key: cord-0856556-vzbqfhgo authors: Ouyang, Jing; Isnard, Stéphane; Lin, John; Fombuena, Brandon; Peng, Xiaorong; Nair Parvathy, Seema; Chen, Yaokai; Silverman, Michael S.; Routy, Jean-Pierre title: Treating From the Inside Out: Relevance of Fecal Microbiota Transplantation to Counteract Gut Damage in GVHD and HIV Infection date: 2020-08-06 journal: Front Med (Lausanne) DOI: 10.3389/fmed.2020.00421 sha: 759793a0d366595a144edf5c0f651d5b7f059b77 doc_id: 856556 cord_uid: vzbqfhgo The gastrointestinal (GI) tract is a complex and well-balanced milieu of anatomic and immunological barriers. The epithelial surface of the GI tract is colonized by trillions of microorganisms, known as the gut microbiota, which is considered an “organ” with distinctive endocrine and immunoregulatory functions. Dysregulation of the gut microbiota composition, termed dysbiosis, has been associated with epithelial damage and translocation of microbial products into the circulating blood. Dysbiosis, increased gut permeability and chronic inflammation play a major role on the clinical outcome of inflammatory bowel diseases, graft-vs.-host disease (GVHD) and HIV infection. In this review, we focus on GVHD and HIV infection, conditions sharing gut immune damage leading to dysbiosis. The degree of dysbiosis and level of epithelial gut damage predict poor clinical outcome in both conditions. Emerging interventions are therefore warranted to promote gut microbiota homeostasis and improve intestinal barrier function. Interventions such as anti-inflammatory medications, and probiotics have toxicity and/or limited transitory effects, justifying innovative approaches. Fecal microbiota transplantation (FMT) is one such approach where fecal microorganisms are transferred from healthy donors into the GI tract of the recipient to restore microbiota composition in patients with Clostridium difficile-induced colitis or inflammatory bowel diseases. Preliminary findings point toward a beneficial effect of FMT to improve GVHD and HIV-related outcomes through the engraftment of beneficial donor bacteria, notably those producing anti-inflammatory metabolites. Herein, we critically review the potential for FMT in alleviating dysbiosis and gut damage in patients with GVHD or HIV-infection. Understanding the underlying mechanism by which FMT restores gut function will pave the way toward novel scalable and targeted interventions. The gastrointestinal (GI) tract is a complex and well-balanced milieu of anatomic and immunological barriers. The epithelial surface of the GI tract is colonized by trillions of microorganisms, known as the gut microbiota, which is considered an "organ" with distinctive endocrine and immunoregulatory functions. Dysregulation of the gut microbiota composition, termed dysbiosis, has been associated with epithelial damage and translocation of microbial products into the circulating blood. Dysbiosis, increased gut permeability and chronic inflammation play a major role on the clinical outcome of inflammatory bowel diseases, graft-vs.-host disease (GVHD) and HIV infection. In this review, we focus on GVHD and HIV infection, conditions sharing gut immune damage leading to dysbiosis. The degree of dysbiosis and level of epithelial gut damage predict poor clinical outcome in both conditions. Emerging interventions are therefore warranted to promote gut microbiota homeostasis and improve intestinal barrier function. Interventions such as anti-inflammatory medications, and probiotics have toxicity and/or limited transitory effects, justifying innovative approaches. Fecal microbiota transplantation (FMT) is one such approach where fecal microorganisms are transferred from healthy donors into the GI tract of the recipient to restore microbiota composition in patients with Clostridium difficile-induced colitis or inflammatory bowel diseases. Preliminary findings point toward a beneficial effect of FMT to improve GVHD and HIV-related outcomes through the engraftment of beneficial donor bacteria, notably Trillions of microorganisms reside in the human gut, encompassing not only bacteria but also fungi, archaea, viruses, and eukaryotic microbes, collectively termed microbiota. The gut microbiota was recently considered as an essential organ, playing a critical role in various host functions such as maintenance of the gut barrier and modulation of systemic immune response (1) . Furthermore, the endocrine function of the gut microbiota was demonstrated through the production of vitamins and immunoregulatory short chain fatty acids (SCFA) (2) . Dysregulation of gut microbiota composition, also known as dysbiosis, can lead to barrier dysfunction and translocation of microbial products leading to systemic inflammation (3) . Recent evidence has shown that patients with diabetes, inflammatory bowel diseases (IBD), cancer, graft-vs.