12,242
Views
68
CrossRef citations to date
0
Altmetric
Review

Efficacy and safety of fecal microbiota transplantation for the treatment of diseases other than Clostridium difficile infection: a systematic review and meta-analysis

, , ORCID Icon, , ORCID Icon, ORCID Icon, , , ORCID Icon, ORCID Icon & show all
Article: 1854640 | Received 09 Sep 2020, Accepted 11 Nov 2020, Published online: 19 Dec 2020

ABSTRACT

The intestinal microbiome has been identified as a key modifier for a variety of health conditions. Fecal Microbiota Transplantation (FMT) has emerged as a fast, safe, and effective means by which to modify the intestinal microbiome and potentially treat a variety of health conditions. Despite extensive research of FMT for CDI, there is a lack of clarity informed by systematic synthesis of data regarding the safety and efficacy of FMT for other health conditions. This systematic review used PRISMA guidelines and was prospectively registered with PROSPERO (CRD42018104243). In March 2020, a search of MEDLINE, EMBASE, and PsycINFO was conducted. We identified 26 eligible studies. A meta-analysis of FMT for active Ulcerative Colitis (UC) showed that FMT significantly improved rates of clinical remission (OR = 3.634, 95% CI = 1.940 to 6.808, I2 = 0%, p < .001), clinical response (OR = 2.634, 95% CI = 1.441 to 4.815, I2 = 33%, p = .002) and endoscopic remission (OR = 4.431, 95% CI = 1.901 to 10.324, I2 = 0%, p = .001). With respect to Irritable Bowel Syndrome, a meta-analysis showed no significant change in symptoms following FMT (p = .739). Hepatic disorders, metabolic syndrome, and antibiotic-resistant organisms were conditions with emerging data on FMT. Serious adverse events (AE) were more often reported in control group participants (n = 43) compared with FMT group participants (n = 26). There were similar rates of mild to moderate AE in both groups. Preliminary data suggest that FMT is a potentially safe, well-tolerated and efficacious treatment for certain conditions other than CDI, with evidence for active UC being the most compelling.

Introduction

The intestinal microbiome has emerged as a modifiable target for treating a variety of health conditions thought to be associated with dysregulated microbiome profiles.Citation1 The intestinal microbiome is believed to have a key role in modifying immunity, inflammation, and – by extension – a plethora of health conditions.Citation2–4 There is now substantial research interestCitation5 into interventions that might target the gut microbiome to improve chronic diseases, including diet, supplementary prebiotics, probiotics, antibiotics, short-chain fatty acids, and Fecal Microbiota Transplantation (FMT).Citation6,Citation7

FMT is a technique in which gut bacteria are transferred from a healthy donor to a patient, with the goal of introducing or restoring a stable microbial community in the gut. FMT has been established as an effective means of rapidly modifying the intestinal microbiota and may therefore have potential as a treatment for the many health conditions linked with the intestinal microbiome.Citation8 FMT is already widely practiced as a highly effective treatment for recurrent Clostridium difficile infection (CDI).Citation9,Citation10

A wealth of new research is investigating whether FMT may be used to treat other health conditions linked to the intestinal microbiome,Citation11,Citation12 including gastrointestinal,Citation13–17 autoimmune,Citation18,Citation19 metabolic,Citation20,Citation21 and neuropsychiatricCitation22–24 conditions. There is also promising preclinical evidence supporting the use of FMT in conditions other than CDI, including Major Depressive Disorder,Citation25,Citation26 schizophrenia,Citation27 and cardiometabolic syndrome.Citation28

While reviews of FMT for specific indications such as IBSCitation29–31 and IBD exist,Citation32–34 to date there have been no comprehensive reviews evaluating and synthesizing the entire body of data for both the efficacy and safety of FMT for all conditions other than CDI. This systematic review and meta-analysis addresses the question of whether FMT is safe and effective at treating health conditions other than CDI in humans.

Methods

Protocol and registration

The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines were adhered to as a methodological template for this review. The protocol for this Systematic Review was prospectively registered with PROSPERO (CRD42018104243).

Search strategy and eligibility criteria

The PICO approach (population, intervention, comparator, outcomes) was used to guide the search strategy for this review. The PICO criteria used are outlined below:

  • Population: Humans participants of any age with any acute or chronic health condition other than CDI. Studies were included only if participants were followed up for at least two weeks post-FMT.

  • Intervention: All possible variations of human FMT were included. For the purposes of this review, FMT was defined as any process by which a fecal microbiota suspension was transferred from the gastrointestinal tract of a healthy individual into another person with the aim of treating a health condition.Citation35

  • Comparator: Studies were included if they utilized a control group.

  • Outcomes: When reporting on efficacy, this review used primary outcome measures as described by each study. When the primary outcome did not relate to efficacy, the secondary outcomes relating to clinical efficacy were noted, but results were only considered significant when the primary outcome measure related to clinical efficacy and was statistically significant vs the control intervention. Adverse events (AE) were reported as presented by the included study.

In March 2020, searches were carried out using MEDLINE, EMBASE, Cochrane Central Register of Controlled Trials, Health Technology Assessment Database, Allied and Complementary Medicine (AMED) and PsycINFO. Reference sections of previously published randomized trials, systematic reviews, and meta-analyses on this and related topics were also searched.

Forty-two iterations of the term “FMT” were identified and used as search terms:

FMT or fecal microbiota transplant* or fecal microbiota transplant* or microbiota transfer or microbiome transfer or microbiota transplant* OR microbiome transplant* or microbial transplant* or microbial transfer or fecal transplant* OR fecal transplant* or feces transplant* OR feces transplant* or stool transplant* or stool transfer or fecal flora transplant* OR fecal flora transplant* or microflora transplant* OR fecal flora transfer or fecal flora transfer OR fecal bacteriotherapy OR fecal bacteriotherapy OR feces bacteriotherapy OR feces bacteriotherapy OR rectal bacteriotherapy OR fecal flora bacteriotherapy OR donor fecal OR donor stool OR donor feces OR donor fecal or donor feces fecal transfer OR fecal transfer OR fecal reconstitution OR fecal reconstitution OR flora reconstitution OR microbiome reconstitution OR feces reconstitution or feces reconstitution. The following modifiers were applied: studies relating to humans and published in English.

Study selection

The following study types were included: randomized controlled trials (RCTs), non-randomized-controlled studies, and observational studies with a comparator arm. In the case of observational studies with a comparator arm, only prospective cohort studies were included in order to assess temporality. Reviews, abstracts, conference papers, and posters were excluded.

Two investigators (JG and JD) independently performed the searches using Rayyan software. JG performed initial screening to identify potentially eligible studies. Articles were first screened by title and abstract. Remaining articles were further scrutinized by full-text review. JD acted as a secondary reviewer and was blind to JG’s screening outcomes. Where there was a lack of consensus between the two reviewers, the senior author (WM) acted as a third reviewer to make a final decision on whether the study met inclusion criteria.

