1,213
Views
2
CrossRef citations to date
0
Altmetric
Review

Fecal microbiota transplantation in hematopoietic cell transplant and cellular therapy recipients: lessons learned and the path forward

&
Article: 2229567 | Received 22 Apr 2023, Accepted 21 Jun 2023, Published online: 29 Jun 2023

ABSTRACT

Disruptions to the gut microbiota have been associated with adverse outcomes including graft-versus-host disease, infections, and mortality after hematopoietic cell transplantation and cellular therapy. Evidence for causal links is accumulating, thus supporting therapeutic interventions targeting the microbiota with the goal of preventing and treating adverse outcomes. One such intervention is fecal microbiota transplantation (FMT) by which an entire community of gut microbiota is transferred to the patient with dysbiosis. As this approach in transplant and cellular therapy recipients is still in its infancy, no best approach has been defined and many open questions need to be addressed before FMT becomes a standard treatment. In this review, we highlight microbiota-outcome associations with the highest level of evidence, provide an overview of the main FMT trials, and suggest some paths forward.

This article is part of the following collections:
Gut Microbiota in Cancer Development and Treatment

Introduction

Critical role of the gut microbiota in tissue and organismal homeostasis

Harboring 1013 to 1014 microbial organisms, the gut represents the most heavily colonized organ in the human body.Citation1 This community of microbes, referred to as the gut microbiota, plays numerous roles in host physiology. To name a few, the gut microbiota is critical in (i) synthesis of essential amino acids and vitamins, and metabolism of indigestible dietary polysaccharides,Citation2 (ii) colonization resistance, the process by which the gut microbiota resists invasion by other microorganisms, thus protecting the host from infections,Citation3 (iv) maintenance of a robust intestinal barrier and its repair after injury,Citation4,Citation5 (v) regulation of intestinal and systemic immunity,Citation6,Citation7 and (vi) production of many metabolites that circulate in the bloodstream, with distant and organism-level physiological effects.Citation8 Not surprisingly, disruptions to the gut microbiota have been implicated in common diseases such as obesity,Citation9 depression,Citation10 and type 2 diabetes.Citation11 Recent evidence for the presence of axes such as the gut-brain axis,Citation12 gut-lung axis,Citation13 and even brain-gut-kidney axisCitation14 attest to the broad involvement of the gut microbiota in tissue and organismal homeostasis.

Hematopoietic cell transplantation and chimeric antigen receptor T-cell therapy

Hematopoietic cell transplantation (HCT) and chimeric antigen receptor (CAR) T-cells are potentially curative treatments for patients with hematologic malignancies. The HCT process starts with administration of a conditioning regimen consisting of chemotherapy with or without total body irradiation, followed by infusion of hematopoietic cells. Hematopoietic cells may be derived from the same patient (autologous) or another individual (allogeneic). Following the infusion of the graft, there is a period of profound immunosuppression lasting weeks to months, during which patients receive antibiotics to prevent and treat infections. Enteral nutrition is often compromised due to the toxicity of conditioning regimens to the upper and lower intestinal tract. Infections and graft-versus-host disease (GVHD; acute or chronic) are common complications of HCT.

Acute GVHD is the clinical outcome of an early alloimmune attack by donor T cells against host tissues. In intestinal acute GVHD, intestinal stem cells seem to be the main target in this attack.Citation15 Detailed reviews of the classic pathophysiology of acute GVHD have been publishedCitation16 and more recent translational aspects of this process have been highlighted.Citation17 A critical pathway in acute GVHD pathogenesis involves the effects of the conditioning regimen (esp. high-dose total body irradiation) used before HCT on the intestinal microenvironment. Conditioning regimens damage the intestinal barrier separating the intestinal microbiota from subepithelial immune cells. Intestinal tissue injury leads to the recruitment and activation of neutrophils, triggering acute GVHD. Tissue damage may occur directly by activated neutrophils.Citation18 In addition, activated neutrophils can migrate to mesenteric lymph nodes, a process dependent on the intestinal microbiota. Once in the mesenteric lymph nodes, these neutrophils present antigens to T cells via MHC class II and lead to their activation.Citation19 Conditioning also augments expression of microbiota-derived Toll-like receptor ligands and MHC class II on intestinal epithelial cells, triggering downstream pro-inflammatory pathways via macrophages, innate lymphoid type 1 cells, and conventional T cells.Citation20 The intestinal microbiota regulates intestinal tissue sensitivity to chemotherapy and radiation.Citation4,Citation5

CAR-T cells are a more recent immunotherapeutic treatment modality for patients with specific lymphoid malignancies.Citation21 CARs are synthetic receptors expressed by an immune effector cell, typically T cells, which recognize cognate antigens on target cancer cells. Before CAR T-cell infusion, low-dose lymphodepletion chemotherapy is administered. The consequent immunosuppression triggers the use of prophylactic and therapeutic antibiotics. Infection, cytokine release syndrome (CRS), and immune effector cell – associated neurotoxicity syndrome (ICANS) are common complications of CAR-T cell therapy.

In this review, we first discuss gut microbiota disruptions in HCT and CAR-T recipients and the potential clinical impact of these disruptions. Next, we focus on one of the strategies – fecal microbiota transplantation (FMT) – to repair microbiota injuries and improve clinical outcomes in these patients. We synthesize the strongest pieces of evidence from the rapidly expanding literature.

Gut microbiota disruptions in HCT and CAR-T therapy

The two most consistent features of dysbiosis repeatedly observed in HCT recipients are loss of alpha diversity and pathobiont overgrowth. Alpha diversity is measured by various indices of microbial richness (number of different taxa) and evenness (taxa relative abundances).Citation22 As HCT recipients frequently have a history of treatment for their underlying blood disorder, the process of dysbiosis may have already started by the time they are referred for HCT. As a result, when they arrive for HCT, the gut microbiota may show a “scar” of prior injuries.Citation23–25 In addition, after allo-HCT, the gut microbiota diversity further declines.Citation23,Citation26–28 Diversity loss is commonly associated with decreased abundance of obligate anaerobic commensalsCitation29,Citation30 and domination by a few species. Intestinal domination has been most consistently demonstrated for Enterococcus.Citation31–34

After the removal of various causes of gut microbiota injury, partial though slow resolution of damage seems to occur.Citation27,Citation35 However, published data on long-term trajectory of microbiota after HCT are limited. In the longest follow-up (median >6 years) thus far of the gut microbiota in allogeneic HCT recipients (59 patients), gut microbial diversity and the abundance of butyrate-producing bacteria continued to be lower than the age- and sex-matched healthy control,Citation36 supporting the microbiota scar theory.

Causes of gut dysbiosis in HCT and CAR-T therapy

Antibiotic exposure is arguably the leading cause of dysbiosis in HCT recipients. Piperacillin-tazobactam and carbapenems are the antibiotics most consistently associated with microbiota injury in HCT patients.Citation37–43 Potent activity against obligate anaerobic bacteria is a shared feature of these antibiotics. In murine studies, carbapenem use led to the expansion of mucus-degrading species Akkermansia muciniphilaCitation38 and Bacteroides thetaiotaomicron.Citation37 The use of anti-anaerobic antibiotics is also associated with Enterococcus expansion.Citation33

Conditioning regimensCitation44 and nutrition (enteral vs. parenteral, calorie-rich vs. calorie-restricted)Citation45–47 may influence the gut microbiota in HCT patients. Enteral nutrition is possibly beneficial for clinical outcomes and may improve microbiota indices but the evidence is weak and more rigorous trials are needed.Citation48 In a large study of allogeneic HCT patients combined with mechanistic murine studies, lactose drove Enterococcus expansion in the gut which was associated with lower overall survival and higher acute GVHD-related mortality.Citation32

The gut microbiota is modulated by several other intrinsic and extrinsic factors. Although evidence in the setting of HCT and CAR-T therapy is limited,Citation49 large population-based studies have found relations between the gut microbiota and variables including anthropometric measures (age, sex, body mass index), stool consistency, common diseases (e.g. irritable bowel syndrome, coronary heart disease, depression, anemia), non-antibiotic drugs (e.g. proton pump inhibitors, statins, laxatives, opioids, antiemetics), and smoking history (direct or indirect exposure).Citation50–53 Some of these variables are frequently present or relevant to HCT and cellular therapy recipients.

Gut dysbiosis and outcomes of HCT and CAR-T therapy

Associations have been reported between dysbiosis and various outcomes of HCT and CAR-T therapy. However, the most consistent associations have been for acute GVHD and transplant-related mortality (TRM; mortality of any cause other than relapse of the underlying malignancy).

