Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 787
Filter
1.
Mayo Clin Proc ; 99(6): 971-979, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38839189

ABSTRACT

Clostridioides difficile infection (CDI) is a significant public health challenge in the developed world. Although previously CDI was primarily a health care-acquired infection, there are now rising numbers of community-acquired cases in patients without traditional risk factors, such as antibiotic exposure. The landscape for the treatment of CDI has changed significantly during the past decade, including newer diagnostic tests, novel antibiotic regimens, and strategies for microbiome restoration in the form of traditional fecal microbiota transplant and approved live biotherapeutics in an effort to address the underlying pathophysiologic process of gut microbial dysbiosis. We present a concise review for clinicians on the diagnosis and management of both primary and recurrent CDI.


Subject(s)
Anti-Bacterial Agents , Clostridium Infections , Fecal Microbiota Transplantation , Humans , Clostridium Infections/therapy , Clostridium Infections/diagnosis , Anti-Bacterial Agents/therapeutic use , Fecal Microbiota Transplantation/methods , Clostridioides difficile , Gastrointestinal Microbiome , Risk Factors
2.
Curr Opin Gastroenterol ; 40(4): 258-267, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38841848

ABSTRACT

PURPOSE OF REVIEW: The role of the microbiome and dysbiosis is increasingly recognized in the pathogenesis of inflammatory bowel disease (IBD). Intestinal microbiota transplant (IMT), previously termed fecal microbiota transplant has demonstrated efficacy in restoring a healthy microbiome and promoting gut health in recurrent Clostridioides difficile infection. Several randomized trials (RCTs) highlighted IMT's potential in treating ulcerative colitis, while smaller studies reported on its application in managing Crohn's disease and pouchitis. RECENT FINDINGS: This review delves into the current understanding of dysbiosis in IBD, highlighting the distinctions in the microbiota of patients with IBD compared to healthy controls. It explores the mechanisms by which IMT can restore a healthy microbiome and provides a focused analysis of recent RCTs using IMT for inducing and maintaining remission in IBD. Lastly, we discuss the current knowledge gaps that limit its widespread use. SUMMARY: The body of evidence supporting the use of IMT in IBD is growing. The lack of a standardized protocol impedes its application beyond clinical trials. Further research is needed to identify patient profile and disease phenotypes that benefit from IMT, to delineate key donor characteristics, optimize the delivery route, dosage, and frequency.


Subject(s)
Dysbiosis , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Humans , Fecal Microbiota Transplantation/methods , Dysbiosis/therapy , Dysbiosis/microbiology , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/therapy , Randomized Controlled Trials as Topic
3.
BMC Microbiol ; 24(1): 160, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38724904

ABSTRACT

BACKGROUND: Antibiotic-associated diarrhea (AAD) refers to symptoms of diarrhea that cannot be explained by other causes after the use of antibiotics. AAD is thought to be caused by a disruption of intestinal ecology due to antibiotics. Fecal Microbiota Transplantation (FMT) is a treatment method that involves transferring microbial communities from the feces of healthy individuals into the patient's gut. METHOD: We selected 23 AAD patients who received FMT treatment in our department. Before FMT, we documented patients' bowel movement frequency, abdominal symptoms, routine blood tests, and inflammatory markers, and collected fecal samples for 16S rRNA sequencing to observe changes in the intestinal microbiota. Patients' treatment outcomes were followed up 1 month and 3 months after FMT. RESULTS: Out of the 23 AAD patients, 19 showed a clinical response to FMT with alleviation of abdominal symptoms. Among them, 82.61% (19/23) experienced relief from diarrhea, 65% (13/20) from abdominal pain, 77.78% (14/18) from abdominal distension, and 57.14% (4/7) from bloody stools within 1 month after FMT. Inflammatory markers IL-8 and CRP significantly decreased after FMT, but there were no noticeable changes in WBC, IL-6, and TNF-α before and after transplantation. After FMT, the abundance of Bacteroides and Faecalibacterium increased in patients' fecal samples, while the abundance of Escherichia-Shigella and Veillonella decreased. CONCLUSION: FMT has a certain therapeutic effect on AAD, and can alleviate abdominal symptoms and change the intestinal microbiota of patients.