Risk of bias assessment

Methodological heterogeneity was evaluated by comparing included data using the ‘risk of bias’ tables. The Cochrane Risk of Bias tool was used to assess the risk of bias in randomized trials. JG and JD independently evaluated risk of bias.Citation36

Statistical analysis

Data from individual trials were to be combined, and a meta-analysis performed only if the data were amenable. Patient groups, disease entity, and outcome measures needed to be sufficiently similar in order for synthesis to occur. Our meta-analyses were conducted in Comprehensive Meta-Analysis (version 3.3.070)Citation37 using a Mantel-Haenszel random-effects model to account for heterogeneity between studies. The ICitation2-statistic was used as an indicator of heterogeneity. A value of 0% indicates no observed heterogeneity, and larger values indicate increasing heterogeneity. Due to the limited number of studies included in each meta-analysis, no sensitivity or subgroup analyses were performed. Furthermore, no test of publication bias was performed due to the limited number of trials.Citation38

Assessment of microbial “engraftment”

This review also assessed whether successful “engraftment” occurred of the donor microbiome in the recipient. For the purposes of this review, the term “engraftment” was ascertained according to the following key concepts:

  • Was a change in recipient microbiota observed following FMT?

If a change in recipient microbiota was observed, then:

  • Was this change toward the donor microbiota?

  • To what extent/how significant was that change?

  • For how long did the microbiota changes persist following cessation of FMT?

Results

Study selection

The systematic search identified 5,495 de-duplicated studies, of which 26 met eligibility criteria for inclusion (see ).

Figure 1. PRISMA flowchart of included studies

Figure 1. PRISMA flowchart of included studies

Study characteristics

As per , of the 26 articles that were included in the final review, 20 were Randomized Controlled Trials (RCTs), two of which were open-label, three single-blind and 15 double-blind. The remaining six studies encompassed non-randomized-controlled studies (n = 3), case–control studies (n = 2), and cohort studies (n = 1). Eight studies investigated the use of FMT for Inflammatory Bowel Disease (IBD), six for functional gut disorders, four for hepatic disorders, three for metabolic syndrome, two for antibiotic-resistant organisms, and one each for “pouchitis,” obesity without metabolic syndrome, and Human Immunodeficiency Virus (HIV).

Table 1. Summary of FMT methodological factors

A total of 1149 participants were enrolled in the included studies, with a mean of 44 participants per study (sample sizes ranged from 6 to 165). When broken down by disorder, there were a total of 463 participants in studies relating to IBD, 424 for functional gut disorders, 104 for metabolic syndrome/obesity, 109 for hepatic disorders, 60 for antibiotic-resistant organisms, and 14 for other disorders. Study follow-up periods varied from 2 weeks to 12 months. The largest sub-groups by disorder were UC (n = 6) and irritable bowel syndrome (IBS) (n = 5). These groups were large enough to allow for meta-analyses.

Seventeen studies included both males and females, eight included males only, and one did not provide demographic data. Studies were conducted in US (n = 6),Citation41,Citation44,Citation47–51 The Netherlands (n = 4),Citation39,Citation42,Citation43,Citation52 Australia (n = 2),Citation53,Citation54 China (n = 2), Norway (n = 2),Citation14,Citation40 France (n = 2),Citation45,Citation46 India (n = 2), Denmark (n = 1), Japan (n = 1),Citation55 Austria (n = 1),Citation56 Canada (n = 1),Citation57 Sweden (n = 1),Citation58 and one was an international multi-site collaborative study between Switzerland, the Netherlands, Israel, and France.Citation59

Methodological factors for FMT manufacture process

The FMT manufacture process varied significantly between studies (see ); indeed, no two studies used the same process. Of the 26 included FMT studies, 11 delivered FMT via colonoscopy, enema, or both,Citation14,Citation45,Citation47,Citation48,Citation53–58,Citation60 nine were delivered endoscopically, either via nasojejunal tube,Citation61nasoduodenal tubeCitation39,Citation40,Citation42,Citation43,Citation52,Citation62,Citation63 or nasogastric tube;Citation46 four were delivered orally via encapsulated FMT;Citation41,Citation49,Citation50,Citation64 and two studies used a mixed methodology of endoscopic delivery or encapsulated FMT,Citation59 or endoscopic route followed by encapsulated FMT.Citation51 Twenty two studies used an aerobic preparation of FMT,Citation14,Citation40,Citation42,Citation43,Citation45–50,Citation52,Citation53,Citation55–63 one used an anaerobic preparation,Citation54 and two did not specify.Citation41,Citation51 Two used a semi-anaerobic preparation of FMT (33, 37) in which feces was exposed to some oxygen during the procedure but attempts were made to minimize this; for example, prior to preparation, feces was stored in oxygen-depleted saline solution (36). Five studies used fresh feces, four used frozen feces, and two protocols allowed for use of fresh or frozen feces. One protocol did not describe whether feces were fresh or frozen.Citation63

Dose

Dosing was inconsistently described. Twelve studies did not provide clear information regarding amount of stool used. Fourteen studies reported on the initial sample of fresh stool, whilst eight described the amount of “suspension” used, which consisted of filtered stool diluted with normal saline and sometimes mixed with a cryoprotectant such as glycerol. Doses of 12 g-250 g of fresh stool were reported in the 14 studies that did provide these data.

Adjunctive treatments

A wide range of adjunctive treatments were employed. Fourteen studies used bowel preparation,Citation14,Citation39,Citation42,Citation43,Citation45,Citation47,Citation52–56,Citation58,Citation60 and six studies used antibiotics.Citation44,Citation46,Citation48,Citation55,Citation56,Citation59,Citation62 Ren et alCitation63 did not state whether bowel preparation was used and Herfarth et alCitation51 did not report whether any adjunctive treatments were used.

Donor methods

Four studies used multiple donors (i.e. a pooled sample),Citation14,Citation53,Citation54,Citation64 two did not adequately describe whether single or multiple donors were used,Citation46,Citation63 and the remaining 20 studies used single donors. Nineteen studies used non-related donors, two used related donors only,Citation55,Citation62 one used either,Citation56 and four did not specify.Citation46,Citation57,Citation63,Citation64

Screening protocol

Donor screening protocols overall were incompletely and poorly described. Where screening was stated as occurring, the methods for screening were frequently not provided. However, the more recent studies tended to have better reporting of screening protocols and more comprehensive screening. Fourteen studies specifically screened for metabolic risk factors, but only seven specifically described screening for mental illnesses.

Study results

Efficacy

Results were categorized by disorder and are summarized in . Of the 26 included studies, 10 reported significant results for their primary outcome measures, where these related to clinical efficacy. These 10 studies related to functional gut disorders,Citation14,Citation40,Citation61,Citation64 Hepatitis B,Citation63 IBD,Citation53,Citation54,Citation57 antibiotic-resistant organisms,Citation46 and metabolic syndrome.Citation52 The evaluated conditions were highly heterogeneous, even within groups. Nonetheless, it was possible to perform meta-analyses for two groups of disorders: IBS, and active UC.

Table 2. Summary of primary outcomes of included studies

Inflammatory bowel disease

There were eight studies of IBD, six of which were of active UC, the remaining two being of Crohn’s Disease (CD),Citation45 and maintenance of remission in UC.Citation60 Sokol et alCitation45 conducted a randomized, single-blind, controlled trial comparing colonoscopic FMT with placebo in 17 adults with CD. There was a significant decrease in CD symptoms in the FMT group compared with placebo (p = .03).Citation45 Sood et alCitation60 conducted a double-blind, randomized-controlled trial (RCT) of colonoscopic FMT compared with placebo as maintenance treatment for inactive UC. The study did not find a significant difference in the primary outcome measure (steroid-free clinical remission) between groups (p = .111); however, significant between-group differences were reported in endoscopic remission (p = .026), histological remission (0.033), and change in inflammatory markers (p < .001) favoring FMT.Citation60

Meta-analysis for ulcerative colitis subgroup

Six studies reported on active UC, which was sufficient to perform a meta-analysis for clinical remission, clinical response, endoscopic remission, and endoscopic response. Outcome measures were heterogeneous, were collected at different time points (between 7 weeks and 90 days), and used differing definitions of clinical response/remission and endoscopic remission/response. Five of six used Mayo score, whilst one used Clinical Activity Index (CAI) score. Definitions and data for clinical remission and response are summarized in Supplementary Table 1, and endoscopic remission and response are summarized in Supplementary Table 2.