-host disease (GVHD) or HIV infection present with gut dysbiosis, gut damage, and microbial translocation (4-7). Allo-hematopoietic stem cell transplantation (HSCT) is used in the treatment of hematological cancers where donor derived T-cells and natural killer cells target cancer cells in the recipient (4). Occurring after chemotherapy conditioning and HSCT, GVHD may develop as a serious complication when donor immune cells recognize the recipient as foreign and attack healthy cells in host's tissues. GVHD mostly occurs in the gut through the disruption of epithelial tight junctions, destruction of epithelial cells and inflammation in association with dysbiosis (5) (6) (7) (8) . A large multicenter study showed that gut microbiota composition independently predicted mortality in 1,362 HSCT patients with GVHD (9) (10) (11) . Similarly, immune damage observed in the gut of people living with HIV (PLWH) was associated with gut dysbiosis, inflammation and clinical outcome (12) (13) (14) (15) . Despite long-term antiretroviral therapy (ART), damage to the gut mucosa and dysbiosis persist in PLWH, leading to systemic inflammation (8, 10, 11, 15) . Like for people with GVHD, PLWH present with a disrupted gut epithelial barrier, immune-mediated intestinal damage, and increased gut permeability (15) (16) (17) (18) (19) (20) (21) (22) . Given the association between microbiota composition and clinical outcome in both GVHD and HIV infection (5) (6) (7) (8) (9) (10) (11) , strategies to modify the gut microbiota have come to light through dietary interventions, the antidiabetic drug metformin, selective antibiotics, probiotics, prebiotics, and fecal microbiota transplantation (FMT) (5, 23, 24) . FMT refers to the transfer of fecal microorganisms from healthy donors into the GI tract of patients. It has shown to be effective in Clostridium difficile colitis (CDC), IBDs or obesity (25) (26) (27) (28) . As FMT has been recently shown to improve intestinal barrier function through promotion of gut microbiota homeostasis in GVHD and HIV infection, we discuss its relevance in both conditions in this review (29) . In GVHD or HIV infection, a decrease of gut microbiota diversity is observed and associated with poor clinical outcome (30) (31) (32) (33) . Compared to patients undergoing allogeneic HSCT without GVHD, patients experiencing GVHD had decreased stool microbial diversity (32) . Taur et al. reported that lower bacterial diversity was associated with increased transplantrelated mortality in HSCT recipients (33) . Nowak et al. also reported that the bacterial diversity of the gut microbiota was correlated with CD4 T-cell counts and inversely correlated with markers of microbial translocation and monocyte activation in PLWH (30) . The gut barrier is organized as a multi-layered and complex system which allows nutrient absorption while preventing the translocation of microbes and their products. Epithelial gut damage occurs in patients with GVHD and PLWH, with damaged enterocytes (basal barrier), non-functional Paneth cells (antimicrobial peptide production) and less mucosal-associated invariant T (MAIT) cells (5, (34) (35) (36) (37) . Several proteins have been used as gut damage markers. Plasma concentrations of regenerating islet-derived 3-alpha (REG3α), secreted by Paneth cells, were 3-fold higher in patients with gut GVHD at the onset of the disease compared to other HSCT patients (36, 37) . Lower levels of REG3α at GVHD onset are correlated with higher 1 year survival (37) . In PLWH, we observed that REG3α but not intestinal fatty acid binding protein (I-FABP) plasma levels were correlated with HIV disease progression, microbial translocation and immune activation (36) . Similarly, soluble suppression of tumorigenicity (sST2) was also used to predict gut damage and clinical outcomes in patients with GVHD and PLWH (38) (39) (40) (41) (42) . Epithelial gut damage allows microbial translocation of microbial products from the lumen to the bloodstream, inducing local and systemic inflammation (43) . Circulating levels of lipopolysaccharide (LPS), a pro-inflammatory bacterial cell wall component, is a clinically significant marker to assess the level of microbial translocation (44) . LPS leakage in the circulation could induce innate immune activation, in association with mortality in GVHD (45) (46) (47) . In PLWH, we and others have shown that LPS translocation is correlated with immune dysfunction and increased risk of non-AIDS comorbidities (48) (49) (50) (51) . Additionally, cytomegalovirus (CMV) primarily replicates in mucosal epithelial