Subject(s)
Anti-Bacterial Agents , Diarrhea , Fecal Microbiota Transplantation , Feces , Gastrointestinal Microbiome , RNA, Ribosomal, 16S , Humans , Diarrhea/microbiology , Diarrhea/therapy , Fecal Microbiota Transplantation/methods , Female , Male , Middle Aged , Anti-Bacterial Agents/adverse effects , Feces/microbiology , Adult , RNA, Ribosomal, 16S/genetics , Aged , Treatment Outcome , Bacteria/classification , Bacteria/isolation & purification , Bacteria/genetics
4.
World J Gastroenterol ; 30(16): 2179-2183, 2024 Apr 28.
Article in English | MEDLINE | ID: mdl-38690018

ABSTRACT

In this editorial we comment on the article published in the recent issue of the World journal of Gastroenterology. We focus specifically on the mechanisms un-derlying the effects of fecal microbiota transplantation (FMT) for irritable bowel syndrome (IBS), the factors which affect the outcomes of FMT in IBS patients, and challenges. FMT has emerged as a efficacious intervention for clostridium difficile infection and holds promise as a therapeutic modality for IBS. The utilization of FMT in the treatment of IBS has undergone scrutiny in numerous randomized controlled trials, yielding divergent outcomes. The current frontier in this field seeks to elucidate these variations, underscore the existing knowledge gaps that necessitate exploration, and provide a guideline for successful FMT imple-mentation in IBS patients. At the same time, the application of FMT as a treatment for IBS confronts several challenges.


Subject(s)
Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Irritable Bowel Syndrome , Irritable Bowel Syndrome/therapy , Irritable Bowel Syndrome/microbiology , Fecal Microbiota Transplantation/methods , Humans , Treatment Outcome , Feces/microbiology , Randomized Controlled Trials as Topic , Clostridioides difficile/pathogenicity , Clostridium Infections/therapy , Clostridium Infections/microbiology
5.
Medicine (Baltimore) ; 103(19): e38088, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38728472

ABSTRACT

Microbiota modulation, the intentional change in the structure and function of the microbial community, is an emerging trajectory that holds the promise to mitigate an infinite number of health issues. The present review illustrates the underlying principles of microbiota modulation and the various applications of this fundamental process to human health, healthcare management, and pharmacologic interventions. Different strategies, directing on dietary interventions, fecal microbiota transplantation, treatment with antibiotics, bacteriophages, microbiome engineering, and modulation of the immune system, are described in detail. This therapeutic implication is reflected in clinical applications to gastrointestinal disorders and immune-mediated diseases for microbiota-modulating agents. In addition to this, the review outlines the challenges of translating researched outcomes into clinical practice to consider safety and provides insights into future research directions of this rapidly developing area.


Subject(s)
Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Humans , Fecal Microbiota Transplantation/methods , Gastrointestinal Microbiome/physiology , Anti-Bacterial Agents/therapeutic use , Probiotics/therapeutic use , Gastrointestinal Diseases/therapy , Gastrointestinal Diseases/microbiology
6.
J Int Med Res ; 52(5): 3000605241241000, 2024 May.
Article in English | MEDLINE | ID: mdl-38749910

ABSTRACT

Ileostomy diverts the flow of feces, which can result in malnutrition in the distal part of the intestine. The diversity of the gut microbiota consequently decreases, ultimately leading to intestinal dysbiosis and dysfunction. This condition can readily result in diversion colitis (DC). Potential treatment strategies include interventions targeting the gut microbiota. In this case study, we effectively treated a patient with severe DC by ileostomy and allogeneic fecal microbiota transplantation (FMT). A 69-year-old man presented with a perforated malignant tumor in the descending colon and an iliac abscess. He underwent laparoscopic radical sigmoid colon tumor resection and prophylactic ileostomy. Follow-up colonoscopy 3 months postoperatively revealed diffuse intestinal mucosal congestion and edema along with granular inflammatory follicular hyperplasia, leading to a diagnosis of severe DC. After two rounds of allogeneic FMT, both the intestinal mucosal bleeding and edema significantly improved, as did the diversity of the gut microbiota. The positive outcome of allogeneic FMT in this case highlights the potential advantages that this procedure can offer patients with DC. However, few studies have focused on allogeneic FMT, and more in-depth research is needed to gain a better understanding.