Acute GVHD

Dysbiosis has been associated with more GVHD in several studies.Citation28,Citation54–58 Microbiota changes associated with acute GVHD or its mortality include loss of alpha diversity,Citation28 loss of butyrogenic bacteria,Citation54,Citation58 loss of Clostridia,Citation58 loss of Blautia,Citation28,Citation59 loss of Lachnospiraceae,Citation55 lower ratios of strict-to-facultative anaerobic bacteria,Citation58 colonization with oral Actinobacteria and Firmicutes,Citation55 expansion of picobirnaviruses,Citation56 Enterococcus domination,Citation32 and large fluctuations in microbial community composition (“microbial chaos”) in the early phase after HCT.Citation57 Certain microbiome-associated systemic metabolomic alterations can be detected at the onset of acute GVHD. In one analysis, the concentration of several microbiota-derived metabolites (aryl hydrocarbon receptor ligands, bile acids, and plasmalogens) at the onset of acute GVHD was significantly different from healthy donors.Citation60

We recently analyzed a large single-center database of allogeneic HCT recipients (>2,000 patients between 2010–2021).Citation61 A total of 94 variables including 17 antibacterial antibiotic classes used between 7 days before and 30 days after transplant (weekly intervals) along with relevant clinical and demographic variables were analyzed. We applied three orthogonal statistical methods: conventional Cox proportional hazard regression, marginal structural models, and machine learning. Marginal structural models consider antibiotic exposures on a given day as a function of antibiotic exposures on the previous day and the baseline covariates. By machine learning, we attempted to reveal non-linear, more complex associations between antibiotic use and acute GVHD. Exposure to carbapenems during weeks 1 and 2 after HCT and exposure to combinations of penicillins with a β-lactamase inhibitor (e.g. piperacillin-tazobactam) during week 1 after HCT were most consistently associated with increased risk for acute GVHD. These two classes of antibiotics have been associated with acute GVHD in other studies as well.Citation37–43 Intriguingly, the use of metronidazole, another strong anti-anaerobic antibiotic, led to less acute GVHD in a seminal randomized trial.Citation62

Preservation of the gut microbiota may be involved or even required in the success of novel therapeutics in prevention of acute GVHD. In a phase 2 trial of tocilizumab (an interleukin-6 inhibitor) used for GVHD prophylaxis, the expected loss of microbiota diversity after HCT was less than in a historical control cohort. Similarly, Enterococcus domination was less frequent among patients receiving tocilizumab than historical controls.Citation63 In the therapeutic setting, a phase 2 trial of a novel recombinant human interleukin-22 dimer used in combination with systemic corticosteroids for frontline treatment of lower gastrointestinal acute GVHD showed expansion of commensal anaerobes and greater microbiota diversity in responders.Citation64

The gut virome has also been associated with the risk of acute GVHD. A viral ‘bloom’ seems to occur after HCT, accompanied with an expansion of vertebrate viral sequences following transplantation. Patients developing gastrointestinal acute GVHD had increased presence and abundance of specific DNA viruses (anelloviruses, herpesviruses, papillomaviruses and polyomaviruses) over time and lower phage richness. Picobirnaviruses were predictive of subsequent intestinal acute GVHD.Citation56

Transplant-related mortality

There is a repeatedly observed association between low gut microbiota diversity and TRM after allogeneic HCT.Citation23,Citation65 Acute GVHD and infection are the leading causes of TRM in HCT recipients. A specific timeframe when dysbiosis may impact mortality is weeks 2 and 3 after HCT, coinciding with hematopoietic engraftment.Citation23,Citation30 Loss of Blautia and expansion of Enterococcus have been associated with acute GVHD-related mortality.Citation32,Citation59 Loss of urinary 3-indoxyl sulfate and butyrogenic bacteria, associated with reduction of Lachnospiraceae and Ruminococcaceae and expansion of Bacilli, in the first 10 days after allogeneic HCT was predictive of increased TRM.Citation30,Citation54

Other outcomes after HCT

Some data suggest associations between the gut microbiota and bloodstream infection (BSI),Citation33,Citation66,Citation67 immune reconstitution,Citation68,Citation69 respiratory tract infections,Citation70,Citation71 CDI,Citation72 chronic GVHD,Citation73 and disease relapseCitation74 after HCT.

Outcomes after CAR-T therapy

Microbiota results from CAR-T studies are more recent. In one study, exposure to piperacillin/tazobactam and carbapenems within 4 weeks before CAR-T infusion was associated with worse survival and increased neurotoxicity. Baseline alpha diversity, as well the abundance of Ruminococcus, Bacteroides, and Faecalibacterium were associated with complete response to therapy in patients with B-cell malignancies.Citation75 These associations were independent of several other variables potentially reflecting more aggressive disease and disease-related complications. Whether exposure to oral vancomycin may have the same association could not be evaluated because exposure to this antibiotic in the absence of piperacillin/tazobactam and carbapenems was infrequent. In another study of B-cell lymphoma patients, piperacillin-tazobactam, meropenem, cefepime or ceftazidime use within 3 weeks before CAR-T infusion was associated with lower response to treatment. Focusing on patients not exposed to these antibiotics, the analysis revealed several bacteria associated with CAR-T response. These bacteria included Bifidobacterium longum, Bacteroides eggerthii, Ruminococcus lactaris, Eubacterium spp. CAG 180, Akkermansia muciniphila, Erysipelatoclostridium ramosum, and Bacteroides stercoris.Citation76

Mechanistic evidence

The possible causal link between microbiota and subsequent clinical outcomes is unclear. In the human setting, it is difficult to eliminate confounding with certainty. Conditioning-induced intestinal barrier damage, peak antibiotic exposure, altered nutrition, and maximum microbiota damage tend to occur about the same time after HCT, making it difficult to determine the effect of each one in isolation. As an example, and given the importance of intestinal damage in the pathogenesis of acute intestinal GVHD,Citation77 could intestinal damage be the underlying cause for both microbiota injury and acute GVHD without the latter two being causally linked? Mechanistic evidence for causality is accumulating in murine models. However, whether results from controlled murine experiments can be generalized to the human setting is unclear. Germ-free mice, a common and useful model for microbiota studies, do not represent antibiotic-treated patients undergoing HCT as the gut microbiota of patients often contains a large microbial community even after heavy antibiotic exposure. Through coprophagy, specific pathogen-free (SPF) mice may replenish their gut microbiota from their own stool and the stool of their co-housed mice. Finally, a fundamental unanswered question is whether microbiota effects are primarily mediated by individual taxa (e.g. Enterococcus, Akkermansia) or community-level properties of the microbiota (e.g. diversity). Complicating the matters is the fact that community-level properties such as alpha diversity are frequently associated with the relative abundance of specific taxa. For example, enterococcal domination is very common in low-diversity communities depleted of obligate anaerobic commensals after HCT.Citation26,Citation32,Citation33

Acute GVHD

Loss of short-chain fatty acids (SCFAs; generated from fermentation of dietary fiber) and indole (a tryptophan degradation product) are two metabolic changes that have been repeatedly observed in the setting of dysbiosis in HCT recipients. These microbiota-derived metabolites have immunosuppressiveCitation78–80 and gut barrier enhancing effectsCitation79–84 and may protect against acute (indole)Citation79 and chronic (butyrate/propionate)Citation73 GVHD. Although most results thus far have been in murine studies, cumulative evidence suggests involvement of these metabolites in the outcomes of HCT in patients. In a pilot pharmacomicrobiomics study with 20 adults undergoing allogeneic HCT using mycophenolate mofetil (MMF) and tacrolimus for GVHD prophylaxis, the association between the gut microbiota and enterohepatic recirculation (EHR) of mycophenolic acid was evaluated. Several Bacteroides species (B. vulgatus, B. stercoris, and B. thetaiotaomicron) were more abundant in the high vs. low EHR group,Citation85 introducing the possibility that the gut microbiota may modulate acute GVHD risk via differential metabolism of prophylactic medications. As MMF is a key drug in GVHD prophylaxis in the increasingly more common HLA-haploidentical HCT, more pharmacomicrobiomics studies are warranted.

Meropenem treatment of mice undergoing allogeneic HCT led to the expansion of B. thetaiotaomicron and aggravation of acute colonic GVHD.Citation37 Through its mucolytic activity, B. thetaiotaomicron caused thinning of the colonic mucus layer and decreased xylose levels within the lumen, a process which was prevented by oral supplementation of xylose. Another study attempted to block the successful entry of live bacteria through the intestinal wall by immunizing mice against the conserved microbial surface poly-N-acetylglucosamine. By depleting bacteria penetrating the gut barrier without altering microbiota diversity, this approach reduced bacterial-induced neutrophil activation and acute GVHD.Citation86

In another murine study, Enterococcus expansion, a predictor of higher acute GVHD risk in patients, was dependent on lactose. Oral lactose depletion attenuated Enterococcus outgrowth and reduced the severity of GVHD, supporting a causal link.Citation32 In the opposite direction, the process of immune attack during acute GVHD can cause damage to the gut microbiota by increasing oxygen tension in the normally hypoxic intestinal lumen, leading to a loss of obligate anaerobic commensals. This in turn worsens intestinal pathology. Importantly, oral iron chelation in murine models mitigated dysbiosis and reduced the GVHD severity, supporting a causal link.Citation87

Bloodstream infection

By thinning the intestinal barrier, expanded A. muciniphila may increase the translocation of bacteria, their components (e.g. flagellin, lipopolysaccharide), and metabolites to the bloodstream.Citation46,Citation88,Citation89 Decreased oral intake potentiates expansion of mucolytic bacteria because they can use mucin as a nutrient.Citation46,Citation90 Enterococcus expansion in the gut has been repeatedly associated with the risk of enterococcal BSICitation33,Citation91 and Gram-negative bacteria domination of the gut microbiota with Gram-negative bacteremia.Citation67 The mechanism seems to be a concentration effect, by which dominant bacteria have a higher chance of passing through the intestinal barrier.

CAR-T efficacy

Findings from a small retrospective study in patients with B-cell acute lymphoblastic leukemia treated with CAR-T suggested better CAR-T expansion in patients exposed to oral vancomycin before infusion.Citation92 In a murine CD19+ lymphoma model, microbiota conditioning with vancomycin combined with CD19 CAR-T therapy enhanced cross-presentation of tumor-associated antigens and improved tumor control compared to CAR-T alone.Citation92 In another study, peptidoglycan biosynthesis was the metabolic pathway most strongly associated with disease progression after CAR-T, independently of demographic or clinical variables.Citation76

Fecal microbiota transplantation in HCT

HCT recipients experience significant disruptions to their gut microbiota,Citation93 with implications for clinical outcomes such as mortality,Citation23,Citation65 acute GVHD,Citation28,Citation37,Citation38,Citation54–58,Citation79 poor immune reconstitution,Citation69,Citation94,Citation95 and disease relapse.Citation74 Similar to HCT recipients, specific antibiotic exposures and gut microbiota changes in CAR-T recipients have been associated with treatment response, mortality, and ICANS.Citation75,Citation76 Inspired by these associations, strategies to mitigate dysbiosis to improve clinical outcomes after HCT and CAR-T therapy have been a subject of recent interest.Citation96 Fecal microbiota transplantation (FMT) is one such strategy by which gut microbial communities are transferred as a whole to the patient to prevent or ameliorate dysbiosis. FMT is currently approved only for the treatment of multiply recurrent or refractory Clostridioides difficile infection (CDI).Citation97

Restoring the commensal microbiota and eradicating pathogens including multi-drug resistant organisms (MDROs) is the main idea in the prophylactic use of FMT after HCT. Clinical outcomes that might improve with this approach include infection rate, acute (and perhaps chronic) GVHD, and TRM. In the therapeutic setting after HCT, FMT has been used for the treatment of refractory CDICitation98–101 and steroid-refractory acute GVHD (see below). There has been a large variability in approach, as we will discuss in the following sections. A summary of the key features of the previous FMT trials in the field with at least 10 patients treated with FMT is shown in Citation102–108

Table 1. Study characteristics.