Meta-analysis confirmed that FMT was associated with a significant improvement in clinical remission rates in UC compared to control conditions (OR = 3.634, 95% CI = 1.940 to 6.808, n = 6 studies, ICitation2=0%, p < .001) (see ). FMT was also associated with a significant improvement in clinical response rates in UC compared to control (OR = 2.634, 95% CI = 1.441 to 4.815, n = 6 studies, ICitation2=33%, p = .002) (see ), as well as for endoscopic remission rates (OR = 4.431, 95% CI = 1.901 to 10.324, n = 5 studies, ICitation2=0%, p = .001) (see ). However, FMT showed no significant improvement in endoscopic response rates in UC compared to controls (OR = 1.065, 95% CI = 0.432 to 2.625, n = 2 studies, ICitation2=0%, p = .892) (see Supplementary Fig 1).

Figure 2. Clinical remission results

Figure 2. Clinical remission results

Figure 3. Clinical response results

Figure 3. Clinical response results

Figure 4. Endoscopic remission results

Figure 4. Endoscopic remission results

Functional gut disorders

In a trial of nasojejunally delivered FMT given daily for six days in adjunct to treatment as usual (TAU) for slow-transit constipation, Tian et alCitation61 reported a clinical remission rate of 36.7% for the FMT group compared with 13.3% for the TAU control group (p = .04).

Five studies reported on IBS, which was sufficient to perform a meta-analysis for clinical response and average change in IBS-SSS. Different studies used different definitions of clinical response: four used IBS-SSS, and one used GSRS-IBS, mostly at 3 months. Definitions and data for clinical response and change in IBS-SSS are summarized in Supplementary Table 3.

Meta-analysis revealed no significant difference in IBS-SSS (Hedge’s g = 0.282, 95% CI = −1.373 to 1.937, n = 3 studies, ICitation2=97%, p = .739) or clinical response (OR = 1.699, 95% CI = 0.273 to 10.588, n = 5 studies, ICitation2=92%, p = .739) following FMT compared to control (see Supplementary material; )

Psychiatric outcomes

Only three studies assessed psychiatric outcomes and all three were conducted in IBS populations. Two of the studies used the Hospital Anxiety and Depression Scale (HADS) to measure depression and anxiety symptoms and neither reported a significant change in symptoms between groups post-intervention. The third study used the mental health subscale of the Fatigue Assessment Scale (FAS), and reported a significant difference between the means of the group who received 30 g FMT (13.3, s.d. 3.1) compared with the placebo group (14.7, s.d. 3.4) at 1 month (p < .05), and the group who received 60 g FMT (13.1, s.d. 3.1), compared with the placebo group (14.5, s.d. 2.7) at 3 months (p < .05) in favor of FMT.

Hepatic disorders

Of the four studies in hepatic disorders, three had significant results for clinical efficacy favoring FMT over control, whilst the fourth did not report significant outcomes. In a trial of nasoduodenally delivered FMT every 4 weeks (for 1–7 treatments) plus TAU for Hepatitis B, Ren et alCitation63 reported that four of the five participants achieved clearance of HbeAg, whereas all 13 of the TAU controls continued to have a positive HbeAg titer (p = .0002). Bajaj et alCitation48 conducted an open-label RCT investigating FMT for recurrent hepatic encephalopathy using a retention enema compared with TAU and reported a significant improvement in two measures of cognitive outcomes in favor of FMT (p < .01 for both). Similarly, in a single-blind RCT of encapsulated FMT compared with placebo capsules, Bajaj et alCitation49 found a significant improvement in cognitive outcomes for the FMT group but not the placebo group (p = .02). Philips et alCitation62 conducted a retrospective cohort study comparing FMT with three control groups (steroids, nutritional support or pentoxifylline) for the treatment of severe alcoholic hepatitis, but found no significant improvement in the primary outcome of 90-day survival (p = .179).

Metabolic syndrome or obesity without metabolic syndrome

Four studies evaluated FMT for the treatment of metabolic syndrome or obesity without metabolic syndrome, and only one of these had significant results for clinical efficacy regarding the primary outcome. The remaining three showed significant results for secondary outcome measures, all in favor of FMT. Vrieze et alCitation52 conducted a double-blind pilot RCT of nasoduodenally delivered FMT compared with autologous FMT for metabolic syndrome and reported a significant improvement in week 6 peripheral insulin sensitivity (p < .05) in favor of FMT, but not in hepatic insulin sensitivity (p = .08), diet composition, resting energy expenditure, or counter-regulatory hormones. In a double-blinded RCT examining nasoduodenally delivered FMT for metabolic syndrome, Kootte et alCitation39 did not find significant differences in their primary outcome measure (change in intestinal microbiota in relation to insulin sensitivity at 18 weeks), nor did they observe a significant change in BMI or SCFA levels at any study time point. In terms of secondary outcomes, change in fecal microbiota composition at 6 weeks associated with improved peripheral insulin sensitivity (from 25.8 [19.3–34.7] to 28.8 [21.4–36.9] mmol kg/1 min/1, p < .05) in the allogenic FMT group, whereas autologous FMT had no effect (from 22.5 [16.9–30.2] to 20.8 [17.6–29.5] mmol kg/1 min/1, p > .5).

Smits et alCitation43 conducted a double-blind pilot RCT of nasoduodenally delivered FMT compared with autologous FMT for TMAO production in participants with metabolic syndrome and did not find a significant difference between groups. In a double-blind pilot RCT of encapsulated FMT compared with placebo capsules for obesity-related biomarkers in participants with obesity but without metabolic syndrome, Allegretti et al found a significant between-group difference in area under the curve at week 12 for leptin compared with baseline (p = .001), but no significant change for other biomarkers of obesity.Citation50

Antibiotic-resistant organisms

Two studies evaluated FMT for the treatment of colonization of antibiotic-resistant organisms, one demonstrating significant clinical efficacy of FMT over control and the other without significant outcomes. In a retrospective matched case-control study of nasogastric FMT compared with TAU for Carbapenemase-Producing Enterobacteriaceae (CPE), Saidani et alCitation46 reported a significant delay in negativation of rectal swab cultures 2-weeks post-FMT compared with TAU (p < .001). Huttner et alCitation59 conducted a multicentre, randomized, open-label, superiority trial of nasogastric or encapsulated FMT (treatment was site dependant), compared with TAU for CPE and Extended spectrum beta-lactamase (ESBL), but did not identify a significant between-group difference in the primary outcome measure for clinical efficacy (p-value not provided).

Other conditions

Two studies were not able to be grouped with the others. They evaluated FMT for the treatment of individuals with HIV and antibiotic-dependant pouchitis, respectively. Neither showed clinical efficacy.