Subject(s)
Colitis , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Ileostomy , Humans , Male , Aged , Fecal Microbiota Transplantation/methods , Colitis/microbiology , Colitis/therapy , Transplantation, Homologous/methods , Treatment Outcome , Colonoscopy
7.
World J Gastroenterol ; 30(19): 2603-2611, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38817661

ABSTRACT

BACKGROUND: The gut microbiota is strongly associated with radiation-induced gut damage. This study aimed to assess the effectiveness and safety of intestinal microecological transplantation for treating patients with chronic radiation enteritis. CASE SUMMARY: A 64-year-old female with cervical cancer developed abdominal pain, diarrhea, and blood in the stool 1 year after radiotherapy. An electronic colonoscopy was performed to diagnose chronic radiation enteritis. Two courses of intestinal microecological transplantation and full-length 16S rRNA microbiological analysis were performed. The patient experienced short- and long-term relief from symptoms without adverse effects. Whole 16S rRNA sequencing revealed significant differences in the intestinal flora's composition between patient and healthy donors. Pathogenic bacteria, such as Escherichia fergusonii and Romboutsia timonensis, were more in the patient. Beneficial bacteria such as Faecalibacterium prausnitzii, Fusicatenibacter saccharivorans, Ruminococcus bromii, and Bifidobacterium longum were more in the healthy donors. Intestinal microbiota transplantation resulted in a significant change in the patient's intestinal flora composition. The composition converged with the donor's flora, with an increase in core beneficial intestinal bacteria, such as Eubacterium rectale, and a decrease in pathogenic bacteria. Changes in the intestinal flora corresponded with the patients' alleviating clinical symptoms. CONCLUSION: Intestinal microecological transplantation is an effective treatment for relieving the clinical symptoms of chronic radiation enteritis by altering the composition of the intestinal flora. This study provides a new approach for treating patients with chronic radiation enteritis.


Subject(s)
Enteritis , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Radiation Injuries , Uterine Cervical Neoplasms , Humans , Female , Middle Aged , Enteritis/microbiology , Enteritis/diagnosis , Enteritis/etiology , Enteritis/therapy , Radiation Injuries/diagnosis , Radiation Injuries/microbiology , Radiation Injuries/etiology , Radiation Injuries/surgery , Gastrointestinal Microbiome/radiation effects , Fecal Microbiota Transplantation/methods , Uterine Cervical Neoplasms/radiotherapy , RNA, Ribosomal, 16S/genetics , Treatment Outcome , Chronic Disease , Colonoscopy , Intestines/microbiology , Intestines/radiation effects , Feces/microbiology , Radiotherapy/adverse effects
9.
Exp Neurol ; 376: 114774, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38599367