Table 2. FMT administration details.

Table 3. FMT safety and efficacy.

Eradication of antibiotic-resistant bacteria (ARB)

The efficacy of FMT in eradicating highly refractory cases of CDI has inspired its use in HCT recipients (both before and after HCT) with MDRO colonization. The ultimate goal of such attempts would be to prevent clinical infections with these organisms. In the largest report thus far with clinical outcomes,Citation109 11 patients received third-party FMT at least 2 weeks before their planned HCT. Eight of these patients proceeded to allogeneic HCT. A non-randomized comparator group was used. Compared to the 6-month period before FMT, patients receiving FMT had a shorter hospitalization and fewer days on carbapenems during HCT. This change did not occur in the comparator group. In another retrospective study,Citation110 8 MDRO-colonized patients received third-party FMT before HCT. When compared to their pair-matched controls without MDRO colonization, 1-year survival was not different. In contrast, MDRO-colonized patients who did not receive FMT had a significantly lower 1-year survival compared to their matched controls without MDRO organization. These findings suggested that the mortality increment due to MDRO colonization may be ameliorated by FMT. Based on limited available data, successful decolonization seems to occur in 50–70% of MDRO-colonized HCT recipients.Citation104

Clinical outcomes after MDRO decolonization may improve, but the evidence is weak. Most studies have been small, retrospective or non-randomized, and without a primary clinical endpoint. With improved supportive care and antibiotics, a randomized trial to demonstrate better survival after FMT vs. placebo in MDRO-colonized patients will require a large sample size which may not be feasible. BSI as a clinical endpoint may be easier to study, and it is an important endpoint because BSIs are associated with re-hospitalization, increased healthcare costs, and more dysbiosis due to antibiotic exposure which could in turn increase the risk of GVHD.

Restoration of microbiota diversity and commensals

Two randomized trials thus far have evaluated the effect of FMT on the microbiota after allogeneic HCT. In the first trial, 25 allogeneic HCT recipients were randomized after neutrophil engraftment to receive autologous FMT vs. no intervention.Citation111 A unique aspect of this study was that only patients with a low fecal abundance of Bacteroidetes were treated. FMT improved microbiota alpha diversity and compositional recovery toward baseline including Lachnospiraceae, Ruminococcaceae, and Bacteroidetes which contain many commensal bacteria. Shotgun metagenomic sequencing suggested enrichment in the control group in antimicrobial-peptide resistance and pathways associated with microbial virulence, biofilm formation, and bacterial flagella assembly. In the second trial,Citation108 we randomized 74 patients to receive FMT or placebo (2:1 ratio) after neutrophil engraftment. FMT enhanced recovery of alpha diversity and Collinsella and reduced the abundance of expanded genera Enterococcus and Dialister. Notably, Blautia recovered to the same extent in both arms, arguing against an essential FMT impact. More details about the design of this trial and clinical outcomes are provided in the next section.

FMT as a prophylactic intervention to improve transplant outcomes

We recently reported the results of our randomized placebo-controlled FMT trial in allogeneic HCT recipients with 4-month all-cause infection rate as the primary endpoint.Citation108 The trial also had an independent cohort of patients with acute myeloid leukemia receiving induction chemotherapy, which is not of primary interest for this review. Five standardized third-party oral capsules were administered per dose. Strict donor and product screening were implemented to minimize the risk of pathogen transmission. The first treatment was given at the time of neutrophil recovery and at least 2 days after discontinuation of antibacterial antibiotics. Patients re-exposed to antibacterial antibiotics after dose 1 received up to 2 more doses. Although infection rate was numerically lower after FMT than placebo, the difference did not reach statistical significance (P = 0.49). Acute toxicities were limited to mild gastrointestinal symptoms. The incidence of acute GVHD (secondary endpoint) was higher in the FMT arm, but the small number of events and imbalance in GVHD prophylaxis regimens between the two arms prohibited a firm conclusion. A randomized placebo-controlled study powered to evaluate the effect of FMT on preventing acute GVHD rate is needed.

FMT as a treatment for refractory acute GVHD

Refractory acute GVHD is a life-threatening complication of HCT in need of novel therapeutics. Current treatment is only partially effective and most patients who are refractory to two lines of treatment die. Standard treatment is based on the use of immunosuppressive medications with a wide range of toxicities and does not address other aspects of GVHD pathogenesis such as tissue repair and tolerance.

In the first use of FMT to treat steroid-refractory acute GVHD,Citation112 three patients with steroid-refractory and one with steroid-dependent acute intestinal GVHD received 1–2 sessions of third-party FMT via a nasoduodenal tube (34–307 grams). Three patients achieved a complete response and 1 achieved a partial response, with no major treatment-related toxicity. The 3 complete responders were not exposed to antibacterial antibiotics during the two weeks after FMT, except trimethoprim-sulfamethoxazole in one patient and a brief course of levofloxacin in another. In contrast, the patient with a partial response was heavily exposed to anti-anaerobic antibiotics after FMT. It is tempting to hypothesize that such exposure decreased the efficacy of FMT, but it is also possible that the use of these antibiotics reflected a worse case of GVHD, with less response to FMT even without antibiotic exposure. Effector regulatory T cells increased in the circulation after FMT, suggestive of a systemic anti-inflammatory response. Taxa that increased after FMT in complete responders included Bacteroides, Faecalibacterium, Lactobacillus, and Bifidobacterium. Escherichia was the dominant taxon after FMT in the partial responder. FMT did not normalize microbiota diversity, suggesting that full restoration of the microbiota may not be necessary for clinical response.

Several reports were subsequently published on the use of FMT in patients with refractory acute GVHD.Citation102,Citation103,Citation105,Citation107,Citation113 In the largest prospective study thus far,Citation107 41 patients with steroid-refractory acute gut GVHD received third-party FMT (23 patients). A control group of 18 patients who received other treatments was included in this non-randomized study. In addition to FMT, the FMT cohort received next-line immunosuppressive treatment, thus FMT was an adjunct treatment rather than a replacement for standard immunosuppression. The FMT cohort received 1–6 FMT treatments via a nasojejunal or gastric tube. Response and survival were higher in the FMT cohort, without increased toxicity. The relative abundance of Firmicutes and Bacteroidetes seemed to increase after FMT while the relative abundance of Proteobacteria seemed to decrease, although a robust analysis was not possible due to the small number of stool samples. In another prospective single-arm study,Citation105 15 patients with steroid-refractory or -dependent acute intestinal GVHD safely received one third-party FMT via nasoduodenal infusion 2 hours after a bowel lavage. Prior immunosuppressive therapy was not changed. Ten patients achieved a complete response within a month, associated with signals for improved alpha diversity, partial donor microbiota engraftment, and increased abundance of Clostridiales, Blautia, and predicted butyrate-producing taxa. However, confidence intervals were large, likely contributing to non-significant differences between responders and non-responders.

Although a randomized trial of FMT for the treatment of refractory acute GVHD has not been published, the available evidence suggests efficacy. An important question to be addressed is the mechanism by which microbiota restoration may improve acute GVHD outcomes. Factors other than a poorly controlled alloimmune attack seem to be involved in steroid-refractory acute GVHD. Tissue tolerance, a concept adopted from non-transplant settings and recently introduced in the HCT setting, posits that tissue-intrinsic factors may have a key role in both progression and resolution of acute GVHD.Citation114 Such factors do not oppose the attacking agents (e.g. T cells) directly, but rather help with tissue resistance and repair. Whether and how the microbiota may contribute to this process is unclear, but ideas such as butyrate serving as a growth factor for injured colonocytes could be entertained.Citation81,Citation82,Citation115,Citation116

Toxicity of FMT

Besides occasional and typically transient gastrointestinal side effects (nausea, bloating, abdominal pain, diarrhea) that tend to happen in the first 1–2 days after FMT, the main risk associated with FMT is pathogen transmission. The introduction of millions of additional bacteria to the gut, particularly in the setting of profound immunosuppression and intestinal barrier damage after HCT can potentially result in microbial translocation to the bloodstream infection and life-threatening sepsis.Citation117 The risk is not limited to bacteria. As an example, eosinophilic gastroenteritis due to Norovirus transmitted via FMT in an allogeneic HCT patient has been reported.Citation118 Major regulatory organizations have established strict rules for screening FMT donors and their stool. It is noteworthy that a BSI after FMT does not necessarily point to the FMT product as the origin of infection. Strain-level analysis of 13 BSI events after FMT in patients with steroid-refractory or -dependent acute GVHD did not find the BSI-associated strains in the FMT product.Citation119 Several bacteria isolated from blood cultures (Enterococcus faecium, Escherichia coli, Pseudomonas aeruginosa, and Acinetobacter baumannii) were present in stools sample before BSI, suggesting that the patients’ own microbiota was likely the source of infection. In our randomized trial, we found no bloodstream infections attributable to FMT.Citation108

An arbitrary interval after neutrophil engraftment has been commonly used to administer FMT after HCT, an approach based on the perceived increased risk of infection in a neutropenic patient. However, no compelling evidence indicates the pre-engraftment period as an especially high-risk period for FMT. As pre-engraftment HCT patients are severely immunosuppressed and at high risk for infection even without FMT, it is important to perform FMT in such patients only in the context of clinical trials, and ideally in a randomized setting to permit distinguishing FMT-related safety signals from baseline risks.