Safety data

There were variable quality and completeness of reporting of safety data for both serious adverse events (SAEs) and mild to moderate AEs, across studies (see ). Studies had a follow up period ranging from four weeksCitation55 to 1 year .Citation14 SAEs

Table 3. Completeness of reporting of AE

Of the 26 included studies, 23 provided clear descriptions of SAEs. A total of 69 SAEs were reported from 12 studies; 26 occurred in participants allocated to receive FMT, and 43 in participants in the control groups (see Supplementary Table 4). Of the 26 SAEs that occurred in participants allocated to receive FMT, all but one was deemed unlikely to be related to the intervention. Twenty of these SAEs occurred in participants who received FMT via colonoscopy or enema, and six in those receiving FMT endoscopically or via capsules. When broken down by specific disorder, 17 of these SAEs occurred in participants with inflammatory bowel disease, three in participants with hepatic encephalopathy, five in participants with antibiotic resistant organisms and one in a participant with IBS.

Mild to moderate AEs

Due to the inconsistent quality and completeness of reporting of mild to moderate AE, it was only possible to pool/summarize data for a small number of included studies (see Supplementary Table 5). These studies were related to IBS, (n = 4), UC (n = 2), slow transit constipation (n = 1), hepatic encephalopathy (n = 1), and metabolic syndrome (n = 1). As such a cross-indication assessment of adverse events was not possible as the data were insufficiently reported across disorders.

Similar rates of mild to moderate AEs were observed in participants allocated to FMT compared to the control groups (see Supplementary Table 5). However, the following AEs were more common in participants receiving allogenic FMT compared with those allocated to control groups: nausea (reported in 80% of FMT recipients compared with 72% in control groups), constipation (reported in 17.4% of FMT recipients compared with 2.4% in control groups), diarrhea (reported in 16.8% of FMT recipients compared with 6.7% in control groups), transient, or low-grade fever (reported in 8.4% of FMT recipients compared with 3.0% in control groups) and vomiting (reported in 5.9% of FMT recipients compared with 2.9% in control groups).

Incomplete reporting precluded comparison of AE rates between different routes of FMT; however, encapsulated FMT appears to have been the best-tolerated route.

Successful microbial “engraftment”

Microbiome analysis pre- and post-FMT was performed in 23 of the 26 included studies. All microbiome analyses used 16s RNA sequencing. The data relating to “engraftment” are summarized in . All 23 of 23 studies which measured microbiome analysis reported change in microbiome following FMT. Fourteen of the 23 studies reported whether the change in microbiota was toward the donor and, of these, 11 confirmed that the recipient microbiome did move toward the donor microbiome. The remaining three studies did not report significant results.

Table 4. Summary of “engraftment” of FMT

Reporting on the extent or significance of microbiota changes was inconsistent across studies and the complexity of microbiome data analysis has meant it was not possible to answer the question of the extent to which the recipient microbiome changed toward the donor, as no clear quantification was provided by the included studies. As such, these data are not reported in .

Regarding longevity of the observed changes in recipient microbiota, it was not possible to answer this question in this review, as included studies either did not follow-up recipients for long enough, or did not measure microbiota changes frequently enough to be able to state the duration for which any changes were observed. However, with these limitations in mind, it appears that the demonstrated microbiome changes were transient and appeared to last between 2 weeks to 1 year following the intervention.

Correlation of “engraftment” with clinical findings

Fourteen of 23 studies reported on associations between “engraftment” and clinical outcomes, and of these, 12 studies had statistically significant results with 10 reporting a significant association between successful engraftment and clinical efficacy and two reporting no association between efficacy and engraftment. These data are summarized in .

Risk of bias assessment

According to the Cochrane Risk of Bias toolCitation36 (see Supplementary Table 6), nine studies were evaluated as “low risk,”Citation14,Citation40–42,Citation49,Citation53,Citation54,Citation58,Citation60,Citation64 six as “some concerns,”Citation39,Citation42,Citation43,Citation45,Citation50,Citation52,Citation57 and five as “high risk.”Citation48,Citation57–59,Citation61 Studies rated “high risk” were: Tian et al,Citation61 due to incomplete reporting across most domains and inadequate randomization processes; Moayyedi et alCitation57 due to likely inadequacy of blinding of participants as water enemas were used as placebo, which would likely be easily differentiated from true FMT by recipients; Bajaj et alCitation48 and Huttner et alCitation59 as the studies were open label, with an absence of blinding; and Herfarth et al,Citation51 due to an absence of a statistical pre-analysis plan, and the fact that the trial was ceased after only six participants were randomized.

Discussion

Statement of principal findings

This review identified FMT trials for conditions other than CDI, with promising, albeit mixed, outcomes regarding efficacy and safety. Meta-analysis of UC studies found FMT to be superior to control conditions for active disease in terms of endoscopic remission, clinical remission, and clinical response. In contrast, meta-analysis of the five IBS studies did not yield significant results regarding symptoms or clinical response. Regarding clinical efficacy in other applications of FMT, studies were too heterogeneous to perform meta-analyses, but four yielded evidence of clinical efficacy in slow-transit constipation, Hepatitis B, colonization of CPE, and insulin sensitivity in metabolic syndrome. The impact of FMT on psychiatric outcomes was assessed in three studies of IBS patients, with one of these finding significant improvements.

This review also found that FMT was safe and well tolerated. Similar rates of mild to moderate AEs were observed in participants who received FMT compared to those allocated to control groups, while SAEs were more commonly reported in participants allocated to control/placebo groups.

Not all studies assessed or reported whether FMT results in successful engraftment of the donor microbiome into the recipient, but a majority of those that did report it confirmed a move toward the donor microbiome following FMT and that these changes persisted for up to 1 year. Furthermore, four of the five studies that reported on association between microbiome changes and clinical efficacy, four of five confirmed such an association. This suggests that FMT alters the recipient microbiome, and that it is possible that this change is a contributing factor to clinical efficacy.

Strengths and weaknesses of the review

This review is the first systematic review to evaluate both safety and efficacy of FMT for all disorders other than CDI. This review aimed to recruit higher quality studies by excluding uncontrolled studies, which represent a majority of studies in this field. Whilst other reviews have been conducted with respect to safety of FMT for indications other than CDI, these reviews are either not recent,Citation65 or were restricted to a single indication such as IBSCitation29–31 and IBD.Citation32–34 With respect to efficacy, whilst other reviews have been published for single indications, such as IBSCitation29–31 and IBD,Citation32–34 there have been no holistic reviews looking at all indications other than CDI. As far as the authors of this review are aware, this review also represents the most up to date systematic review and meta-analysis of the safety and efficacy of FMT for IBS.

However, the 26 studies included were heterogeneous and of mixed quality, with several using open-label designs and small samples. Encouragingly, more recently published studies appear to be of higher quality, using more robust study designs (such as double-blinded RCTs), and with clearer and more complete descriptions of study methodology.

It was possible to conduct meta-analyses for both IBS and active UC. However, due to the lack of consensus regarding outcome measures and small number of included studies, the results of these meta-analyses should be considered preliminary at this stage. Further, due to the low numbers, tests for publication bias (e.g. eggers regression and funnel plots) were unable to be carried out.

We were unable to undertake a quantitative analysis on the level of engraftment, given the gaps in data in the included studies. Future studies should evaluate microbial engraftment as a result of FMT, allowing for a systematic assessment.

This paper evaluated safety data across a range of indications, finding broadly that FMT is well tolerated and safe. However, due to the poor quality and incompleteness of reporting in several papers, a cross-indication analysis of safety data was not possible. We recommend future FMT studies report more clearly on mild to moderate and SAE.