ABSTRACT

Peripheral nerve injury (PNI) resulting from trauma or neuropathies can cause significant disability, and its prognosis deteriorates with age. Emerging evidence suggests that gut dysbiosis and reduced fecal short-chain fatty acids (SCFAs) contribute to an age-related systemic hyperinflammation (inflammaging), which hinders nerve recovery after injury. This study thus aimed to evaluate the pro-regenerative effects of a rejuvenating fecal microbiota transplant (FMT) in a preclinical PNI model using aged mice. Aged C57BL/6 mice underwent bilateral crush injuries to their sciatic nerves. Subsequently, they either received FMT from young donors at three and four days after the injury or retained their aged gut microbiota. We analyzed gut microbiome composition and SCFA concentrations in fecal samples. The integrity of the ileac mucosal barrier was assessed by immunofluorescence staining of Claudin-1. Flow cytometry was utilized to examine immune cells and cytokine production in the ileum, spleen, and sciatic nerve. Various assessments, including behavioural tests, electrophysiological studies, and morphometrical analyses, were conducted to evaluate peripheral nerve function and repair following injury. Rejuvenating FMT reversed age-related gut dysbiosis by increasing Actinobacteria, especially Bifidobacteriales genera. This intervention also led to an elevation of gut SCFA levels and mitigated age-related ileac mucosal leakiness in aged recipients. Additionally, it augmented the number of T-helper 2 (Th2) and regulatory T (Treg) cells in the ileum and spleen, with the majority being positive for anti-inflammatory interleukin-10 (IL-10). In sciatic nerves, rejuvenating FMT resulted in increased M2 macrophage counts and a higher IL-10 production by IL-10+TNF-α- M2 macrophage subsets. Ultimately, restoring a youthful gut microbiome in aged mice led to improved nerve repair and enhanced functional recovery after PNI. Considering that FMT is already a clinically available technique, exploring novel translational strategies targeting the gut microbiome to enhance nerve repair in the elderly seems promising and warrants further evaluation.


Subject(s)
Aging , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Mice, Inbred C57BL , Nerve Regeneration , Animals , Mice , Fecal Microbiota Transplantation/methods , Gastrointestinal Microbiome/physiology , Nerve Regeneration/physiology , Male , Peripheral Nerve Injuries/therapy , Inflammation/metabolism , Inflammation/therapy , Dysbiosis/therapy , Sciatic Nerve/injuries
10.
Sci Rep ; 14(1): 9558, 2024 04 26.
Article in English | MEDLINE | ID: mdl-38664425

ABSTRACT

Neurodegenerative diseases (NDDs) are characterized by neuronal damage and progressive loss of neuron function. Microbiome-based interventions, such as dietary interventions, biotics, and fecal microbiome transplant, have been proposed as a novel approach to managing symptoms and modulating disease progression. Emerging clinical trials have investigated the efficacy of interventions modulating the GM in alleviating or reversing disease progression, yet no comprehensive synthesis have been done. A systematic review of the literature was therefore conducted to investigate the efficacy of microbiome-modulating methods. The search yielded 4051 articles, with 15 clinical trials included. The overall risk of bias was moderate in most studies. Most microbiome-modulating interventions changed the GM composition. Despite inconsistent changes in GM composition, the meta-analysis showed that microbiome-modulating interventions improved disease burden (SMD, - 0.57; 95% CI - 0.93 to - 0.21; I2 = 42%; P = 0.002) with a qualitative trend of improvement in constipation. However, current studies have high methodological heterogeneity and small sample sizes, requiring more well-designed and controlled studies to elucidate the complex linkage between microbiome, microbiome-modulating interventions, and NDDs.


Subject(s)
Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Neurodegenerative Diseases , Humans , Neurodegenerative Diseases/microbiology , Neurodegenerative Diseases/therapy , Fecal Microbiota Transplantation/methods , Probiotics/therapeutic use , Microbiota
11.
Front Cell Infect Microbiol ; 14: 1370999, 2024.
Article in English | MEDLINE | ID: mdl-38660489

ABSTRACT

Diabetes mellitus (DM) refers to a group of chronic diseases with global prevalence, characterized by persistent hyperglycemia resulting from various etiologies. DM can harm various organ systems and lead to acute or chronic complications, which severely endanger human well-being. Traditional treatment mainly involves controlling blood sugar levels through replacement therapy with drugs and insulin; however, some patients still find a satisfactory curative effect difficult to achieve. Extensive research has demonstrated a close correlation between enteric dysbacteriosis and the pathogenesis of various types of DM, paving the way for novel therapeutic approaches targeting the gut microbiota to manage DM. Fecal microbiota transplantation (FMT), a method for re-establishing the intestinal microbiome balance, offers new possibilities for treating diabetes. This article provides a comprehensive review of the correlation between DM and the gut microbiota, as well as the current advancements in FMT treatment for DM, using FMT as an illustrative example. This study aims to offer novel perspectives and establish a theoretical foundation for the clinical diagnosis and management of DM.