Concluding remarks

What are the lessons learned?

Several associations have been reported between intestinal dysbiosis and poor clinical outcomes after HCT and cellular therapy. The evidence is currently stronger in the HCT setting, but recent studies have found similar associations after CAR-T therapy. In particular, early loss of obligate anaerobic commensal bacteria and expansion of Enterococcus and mucolytic species may potentiate complications and lead to poor outcomes. The multitude of factors mediating both clinical outcomes and gut microbiota in these settings makes causal links difficult to establish in humans. Controlled murine experiments have suggested mechanistic connections, but generalization to the human setting is not trivial. In particular, procedures to access the mucosal-adherent microbiota in this patient population are risky and stool samples represent luminal microbiota, which is different from the likely more relevant mucosal-adherent microbiota. One approach to demonstrate a causal link in humans is by randomized trials. Randomization reduces the imbalance between the arms in measured and unmeasured variables. Different outcomes between the arms in such a design would strongly suggest causality. The only completed randomized controlled FMT trial after allogeneic HCT with primary (infections) and secondary (acute GVHD) clinical endpoints did not improve outcomes despite substantial amelioration of dysbiosis.Citation108

What is the path forward?

Novel aspects of microbiota and its role in tissue homeostasis need to be translated to clinical trials. Although most attention has been paid to the bacteria in the setting of FMT, the possible role of other microorganisms such as viruses (e.g. phages) is becoming more clear. Removing the bacterial component of donor stool by sterile filtration maintains FMT efficacy on gut microbiota composition and host metabolome.Citation120 Sterile fecal filtrates contain bacterial debris, proteins, antimicrobial compounds, metabolic products, oligonucleotides/DNA, and viruses, but not intact bacteria. Such filtrates (a.k.a. fecal virome transplantation) have been tested with success for the treatment of recurrent CDI in small seriesCitation121 and may have a role in HCT recipients too. Currently, there are no published FMT trials in CAR-T recipients, and to our knowledge, no such trial is ongoing in this setting. Given recent findings about microbiota associations with CAR-T efficacy and toxicity, FMT should be tested in clinical trials in this setting. The interval between CAR-T referral and lymphodepletion chemotherapy is often several weeks long, providing sufficient opportunity for FMT to modulate the microbiota in patients deemed high-risk based on the available associational data. If the association between microbiota injury and disease progression after CAR-T therapy is confirmed in more cohorts, pre- or post-CAR FMT may be a testable strategy to boost treatment effect and improve cure rates.

What are the open questions that need to be addressed before FMT becomes a standard therapeutic regimen?

From a mechanistic standpoint, the determinants of donor microbiota engraftment in the HCT setting are unknown. In addition, the effect of FMT on non-bacterial compartments of the microbiome (e.g. virome, mycobiome) is rather unexplored.Citation122

The optimal timing and schedule of administration and minimum required dose of FMT are unknown. With rigorous screening of donors and stools to minimize the risk of pathogen transmission, the existence of a specific neutrophil count threshold for FMT is questionable. Careful examination of FMT safety during severe neutropenia in a properly designed clinical trial with safety as the primary endpoint would be informative as the pre-engraftment period when antibiotic pressure tends to be maximal may offer a new window of opportunity for intervention on the microbiota. Most studies have administered FMT on a single day or on 2 consecutive days. Whether a more sustained schedule extending for several days and potentially delivering a larger dose of microbiota is beneficial is unknown. The route of administration is another unknown factor potentially influencing efficacy. Besides the oral encapsulated form,Citation103,Citation106,Citation108,Citation113 FMT has been administered by enema,Citation104,Citation111,Citation123 via nasogastric/nasoduodenal tube,Citation104,Citation105,Citation109,Citation112,Citation124 or by upper127 or lower endoscopy.Citation102 Another open question concerns the FMT source. Both third-partyCitation102,Citation104,Citation105,Citation108,Citation109,Citation124,Citation125 and autologousCitation111 products have been used. While an autologous product may be simpler and less expensive to manufacture, most HCT recipients have had microbiota insults before referral for transplant and the efficacy of a product manufactured from a “scarred” microbiota needs further investigation. Similarly, whether a single “super donor”,Citation126 several donors (one used for each patient), or pooled donors provide better outcomes is unknown. Pooling stool from multiple donors may generate a more diverse microbiota in the product, but the value of an extra-high diversity beyond what an average healthy individual has is unknown. Whether concurrent use of antibiotics may be detrimental to FMT efficacy and safety is also unclear. Related to this point, the optimal washout after the last antibiotic use has not been established. Finally, the efficacy of “microbiota conditioning” by antibiotic gut decontamination before FMT to increase donor microbiota engraftment has not been determined.

Disclosure statement

No potential conflict of interest was reported by the author(s).

Additional information

Funding

The author(s) reported there is no funding associated with the work featured in this article.

