1
|
Hoops SL, Moutsoglou D, Vaughn BP, Khoruts A, Knights D. Metagenomic source tracking after microbiota transplant therapy. Gut Microbes 2025; 17:2487840. [PMID: 40229213 PMCID: PMC12005403 DOI: 10.1080/19490976.2025.2487840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/07/2025] [Accepted: 03/28/2025] [Indexed: 04/16/2025] Open
Abstract
Reliable engraftment assessment of donor microbial communities and individual strains is an essential component of characterizing the pharmacokinetics of microbiota transplant therapies (MTTs). Recent methods for measuring donor engraftment use whole-genome sequencing and reference databases or metagenome-assembled genomes (MAGs) to track individual bacterial strains but lack the ability to disambiguate DNA that matches both donor and patient microbiota. Here, we describe a new, cost-efficient analytic pipeline, MAGEnTa, which compares post-MTT samples to a database comprised MAGs derived directly from donor and pre-treatment metagenomic data, without relying on an external database. The pipeline uses Bayesian statistics to determine the likely sources of ambiguous reads that align with both the donor and pre-treatment samples. MAGEnTa recovers engrafted strains with minimal type II error in a simulated dataset and is robust to shallow sequencing depths in a downsampled dataset. Applying MAGEnTa to a dataset from a recent MTT clinical trial for ulcerative colitis, we found the results to be consistent with 16S rRNA gene SourceTracker analysis but with added MAG-level specificity. MAGEnTa is a powerful tool to study community and strain engraftment dynamics in the development of MTT-based treatments that can be integrated into frameworks for functional and taxonomic analysis.
Collapse
Affiliation(s)
- Susan L. Hoops
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN, USA
- Biotechnology Institute, University of Minnesota, Minneapolis, MN, USA
| | - Daphne Moutsoglou
- Gastroenterology Section, Minneapolis VA Health Care System, Minneapolis, MN, USA
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Byron P. Vaughn
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
- Division of Gastroenterology, University of Minnesota, Minneapolis, MN, USA
| | - Alexander Khoruts
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
- Division of Gastroenterology, University of Minnesota, Minneapolis, MN, USA
- Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Dan Knights
- Department of Computer Science and Engineering, University of Minnesota, Minneapolis, MN, USA
- Biotechnology Institute, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
2
|
Hoffmann DE, Javitt GH, Kelly CR, Keller JJ, Baunwall SMD, Hvas CL. Fecal microbiota transplantation: a tale of two regulatory pathways. Gut Microbes 2025; 17:2493901. [PMID: 40302307 DOI: 10.1080/19490976.2025.2493901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/04/2025] [Accepted: 04/10/2025] [Indexed: 05/02/2025] Open
Abstract
Fecal microbiota transplantation (FMT) is a procedure involving the transfer of intestinal microbiota from a healthy donor to a patient to restore a functional intestinal microbiome. First described in modern science in 1958, the use of FMT has been practiced for decades, but only during the past dozen years have clinical frameworks and legal regulations from competent authorities been developed. Future development of microbiota-derived medical therapies will be shaped by the regulatory frameworks of various jurisdictions. This review examines the historical development and status of FMT regulations in the United States and Europe, with particular attention to their respective approaches to ensuring the safety and quality of the therapeutic product and patient access.
Collapse
Affiliation(s)
- Diane E Hoffmann
- University of Maryland Francis King Carey School of Law, Baltimore, MD, USA
| | | | - Colleen R Kelly
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Josbert J Keller
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, The Hague, Netherlands
- Department of Gastroenterology, Haaglanden Medical Center, The Hague, Netherlands
- Netherlands Donor Feces Bank, Leiden, Netherlands
| | | | - Christian Lodberg Hvas
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
3
|
Faith JJ. Assessing live microbial therapeutic transmission. Gut Microbes 2025; 17:2447836. [PMID: 39746875 DOI: 10.1080/19490976.2024.2447836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/09/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025] Open
Abstract
The development of fecal microbiota transplantation and defined live biotherapeutic products for the treatment of human disease has been an empirically driven process yielding a notable success of approved drugs for the treatment of recurrent Clostridioides difficile infection. Assessing the potential of this therapeutic modality in other indications with mixed clinical results would benefit from consistent quantitative frameworks to characterize drug potency and composition and to assess the impact of dose and composition on the frequency and duration of strain engraftment. Monitoring these drug properties and engraftment outcomes would help identify minimally sufficient sets of microbial strains to treat disease and provide insights into the intersection between microbial function and host physiology. Broad and correct usage of strain detection methods is essential to this advancement. This article describes strain detection approaches, where they are best applied, what data they require, and clinical trial designs that are best suited to their application.