With respect to SAE, these were observed more frequently in control group participants than those allocated to receive FMT, and of the SAE observed in FMT recipients, most considered to be unrelated to FMT. In understanding this finding, it should be noted that most SAE were likely due to the underlying disease process rather than the FMT procedure, a majority of reported SAE were flares of the disease in inflammatory bowel disease participants. Thus, FMT may have prevented disease flares.

Implications for clinicians and policymakers

With respect to active UC, our meta-analysis revealed that FMT appears to be clinically efficacious compared to control conditions. Four of the six included studies used a gold-standard double-blind placebo-controlled RCT design, and all six included studies favored FMT over control conditions regarding clinical efficacy, notwithstanding limitations described above. Thus, the quality and consistency of outcomes appear to favor FMT in the treatment of active UC, making this is a promising area for research attention.

Evidence supporting the application of FMT in the treatment of IBS is more equivocal. Five studies included in this review showed mixed outcomes, with three reporting that FMT was favorable, and two finding that control conditions were more effective than FMT. Possible reasons for these mixed findings are discussed in depth in other review papers,Citation29–31 which note, inter alia, that route of delivery, choice of placebo (i.e. inert vs autologous FMT), and patient group may have contributed.Citation29–31 Suffice to say, there is no strong evidence at this stage that FMT is efficacious for the treatment of functional gut disorders.

This review also identified two additional studies relating to inflammatory bowel disease yielding significant outcomes regarding clinical efficacy, and four studies that evaluated FMT for metabolic syndrome or obesity that focused on biological outcomes rather than clinical efficacy. As such, these are identified as conditions of interest for further research only.

Regarding safety, FMT appears generally to be a safe and well-tolerated treatment, with orally administered FMT appearing to be the best-tolerated route. However, it is also important to note that on 13th June, 2019 the American-based Food and Drug Administration (FDA) released a statement warning of the risks of FMT. They reported two cases (both immunocompromised patients) in whom antibiotic-resistant organisms, (specifically, Extended Spectrum Beta Lactamase-producing Escherichia coli (E.coli)) were transferred via FMT, resulting in one death. In these cases, donor feces were not screened for antibiotic-resistant organisms.Citation66 A further warning was issued on 12th March 2020 advising of six cases of additional transmission of antibiotic-resistant organisms via FMT provided by a US-based stool bank (enteropathogenic Escherichia coli in two cases and Shigatoxin-producing Escherichia coli in four cases) and two deaths that occurred in recipients of FMT, but in which FMT may not have been the cause of death.Citation67 It is now standard across widely accepted protocols in the United Kingdom, United States, and Australasia to screen thoroughly for antibiotic-resistant organisms. These recent serious incidents highlight the importance of adhering to rigorous screening protocols, such as the Openbiome Protocol in the US,Citation68 the British Guidelines for donor screening,Citation69 or the Australasian guidelines.Citation70

Unanswered questions, challenges for the field of FMT research, and future directions

Of all the uses of FMT for conditions other than CDI, the most promising at this stage is for active flares of UC. Further large scale, high-quality studies, utilizing consistent data points to measure primary outcomes, are urgently called for. Other indications with some promise include metabolic syndrome/obesity, antibiotic-resistant organisms, and certain hepatic disorders. Whilst published outside of the time range of the search performed for this review, a recent Phase I study investigating FMT for Alcohol Use Disorder showed safety and efficacy.Citation71 Thus, we watch with interest the growing field of FMT for hepatic disorders.

However, on the whole, studies in these emerging areas are heterogeneous and generally of poor quality, with most using open-label designs and only one study using clinical efficacy as a primary outcome measure. Again, further high-quality research, using larger sample sizes and double-blinded, placebo-controlled designs, and that use clinical efficacy endpoints as a primary outcome, are needed for these emerging indications. Furthermore, no studies evaluated the use of FMT for psychiatric conditions, an area of great importance given growing interest and data supporting the relationship between mental health and the gut microbiome (“the microbiota-gut-brain axis”).Citation7,Citation72 Much also remains unknown about the ideal methodological design for studies of FMT for conditions other than Clostridium difficile infection. For one, an important question remains around choice of placebo. Nine studies selected an inert placebo, such as water mixed with glycerol and food dye, whilst eight opted for autologous FMT as a placebo. There is some evidence to suggest that even autologous FMT may have an impact on gut microbiota,Citation58 which may confound results. This needs to be further explored but presents an argument against using autologous FMT as a placebo in future studies aimed at determining efficacy of FMT compared with an inactive control. Follow-up periods to assess long-term safety, engraftment, and metagenomics are also an important consideration for study design. The authors of this review suggest a follow up period of at least 6 months to adequately monitor for safety and long-term AE.

Conclusions

This systematic review and meta-analysis provide preliminary data that FMT may be safe and effective for several conditions other than CDI. Preliminary meta-analyses suggest efficacy for outcomes related to active ulcerative colitis but not IBS. Hepatic disorders, metabolic syndrome/obesity, and antibiotic-resistant organisms were also identified as emerging areas of interest for FMT research. Regarding safety, there was little difference in SAEs between participants allocated to receive FMT and those allocated to control groups. With respect to mild to moderate adverse events, similar rates were also observed in treatment and control groups. These encouraging pilot outcomes provide preliminary support for further high-quality research in these areas.

Disclosure of Potential Conflicts of Interest

Michael Berk is supported by an NHMRC Senior Principal Research Fellowship (1156072). MB has received Grant/Research Support from the NIH, Cooperative Research Centre, Simons Autism Foundation, Cancer Council of Victoria, Stanley Medical Research Foundation, Medical Benefits Fund, National Health and Medical Research Council, Medical Research Futures Fund, Beyond Blue, Rotary Health, A2 milk company, Meat and Livestock Board, Woolworths, Avant and the Harry Windsor Foundation, has been a speaker for Astra Zeneca, Lundbeck, Merck, Pfizer, and served as a consultant to Allergan, Astra Zeneca, Bioadvantex, Bionomics, Collaborative Medicinal Development, Lundbeck Merck, Pfizer and Servier – all unrelated to this work. Felice Jacka has received: (1) competitive Grant/Research support from the Brain and Behaviour Research Institute, the National Health and Medical Research Council (NHMRC), Australian Rotary Health, the Geelong Medical Research Foundation, the Ian Potter Foundation, The University of Melbourne; (2) industry support for research from Meat and Livestock Australia, Woolworths Limited, the A2 Milk Company, Be Fit Foods; (3) philanthropic support from the Fernwood Foundation, Wilson Foundation, the JTM Foundation, the Serp Hills Foundation, the Roberts Family Foundation, the Waterloo Foundation and; (4) travel support and speakers honoraria from Sanofi-Synthelabo, Janssen Cilag, Servier, Pfizer, Health Ed, Network Nutrition, Angelini Farmaceutica, Eli Lilly and Metagenics. Wolfgang Marx is currently funded by an Alfred Deakin Postdoctoral Research Fellowship and a Multiple Sclerosis Research Australia early-career fellowship. Wolfgang has previously received funding from the Cancer Council Queensland and university grants/fellowships from La Trobe University, Deakin University, University of Queensland, and Bond University, received industry funding and has attended events funded by Cobram Estate Pty. Ltd, received travel funding from Nutrition Society of Australia, received consultancy funding from Nutrition Research Australia, and has received speakers honoraria from The Cancer Council Queensland and the Princess Alexandra Research Foundation. The Food & Mood Centre has received Grant/Research support from Fernwood Foundation, Wilson Foundation, the A2 Milk Company, and Be Fit Foods.