References

  • Turnbaugh PJ, Ley RE, Hamady M, Fraser-Liggett CM, Knight R, Gordon JI. The human microbiome project. Nature. 2007;449(7164):804–18. doi: 10.1038/nature06244.
  • Backhed F, Ley RE, Sonnenburg JL, Peterson DA, Gordon JI. Host-bacterial mutualism in the human intestine. Science. 2005;307(5717):1915–1920. doi: 10.1126/science.1104816.
  • Kim S, Covington A, Pamer EG. The intestinal microbiota: antibiotics, colonization resistance, and enteric pathogens. Immunol Rev. 2017;279(1):90–105. doi: 10.1111/imr.12563.
  • Crawford PA, Gordon JI. Microbial regulation of intestinal radiosensitivity. Proc Natl Acad Sci USA. 2005;102(37):13254–13259. doi: 10.1073/pnas.0504830102.
  • Guo H, Chou W-C, Lai Y, Liang K, Tam JW, Brickey WJ, Chen L, Montgomery ND, Li X, Bohannon LM, et al. Multi-omics analyses of radiation survivors identify radioprotective microbes and metabolites. Science. 2020;370(6516):eaay9097. doi: 10.1126/science.aay9097.
  • Ivanov II, Atarashi K, Manel N, Brodie EL, Shima T, Karaoz U, Wei D, Goldfarb KC, Santee CA, Lynch SV, et al. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell. 2009;139(3):485–498. doi: 10.1016/j.cell.2009.09.033.
  • Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly-Y M, Glickman JN, Garrett WS. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science. 2013;341(6145):569–573. doi: 10.1126/science.1241165.
  • Diener C, Dai CL, Wilmanski T, Baloni P, Smith B, Rappaport N, Hood L, Magis AT, Gibbons SM. Genome–microbiome interplay provides insight into the determinants of the human blood metabolome. Nat Metab. 2022;4(11):1560–1572. doi: 10.1038/s42255-022-00670-1.
  • Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444(7122):1027–1031. doi: 10.1038/nature05414.
  • Radjabzadeh D, Bosch JA, Uitterlinden AG, Zwinderman AH, Ikram MA, van Meurs JBJ, Luik AI, Nieuwdorp M, Lok A, van Duijn CM, et al. Gut microbiome-wide association study of depressive symptoms. Nat Commun. 2022;13(1):7128. doi: 10.1038/s41467-022-34502-3.
  • Qin J, Li Y, Cai Z, Li S, Zhu J, Zhang F, Liang S, Zhang W, Guan Y, Shen D, et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature. 2012;490(7418):55–60. doi: 10.1038/nature11450.
  • Morais LH, Schreiber HL, Mazmanian SK. The gut microbiota–brain axis in behaviour and brain disorders. Nat Rev Microbiol. 2020;19(4):241–255. doi: 10.1038/s41579-020-00460-0.
  • Dang AT, Marsland BJ. Microbes, metabolites, and the gut–lung axis. Mucosal Immunol. 2019;12(4):843–850. doi: 10.1038/s41385-019-0160-6.
  • Yang T, Richards EM, Pepine CJ, Raizada MK. The gut microbiota and the brain–gut–kidney axis in hypertension and chronic kidney disease. Nat Rev Nephrol. 2018;14(7):442–456. doi: 10.1038/s41581-018-0018-2.
  • Fu Y-Y, Egorova A, Sobieski C, Kuttiyara J, Calafiore M, Takashima S, Clevers H, Hanash AM. T cell recruitment to the intestinal stem cell compartment drives immune-mediated intestinal damage after allogeneic transplantation. Immunity. 2019;51(1):90–103.e3. doi: 10.1016/j.immuni.2019.06.003.
  • Zeiser R, Blazar BR, Longo DL. Acute graft-versus-host disease — biologic process, prevention, and therapy. N Engl J Med. 2017;377(22):2167–2179. doi: 10.1056/NEJMra1609337.
  • Socié G, Kean LS, Zeiser R, Blazar BR. Insights from integrating clinical and preclinical studies advance understanding of graft-versus-host disease. J Clin Invest. 2021;131(12):e149296. doi: 10.1172/JCI149296.
  • Schwab L, Goroncy L, Palaniyandi S, Gautam S, Triantafyllopoulou A, Mocsai A, Reichardt W, Karlsson FJ, Radhakrishnan SV, Hanke K, et al. Neutrophil granulocytes recruited upon translocation of intestinal bacteria enhance graft-versus-host disease via tissue damage. Nat Med. 2014;20(6):648–654. doi: 10.1038/nm.3517.
  • Hülsdünker J, Ottmüller KJ, Neeff HP, Koyama M, Gao Z, Thomas OS, Follo M, Al-Ahmad A, Prinz G, Duquesne S, et al. Neutrophils provide cellular communication between ileum and mesenteric lymph nodes at graft-versus-host disease onset. Blood. 2018;131(16):1858–1869. doi: 10.1182/blood-2017-10-812891.
  • Koyama M, Mukhopadhyay P, Schuster IS, Henden AS, Hülsdünker J, Varelias A, Vetizou M, Kuns RD, Robb RJ, Zhang P, et al. MHC class II antigen presentation by the intestinal epithelium initiates graft-versus-host disease and is influenced by the microbiota. Immunity. 2019;51(5):885–98.e7. doi: 10.1016/j.immuni.2019.08.011.
  • June CH, Sadelain M. Chimeric antigen receptor therapy. N Engl J Med. 2018;379(1):64–73. doi: 10.1056/NEJMra1706169.
  • Walters KE, Martiny JBH, Nabout JC. Alpha-, beta-, and gamma-diversity of bacteria varies across habitats. PLoS One. 2020;15(9):e0233872. doi: 10.1371/journal.pone.0233872.
  • Peled JU, Gomes ALC, Devlin SM, Littmann ER, Taur Y, Sung AD, Weber D, Hashimoto D, Slingerland AE, Slingerland JB, et al. Microbiota as predictor of mortality in allogeneic hematopoietic-cell transplantation. N Engl J Med. 2020;382(9):822–834. doi: 10.1056/NEJMoa1900623.
  • Rashidi A, Maeser D, Kaiser T, Ebadi M, Rehman TU, Holtan SG, Weisdorf DJ, Khoruts A, Staley C. Microbiome swings with repeated insults. Br J Haematol. 2020;189(3):e94–6. doi: 10.1111/bjh.16509.
  • Rashidi A, Kaiser T, Holtan SG, Weisdorf DJ, Khoruts A, Staley C. Pre-transplant recovery of microbiome diversity without recovery of the original microbiome. Bone Marrow Transplant. 2019;54(7):1115–1117. doi: 10.1038/s41409-018-0414-z.
  • Rashidi A, Kaiser T, Graiziger C, Holtan SG, Rehman TU, Weisdorf DJ, Dunny GM, Khoruts A, Staley C. Gut dysbiosis during antileukemia chemotherapy versus allogeneic hematopoietic cell transplantation. Cancer. 2020;126(7):1434–1447. doi: 10.1002/cncr.32641.
  • Ingham AC, Kielsen K, Mordhorst H, Ifversen M, Aarestrup FM, Müller KG, Pamp SJ. Microbiota long-term dynamics and prediction of acute graft-versus-host disease in pediatric allogeneic stem cell transplantation. Microbiome. 2021;9(1):148. doi: 10.1186/s40168-021-01100-2.
  • Ilett EE, Jørgensen M, Noguera-Julian M, Nørgaard JC, Daugaard G, Helleberg M, Paredes R, Murray DD, Lundgren J, MacPherson C, et al. Associations of the gut microbiome and clinical factors with acute GVHD in allogeneic HSCT recipients. Blood Adv. 2020;4(22):5797–5809. doi: 10.1182/bloodadvances.2020002677.
  • Weber D, Hiergeist A, Weber M, Dettmer K, Wolff D, Hahn J, Herr W, Gessner A, Holler E. Detrimental effect of broad-spectrum antibiotics on intestinal microbiome diversity in patients after allogeneic stem cell transplantation: lack of commensal sparing antibiotics. Clin Infect Dis. 2019;68(8):1303–1310. doi: 10.1093/cid/ciy711.
  • Weber D, Oefner PJ, Hiergeist A, Koestler J, Gessner A, Weber M, Hahn J, Wolff D, Stämmler F, Spang R, et al. Low urinary indoxyl sulfate levels early after transplantation reflect a disrupted microbiome and are associated with poor outcome. Blood. 2015;126(14):1723–1728. doi: 10.1182/blood-2015-04-638858.
  • Holler E, Butzhammer P, Schmid K, Hundsrucker C, Koestler J, Peter K, Zhu W, Sporrer D, Hehlgans T, Kreutz M, et al. Metagenomic analysis of the stool microbiome in patients receiving allogeneic stem cell transplantation: loss of diversity is associated with use of systemic antibiotics and more pronounced in gastrointestinal graft-versus-host disease. Biol Blood Marrow Transplant. 2014;20(5):640–645. doi: 10.1016/j.bbmt.2014.01.030.
  • Stein-Thoeringer CK, Nichols KB, Lazrak A, Docampo MD, Slingerland AE, Slingerland JB, Clurman AG, Armijo G, Gomes ALC, Shono Y, et al. Lactose drives Enterococcus expansion to promote graft-versus-host disease. Science. 2019;366(6469):1143–1149. doi: 10.1126/science.aax3760.
  • Taur Y, Xavier JB, Lipuma L, Ubeda C, Goldberg J, Gobourne A, Lee YJ, Dubin KA, Socci ND, Viale A, et al. Intestinal domination and the risk of bacteremia in patients undergoing allogeneic hematopoietic stem cell transplantation. Clin Infect Dis. 2012;55(7):905–914. doi: 10.1093/cid/cis580.
  • van den Brink MRM, Taur Y, Pamer EG. Diversification and evolution of vancomycin-resistant Enterococcus faecium during intestinal domination. Infection. 2019;87(7):e00102–19. doi: 10.1128/IAI.00102-19.
  • Biagi E, Zama D, Nastasi C, Consolandi C, Fiori J, Rampelli S, Turroni S, Centanni M, Severgnini M, Peano C, et al. Gut microbiota trajectory in pediatric patients undergoing hematopoietic SCT. Bone Marrow Transplant. 2015;50(7):992–998. doi: 10.1038/bmt.2015.16.
  • Hino A, Fukushima K, Kusakabe S, Ueda T, Sudo T, Fujita J, Motooka D, Takeda AK, Shinozaki NO, Watanabe S, et al. Prolonged gut microbial alterations in post-transplant survivors of allogeneic haematopoietic stem cell transplantation. Br J Haematol. 2023;201(4):725–737. doi: 10.1111/bjh.18574.
  • Hayase E, Hayase T, Jamal MA, Miyama T, Chang C-C, Ortega MR, Ahmed SS, Karmouch JL, Sanchez CA, Brown AN, et al. Mucus-degrading Bacteroides link carbapenems to aggravated graft-versus-host disease. Cell. 2022;185(20):3705–19.e14. doi: 10.1016/j.cell.2022.09.007.