Collapse
Affiliation(s)
- Jeremiah J Faith
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
4
|
Theuretzbacher U. The global resistance problem and the clinical antibacterial pipeline. Nat Rev Microbiol 2025:10.1038/s41579-025-01169-8. [PMID: 40210708 DOI: 10.1038/s41579-025-01169-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2025] [Indexed: 04/12/2025]
Abstract
A comprehensive analysis of the clinical antibacterial pipeline demonstrates that there is a limited range of strategies that are primarily focused on modified versions of widely used chemical classes. These modifications aim to circumvent class-specific resistance mechanisms and reduce resistance rates in certain multidrug-resistant pathogens. Owing to the great variation in resistance rates and mechanisms, the clinical success of current approaches varies substantially across different countries, regions, and economic and environmental conditions, which affects the global societal value of these antibiotics that remain vulnerable to cross-resistance. Although there has been some progress in developing urgently needed antibiotics with novel targets and chemical structures, some of which have advanced to phase I/II trials, further breakthroughs are required. Additionally, adjunctive agents designed to enhance the outcome of conventional antibiotic therapies, along with bacteriophages that offer targeted and personalized treatments, are also under investigation. However, the potential of adjunctive therapeutics, such as antivirulence agents, and bacteriophages has yet to be realized in terms of feasibility and global societal impact.
Collapse
|
5
|
Raeisi H, Leeflang J, Hasan S, Woods SL. Bioengineered Probiotics for Clostridioides difficile Infection: An Overview of the Challenges and Potential for This New Treatment Approach. Probiotics Antimicrob Proteins 2025; 17:763-780. [PMID: 39531149 DOI: 10.1007/s12602-024-10398-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
The rapid increase in microbial antibiotic resistance in Clostridioides difficile (C. difficile) strains and the formation of hypervirulent strains have been associated with a global increase in the incidence of C. difficile infection (CDI) and subsequently, an increase in the rate of recurrence. These consequences have led to an urgent need to develop new and promising alternative strategies to control this pathogen. Engineered probiotics are exciting new bacterial strains produced by editing the genome of the original probiotics. Recently, engineered probiotics have been used to develop delivery vehicles for vaccines, diagnostics, and therapeutics. Recent studies have demonstrated engineered probiotics may potentially be an effective approach to control or treat CDI. This review provides a brief overview of the considerations for engineered probiotics for medicinal use, with a focus on recent preclinical research using engineered probiotics to prevent or treat CDI. We also address the challenges faced in the production of engineered strains and how they may be overcome in the application of these agents to meet patient needs in the future.