cells, decreasing gut barrier integrity. In patients with GVHD and PLWH, CMV latent infection or reactivation is associated with poor clinical outcomes (52) (53) (54) (55) (56) (57) . These findings suggest that patients with GVHD and HIV infection share similar features in gut damage and related microbial translocation. Moreover, the gut microbiota can influence host cell physiology via production of metabolites such as SCFAs and bile acids. SCFAs, especially butyrate, constitute the primary energy source for colon epithelial cells. SCFAs play an important role in protecting intestinal barrier function, preventing microbial translocation and reducing inflammation through regulation of host epigenetics (58) (59) (60) . GVHD patients or PLWH present with a lower abundance of SCFA-producing bacteria and a lower level of SCFAs, compared to non-GVHD HSCT patients or HIVnegative individuals, respectively (61) (62) (63) (64) . In both conditions, lower levels of SCFAs have been associated with gut damage and inflammation (62, (64) (65) (66) . Conflicting results exist on the role of butyrate in GVHD as one report shows that patients developing GVHD had higher butyrate production (67) . Furthermore, microbiota modulation leading to poor bile acids reabsorption could also be associated with gut damage in both patients with GVHD or PLWH (68) (69) (70) (71) (72) . Globally, gut dysbiosis, increased gut permeability, inflammation and systemic immune activation are common features of patients with GVHD or PLWH. Given the dysbiosis and gut permeability in patients with GVHD, and regarding the vital role of gut microbiota in intestinal barrier and homeostasis, strategies targeting the microbiota offer one promising avenue for preventing or treating this condition. In the 1990s, investigators attempted to prevent the development of acute GVHD by drastically reducing the gut microbiota mass with antibiotics, removing the triggers of inflammation (73) (74) (75) . However, newer studies have proven that gut microbiota-depleted patients had a higher risk of developing acute GVHD following HSCT than non-depleted patients (76, 77) . Therefore, strategies promoting a "healthy" microbiota including FMT have attracted recent attention. Kakihana et al. (78) conducted a pilot study on four patients with steroidresistant or steroid-dependent gut GVHD to observe the effects of FMT from spouses or relatives via nasoduodenal tube. All patients responded to FMT with three complete responses, one partial response, all in absence of severe adverse events. Spindelboeck et al. (79) reported successful FMT in three patients with severe acute GVHD. After one to six FMTs delivered via colonoscopy, all three patients showed increased diversity of the gut microbiota, with two complete remissions of GVHD and one partial remission. Qi et al. (80) reported eight patients with steroid-refractory gut GVHD receiving FMT through a nasoduodenal tube, from a stool bank. After FMT, all patients' clinical symptoms were relieved, bacteria diversity was enriched, and the gut microbiota diversity was restored. Compared to those who did not receive FMT, these eight patients achieved a longer progression-free survival. These case studies suggest that FMT can serve as a promising therapeutic option for gut GVHD, however larger controlled studies are required to confirm these effects. In PLWH, the mucosal immune system is disturbed by HIV infection. Th17 and Th22 cells, important components of mucosal immunity, are rapidly depleted following HIV or simian immunodeficiency virus (SIV) infection, contributing to a reduced barrier integrity, microbial translocation, and systemic immune activation (81) (82) (83) . In a pilot study, Hensley-McBain et al. (84) reported that FMT significantly increases the number of peripheral Th17 and Th22 cells and reduced CD4 T-cell activation in the gut in SIV-infected macaques receiving ART. Moreover, the transplant was well-tolerated and no side effects were observed (84) . A pilot study in ART-suppressed individuals who received one-time FMT from stool bank via colonoscopy reported no serious adverse effects during the 24 weeks of follow-up. Microbial engraftment occurred but was partial, and limited to specific bacterial taxa including an increase of Faecalibacterium (85) , which has been shown to exert anti-inflammatory effects in murine experimental colitis (86, 87) . The authors considered that the limited effects of FMT might be related to the single dose of FMT given and the absence of antibiotic pretreatment to "provide space" before FMT (85) . Serrano-Villar et al. reported that repeated oral capsular FMT was one way to safely introduce incremental compositional changes