  • Shono Y, Docampo MD, Peled JU, Perobelli SM, Velardi E, Tsai JJ, Slingerland AE, Smith OM, Young LF, Gupta J, et al. Increased GVHD-related mortality with broad-spectrum antibiotic use after allogeneic hematopoietic stem cell transplantation in human patients and mice. Sci Transl Med. 2016;8(339):339ra71. doi: 10.1126/scitranslmed.aaf2311.
  • Elgarten CW, Li Y, Getz KD, Hemmer M, Huang Y-S, Hall M, Wang T, Kitko CL, Jagasia MH, Nishihori T, et al. Broad-spectrum antibiotics and risk of graft-versus-host disease in pediatric patients undergoing transplantation for acute leukemia: association of carbapenem use with the risk of acute graft-versus-host disease. Transplant Cell Ther. 2021;27(2):.e177.1–.e177.8. doi: 10.1016/j.jtct.2020.10.012.
  • Lee S-E, Lim J-Y, Ryu D-B, Kim TW, Park SS, Jeon Y-W, Yoon J-H, Cho B-S, Eom K-S, Kim Y-J, et al. Alteration of the intestinal microbiota by broad-spectrum antibiotic use correlates with the occurrence of intestinal graft-versus-host disease. Biol Blood Marrow Transplant. 2019;25(10):1933–1943. doi: 10.1016/j.bbmt.2019.06.001.
  • Tanaka JS, Young RR, Heston SM, Jenkins K, Spees LP, Sung AD, Corbet K, Thompson JC, Bohannon L, Martin PL, et al. Anaerobic antibiotics and the risk of graft-versus-host disease after allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2020;26(11):2053–2060. doi: 10.1016/j.bbmt.2020.07.011.
  • Hidaka D, Hayase E, Shiratori S, Hasegawa Y, Ishio T, Tateno T, Okada K, Goto H, Sugita J, Onozawa M, et al. The association between the incidence of intestinal graft-vs-host disease and antibiotic use after allogeneic hematopoietic stem cell transplantation. Clin Transplant. 2018;32(9):e13361. doi: 10.1111/ctr.13361.
  • Hanks CR, Slain D, Kanate AS, Wen S, Cumpston A. Impact of anti-anaerobic antibiotic activity on graft-versus-host disease in allogeneic hematopoietic stem cell transplant (HSCT) recipients at an institution that utilizes antibiotic cycling. Transpl Infect Dis. 2021;23(4):e13676. doi: 10.1111/tid.13676.
  • Shouval R, Waters NR, Gomes ALC, Zuanelli Brambilla C, Fei T, Devlin SM, Nguyen CL, Markey KA, Dai A, Slingerland JB, et al. Conditioning regimens are associated with distinct patterns of microbiota injury in allogeneic hematopoietic cell transplantation. Clin Cancer Res. 2023;29(1):165–173. doi: 10.1158/1078-0432.CCR-22-1254.
  • Andersen S, Staudacher H, Weber N, Kennedy G, Varelias A, Banks M, Bauer J. Pilot study investigating the effect of enteral and parenteral nutrition on the gastrointestinal microbiome post-allogeneic transplantation. Br J Haematol. 2020;188(4):570–581. doi: 10.1111/bjh.16218.
  • Schwabkey ZI, Wiesnoski DH, Chang C-C, Tsai W-B, Pham D, Ahmed SS, Hayase T, Ortega Turrubiates MR, El-Himri RK, Sanchez CA, et al. Diet-derived metabolites and mucus link the gut microbiome to fever after cytotoxic cancer treatment. Sci Transl Med. 2022;14(671):eabo3445. doi: 10.1126/scitranslmed.abo3445.
  • D’Amico F, Biagi E, Rampelli S, Fiori J, Zama D, Soverini M, Barone M, Leardini D, Muratore E, Prete A, et al. Enteral nutrition in pediatric patients undergoing hematopoietic sct promotes the recovery of gut microbiome homeostasis. Nutrients. 2019;11(12):11. doi: 10.3390/nu11122958.
  • Zama D, Gori D, Muratore E, Leardini D, Rallo F, Turroni S, Prete A, Brigidi P, Pession A, Masetti R. Enteral versus parenteral nutrition as nutritional support after allogeneic hematopoietic stem cell transplantation: a systematic review and meta-analysis. Transplant Cell Ther. 2021;27(2):e180.1–e180.8. doi: 10.1016/j.jtct.2020.11.006.
  • Nguyen CL, Markey KA, Miltiadous O, Dai A, Waters N, Sadeghi K, Fei T, Shouval R, Taylor BP, Liao C, et al. High-resolution analyses of associations between medications, microbiome, and mortality in cancer patients. Cell. 2023;186(12):2705–18.e17. doi: 10.1016/j.cell.2023.05.007.
  • Zhernakova A, Kurilshikov A, Bonder MJ, Tigchelaar EF, Schirmer M, Vatanen T, Mujagic Z, Vila AV, Falony G, Vieira-Silva S, et al. Population-based metagenomics analysis reveals markers for gut microbiome composition and diversity. Science. 2016;352(6285):565–569. doi: 10.1126/science.aad3369.
  • Falony G, Joossens M, Vieira-Silva S, Wang J, Darzi Y, Faust K, Kurilshikov A, Bonder MJ, Valles-Colomer M, Vandeputte D, et al. Population-level analysis of gut microbiome variation. Science. 2016;352(6285):560–564. doi: 10.1126/science.aad3503.
  • Maier L, Pruteanu M, Kuhn M, Zeller G, Telzerow A, Anderson EE, Brochado AR, Fernandez KC, Dose H, Mori H, et al. Extensive impact of non-antibiotic drugs on human gut bacteria. Nature. 2018;555(7698):623–628. doi: 10.1038/nature25979.
  • Vich Vila A, Collij V, Sanna S, Sinha T, Imhann F, Bourgonje AR, Mujagic Z, DMAE J, Masclee AAM, Fu J, et al. Impact of commonly used drugs on the composition and metabolic function of the gut microbiota. Nat Commun. 2020;11(1):362. doi: 10.1038/s41467-019-14177-z.
  • Meedt E, Hiergeist A, Gessner A, Dettmer K, Liebisch G, Ghimire S, Poeck H, Edinger M, Wolff D, Herr W, et al. Prolonged suppression of butyrate-producing bacteria is associated with acute gastrointestinal graft-vs-host disease and transplantation-related mortality after allogeneic stem cell transplantation. Clin Infect Dis. 2022;74(4):614–621. doi: 10.1093/cid/ciab500.
  • Golob JL, Pergam SA, Srinivasan S, Fiedler TL, Liu C, Garcia K, Mielcarek M, Ko D, Aker S, Marquis S, et al. Stool microbiota at neutrophil recovery is predictive for severe acute graft vs host disease after hematopoietic cell transplantation. Clin Infect Dis. 2017;65(12):1984–1991. doi: 10.1093/cid/cix699.
  • Legoff J, Resche-Rigon M, Bouquet J, Robin M, Naccache SN, Mercier-Delarue S, Federman S, Samayoa E, Rousseau C, Piron P, et al. The eukaryotic gut virome in hematopoietic stem cell transplantation: new clues in enteric graft-versus-host disease. Nat Med. 2017;23(9):1080–1085. doi: 10.1038/nm.4380.
  • Jenq RR, Ubeda C, Taur Y, Menezes CC, Khanin R, Dudakov JA, Liu C, West ML, Singer NV, Equinda MJ, et al. Regulation of intestinal inflammation by microbiota following allogeneic bone marrow transplantation. J Exp Med. 2012;209(5):903–911. doi: 10.1084/jem.20112408.
  • Burgos da Silva M, Ponce DM, Dai A, Devlin SM, Gomes AL, Moore GF, Slingerland J, Shouval R, Armijo GK, DeWolf S, et al. Preservation of the fecal microbiome is associated with reduced severity of graft-versus-host disease. Blood. 2022;140(22):2385–2397. doi: 10.1182/blood.2021015352.
  • Jenq RR, Taur Y, Devlin SM, Ponce DM, Goldberg JD, Ahr KF, Littmann ER, Ling L, Gobourne AC, Miller LC, et al. Intestinal blautia is associated with reduced death from graft-versus-host disease. Biol Blood Marrow Transplant. 2015;21(8):1373–1383. doi: 10.1016/j.bbmt.2015.04.016.
  • Michonneau D, Latis E, Curis E, Dubouchet L, Ramamoorthy S, Ingram B, de Latour RP, Robin M, de Fontbrune FS, Chevret S, et al. Metabolomics analysis of human acute graft-versus-host disease reveals changes in host and microbiota-derived metabolites. Nat Commun. 2019;10(1):5695. doi: 10.1038/s41467-019-13498-3.
  • Rashidi A, Gao F, Fredricks DN, Pergam SA, Mielcarek M, Milano F, Sandmaier BM, Lee SJ. Analysis of antibiotic exposure and development of acute graft-vs-host disease following allogeneic hematopoietic cell transplantation. JAMA Netw Open. 2023;6:e2317188. doi: 10.1001/jamanetworkopen.2023.17188.
  • Beelen DW, Elmaagacli A, Müller K-D, Hirche H, Schaefer UW. Influence of intestinal bacterial decontamination using metronidazole and ciprofloxacin or ciprofloxacin alone on the development of acute graft-versus-host disease after marrow transplantation in patients with hematologic malignancies: final results and long-term follow-up of an open-label prospective randomized trial. Blood J Am Society Hematol. 1999;93:3267–3275.
  • Chhabra S, Szabo A, Clurman A, McShane K, Waters N, Eastwood D, Samanas L, Fei T, Armijo G, Abedin S, et al. Mitigation of gastrointestinal graft versus host disease with tocilizumab prophylaxis is accompanied by preservation of microbial diversity and attenuation of enterococcal domination. Haemaologica. 2023;108(1):250–256. doi: 10.3324/haematol.2022.281309.
  • Ponce DM, Alousi AM, Nakamura R, Slingerland J, Calafiore M, Sandhu KS, Barker JN, Devlin S, Shia J, Giralt S, et al. A phase 2 study of interleukin-22 and systemic corticosteroids as initial treatment for acute GVHD of the lower GI tract. Blood. 2023;141(12):1389–1401. doi: 10.1182/blood.2021015111.
  • Taur Y, Jenq RR, Perales M-A, Littmann ER, Morjaria S, Ling L, No D, Gobourne A, Viale A, Dahi PB, et al. The effects of intestinal tract bacterial diversity on mortality following allogeneic hematopoietic stem cell transplantation. Blood. 2014;124(7):1174–1182. doi: 10.1182/blood-2014-02-554725.
