Infect. 100, S1–S31.
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.Crossref | GoogleScholarGoogle Scholar | 30173851PubMed |

[13]  Cammarota, G. et al. (2017) European consensus conference on faecal microbiota transplantation in clinical practice. Gut 66, 569–580.
European consensus conference on faecal microbiota transplantation in clinical practice.Crossref | GoogleScholarGoogle Scholar | 28087657PubMed |

[14]  McDonald, L.C. et al. (2018) Clinical practice guidelines for Clostridium difficile infection in adults and children: 2017 update by the Infectious Diseases Society of America (IDSA) and Society for Healthcare Epidemiology of America (SHEA). Clin. Infect. Dis. 66, 987–994.
Clinical practice guidelines for Clostridium difficile infection in adults and children: 2017 update by the Infectious Diseases Society of America (IDSA) and Society for Healthcare Epidemiology of America (SHEA).Crossref | GoogleScholarGoogle Scholar | 29562266PubMed |

[15]  Baxter, M. and Colville, A. (2016) Adverse events in faecal microbiota transplant: a review of the literature. J. Hosp. Infect. 92, 117–127.
Adverse events in faecal microbiota transplant: a review of the literature.Crossref | GoogleScholarGoogle Scholar | 26803556PubMed |

[16]  van Beurden, Y.H. et al. (2017) Complications, effectiveness, and long term follow-up of fecal microbiota transfer by nasoduodenal tube for treatment of recurrent Clostridium difficile infection. United European Gastroenterol. J. 5, 868–879.
Complications, effectiveness, and long term follow-up of fecal microbiota transfer by nasoduodenal tube for treatment of recurrent Clostridium difficile infection.Crossref | GoogleScholarGoogle Scholar | 29026601PubMed |

[17]  Baxter, M. et al. (2015) Fatal aspiration pneumonia as a complication of fecal microbiota transplant. Clin. Infect. Dis. 61, 136–137.
Fatal aspiration pneumonia as a complication of fecal microbiota transplant.Crossref | GoogleScholarGoogle Scholar | 25805303PubMed |

[18]  Friedman-Korn, T. et al. (2018) Fecal transplantation for treatment of Clostridium difficile infection in elderly and debilitated patients. Dig. Dis. Sci. 63, 198–203.
Fecal transplantation for treatment of Clostridium difficile infection in elderly and debilitated patients.Crossref | GoogleScholarGoogle Scholar | 29134299PubMed |

[19]  DeFilipp, Z. et al. (2019) Drug-resistant E. coli bacteremia transmitted by fecal microbiota transplant. N. Engl. J. Med. 381, 2043–2050.
Drug-resistant E. coli bacteremia transmitted by fecal microbiota transplant.Crossref | GoogleScholarGoogle Scholar | 31665575PubMed |

[20]  Luo, Y. et al. (2020) Fecal microbiota transplantation for Clostridioides difficile in high-risk older adults is associated with early recurrence. Dig. Dis. Sci. , .
Fecal microbiota transplantation for Clostridioides difficile in high-risk older adults is associated with early recurrence.Crossref | GoogleScholarGoogle Scholar | 32078714PubMed |

[21]  Jalanka, J. et al. (2018) The long-term effects of faecal microbiota transplantation for gastrointestinal symptoms and general health in patients with recurrent Clostridium difficile infection. Aliment. Pharmacol. Ther. 47, 371–379.
The long-term effects of faecal microbiota transplantation for gastrointestinal symptoms and general health in patients with recurrent Clostridium difficile infection.Crossref | GoogleScholarGoogle Scholar | 29226561PubMed |

[22]  Costello, S.P. et al. (2016) Establishing a fecal microbiota transplant service for the treatment of Clostridium difficile infection. Clin. Infect. Dis. 62, 908–914.
Establishing a fecal microbiota transplant service for the treatment of Clostridium difficile infection.Crossref | GoogleScholarGoogle Scholar | 26628567PubMed |

[23]  Haifer, C. et al. (2020) Australian consensus statements for the regulation, production and use of faecal microbiota transplantation in clinical practice. Gut 69, 801–810.
Australian consensus statements for the regulation, production and use of faecal microbiota transplantation in clinical practice.Crossref | GoogleScholarGoogle Scholar | 32047093PubMed |

[24]  Papanicolas, L.E. et al. (2019) Bacterial viability in faecal transplants: which bacteria survive? EBioMedicine 41, 509–516.
Bacterial viability in faecal transplants: which bacteria survive?Crossref | GoogleScholarGoogle Scholar | 30796005PubMed |