Collapse
Affiliation(s)
- Hamideh Raeisi
- Gastroenterology and Liver Diseases Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Julia Leeflang
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5000, Australia
| | - Sadia Hasan
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5000, Australia
| | - Susan L Woods
- Adelaide Medical School, University of Adelaide, Adelaide, SA, 5000, Australia
- Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, 5000, Australia
| |
Collapse
|
6
|
Alonso CD, Tillotson GS, Bidell MR, Guthmueller B, Hoeyer F, Fischer M, Dubberke ER. Safety and Efficacy of Fecal Microbiota, Live-jslm, in Preventing Recurrent Clostridioides difficile Infection in Participants Who Were Mildly to Moderately Immunocompromised in the Phase 3 PUNCH CD3-OLS Study. Open Forum Infect Dis 2025; 12:ofaf117. [PMID: 40177588 PMCID: PMC11961354 DOI: 10.1093/ofid/ofaf117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 02/28/2025] [Indexed: 04/05/2025] Open
Abstract
Background Fecal microbiota, live-jslm (RBL; Rebyota), is the first Food and Drug Administration-approved, single-dose, microbiota-based live biotherapeutic to prevent recurrent Clostridioides difficile infection (rCDI) in adults following standard-of-care antimicrobials. Patients who are immunocompromised are often considered at higher risk for C difficile infection, including recurrence, as compared with those who are immunocompetent. This subgroup analysis of PUNCH CD3-OLS (NCT03931941) evaluated RBL safety and efficacy in participants with rCDI who were considered mildly to moderately immunocompromised. Methods Participants with rCDI who had immunocompromising conditions and/or were taking immunosuppressive medications were included. Treatment-emergent adverse events (TEAEs) were collected for up to 6 months following RBL administration. Efficacy outcomes included treatment success at 8 weeks and sustained clinical response at 6 months. Results Overall, 793 participants were enrolled in PUNCH CD3-OLS and 697 received RBL; 141 were included in the immunocompromised subgroup. TEAEs within 8 weeks were reported by 44.7% and 48.0% of participants in the immunocompromised and nonimmunocompromised subgroups, respectively; most events were mild or moderate gastrointestinal disorders. Serious TEAEs within 8 weeks were reported by 4.3% and 3.8% of participants in the immunocompromised and nonimmunocompromised subgroups. No RBL-related systemic infections occurred. In the immunocompromised subgroup, the treatment success rate at 8 weeks was 75.7% and the sustained clinical response rate at 6 months was 88.7%; similar rates were observed in the nonimmunocompromised subgroup (73.3% and 91.6%). Conclusions Results of this subgroup analysis of PUNCH CD3-OLS suggest that RBL is safe and efficacious for the prevention of rCDI in participants with mildly to moderately immunocompromising conditions. Clinical Trials Registration NCT03931941.
Collapse
Affiliation(s)
- Carolyn D Alonso
- Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | | - Erik R Dubberke
- Washington University School of Medicine, St Louis, Missouri, USA
| |
Collapse
|
7
|
Khanna S, Seo S, Yang M, Garcia-Horton V, Gao Y, Kim HH, Ormenaj L, Guo A. Characteristics and Real-World Outcomes of Patients Treated with Fecal Microbiota, Live-jslm (RBL) for the Prevention of Recurrent Clostridioides difficile Infection. Infect Dis Ther 2025; 14:793-802. [PMID: 40119029 PMCID: PMC11993521 DOI: 10.1007/s40121-025-01130-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 03/07/2025] [Indexed: 03/24/2025] Open
Abstract
INTRODUCTION Recurrent Clostridioides difficile infection (rCDI) is common, with symptoms ranging from diarrhea to life-threatening sepsis. This study aimed to assess the real-world outcomes of patients with rCDI in the United States (US) who received fecal microbiota, live-jslm (RBL), the first US Food and Drug Administration-approved microbiota-based therapy for the prevention of rCDI after antibiotic treatment. METHODS Adults with rCDI who received RBL between July 2023 and August 2024 at home or in a clinic and had ≥ 8 weeks of follow-up or experienced CDI recurrence at any time after RBL administration were included. Treatment success, defined as no CDI recurrence within 8 weeks of RBL, was assessed overall and in subgroups stratified by age, number of prior CDI recurrences, duration of the antibiotic washout period, prior bezlotoxumab use, and RBL administration setting. RESULTS Among 196 patients who received RBL, 176 had either ≥ 8 weeks of follow-up or had < 8 weeks of follow-up but experienced CDI recurrence during that period. The treatment success rate at 8 weeks was 83.0%. No significant differences were observed in treatment success rates among subgroups based on age (< 65 years old vs. ≥ 65 years old: 85.9% vs. 80.2%, p = 0.20), duration of the antibiotic washout period (24 h: 80.0%, 48 h: 84.5%, 72 h: 85.0%, p = 0.68), number of prior CDI recurrences (< 3 vs. ≥ 3: 82.5% vs. 83.1%, p = 0.60), or prior bezlotoxumab use (86.4% vs. 83.7%, p = 1.00). Patients receiving RBL at home had a higher treatment success rate compared to those receiving RBL in a clinic (87.3% vs. 62.5%, p < 0.01). CONCLUSIONS RBL was highly effective in preventing rCDI in a real-world setting, including at-home administration. The effectiveness was also observed among high-risk subgroups, such as patients ≥ 65 years old and those with ≥ 3 prior CDI recurrences.