  • Ubeda C, Taur Y, Jenq RR, Equinda MJ, Son T, Samstein M, Viale A, Socci ND, van den Brink MRM, Kamboj M, et al. Vancomycin-resistant Enterococcus domination of intestinal microbiota is enabled by antibiotic treatment in mice and precedes bloodstream invasion in humans. J Clin Invest. 2010;120(12):4332–4341. doi: 10.1172/JCI43918.
  • Stoma I, Littmann ER, Peled JU, Giralt S, van den Brink MRM, Pamer EG, Taur Y. Compositional flux within the intestinal microbiota and risk for bloodstream infection with gram-negative bacteria. Clin Infect Dis. 2021;73(11):e4627–35. doi: 10.1093/cid/ciaa068.
  • Schluter J, Peled JU, Taylor BP, Markey KA, Smith M, Taur Y, Niehus R, Staffas A, Dai A, Fontana E, et al. The gut microbiota is associated with immune cell dynamics in humans. Nature. 2020;588(7837):303–307. doi: 10.1038/s41586-020-2971-8.
  • Miltiadous O, Waters NR, Andrlová H, Dai A, Nguyen CL, Burgos da Silva M, Lindner S, Slingerland J, Giardina P, Clurman A, et al. Early intestinal microbial features are associated with CD4 T-cell recovery after allogeneic hematopoietic transplant. Blood J Am Society Hematol. 2022;139(18):2758–2769. doi: 10.1182/blood.2021014255.
  • Haak BW, Littmann ER, Chaubard J-L, Pickard AJ, Fontana E, Adhi F, Gyaltshen Y, Ling L, Morjaria SM, Peled JU, et al. Impact of gut colonization with butyrate-producing microbiota on respiratory viral infection following allo-HCT. Blood. 2018;131:2978–2986. doi: 10.1182/blood-2018-01-828996.
  • Harris B, Morjaria SM, Littmann ER, Geyer AI, Stover DE, Barker JN, Giralt SA, Taur Y, Pamer EG. Gut microbiota predict pulmonary infiltrates after allogeneic hematopoietic cell transplantation. Am J Respir Crit Care Med. 2016;194(4):450–463. doi: 10.1164/rccm.201507-1491OC.
  • Lee YJ, Arguello ES, Jenq RR, Littmann E, Kim GJ, Miller LC, Ling L, Figueroa C, Robilotti E, Perales M-A, et al. Protective factors in the intestinal microbiome against clostridium difficile infection in recipients of allogeneic hematopoietic stem cell transplantation. J Infect Dis. 2017;215(7):1117–1123. doi: 10.1093/infdis/jix011.
  • Markey KA, Schluter J, Gomes ALC, Littmann ER, Pickard AJ, Taylor BP, Giardina PA, Weber D, Dai A, Docampo MD, et al. The microbe-derived short-chain fatty acids butyrate and propionate are associated with protection from chronic GVHD. Blood. 2020;136(1):130–136. doi: 10.1182/blood.2019003369.
  • Peled JU, Devlin SM, Staffas A, Lumish M, Khanin R, Littmann ER, Ling L, Kosuri S, Maloy M, Slingerland JB, et al. Intestinal microbiota and relapse after hematopoietic-cell transplantation. J Clin Oncol. 2017;35(15):1650–1659. doi: 10.1200/JCO.2016.70.3348.
  • Smith M, Dai A, Ghilardi G, Amelsberg KV, Devlin SM, Pajarillo R, Slingerland JB, Beghi S, Herrera PS, Giardina P, et al. Gut microbiome correlates of response and toxicity following anti-CD19 CAR T cell therapy. Nat Med. 2022;28(4):713–723. doi: 10.1038/s41591-022-01702-9.
  • Stein-Thoeringer CK, Saini NY, Zamir E, Blumenberg V, Schubert M-L, Mor U, Fante MA, Schmidt S, Hayase E, Hayase T, et al. A non-antibiotic-disrupted gut microbiome is associated with clinical responses to CD19-CAR-T cell cancer immunotherapy. Nat Med. 2023;29(4):906–916. doi: 10.1038/s41591-023-02234-6.
  • Hill GR, Ferrara JLM. The primacy of the gastrointestinal tract as a target organ of acute graft-versus-host disease: rationale for the use of cytokine shields in allogeneic bone marrow transplantation. Blood J Am Society Hematol. 2000;95(9):2754–2759. doi: 10.1182/blood.V95.9.2754.009k25_2754_2759.
  • Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, deRoos P, Liu H, Cross JR, Pfeffer K, Coffer PJ, et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature. 2013;504(7480):451–455. doi: 10.1038/nature12726.
  • Swimm A, Giver CR, DeFilipp Z, Rangaraju S, Sharma A, Ulezko Antonova A, Sonowal R, Capaldo C, Powell D, Qayed M, et al. Indoles derived from intestinal microbiota act via type I interferon signaling to limit graft-versus-host disease. Blood. 2018;132(23):2506–2519. doi: 10.1182/blood-2018-03-838193.
  • Wlodarska M, Luo C, Kolde R, d’Hennezel E, Annand JW, Heim CE, Krastel P, Schmitt EK, Omar AS, Creasey EA, et al. Indoleacrylic acid produced by commensal peptostreptococcus species suppresses inflammation. Cell Host & Microbe. 2017;22(1):25–37.e6. doi: 10.1016/j.chom.2017.06.007.
  • Sakata T, von Engelhardt W. Stimulatory effect of short chain fatty acids on the epithelial cell proliferation in rat large intestine. Comp Biochem Physiol A Comp Physiol. 1983;74(2):459–462. doi: 10.1016/0300-9629(83)90631-X.
  • Roediger WE. Utilization of nutrients by isolated epithelial cells of the rat colon. Gastroenterology. 1982;83(2):424–429. doi: 10.1016/S0016-5085(82)80339-9.
  • Bansal T, Alaniz RC, Wood TK, Jayaraman A. The bacterial signal indole increases epithelial-cell tight-junction resistance and attenuates indicators of inflammation. Proc Natl Acad Sci USA. 2010;107(1):228–233. doi: 10.1073/pnas.0906112107.
  • Alexeev EE, Lanis JM, Kao DJ, Campbell EL, Kelly CJ, Battista KD, Gerich ME, Jenkins BR, Walk ST, Kominsky DJ, et al. Microbiota-derived indole metabolites promote human and murine intestinal homeostasis through regulation of interleukin-10 receptor. Am J Pathol. 2018;188(5):1183–1194. doi: 10.1016/j.ajpath.2018.01.011.
  • Saqr A, Carlson B, Staley C, Rashidi A, Al-Kofahi M, Kaiser T, Holtan S, MacMillan M, Young J-A, Jurdi NE, et al. Reduced enterohepatic recirculation of mycophenolate and lower blood concentrations are associated with the stool bacterial microbiome after hematopoietic cell transplantation. Transplant Cell Ther. 2022;28(7):.e372.1–.e372.9. doi: 10.1016/j.jtct.2022.04.018.
  • Hülsdünker J, Thomas OS, Haring E, Unger S, Gonzalo Núñez N, Tugues S, Gao Z, Duquesne S, Cywes-Bentley C, Oyardi O, et al. Immunization against poly-N-acetylglucosamine reduces neutrophil activation and GVHD while sparing microbial diversity. Proc Natl Acad Sci USA. 2019;116(41):20700–20706. doi: 10.1073/pnas.1908549116.
  • Seike K, Kiledal A, Fujiwara H, Henig I, Burgos da Silva M, van den Brink MRM, Hein R, Hoostal M, Liu C, Oravecz-Wilson K, et al. Ambient oxygen levels regulate intestinal dysbiosis and GVHD severity after allogeneic stem cell transplantation. Immunity. 2023;56(2):353–368. doi: 10.1016/j.immuni.2023.01.007.
  • Rashidi A, Ebadi M, Rehman TU, Elhusseini H, Nalluri H, Kaiser T, Ramamoorthy S, Holtan SG, Khoruts A, Weisdorf DJ, et al. Altered microbiota-host metabolic cross talk preceding neutropenic fever in patients with acute leukemia. Blood Adv. 2021;5(20):3937–3950. doi: 10.1182/bloodadvances.2021004973.
  • Rashidi A, Kaiser T, Graiziger C, Holtan SG, Rehman TU, Weisdorf DJ, Khoruts A, Staley C. Specific gut microbiota changes heralding bloodstream infection and neutropenic fever during intensive chemotherapy. Leukemia. 2020;34(1):312–316. doi: 10.1038/s41375-019-0547-0.
  • Desai MS, Seekatz AM, Koropatkin NM, Kamada N, Hickey CA, Wolter M, Pudlo NA, Kitamoto S, Terrapon N, Muller A, et al. A dietary fiber-deprived gut microbiota degrades the colonic mucus barrier and enhances pathogen susceptibility. Cell. 2016;167(5):1339–53.e21. doi: 10.1016/j.cell.2016.10.043.
  • Ubeda C, Taur Y, Jenq RR, Equinda MJ, Son T, Samstein M, Viale A, Socci ND, van den Brink MRM, Kamboj M, et al. Vancomycin-resistant Enterococcus domination of intestinal microbiota is enabled by antibiotic treatment in mice and precedes bloodstream invasion in humans. J Clin Invest. 2010;120(12):4332–4341. doi: 10.1172/JCI43918.
  • Uribe-Herranz M, Beghi S, Ruella M, Parvathaneni K, Salaris S, Kostopoulos N, George SS, Pierini S, Krimitza E, Costabile F, et al. Modulation of the gut microbiota engages antigen cross-presentation to enhance antitumor effects of CAR T cell immunotherapy. Mol Ther. 2023;31(3):686–700. doi: 10.1016/j.ymthe.2023.01.012.
  • Shono Y, van den Brink MRM. Gut microbiota injury in allogeneic haematopoietic stem cell transplantation. Nat Rev Cancer. 2018;18:283–295. doi: 10.1038/nrc.2018.10.
  • Staffas A, Burgos da Silva M, Slingerland AE, Lazrak A, Bare CJ, Holman CD, Docampo MD, Shono Y, Durham B, Pickard AJ, et al. Nutritional support from the intestinal microbiota improves hematopoietic reconstitution after bone marrow transplantation in mice. Cell Host & Microbe. 2018;23(4):447–57.e4. doi: 10.1016/j.chom.2018.03.002.
  • Andrlová H, Miltiadous O, Kousa AI, Dai A, DeWolf S, Violante S, Park H-Y, Janaki-Raman S, Gardner R, Daker SE, et al. MAIT and Vδ2 unconventional T cells are supported by a diverse intestinal microbiome and correlate with favorable patient outcome after allogeneic HCT. Sci Transl Med. 2022;14(646):eabj2829. doi: 10.1126/scitranslmed.abj2829.
  • Rashidi A, Weisdorf DJ. Microbiota-based approaches to mitigate infectious complications of intensive chemotherapy in patients with acute leukemia. Transl Res. 2020;220:167–181. doi: 10.1016/j.trsl.2020.03.011.