Subject(s)
Diabetes Mellitus , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Fecal Microbiota Transplantation/methods , Humans , Diabetes Mellitus/therapy , Diabetes Mellitus/microbiology , Dysbiosis/therapy , Animals , Feces/microbiology
12.
Medicina (Kaunas) ; 60(4)2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38674209

ABSTRACT

The gastrointestinal tract is inhabited by the gut microbiota. The main phyla are Firmicutes and Bacteroidetes. In non-alcoholic fatty liver disease, now renamed metabolic dysfunction-associated fatty liver disease (MAFLD), an alteration in Firmicutes and Bacteroidetes abundance promotes its pathogenesis and evolution into non-alcoholic steatohepatitis, liver cirrhosis, and hepatocellular carcinoma. For this reason, early treatment is necessary to counteract its progression. The aim of the present narrative review is to evaluate the different therapeutic approaches to MAFLD. The most important treatment for MAFLD is lifestyle changes. In this regard, the Mediterranean diet could be considered the gold standard in the prevention and treatment of MAFLD. In contrast, a Western diet should be discouraged. Probiotics and fecal microbiota transplantation seem to be valid, safe, and effective alternatives for MAFLD treatment. However, more studies with a longer follow-up and with a larger cohort of patients are needed to underline the more effective approaches to contrasting MAFLD.


Subject(s)
Diet, Mediterranean , Fecal Microbiota Transplantation , Non-alcoholic Fatty Liver Disease , Humans , Fecal Microbiota Transplantation/methods , Non-alcoholic Fatty Liver Disease/therapy , Non-alcoholic Fatty Liver Disease/microbiology , Probiotics/therapeutic use , Probiotics/administration & dosage , Gastrointestinal Microbiome/physiology
13.
J Hosp Infect ; 148: 189-219, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38609760

ABSTRACT

The first British Society of Gastroenterology (BSG) and Healthcare Infection Society (HIS)-endorsed faecal microbiota transplant (FMT) guidelines were published in 2018. Over the past 5 years, there has been considerable growth in the evidence base (including publication of outcomes from large national FMT registries), necessitating an updated critical review of the literature and a second edition of the BSG/HIS FMT guidelines. These have been produced in accordance with National Institute for Health and Care Excellence-accredited methodology, thus have particular relevance for UK-based clinicians, but are intended to be of pertinence internationally. This second edition of the guidelines have been divided into recommendations, good practice points and recommendations against certain practices. With respect to FMT for Clostridioides difficile infection (CDI), key focus areas centred around timing of administration, increasing clinical experience of encapsulated FMT preparations and optimising donor screening. The latter topic is of particular relevance given the COVID-19 pandemic, and cases of patient morbidity and mortality resulting from FMT-related pathogen transmission. The guidelines also considered emergent literature on the use of FMT in non-CDI settings (including both gastrointestinal and non-gastrointestinal indications), reviewing relevant randomised controlled trials. Recommendations are provided regarding special areas (including compassionate FMT use), and considerations regarding the evolving landscape of FMT and microbiome therapeutics.


Subject(s)
Clostridium Infections , Fecal Microbiota Transplantation , Fecal Microbiota Transplantation/methods , Humans , Clostridium Infections/therapy , United Kingdom , Clostridioides difficile , COVID-19/therapy , Recurrence , Gastroenterology/standards , Gastroenterology/methods , SARS-CoV-2 , Societies, Medical
14.
Gut ; 73(7): 1052-1075, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38609165