into the gut microbiota in ART-treated PLWH (88) . Compared to placebo, FMT significantly decreased the gut damage marker I-FABP 4 weeks after initiating FMT. Furthermore, mild engraftment of the donor's microbiota persisted until week 36 after initiating FMT and greater engraftment was observed among the four subjects who had received antibiotics in the 12 week period before FMT (88) (Figure 1) . Safety should be the primary focus of any intervention. Concern persists on the safety of FMT administration, even more so in immunosuppressed recipients. However, PLWH with low CD4 T-cell count were shown to have the most profound modification of their gut microbiota and therefore would benefit greatly from FMT (89) . As reviewed by Shogbesan et al. (27) , FMT is successful in the treatment of recurrent CDC in immunocompromised patients including organ transplant recipients and PLWH. Encouragingly, FMT showed similar rates of adverse events in immunocompromised participants compared to immunocompetent ones including PLWH with CD4 counts lower than 200 cells/mm 3 (90) (91) (92) (93) . Additionally, Schunemann et al. showed that FMT increased CD4 counts in an individual with HIV (94) . To better assess the efficacy and safety of FMT, well-designed RCT clinical trials are ongoing and presented in Table 1 . However, large studies assessing the influence of FMT in PLWH are still needed. FMT needs further confirmation of its efficacy in decreasing gut damage in patient with GVHD or PLWH since studies assessing FMT with GVHD or PLWH involved a small number of participants. Moreover, safety needs to be validated as rare side effects may not have been observed in small studies. Therefore, challenges in designing formulations, preventing potential risks and implementing application in clinic for patients with GVHD and PLWH still remain. Firstly, both healthy donors and patients have a microbiota composition with a high inter-person variability, and the key factors causing microbiota composition variation over time are not fully characterized. The precise influence of different microbiota composition and metabolites on epithelial barrier and clinical outcomes remain poorly understood and need further studies to define their distinctive role on the development of GVHD and HIV infection. Therefore, it remains difficult to select donors and special products for FMT formulation. Moreover, FMT treatment may carry pathogens for digestive and bloodstream infection, as DeFilipp et al. recently reported two cases of drug-resistant Escherichia coli bacteremia transmitted by FMT (95) . Therefore, despite the absence of a uniform standard for "qualified" microbial communities, donors have to be thoroughly screened for transmissible diseases (e.g., HIV and hepatitis) and other noninfectious conditions (e.g., obesity and diabetes) that may be influenced by changes in the microbiome. In the light of Coronavirus Disease 2019, efforts to screen for novel infectious diseases should be implemented in the future. SARS-CoV-2, the virus that causes this disease, was found in stools even after diminution of respiratory symptoms and could be transmitted through a fecal-oral route (96, 97) . Donors who may transfer undesirable agents (e.g., antibiotics, anti-acid proton pump inhibitors, systemic immunosuppressive agents, antineoplastic agents, and glucocorticoids) which can affect the safety or efficacy of FMT should also be excluded (98) . Hence, screening for potential donors is costly and time consuming (99) . Fortunately, new techniques allow freezing and storage of donor stools for extended periods of time, possibly facilitating FMT implementation (100) . As donor selection is a difficult process, and in order to favor clinical improvement, engraftment of the donor's microbiota should be optimal. Antibiotic conditioning given to the recipient just before FMT seems to improve microbiota engraftment (88) . This procedure may destabilize the existing microbial community and promote engraftment of another community. By preventing niche competition in the mucosa between the xenomicrobiota and indigenous microbiota, preparing the gut with antibiotics was shown to facilitate xenomicrobiota colonization, thus enhancing the overall gut microbiota modification efficiency (101) . Preliminary results by Serrano-villar et al. showed greater engraftment in four PLWH who had received antibiotics before FMT (88) . Pre-therapy with antibiotics before FMT to alleviate GVHD is currently under study (NCT03862079 , Table 1 ). Encouragingly, multiple clinical trials studying the potential of FMT as a treatment for GVHD or HIV-related gut damage are ongoing ( Table 1) . In these trials, several routes of administration