Author contributions statement

JG wrote the protocol document, performed the primary and secondary searches, data extraction, data analysis, and drafted and edited the manuscript. JD was the second reviewer for primary and secondary searches and data extraction. WM was senior author, provided oversight, edited the manuscript, and completed data analysis for the meta-analyses. FJ was a second senior author and provided oversight and input into the manuscript. All authors read, edited, and approved the final draft.

Supplemental material

Supplemental Material

Download MS Word (505.1 KB)

Supplementary material

Supplemental data for this article can be accessed on the publisher’s website.

Additional information

Funding

No funding was received for this work.

References

  • Cani PD. Human gut microbiome: hopes, threats and promises. Gut. 2018;67(9):1716–1725. doi:10.1136/gutjnl-2018-316723.
  • Clemente JC, Manasson J, Scher JU. The role of the gut microbiome in systemic inflammatory disease. BMJ. 2018;360:j5145. doi:10.1136/bmj.j5145.
  • Thaiss CA, Zmora N, Levy M, Elinav E. The microbiome and innate immunity. Nature. 2016;535(7610):65–74. doi:10.1038/nature18847.
  • Slyepchenko A, Maes M, Jacka FN, Köhler CA, Barichello T, McIntyre RS, Berk M, Grande I, Foster JA, Vieta E, et al. Gut microbiota, bacterial translocation, and interactions with diet: pathophysiological links between major depressive disorder and non-communicable medical comorbidities. Psychother Psychosom. 2017;86(1):31–46. doi:10.1159/000448957.
  • Peterson J, Garges S, Giovanni M, McInnes P, Wang L, Schloss JA, Bonazzi V, McEwen JE, Wetterstrand KA, Deal C, et al. The NIH human microbiome project. Genome Res. 2009;19(12):2317–2323. doi:10.1101/gr.096651.109.
  • Lynch SV, Pedersen O. The human intestinal microbiome in health and disease. N Engl J Med. 2016;375(24):2369–2379. doi:10.1056/NEJMra1600266.
  • Green J, Castle D, Berk M, Hair C, Loughman A, Cryan J, Nierenberg A, Athan E, Jacka F. Faecal microbiota transplants for depression – who gives a capsule? Aust N Z J Psychiatry. 2019;53(8):732–734. doi:10.1177/0004867419839776.
  • Khoruts A, Sadowsky MJ. Understanding the mechanisms of faecal microbiota transplantation. Nat Rev Gastroenterol Hepatol. 2016;13(9):508–516. doi:10.1038/nrgastro.2016.98.
  • Gough E, Shaikh H, Manges AR. Systematic review of intestinal microbiota transplantation (fecal bacteriotherapy) for recurrent Clostridium difficile infection. Clin Infect Dis. 2011;53(10):994–1002. doi:10.1093/cid/cir632.
  • Drekonja D, Reich J, Gezahegn S, Greer N, Shaukat A, MacDonald R, Rutks I, Wilt TJ. Fecal microbiota transplantation for clostridium difficile infection. Ann Intern Med. 2015;162(9):630–638. doi:10.7326/M14-2693.
  • Bafeta A, Yavchitz A, Riveros C, Batista R, Methods RP. Reporting studies assessing fecal microbiota transplantation: a systematic review. Ann Intern Med. 2017;167(1):34–39. doi:10.7326/M16-2810.
  • Sha S, Liang J, Chen M, Xu B, Liang C, Wei N, Wu K. Systematic review: faecal microbiota transplantation therapy for digestive and nondigestive disorders in adults and children. Aliment Pharmacol Ther. 2014;39(10):1003–1032. doi:10.1111/apt.12699.
  • Qazi T, Amaratunga T, Barnes EL, Fischer M, Kassam Z, Allegretti JR. The risk of inflammatory bowel disease flares after fecal microbiota transplantation: systematic review and meta-analysis. Gut Microbes. 2017;8(6):574–588. doi:10.1080/19490976.2017.1353848.
  • Johnsen PH, Hilpusch F, Cavanagh JP, Leikanger IS, Kolstad C, Valle PC, Goll R. Faecal microbiota transplantation versus placebo for moderate-to-severe irritable bowel syndrome: a double-blind, randomised, placebo-controlled, parallel-group, single-centre trial. Lancet Gastroenterol Hepatol. 2018;3(1):17–24. doi:10.1016/S2468-1253(17)30338-2.
  • Pinn D, Aroniadis O, Brandt L. Follow-up study of fecal microbiota transplantation (FMT) for the treatment of refractory irritable bowel syndrome (IBS). Am J Gastroenterol. 2013;108:S563. doi:10.14309/00000434-201310001-01862.
  • Borody TJ, George L, Andrews P, Brandl S, Noonan S, Cole P, Hyland L, Morgan A, Maysey J, Moore‐Jones D, et al. Bowel-flora alteration: a potential cure for inflammatory bowel disease and irritable bowel syndrome? Med J Aust. 1989;150(10):604. doi:10.5694/j.1326-5377.1989.tb136704.x.
  • Borody TJ, Warren EF, Leis SM, Surace R, Ashman O, Siarakas S. Bacteriotherapy using fecal flora: toying with human motions. J Clin Gastroenterol. 2004;38(6):475–483. doi:10.1097/01.mcg.0000128988.13808.dc.
  • Xu M-Q, Cao H-L, Wang W-Q, Wang S, Cao X-C, Yan F, Wang B-M. Fecal microbiota transplantation broadening its application beyond intestinal disorders. WJG. 2015;21(1):102–111.
  • Luckey D, Gomez A, Murray J, White B, Bugs TV. us: the role of the gut in autoimmunity. Indian J Med Res. 2013;138:732–743.
  • Greenhill C. Microbiota: FMT transiently improves insulin sensitivity. Nat Rev Endocrinol. 2017;13:688.
  • Marotz CA, Zarrinpar A. Treating obesity and metabolic syndrome with fecal microbiota transplantation. Yale J Biol Med. 2016;89:383–388.
  • Kurokawa S, Kishimoto T, Mizuno S, Masaoka T, Naganuma M, Liang K-C, Kitazawa M, Nakashima M, Shindo C, Suda W, et al. The effect of fecal microbiota transplantation on psychiatric symptoms among patients with irritable bowel syndrome, functional diarrhea and functional constipation: an open-label observational study. J Affect Disord. 2018;235:506–512. doi:10.1016/j.jad.2018.04.038.
  • Kang DW, Adams JB, Gregory AC, Borody T, Chittick L, Fasano A, Khoruts A, Geis E, Maldonado J, McDonough-Means S, et al. Microbiota transfer therapy alters gut ecosystem and improves gastrointestinal and autism symptoms: an open-label study. Microbiome. 2017;5:1.
  • de Clercq NC, Frissen MN, Davids M, Groen AK, Nieuwdorp M. Weight gain after fecal microbiota transplantation in a patient with recurrent underweight following clinical recovery from anorexia nervosa. Psychother Psychosom. 2019;88(1):58–60. doi:10.1159/000495044.