Collapse
Affiliation(s)
- Sahil Khanna
- Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| | - Sanghyuk Seo
- Ferring Pharmaceuticals, Inc., 100 Interpace Parkway, Parsippany, NJ, 07054, USA.
| | - Min Yang
- Analysis Group, Inc., 111 Huntington Ave, Boston, MA, 02199, USA
| | | | - Yipeng Gao
- Analysis Group, Inc., 111 Huntington Ave, Boston, MA, 02199, USA
| | - Hannah H Kim
- Analysis Group, Inc., 151 W 42nd St, New York, NY, 10036, USA
| | - Loren Ormenaj
- Analysis Group, Inc., 151 W 42nd St, New York, NY, 10036, USA
| | - Amy Guo
- Ferring Pharmaceuticals, Inc., 100 Interpace Parkway, Parsippany, NJ, 07054, USA
| |
Collapse
|
8
|
Rodriguez J, Cordaillat-Simmons M, Pot B, Druart C. The regulatory framework for microbiome-based therapies: insights into European regulatory developments. NPJ Biofilms Microbiomes 2025; 11:53. [PMID: 40155609 PMCID: PMC11953238 DOI: 10.1038/s41522-025-00683-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 03/15/2025] [Indexed: 04/01/2025] Open
Abstract
The emergence of a broad spectrum of microbiome-based therapies has triggered changes in European regulatory frameworks. The first part of the review describes these innovative therapies. The second part provides an overview of the current framework and key changes introduced by the Regulation on substances of human origin (SoHO) for the development of microbiome-based therapies, highlighting the need of microbiome regulatory science to unlock the full potential of microbiome-based therapies.
Collapse
Affiliation(s)
| | | | - Bruno Pot
- Pharmabiotic Research Institute (PRI), Narbonne, France
- Research Group of Industrial Microbiology and Food Biotechnology (IMDO), Faculty of Sciences and Bioengineering Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Céline Druart
- Pharmabiotic Research Institute (PRI), Narbonne, France
| |
Collapse
|
9
|
McDonnell KJ. Operationalizing Team Science at the Academic Cancer Center Network to Unveil the Structure and Function of the Gut Microbiome. J Clin Med 2025; 14:2040. [PMID: 40142848 PMCID: PMC11943358 DOI: 10.3390/jcm14062040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/28/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
Oncologists increasingly recognize the microbiome as an important facilitator of health as well as a contributor to disease, including, specifically, cancer. Our knowledge of the etiologies, mechanisms, and modulation of microbiome states that ameliorate or promote cancer continues to evolve. The progressive refinement and adoption of "omic" technologies (genomics, transcriptomics, proteomics, and metabolomics) and utilization of advanced computational methods accelerate this evolution. The academic cancer center network, with its immediate access to extensive, multidisciplinary expertise and scientific resources, has the potential to catalyze microbiome research. Here, we review our current understanding of the role of the gut microbiome in cancer prevention, predisposition, and response to therapy. We underscore the promise of operationalizing the academic cancer center network to uncover the structure and function of the gut microbiome; we highlight the unique microbiome-related expert resources available at the City of Hope of Comprehensive Cancer Center as an example of the potential of team science to achieve novel scientific and clinical discovery.