ABSTRACT

The first British Society of Gastroenterology (BSG) and Healthcare Infection Society (HIS)-endorsed faecal microbiota transplant (FMT) guidelines were published in 2018. Over the past 5 years, there has been considerable growth in the evidence base (including publication of outcomes from large national FMT registries), necessitating an updated critical review of the literature and a second edition of the BSG/HIS FMT guidelines. These have been produced in accordance with National Institute for Health and Care Excellence-accredited methodology, thus have particular relevance for UK-based clinicians, but are intended to be of pertinence internationally. This second edition of the guidelines have been divided into recommendations, good practice points and recommendations against certain practices. With respect to FMT for Clostridioides difficile infection (CDI), key focus areas centred around timing of administration, increasing clinical experience of encapsulated FMT preparations and optimising donor screening. The latter topic is of particular relevance given the COVID-19 pandemic, and cases of patient morbidity and mortality resulting from FMT-related pathogen transmission. The guidelines also considered emergent literature on the use of FMT in non-CDI settings (including both gastrointestinal and non-gastrointestinal indications), reviewing relevant randomised controlled trials. Recommendations are provided regarding special areas (including compassionate FMT use), and considerations regarding the evolving landscape of FMT and microbiome therapeutics.


Subject(s)
Clostridium Infections , Fecal Microbiota Transplantation , Gastroenterology , Fecal Microbiota Transplantation/methods , Humans , Clostridium Infections/therapy , Gastroenterology/standards , COVID-19/therapy , SARS-CoV-2 , Recurrence , Clostridioides difficile , United Kingdom , Societies, Medical
15.
Anaerobe ; 86: 102841, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38521227

ABSTRACT

OBJECTIVES: Clostridioides difficile infection (CDI) is the leading hospital-acquired infection in North America. While previous work on fecal microbiota transplantation (FMT), a highly effective treatment for CDI, has focused on colonization resistance mounted against C. difficile by FMT-delivered commensals, the effects of FMT on host gene expression are relatively unexplored. This study aims to identify transcriptional changes associated with FMT, particularly changes associated with protective immune responses. METHODS: Gene expression was assessed on day 2 and day 7 after FMT in mice after antibiotic-induced dysbiosis. Flow cytometry was also performed on colon and mesenteric lymph nodes at day 7 to investigate changes in immune cell populations. RESULTS: FMT administration after antibiotic-induced dysbiosis successfully restored microbial alpha diversity to levels of donor mice by day 7 post-FMT. Bulk RNA sequencing of cecal tissue at day 2 identified immune genes, including both pro-inflammatory and Type 2 immune pathways as upregulated after FMT. RNA sequencing was repeated on day 7 post-FMT, and expression of these immune genes was decreased along with upregulation of genes associated with restoration of intestinal homeostasis. Immunoprofiling on day 7 identified increased colonic CD45+ immune cells that exhibited dampened Type 1 and heightened regulatory and Type 2 responses. These include an increased abundance of eosinophils, alternatively activated macrophages, Th2, and T regulatory cell populations. CONCLUSION: These results highlight the impact of FMT on host gene expression, providing evidence that FMT restores intestinal homeostasis after antibiotic treatment and facilitates tolerogenic and Type 2 immune responses.


Subject(s)
Clostridium Infections , Disease Models, Animal , Fecal Microbiota Transplantation , Animals , Fecal Microbiota Transplantation/methods , Mice , Clostridium Infections/therapy , Clostridium Infections/immunology , Clostridium Infections/microbiology , Gastrointestinal Microbiome , Dysbiosis/therapy , Clostridioides difficile/immunology , Immune Tolerance , Mice, Inbred C57BL
16.
J Clin Lab Anal ; 38(7): e25023, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38544348

ABSTRACT

BACKGROUND: Faecal microbiota transplantation (FMT) is an established treatment for Clostridioides difficile infection and is under investigation for other conditions. The availability of suitable donors and the logistics of fresh stool preparation present challenges, making frozen, biobanked stools an attractive alternative. AIMS: This study aimed to evaluate the long-term viability of bacterial populations in faecal samples stored at -80°C for up to 12 months, supporting the feasibility of using frozen grafts for FMT. METHODS: Fifteen faecal samples from nine healthy donors were processed, mixed with cryoprotectants and stored at -80°C. Samples were assessed at baseline and after 3, 6 and 12 months using quantitative culturing methods to determine the concentration of live bacteria. RESULTS: Quantitative analysis showed no significant decrease in bacterial viability over the 12-month period for both aerobic and anaerobic cultures (p = 0.09). At all timepoints, the coefficients of variability in colony-forming unit (CFU) counts were greater between samples (102 ± 21% and 100 ± 13% for aerobic and anaerobic cultures, respectively) than the variability between measurements of the same sample (30 ± 22% and 30 ± 19%). CONCLUSIONS: The study confirmed that faecal microbiota can be preserved with high viability in deep-freeze storage for up to a year, making allogenic FMT from biobanked samples a viable and safer option for patients. However, a multidonor approach may be beneficial to mitigate the risk of viability loss in any single donor sample.