  • Kelly JR, Borre Y, Patterson E, El AS, Deane J, Deane J, Kennedy PJ, Beers S, Scott K, Moloney G, et al. Transferring the blues: depression-associated gut microbiota induces neurobehavioural changes in the rat. J Psychiatr Res. 2016;82:109–118. doi:10.1016/j.jpsychires.2016.07.019.
  • Zheng P, Zeng B, Zhou C, Liu M, Fang Z, Xu X, Zeng L, Chen J, Fan S, Du X, et al. Gut microbiome remodeling induces depressive-like behaviors through a pathway mediated by the host’s metabolism. Mol Psychiatry. 2016;21(6):786–796. doi:10.1038/mp.2016.44.
  • Zheng P, Zeng B, Liu M, Chen J, Pan J, Han Y, Liu Y, Cheng K, Zhou C, Wang H, et al. The gut microbiome from patients with schizophrenia modulates the glutamate-glutamine-GABA cycle and schizophrenia-relevant behaviors in mice. Sci Adv. 2019;5(2):eaau8317. doi:10.1126/sciadv.aau8317.
  • Leshem A, Horesh N, Elinav E. Fecal microbial transplantation and its potential application in cardiometabolic syndrome. Front Immunol. 2019;10:1341.
  • Xu D, Chen VL, Steiner CA, Berinstein JA, Eswaran S, Waljee AK, Higgins P, Owyang C. Efficacy of fecal microbiota transplantation in irritable bowel syndrome: a systematic review and meta-analysis. Off J Am Coll Gastroenterol. 2019;114(7):1043-1050.
  • Ianiro G, Eusebi LH, Black CJ, Gasbarrini A, Cammarota G, Ford AC. 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. doi:10.1111/apt.15330.
  • Myneedu K, Deoker A, Schmulson MJ, Bashashati M. Fecal microbiota transplantation in irritable bowel syndrome: a systematic review and meta-analysis. United Eur Gastroenterol J. 2019;7:1033–1041.
  • LdF C, Borba HH, Tonin FS, Wiens A, Fernandez-Llimos F, Pontarolo R. Fecal microbiota transplantation in inflammatory bowel disease patients: a systematic review and meta-analysis. Plos One. 2020;15(9):e0238910. doi:10.1371/journal.pone.0238910.
  • Tang L-L, Feng W-Z, Cheng -J-J, Gong Y-N. Clinical remission of ulcerative colitis after different modes of faecal microbiota transplantation: a meta-analysis. Int J Colorectal Dis. 2020;35(6):1025–1034. doi:10.1007/s00384-020-03599-7.
  • Zhao HL, Chen SZ, Xu HM, Zhou YL, He J, Huang HL, Xu J, Nie YQ. Efficacy and safety of fecal microbiota transplantation for treating patients with ulcerative colitis: a systematic review and meta-analysis. J Dig Dis. 2020;21(10):534–548. doi:10.1111/1751-2980.12933.
  • Aroniadis OC, Brandt LJ. Fecal microbiota transplantation: past, present and future. Curr Opin Gastroenterol. 2013;29(1):79–84. doi:10.1097/MOG.0b013e32835a4b3e.
  • Higgins JPT, Altman DG, Gøtzsche PC, Jüni P, Moher D, Oxman AD, Savovic J, Schulz KF, Weeks L, Sterne JAC, et al. The Cochrane collaboration’s tool for assessing risk of bias in randomised trials. BMJ. 2011;343:d5928. doi:10.1136/bmj.d5928.
  • Englewood. Comprehensive meta-analysis version 3. 3 ed.: Biostat 104; 2011.
  • Collaboration C. Recommendations on testing for funnel plot asymmetry. Cochrane handbook for systematic reviews of interventions version. Cochrane handbook for systematic reviews of interventions version [Internet]. Cochrane Collaboration. 5; 2011.
  • Kootte RS, Levin E, Salojarvi J, Smits LP, Hartstra AV, Udayappan SD, Hermes G, Bouter KE, Koopen AM, Holst JJ, 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. doi:10.1016/j.cmet.2017.09.008.
  • El-Salhy M, Hatlebakk JG, Gilja OH, Brathen Kristoffersen A, Hausken T. Efficacy of faecal microbiota transplantation for patients with irritable bowel syndrome in a randomised, double-blind, placebo-controlled study. Gut. 2020;69(5):859–867. doi:10.1136/gutjnl-2019-319630.
  • Aroniadis OC, Brandt LJ, Oneto C, Feuerstadt P, Sherman A, Wolkoff AW, Kassam Z, Sadovsky RG, Elliott RJ, Budree S, et al. Faecal microbiota transplantation for diarrhoea-predominant irritable bowel syndrome: a double-blind, randomised, placebo-controlled trial. Lancet Gastroenterol Hepatol. 2019;4(9):675–685. doi:10.1016/S2468-1253(19)30198-0.
  • Rossen NG, Fuentes S, van der Spek MJ, Tijssen JG, Hartman JH, Duflou A, Löwenberg M, van den Brink GR, Mathus-Vliegen EM, de Vos WM, et al. Findings from a randomized controlled trial of fecal transplantation for patients with ulcerative colitis. Gastroenterology. 2015;149(1):110–118. doi:10.1053/j.gastro.2015.03.045.
  • Smits LP, Kootte RS, Levin E, Prodan A, Fuentes S, Zoetendal EG, Wang Z, Levison B, Cleophas M, Kemper 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):e008342.
  • Kang DW, Adams JB, Gregory AC, Borody T, Chittick L, Fasano A, Khoruts A, Geis E, Maldonado J, McDonough-Means S, et al. Microbiota transfer therapy alters gut ecosystem and improves gastrointestinal and autism symptoms: an open-label study. Microbiome. 2017;5(1).
  • Sokol H, Landman C, Seksik P, Berard L, Montil M, Nion-Larmurier I, Bourrier A, Le Gall G, Lalande V, De Rougemont A, et al. Fecal microbiota transplantation to maintain remission in Crohn’s disease: a pilot randomized controlled study. Microbiome. 2020;8(1):12. doi:10.1186/s40168-020-0792-5.
  • Saidani N, Lagier J-C, Cassir N, Million M, Baron S, Dubourg G, Eldin C, Kerbaj J, Valles C, Raoult D, et al. Faecal microbiota transplantation shortens the colonisation period and allows re-entry of patients carrying carbapenamase-producing bacteria into medical care facilities. Int J Antimicrob Agents. 2019;53(4):355–361. doi:10.1016/j.ijantimicag.2018.11.014.
  • Vujkovic-Cvijin I, Rutishauser RL, Pao M, Hunt PW, Lynch SV, McCune JM, Somsouk M. Limited engraftment of donor microbiome via one-time fecal microbial transplantation in treated HIV-infected individuals. Gut Microbes. 2017;8:440–450.
  • Bajaj JS, Kassam Z, Fagan A, Gavis EA, Liu E, Cox IJ, Kheradman R, Heuman D, Wang J, Gurry T, et al. Fecal microbiota transplant from a rational stool donor improves hepatic encephalopathy: a randomized clinical trial. Hepatology. 2017;66(6):1727–1738. doi:10.1002/hep.29306.
  • Bajaj JS, Salzman NH, Acharya C, Sterling RK, White MB, Gavis EA, Fagan A, Hayward M, Holtz ML, Matherly S, et al. Fecal microbial transplant capsules are safe in hepatic encephalopathy: a phase 1, randomized, placebo-controlled trial. Hepatology. 2019;70(5):1690–1703. doi:10.1002/hep.30690.