for FMT are under investigations, including oral capsules, nasal tube, colonoscopy, or enema. The optimal administration route may depend on the characteristics of the disease, and general condition of the patient. Compared with enema, colonoscopy could deliver the FMT to deep cecum, and increase engraftment while the donor stools are expelled less rapidly. However, colonoscopy remains a relatively invasive procedure (102): Kelly et al. reported one case of death from lung-aspiration injury during sedation for FMT administered via colonoscopy (103) . Furthermore, nasal administration is considered inconvenient as some cases of intestinal bleeding and rare peritonitis have been reported (104) . However, oral capsules have been developed to pass through the acidic environment of the stomach and ensure a delayed delivery of live microbial communities into the intestine (105) . By using questionnaires, this route is considered to be most convenient for patients. Kao et al. compared oral capsule and colonoscopy delivered FMT on recurrent CDC showed similar efficacy, with less adverse events (106) . Further studies should analyze the preferential route of FMT to alleviate gut damage patients in GVHD and PLWH. Both gut GVHD and HIV infection have been associated with dysbiosis and increased gut permeability, contributing to microbial translocation, inflammation, and poor clinical outcomes. Progress has been made in discerning the role of the microbiota in GVHD patients and PLWH. Manipulating the gut microbiota with FMT has been successfully used to treat CDC through microbiota restoration and has paved the way as a novel strategy to improve the outcomes of GVHD patients and PLWH. Several clinical trials are ongoing to assess the efficacy and safety of treating GVHD and HIV-induced gut damage with FMT. However, most trials and published studies are pilot or case series, thus making it difficult to confirm its efficacy and safety. Only large multicentre RCT studies will address the merit of such intervention. Moreover, a standard FMT procedure needs to be implemented and described, including pre-treatment with antibiotics and delivery with oral capsules to favor engraftment. Overall, collaborative efforts encompassing microbiology, clinical care, and pharmacy will define the optimal procedure and number of FMT to obtain a significant and lasting benefit from FMT for individuals with GVHD and HIV. In the future, FMT will pave avenues toward the characterization of important species and their metabolites in modulation of gut damage in patient with GVHD or PLWH, leading to more effective interventions. JO and SI wrote the first draft of the manuscript. JL, BF, XP, SN, YC, and MS provided critical revision of the manuscript. J-PR conceived and designed the manuscript. All authors approved it for publication. Altered gut microbiota in type 2 diabetes: just a coincidence? The human intestinal microbiome in health and disease Regulation of inflammation by short chain fatty acids Specific gut microbiome members are associated with distinct immune markers in pediatric allogeneic hematopoietic stem cell transplantation The gut microbiota and graft-versus-host disease Pathophysiology of GvHD and other HSCT-related major complications. Front Immunol Graft-versus-host disease propagation depends on increased intestinal epithelial tight junction permeability Apoptosis is the predominant form of epithelial target cell injury in acute experimental graft-versus-host disease Microbiota as predictor of mortality in allogeneic hematopoietic-cell transplantation Immunostimulatory gut bacteria Lactose drives Enterococcus expansion to promote graft-versus-host disease Plasma (1 -> 3)-beta-D-glucan and suPAR levels correlate with neurocognitive performance in people living with HIV on antiretroviral therapy: a CHARTER analysis ->3)-beta-D-Glucan levels correlate with neurocognitive functioning in HIV-infected persons on suppressive antiretroviral therapy: a cohort study Impact of HIV-related gut microbiota alterations on metabolic comorbidities HIV-associated gut dysbiosis is independent of sexual practice and correlates with noncommunicable diseases The gastrointestinal tract is critical to the pathogenesis of acute HIV-1 infection HIV infection and the gastrointestinal immune system Defining total-body AIDS-virus burden with implications for curative strategies Intestinal damage precedes mucosal immune dysfunction in SIV infection Jejunal enteropathy associated with human immunodeficiency virus infection: quantitative histology HIV enteropathy: crypt stem and transit cell hyperproliferation induces villous atrophy in HIV/Microsporidia-infected jejunal mucosa Human immunodeficiency