[25]  Costello, S.P. et al. (2015) Faecal microbiota transplant for recurrent Clostridium difficile infection using long-term frozen stool is effective: clinical efficacy and bacterial viability data. Aliment. Pharmacol. Ther. 42, 1011–1018.
Faecal microbiota transplant for recurrent Clostridium difficile infection using long-term frozen stool is effective: clinical efficacy and bacterial viability data.Crossref | GoogleScholarGoogle Scholar | 26264455PubMed |

[26]  Staley, C. et al. (2017) Successful resolution of recurrent Clostridium difficile infection using freeze-dried, encapsulated fecal microbiota; pragmatic cohort study. Am. J. Gastroenterol. 112, 940–947.
Successful resolution of recurrent Clostridium difficile infection using freeze-dried, encapsulated fecal microbiota; pragmatic cohort study.Crossref | GoogleScholarGoogle Scholar | 28195180PubMed |

[27]  Jiang, Z.D. et al. (2017) 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. 45, 899–908.
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.Crossref | GoogleScholarGoogle Scholar | 28220514PubMed |

[28]  Jiang, Z.D. et al. (2018) 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 13, e0205064.
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.Crossref | GoogleScholarGoogle Scholar | 30388112PubMed |

[29]  Kao, D. et al. (2017) Effect of Oral capsule- vs colonoscopy-delivered fecal microbiota transplantation on recurrent Clostridium difficile infection: a randomized clinical trial. JAMA 318, 1985–1993.
Effect of Oral capsule- vs colonoscopy-delivered fecal microbiota transplantation on recurrent Clostridium difficile infection: a randomized clinical trial.Crossref | GoogleScholarGoogle Scholar | 29183074PubMed |

[30]  Ianiro, G. et al. (2018) Efficacy of different faecal microbiota transplantation protocols for Clostridium difficile infection: a systematic review and meta-analysis. United European Gastroenterol. J. 6, 1232–1244.
Efficacy of different faecal microbiota transplantation protocols for Clostridium difficile infection: a systematic review and meta-analysis.Crossref | GoogleScholarGoogle Scholar | 30288286PubMed |

[31]  Kelly, C.R. et al. (2017) The AGA’s Fecal Microbiota Transplantation National Registry: an important step toward understanding risks and benefits of microbiota therapeutics. Gastroenterology 152, 681–684.
The AGA’s Fecal Microbiota Transplantation National Registry: an important step toward understanding risks and benefits of microbiota therapeutics.Crossref | GoogleScholarGoogle Scholar | 28143773PubMed |

[32]  Cammarota, G. et al. (2019) International consensus conference on stool banking for faecal microbiota transplantation in clinical practice. Gut 68, 2111–2121.
International consensus conference on stool banking for faecal microbiota transplantation in clinical practice.Crossref | GoogleScholarGoogle Scholar | 31563878PubMed |

[33]  Laffin, M. et al. (2017) Fecal microbial transplantation as a therapeutic option in patients colonized with antibiotic resistant organisms. Gut Microbes 8, 221–224.
Fecal microbial transplantation as a therapeutic option in patients colonized with antibiotic resistant organisms.Crossref | GoogleScholarGoogle Scholar | 28059612PubMed |

[34]  Huttner, B.D. et al. (2019) A 5-day course of oral antibiotics followed by faecal transplantation to eradicate carriage of multidrug-resistant Enterobacteriaceae: a randomized clinical trial. Clin. Microbiol. Infect. 25, 830–838.
A 5-day course of oral antibiotics followed by faecal transplantation to eradicate carriage of multidrug-resistant Enterobacteriaceae: a randomized clinical trial.Crossref | GoogleScholarGoogle Scholar | 30616014PubMed |

[35]  Tacconelli, E. et al. (2019) ESCMID-EUCIC clinical guidelines on decolonization of multidrug-resistant Gram-negative bacteria carriers. Clin. Microbiol. Infect. 25, 807–817.
ESCMID-EUCIC clinical guidelines on decolonization of multidrug-resistant Gram-negative bacteria carriers.Crossref | GoogleScholarGoogle Scholar | 30708122PubMed |


Biographies

Dr Holly A Sinclair, BMedSc, MBBS(hons), is an infectious diseases and microbiology advanced trainee with the Royal Australasian College of Physicians and the Royal College of Pathologists of Australasia.

Dr Paul Chapman, MBBS, FRACP, MPHTM, is an infectious diseases physician with interests in hospital-acquired infection, parasitology, microbiome and device-related infections.