  • Allegretti JR, Kassam Z, Mullish BH, Chiang A, Carrellas M, Hurtado J, Marchesi JR, McDonald JAK, Pechlivanis A, Barker GF, et al. Effects of fecal microbiota transplantation with oral capsules in obese patients. Clin Gastroenterol Hepatol. 2020;18(4):855–63.e2. doi:10.1016/j.cgh.2019.07.006.
  • Herfarth H, Barnes EL, Long MD, Isaacs KL, Leith T, Silverstein M, Gerardin Y, Kassam Z. Combined endoscopic and oral fecal microbiota transplantation in patients with antibiotic-dependent pouchitis: low clinical efficacy due to low donor microbial engraftment. Inflammatory Intestinal Diseases. 2019;4(1):1–6. doi:10.1159/000497042.
  • Vrieze A, Van Nood E, Holleman F, Salojarvi J, Kootte RS, Bartelsman JF, Dallinga–Thie GM, Ackermans MT, Serlie MJ, Oozeer R, et al. Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Gastroenterology. 2012;143(4):913–6.e7. doi:10.1053/j.gastro.2012.06.031.
  • Paramsothy S, Kamm MA, Kaakoush NO, Walsh AJ, van den Bogaerde J, Samuel D, Leong RWL, Connor S, Ng W, Paramsothy R, et al. Multidonor intensive faecal microbiota transplantation for active ulcerative colitis: a randomised placebo-controlled trial. Lancet. 2017;389(10075):1218–1228. doi:10.1016/S0140-6736(17)30182-4.
  • Costello SP, Hughes PA, Waters O, Bryant RV, Vincent AD, Blatchford P, Katsikeros R, Makanyanga J, Campaniello MA, Mavrangelos C, et al. Effect of fecal microbiota transplantation on 8-week remission in patients with ulcerative colitis: a randomized clinical trial. Jama. 2019;321(2):156–164. doi:10.1001/jama.2018.20046.
  • Ishikawa D, Sasaki T, Osada T, Kuwahara-Arai K, Haga K, Shibuya T, Hiramatsu K, Watanabe S. Changes in intestinal microbiota following combination therapy with fecal microbial transplantation and antibiotics for ulcerative colitis. Inflammatory Bowel Dis. 2017;23(1):116–125. doi:10.1097/MIB.0000000000000975.
  • Kump P, Wurm P, Gröchenig HP, Wenzl H, Petritsch W, Halwachs B, Wagner M, Stadlbauer V, Eherer A, Hoffmann KM, 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(1):67–77. doi:10.1111/apt.14387.
  • Moayyedi P, Surette MG, Kim PT, Libertucci J, Wolfe M, Onischi C, Armstrong D, Marshall JK, Kassam Z, Reinisch W, et al. Fecal microbiota transplantation induces remission in patients with active ulcerative colitis in a randomized controlled trial. Gastroenterology. 2015;149(1):102–109. doi:10.1053/j.gastro.2015.04.001.
  • Holster S, Lindqvist CM, Repsilber D, Salonen A, de Vos WM, Konig J, et al. The effect of allogenic versus autologous fecal microbiota transfer on symptoms, visceral perception and fecal and mucosal microbiota in irritable bowel syndrome: a randomized controlled study. Clin Transl Gastroenterol. 2019;10(4):e00034.
  • Huttner BD, de Lastours V, Wassenberg M, Maharshak N, Mauris A, Galperine T, Zanichelli V, Kapel N, Bellanger A, Olearo F, et al. 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. 2019;25(7):830–838. doi:10.1016/j.cmi.2018.12.009.
  • Sood A, Mahajan R, Singh A, Midha V, Mehta V, Narang V, Singh T, Singh Pannu A. Role of faecal microbiota transplantation for maintenance of remission in patients with ulcerative colitis: a pilot study. J Crohns Colitis. 2019;13(10):1311–1317. doi:10.1093/ecco-jcc/jjz060.
  • Tian H, Ge X, Nie Y, Yang L, Ding C, McFarland LV, Zhang X, Chen Q, Gong J, Li N. Fecal microbiota transplantation in patients with slow-transit constipation: A randomized, clinical trial. PLoS ONE. 2017;12(2):e0171308.
  • Philips CA, Phadke N, Ganesan K, Ranade S, Augustine P. Corticosteroids, nutrition, pentoxifylline, or fecal microbiota transplantation for severe alcoholic hepatitis. Indian J Gastroenterol. 2018;37(3):215-225.
  • Ren YD, Ye ZS, Yang LZ, Jin LX, Wei WJ, Deng YY, Chen XX, Xiao CX, Yu XF, Xu HZ, et al. Fecal microbiota transplantation induces hepatitis B virus e-antigen (HBeAg) clearance in patients with positive HBeAg after long-term antiviral therapy. Hepatology. 2017;65(5):1765–1768. doi:10.1002/hep.29008.
  • Halkjær SI, Christensen AH, Lo BZS, Browne PD, Günther S, Hansen LH, Petersen AM. Faecal microbiota transplantation alters gut microbiota in patients with irritable bowel syndrome: results from a randomised, double-blind placebo-controlled study. Gut. 2018;67(12):2107–2115. doi:10.1136/gutjnl-2018-316434.
  • Wang S, Xu M, Wang W, Cao X, Piao M, Khan S, Yan F, Cao H, Wang B. Systematic review: adverse events of fecal microbiota transplantation. PLoS One. 2016;11(8):e0161174. doi:10.1371/journal.pone.0161174.
  • Administration FaD. Important safety alert regarding use of fecal microbiota for transplantation and risk of serious adverse reactions due to transmission of multi-drug resistant organisms. FDA; 2019 [updated 13/06/19.
  • FDA US. Safety alert regarding use of fecal microbiota for transplantation and risk of serious adverse events likely due to transmission of pathogenic organisms: FDA; 2020 [accessed 2020 Mar 12]. https://www.fda.gov/vaccines-blood-biologics/safety-availability-biologics/safety-alert-regarding-use-fecal-microbiota-transplantation-and-risk-serious-adverse-events-likely.
  • Openbiome. Openbiome quality and safety program openbiome; 2019. [accessed 2019 Dec 19] https://www.openbiome.org/safety.
  • Cammarota G, Ianiro G, Tilg H, Rajilic-Stojanovic M, Kump P, Satokari R, Harry S, Perttu A, Cristina P, Ailsa H, Jonathan S, Marina Aloi, et al. European consensus conference on faecal microbiota transplantation in clinical practice. Gut. 2017;66(4):569–580. doi:10.1136/gutjnl-2016-313017.
  • Trubiano JA, Cheng AC, Korman TM, Roder C, Campbell A, May ML, Blyth CC, Ferguson JK, Blackmore TK, Riley TV, et al. 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. 2016;46(4):479–493. doi:10.1111/imj.13027.
  • Bajaj JS, Gavis EA, Fagan A, Wade JB, Thacker LR, Fuchs M, Patel S, Davis B, Meador J, Puri P, et al. A randomized clinical trial of fecal microbiota transplant for alcohol use disorder. Hepatology. 2020. doi:10.1002/hep.31496. Epub ahead of print. PMID: 32750174..
  • Dinan TG, Cryan JF. Gut microbiota: a missing link in psychiatry. World Psychiat. 2020;19(1):111–112. doi:10.1002/wps.20726.