virus infection of enterocytes and mononuclear cells in human jejunal mucosa Strategies to increase the efficacy of using gut microbiota for the modulation of obesity HIV and the gut microbiota: composition, consequences, and avenues for amelioration European consensus conference on faecal microbiota transplantation in clinical practice The use of faecal microbiota transplant as treatment for recurrent or refractory Clostridium difficile infection and other potential indications: joint British society of gastroenterology (BSG) and healthcare infection society (HIS) guidelines a systematic review of the efficacy and safety of fecal microbiota transplant for Clostridium difficile infection in immunocompromised patients Successful therapy of Clostridium difficile infection with fecal microbiota transplantation Altered gut microbiota in hiv infection: future perspective of fecal microbiota transplantation therapy Gut microbiota diversity predicts immune status in HIV-1 infection The impact of human immunodeficiency virus infection on gut microbiota alpha-diversity: an individual-level meta-analysis Regulation of intestinal inflammation by microbiota following allogeneic bone marrow transplantation The effects of intestinal tract bacterial diversity on mortality following allogeneic hematopoietic stem cell transplantation Epithelial barrier dysfunction as permissive pathomechanism in human intestinal graft-versus-host disease Endothelial and epithelial barriers in graft-versus-host disease Plasma levels of C-type lectin REG3alpha and gut damage in people with human immunodeficiency virus Regenerating islet-derived 3-alpha is a biomarker of gastrointestinal graft-versus-host disease ST2 as a marker for risk of therapy-resistant graft-versus-host disease and death Assessment of ST2 for risk of death following graft-versus-host disease in the pediatric and adult age groups Plasma level of soluble ST2 in chronically infected HIV-1 patients with suppressed viremia The plasma levels of soluble ST2 as a marker of gut mucosal damage in early HIV infection The dynamic role of the IL-33/ST2 axis in chronic viral-infections: alarming and adjuvanting the immune response Influence of gut microbiota and intestinal barrier on enterogenic infection after liver transplantation Microbial translocation is a cause of systemic immune activation in chronic HIV infection Progressive accumulation of bacterial lipopolysaccharide in vivo during murine acute graft-versus-host disease Is it time to reconsider the lipopolysaccharide paradigm in acute graft-versus-host disease? Front Immunol Total body irradiation and acute graft-versus-host disease: the role of gastrointestinal damage and inflammatory cytokines Immunosuppressive tryptophan catabolism and gut mucosal dysfunction following early HIV infection Circulating LPS and (1->3)-beta-D-Glucan: a folie a deux contributing to HIV-associated immune activation Serum (1->3)-beta-D-glucan levels in HIV-infected individuals are associated with immunosuppression, inflammation, and cardiopulmonary function Distinct systemic microbiome and microbial translocation are associated with plasma level of anti-CD4 autoantibody in HIV infection Replication of CMV in the gut of HIV-infected individuals and epithelial barrier dysfunction CMV seropositivity is associated with increased microbial translocation in people living with HIV and uninfected controls Molecular determinants and the regulation of human cytomegalovirus latency and reactivation Deep functional immunophenotyping predicts risk of cytomegalovirus reactivation after hematopoietic cell transplantation Evidence for a bidirectional relationship between cytomegalovirus replication and acute graft-versus-host disease Increased transplant-related morbidity and mortality in CMV-seropositive patients despite highly effective prevention of CMV disease after allogeneic T-cell-depleted stem cell transplantation Host-microbiota interactions: epigenomic regulation Butyrate enhances intestinal epithelial barrier function via up-regulation of tight junction protein Claudin-1 transcription The role of short-chain fatty acids in health and disease Alterations in bacterial communities, SCFA and biomarkers in an elderly HIV-positive and HIV-negative population in western Mexico Low abundance of colonic butyrate-producing bacteria in HIV infection is associated with microbial translocation and immune activation Gut microbiota trajectory in pediatric patients undergoing hematopoietic SCT Gut microbiome-derived metabolites modulate intestinal epithelial cell damage and mitigate graft-versus-host disease Intestinal microbiota influence immune tolerance post allogeneic hematopoietic cell transplantation and