Subject(s)
Fecal Microbiota Transplantation , Feces , Microbial Viability , Humans , Fecal Microbiota Transplantation/methods , Feces/microbiology , Freezing , Cryopreservation/methods , Male
17.
Dig Dis Sci ; 69(5): 1778-1784, 2024 May.
Article in English | MEDLINE | ID: mdl-38457115

ABSTRACT

BACKGROUND: Fecal microbiota transplants can be administered orally in encapsulated form or require invasive procedures to administer liquid formulations. There is a need for an oral liquid formulation of fecal microbiota for patients who are unable to swallow capsules, especially if they require multiple, repeated administrations. AIMS: These studies were conducted to develop a protocol to manufacture an organoleptically acceptable powdered fecal microbiota formulation that can be suspended in a liquid carrier and used for fecal microbiota transplantation. METHODS: Several processing steps were investigated, including extra washes of microbiota prior to lyophilization and an addition of a flavoring agent. The viability of bacteria in the transplant formulation was tested using live/dead microscopy staining and engraftment into antibiotic-treated mice. After development of a clinical protocol for suspension of the powdered microbiota, the new formulation was tested in three elderly patients with recurrent Clostridioides difficile infections and who have difficulties in swallowing capsules. Changes in the microbial community structure in one of the patients were characterized using 16S rRNA gene profiling and engraftment analysis. RESULTS: The processing steps used to produce an organoleptically acceptable suspension of powdered fecal microbiota did not result in loss of its viability. The powder could be easily suspended in a liquid carrier. The use of the new formulation was associated with abrogation of the cycle of C. difficile infection recurrences in the three patients. CONCLUSION: We developed a novel organoleptically acceptable liquid formulation of fecal microbiota that is suitable for use in clinical trials for patients with difficulties in swallowing capsules.


Subject(s)
Fecal Microbiota Transplantation , Fecal Microbiota Transplantation/methods , Humans , Animals , Administration, Oral , Clostridium Infections/therapy , Clostridium Infections/microbiology , Mice , Aged , Feces/microbiology , Clostridioides difficile/isolation & purification , Recurrence , Male , Female , Gastrointestinal Microbiome/drug effects , Powders , Treatment Outcome , Aged, 80 and over
18.
Blood Adv ; 8(9): 2074-2084, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38471063

ABSTRACT

ABSTRACT: Disruption of the intestinal microbiome is observed with acute graft-versus-host disease (GVHD) of the lower gastrointestinal (LGI) tract, and fecal microbiota transplantation (FMT) has successfully cured steroid-refractory cases. In this open-label, single-arm, pilot study, third-party, single-donor FMT was administered in combination with systemic corticosteroids to participants with high-risk acute LGI GVHD, with a focus on treatment-naïve cases. Participants were scheduled to receive 1 induction dose (15 capsules per day for 2 consecutive days), followed by 3 weekly maintenance doses, consisting of 15 capsules per dose. The primary end point of the study was feasibility, which would be achieved if ≥80% of participants able to swallow ≥40 of the 75 scheduled capsules. Ten participants (9 treatment-naïve; 1 steroid-refractory) were enrolled and treated. The study met the primary end point, with 9 of 10 participants completing all eligible doses. Organ-specific LGI complete response rate at day 28 was 70%. Initial clinical response was observed within 1 week for all responders, and clinical responses were durable without recurrent LGI GVHD in complete responders. Exploratory analyses suggest that alpha diversity increased after FMT. Although recipient microbiome composition never achieved a high degree of donor similarity, expansion of donor-derived species and increases in tryptophan metabolites and short-chain fatty acids were observed within the first 7 days after FMT. Investigation into the use of microbiome-targeted interventions earlier in the treatment paradigm for acute LGI GVHD is warranted. This trial was registered at www.ClinicalTrials.gov as #NCT04139577.