  • Kelly CR, Khoruts A, Staley C, Sadowsky MJ, Abd M, Alani M, Bakow B, Curran P, McKenney J, Tisch A, et al. Effect of fecal microbiota transplantation on recurrence in multiply recurrent clostridium difficile infection: a randomized trial. Ann Intern Med. 2016;165(9):609–616. doi: 10.7326/M16-0271.
  • Neemann K, Eichele DD, Smith PW, Bociek R, Akhtari M, Freifeld A. Fecal microbiota transplantation for fulminant Clostridium difficile infection in an allogeneic stem cell transplant patient. Transpl Infect Dis. 2012;14(6):E161–5. doi: 10.1111/tid.12017.
  • de Castro CG, Ganc AJ, Ganc RL, Petrolli MS, Hamerschlack N. Fecal microbiota transplant after hematopoietic SCT: report of a successful case. Bone Marrow Transplant. 2015;50:145. doi: 10.1038/bmt.2014.212.
  • Webb BJ, Brunner A, Ford CD, Gazdik MA, Petersen FB, Hoda D. Fecal microbiota transplantation for recurrent Clostridium difficile infection in hematopoietic stem cell transplant recipients. Transpl Infect Dis. 2016;18(4):628–633. doi: 10.1111/tid.12550.
  • Bluestone H, Kronman MP, Suskind DL. Fecal microbiota transplantation for recurrent clostridium difficile infections in pediatric hematopoietic stem cell transplant recipients. J Pediatric Infect Dis Soc. 2018;7(1):e6–8. doi: 10.1093/jpids/pix076.
  • Spindelboeck W, Halwachs B, Bayer N, Huber-Krassnitzer B, Schulz E, Uhl B, Gaksch L, Hatzl S, Bachmayr V, Kleissl L, et al. Antibiotic use and ileocolonic immune cells in patients receiving fecal microbiota transplantation for refractory intestinal GvHD: a prospective cohort study. Ther Adv Hematol. 2021;12:20406207211058333. doi: 10.1177/20406207211058333.
  • Goeser F, Sifft B, Stein-Thoeringer C, Farowski F, Strassburg CP, Brossart P, Higgins PG, Scheid C, Wolf D, Holderried TAW, et al. Fecal microbiota transfer for refractory intestinal graft-versus-host disease — Experience from two German tertiary centers. Eur J Haematol. 2021;107(2):229–245. doi: 10.1111/ejh.13642.
  • Battipaglia G, Malard F, Rubio MT, Ruggeri A, Mamez AC, Brissot E, Giannotti F, Dulery R, Joly AC, Baylatry MT, et al. Fecal microbiota transplantation before or after allogeneic hematopoietic transplantation in patients with hematologic malignancies carrying multidrug-resistance bacteria. Haematologica. 2019;104(8):1682–1688. doi: 10.3324/haematol.2018.198549.
  • van Lier YF, Davids M, Haverkate NJE, de Groot PF, Donker ML, Meijer E, Heubel-Moenen FCJI, Nur E, Zeerleder SS, Nieuwdorp M, et al. Donor fecal microbiota transplantation ameliorates intestinal graft-versus-host disease in allogeneic hematopoietic cell transplant recipients. Sci Transl Med. 2020;12(556):eaaz8926. doi: 10.1126/scitranslmed.aaz8926.
  • DeFilipp Z, Peled JU, Li S, Mahabamunuge J, Dagher Z, Slingerland AE, Del Rio C, Valles B, Kempner ME, Smith M, et al. Third-party fecal microbiota transplantation following allo-HCT reconstitutes microbiome diversity. Blood Adv. 2018;2(7):745–753. doi: 10.1182/bloodadvances.2018017731.
  • Zhao Y, Li X, Zhou Y, Gao J, Jiao Y, Zhu B, Wu D, Qi X. Safety and efficacy of fecal microbiota transplantation for grade iv steroid refractory gi-gvhd patients: interim results from FMT2017002 trial. Front Immunol. 2021;12:678476. doi: 10.3389/fimmu.2021.678476.
  • Rashidi A, Ebadi M, Rehman TU, Elhusseini H, Kazadi D, Halaweish H, Khan MH, Hoeschen A, Cao Q, Luo X, et al. Randomized double-blind phase II trial of fecal microbiota transplantation versus placebo in allogeneic hematopoietic cell transplantation and AML. J Clin Oncol. 2023:JCO2202366. doi: 10.1200/JCO.22.02366
  • Ghani R, Mullish BH, McDonald JAK, Ghazy A, Williams HRT, Brannigan ET, Mookerjee S, Satta G, Gilchrist M, Duncan N, et al. Disease prevention not decolonization: a model for fecal microbiota transplantation in patients colonized with multidrug-resistant organisms. Clin Infect Dis. 2021;72(8):1444–1447. doi: 10.1093/cid/ciaa948.
  • Innes AJ, Mullish BH, Ghani R, Szydlo RM, Apperley JF, Olavarria E, Palanicawandar R, Kanfer EJ, Milojkovic D, McDonald JAK, et al. Fecal microbiota transplant mitigates adverse outcomes seen in patients colonized with multidrug-resistant organisms undergoing allogeneic hematopoietic cell transplantation. Front Cell Infect Microbiol. 2021;11:684659. doi: 10.3389/fcimb.2021.684659.
  • Taur Y, Coyte K, Schluter J, Robilotti E, Figueroa C, Gjonbalaj M, Littmann ER, Ling L, Miller L, Gyaltshen Y, et al. Reconstitution of the gut microbiota of antibiotic-treated patients by autologous fecal microbiota transplant. Sci Transl Med. 2018;10(460):eaap9489. doi: 10.1126/scitranslmed.aap9489.
  • Kakihana K, Fujioka Y, Suda W, Najima Y, Kuwata G, Sasajima S, Mimura I, Morita H, Sugiyama D, Nishikawa H, et al. Fecal microbiota transplantation for patients with steroid-resistant acute graft-versus-host disease of the gut. Blood. 2016;128(16):2083–2088. doi: 10.1182/blood-2016-05-717652.
  • Kaito S, Toya T, Yoshifuji K, Kurosawa S, Inamoto K, Takeshita K, Suda W, Kakihana K, Honda K, Hattori M, et al. Fecal microbiota transplantation with frozen capsules for a patient with refractory acute gut graft-versus-host disease. Blood Adv. 2018;2(22):3097–3101. doi: 10.1182/bloodadvances.2018024968.
  • Wu S-R, Reddy P. Tissue tolerance: a distinct concept to control acute GVHD severity. Blood. 2017;129(13):1747–1752. doi: 10.1182/blood-2016-09-740431.
  • Köhler N, Zeiser R. Intestinal microbiota influence immune tolerance post allogeneic hematopoietic cell transplantation and intestinal GVHD. Front Immunol. 2018;9:3179. doi: 10.3389/fimmu.2018.03179.
  • Soares MP, Teixeira L, Moita LF. Disease tolerance and immunity in host protection against infection. Nat Rev Immunol. 2017;17(2):83–96. doi: 10.1038/nri.2016.136.
  • DeFilipp Z, Bloom PP, Torres Soto M, Mansour MK, Sater MRA, Huntley MH, Turbett S, Chung RT, Chen Y-B, Hohmann EL. Drug-resistant E. coli bacteremia transmitted by fecal microbiota transplant. N Engl J Med. 2019;381:2043–2050. doi: 10.1056/NEJMoa1910437.
  • Bilinski J, Lis K, Tomaszewska A, Pechcinska A, Grzesiowski P, Dzieciatkowski T, Walesiak A, Gierej B, Ziarkiewicz-Wróblewska B, Tyszka M, et al. Eosinophilic gastroenteritis and graft-versus-host disease induced by transmission of Norovirus with fecal microbiota transplant. Transpl Infect Dis. 2021;23(1):e13386. doi: 10.1111/tid.13386.
  • Eshel A, Sharon I, Nagler A, Bomze D, Danylesko I, Fein JA, Geva M, Henig I, Shimoni A, Zuckerman T, et al. Origins of bloodstream infections following fecal microbiota transplantation: a strain-level analysis. Blood Adv. 2022;6(2):568–573. doi: 10.1182/bloodadvances.2021005110.
  • Rasmussen TS, Koefoed AK, Jakobsen RR, Deng L, Castro-Mejía JL, Brunse A, Neve H, Vogensen FK, Nielsen DS. Bacteriophage-mediated manipulation of the gut microbiome – promises and presents limitations. FEMS Microbiol Rev. 2020;44(4):507–521. doi: 10.1093/femsre/fuaa020.
  • Ott SJ, Waetzig GH, Rehman A, Moltzau-Anderson J, Bharti R, Grasis JA, Cassidy L, Tholey A, Fickenscher H, Seegert D, et al. Efficacy of sterile fecal filtrate transfer for treating patients with clostridium difficile infection. Gastroenterology. 2017;152(4):799–811.e7. doi: 10.1053/j.gastro.2016.11.010.
  • Zhang F, Zuo T, Yeoh YK, Cheng FWT, Liu Q, Tang W, Cheung KCY, Yang K, Cheung CP, Mo CC, et al. Longitudinal dynamics of gut bacteriome, mycobiome and virome after fecal microbiota transplantation in graft-versus-host disease. Nat Commun. 2021;12(1):65. doi: 10.1038/s41467-020-20240-x.
  • Malard F, Vekhoff A, Lapusan S, Isnard F, D’incan-Corda E, Rey J, Saillard C, Thomas X, Ducastelle-Lepretre S, Paubelle E, et al. Gut microbiota diversity after autologous fecal microbiota transfer in acute myeloid leukemia patients. Nat Commun. 2021;12(1):3084. doi: 10.1038/s41467-021-23376-6.
  • Bilinski J, Grzesiowski P, Sorensen N, Madry K, Muszynski J, Robak K, Wroblewska M, Dzieciatkowski T, Dulny G, Dwilewicz-Trojaczek J, et al. Fecal microbiota transplantation in patients with blood disorders inhibits gut colonization with antibiotic-resistant bacteria: results of a prospective, single-center study. Clin Infect Dis. 2017;65(3):364–370. doi: 10.1093/cid/cix252.
  • Merli P, Putignani L, Ruggeri A, Del Chierico F, Gargiullo L, Galaverna F, Gaspari S, Pagliara D, Russo A, Pane S, et al. Decolonization of multi-drug resistant bacteria by fecal microbiota transplantation in five pediatric patients before allogeneic hematopoietic stem cell transplantation: gut microbiota profiling, infectious and clinical outcomes. Haematologica. 2020;105(11):2686–2690. doi: 10.3324/haematol.2019.244210.
  • Olesen SW, Leier MM, Alm EJ, Kahn SA. Searching for superstool: maximizing the therapeutic potential of FMT. Nat Rev Gastroenterol Hepatol. 2018;15(7):387–388. doi: 10.1038/s41575-018-0019-4.