intestinal GVHD The effects of prebiotics on microbial dysbiosis, butyrate production and immunity in HIV-infected subjects Butyrogenic bacteria after acute graft-versus-host disease (GVHD) are associated with the development of steroid-refractory GVHD Gut microbiota regulates bile acid metabolism by reducing the levels of taurobeta-muricholic acid, a naturally occurring FXR antagonist Regulation of antibacterial defense in the small intestine by the nuclear bile acid receptor Bile acid malabsorption in HIV infected patients with chronic diarrhoea HIVenteropathy and bile acid malabsorption: response to cholestyramine Bile acid malabsorption in patients with graft-versus-host disease of the gastrointestinal tract Prevention of infection and graft-versus-host disease by suppression of intestinal microflora in children treated with allogeneic bone marrow transplantation Evidence that sustained growth suppression of intestinal anaerobic bacteria reduces the risk of acute graft-versus-host disease after sibling marrow transplantation 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 The influence of gut-decontamination prophylactic antibiotics on acute graft-versus-host disease and survival following allogeneic hematopoietic stem cell transplantation Gut decontamination during allogeneic hematopoietic stem cell transplantation and the risk of acute graft-versus-host disease Fecal microbiota transplantation for patients with steroid-resistant acute graft-versus-host disease of the gut Repeated fecal microbiota transplantations attenuate diarrhea and lead to sustained changes in the fecal microbiota in acute, refractory gastrointestinal graft-versus-host-disease Treating steroid refractory intestinal acute graft-vs.-host disease with fecal microbiota transplantation: a pilot study Loss of mucosal CD103+ DCs and IL-17+ and IL-22+ lymphocytes is associated with mucosal damage in SIV infection Th17 cell dynamics in HIV infection Differential Th17 CD4 T-cell depletion in pathogenic and nonpathogenic lentiviral infections Effects of fecal microbial transplantation on microbiome and immunity in simian immunodeficiency virus-infected macaques Bacteroides are associated with GALT iNKT cell function and reduction of microbial translocation in HIV-1 infection Faecalibacterium prausnitzii is an antiinflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases Rational donor fecal microbiota transplantation in HIV (refresh study) Association between gut microbiota and CD4 recovery in HIV-1 infected patients Fecal transplant is as effective and safe in immunocompromised as non-immunocompromised patients for Clostridium difficile Efficacy of sterile fecal filtrate transfer for treating patients with Clostridium difficile infection Fecal microbiota therapy for recurrent Clostridium difficile infection in HIV-infected persons Fecal transplantation for Clostridium difficile-"all stool may not be created equal Fecal microbiota transplantation for Clostridium difficile-associated colitis in a severely immunocompromized critically ill AIDS patient: a case report Drug-resistant E. coli bacteremia transmitted by fecal microbiota transplant SARS-CoV-2 induced diarrhoea as onset symptom in patient with COVID-19 Prolonged presence of SARS-CoV-2 viral RNA in faecal samples Fecal Microbiota Therapy Used in the Treatment of Clostridium difficile Infection Not Responsive to Conventional Therapies: Health Canada Extended screening costs associated with selecting donors for fecal microbiota transplantation for treatment of metabolic syndrome-associated diseases. Open Forum Infect Dis Standardized frozen preparation for transplantation of fecal microbiota for recurrent Clostridium difficile infection Preparing the gut with antibiotics enhances gut microbiota reprogramming efficiency by promoting xenomicrobiota colonization. Front Microbiol Consensus report: faecal microbiota transferclinical applications and procedures Fecal microbiota transplant for treatment of Clostridium difficile infection in immunocompromised patients Fecal microbiota transplantation for Clostridium difficile infection: systematic review and meta-analysis Oral, capsulized, frozen fecal microbiota transplantation for relapsing Clostridium difficile infection Effect of oral capsule-vs colonoscopy-delivered fecal microbiota transplantation on recurrent Clostridium difficile infection: a randomized clinical trial We are highly grateful to Angie Massicotte, Josée Girouard, and Cezar Iovi for coordination and assistance. The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.Copyright © 2020 Ouyang, Isnard, Lin, Fombuena, Peng, Nair Parvathy, Chen, Silverman and Routy. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.