Subject(s)
Fecal Microbiota Transplantation , Graft vs Host Disease , Humans , Graft vs Host Disease/therapy , Graft vs Host Disease/etiology , Fecal Microbiota Transplantation/methods , Male , Female , Middle Aged , Adult , Gastrointestinal Microbiome , Aged , Pilot Projects , Acute Disease , Treatment Outcome
19.
J Affect Disord ; 354: 309-317, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38499070

ABSTRACT

BACKGROUND: The gut-brain-axis has a role in mental health disorders. In people with generalized anxiety disorder, GAD,1 normal flora Escherichia-Shigella, are significantly elevated. Fecal microbiota transplant, FMT,2 has been used to alter the gut composition in unhealthy individuals. There may be a role for FMT in the treatment of GAD to improve the gut-brain-axis. METHODS: A systematic review of literature was conducted on articles published in PubMed, CINAHL Plus, Scopus, Cochrane Library, and Wed of Science from 2000 to 2022 that analyzed FMT as a modality to alter the gut microbiome in which Escherichia-Shigella levels were quantified and reported. RESULTS: Of 1916 studies identified, 14 fit criteria and were included. Recipients undergoing FMT procedures had at least one enteric diagnosis and increased percentages of Escherichia-Shigella pre-FMT. Five studies on recurrent Clostridioides difficile infection, three irritable bowel syndrome, two ulcerative colitis, one ulcerative colitis and recurrent Clostridioides difficile infection, one acute intestinal and chronic graft-vs-host disease, one pouchitis, and one slow transit constipation. 10 articles (71.4 %) showed decreased levels of Escherichia-Shigella post-FMT compared to pre-FMT. Four studies claimed the results were significant (40 %). LIMITATIONS: Limitations include potential bias in study selection, study methods of analysis, and generalization of results. CONCLUSIONS: The gut-brain-axis has a role in GAD. Those with GAD have significantly higher Escherichia-Shigella compared to those without GAD. FMT has the potential to decrease Escherichia-Shigella in patients with GAD to positively alter the gut-brain-axis as a potential for future GAD treatment.


Subject(s)
Clostridium Infections , Colitis, Ulcerative , Humans , Fecal Microbiota Transplantation/methods , Feces , Anxiety Disorders/therapy , Clostridium Infections/therapy , Treatment Outcome
20.
Gut Microbes ; 16(1): 2327442, 2024.
Article in English | MEDLINE | ID: mdl-38478462

ABSTRACT

In small series, third-party fecal microbiota transplantation (FMT) has been successful in decolonizing the gut from clinically relevant antibiotic resistance genes (ARGs). Less is known about the short- and long-term effects of FMT on larger panels of ARGs. We analyzed 226 pre- and post-treatment stool samples from a randomized placebo-controlled trial of FMT in 100 patients undergoing allogeneic hematopoietic cell transplantation or receiving anti-leukemia induction chemotherapy for 47 ARGs. These patients have heavy antibiotic exposure and a high incidence of colonization with multidrug-resistant organisms. Samples from each patient spanned a period of up to 9 months, allowing us to describe both short- and long-term effects of FMT on ARGs, while the randomized design allowed us to distinguish between spontaneous changes vs. FMT effect. We find an overall bimodal pattern. In the first phase (days to weeks after FMT), low-level transfer of ARGs largely associated with commensal healthy donor microbiota occurs. This phase is followed by long-term resistance to new ARGs as stable communities with colonization resistance are formed after FMT. The clinical implications of these findings are likely context-dependent and require further research. In the setting of cancer and intensive therapy, long-term ARG decolonization could translate into fewer downstream infections.


Subject(s)
Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Humans , Fecal Microbiota Transplantation/methods , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Gastrointestinal Microbiome/genetics , Treatment Outcome , Drug Resistance, Microbial , Feces
SELECTION OF CITATIONS
SEARCH DETAIL
...