Collapse
Affiliation(s)
- Kevin J McDonnell
- Center for Precision Medicine, Department of Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| |
Collapse
|
10
|
Cao Z, Gao T, Bajinka O, Zhang Y, Yuan X. Fecal microbiota transplantation-current perspective on human health. Front Med (Lausanne) 2025; 12:1523870. [PMID: 40160324 PMCID: PMC11949973 DOI: 10.3389/fmed.2025.1523870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 02/28/2025] [Indexed: 04/02/2025] Open
Abstract
Recently, microbiome medicine has attracted the attention of researchers. While this rapidly growing medical approach for various diseases and disorders is changing the paradigm, it is imperative to weigh both its benefits and the associated risk factors. For instance, manipulation of the gut microbiota (GM) has positive effects on metabolic and neurodegenerative diseases. Notably, fecal microbiota transplantation (FMT), a complex method, has shown promise; however, many doubt its feasibility without adverse effects on human health. Given the number of human clinical trials investigating FMT for the treatment of various disorders, this review summarizes recent findings on its impact on human health. This review summarizes the metabolic responses associated with FMT and their reversal effects on gastrointestinal infections, behavioral changes, and immune responses. Additionally, this review discusses the role of FMT in antimicrobial resistance and its co-supplementation effects on human health, safety, potential risks, limitations, prospects, and recommendations. Although this review does not cover all the studies in the database, the searched terms for FMT and human health in clinical trials are sufficient to provide a summary of the current perspective.
Collapse
Affiliation(s)
- Zixuan Cao
- Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
| | - Tingting Gao
- Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
| | - Ousman Bajinka
- Country School of Medicine and Allied Health Sciences, University of The Gambia, Banjul, Gambia
| | - Yali Zhang
- Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
| | - Xingxing Yuan
- Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
| |
Collapse
|
11
|
Tian S, Kim MS, Zhao J, Heber K, Hao F, Koslicki D, Tian S, Singh V, Patterson AD, Bisanz JE. A designed synthetic microbiota provides insight to community function in Clostridioides difficile resistance. Cell Host Microbe 2025; 33:373-387.e9. [PMID: 40037353 PMCID: PMC11911935 DOI: 10.1016/j.chom.2025.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 01/19/2025] [Accepted: 02/10/2025] [Indexed: 03/06/2025]
Abstract
Clostridioides difficile, a major cause of antibiotic-associated diarrhea, is suppressed by the gut microbiome, but the precise mechanisms are not fully described. Through a meta-analysis of 12 human studies, we designed a synthetic fecal microbiota transplant (sFMT1) by reconstructing microbial networks negatively associated with C. difficile colonization. This lab-built 37-strain consortium formed a functional community suppressing C. difficile in vitro and in animal models. Using sFMT1 as a tractable model system, we find that bile acid 7α-dehydroxylation is not a determinant of sFMT1 efficacy while one strain performing Stickland fermentation-a pathway of competitive nutrient utilization-is both necessary and sufficient for the suppression of C. difficile, replicating the efficacy of a human fecal transplant in a gnotobiotic mouse model. Our data illustrate the significance of nutrient competition in suppression of C. difficile and a generalizable approach to interrogating complex community function through robust methods to leverage publicly available sequencing data.
Collapse
Affiliation(s)
- Shuchang Tian
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA
| | - Min Soo Kim
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA
| | - Jingcheng Zhao
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA
| | - Kerim Heber
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA
| | - Fuhua Hao
- One Health Microbiome Center, Huck Life Sciences Institute, The Pennsylvania State University, University Park, PA 16802, USA; Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802, USA
| | - David Koslicki
- One Health Microbiome Center, Huck Life Sciences Institute, The Pennsylvania State University, University Park, PA 16802, USA; Department of Computer Science and Engineering, The Pennsylvania State University, University Park, PA 16802, USA; Department of Biology, The Pennsylvania State University, University Park, PA 16802, USA
| | - Sangshan Tian
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Vishal Singh
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Andrew D Patterson
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA; One Health Microbiome Center, Huck Life Sciences Institute, The Pennsylvania State University, University Park, PA 16802, USA; Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA 16802, USA
| | - Jordan E Bisanz
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, University Park, PA 16802, USA; One Health Microbiome Center, Huck Life Sciences Institute, The Pennsylvania State University, University Park, PA 16802, USA.
| |
Collapse
|
12
|
Ekwudo MN, Gubert C, Hannan AJ. The microbiota-gut-brain axis in Huntington's disease: pathogenic mechanisms and therapeutic targets. FEBS J 2025; 292:1282-1315. [PMID: 38426291 PMCID: PMC11927060 DOI: 10.1111/febs.17102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 01/08/2024] [Accepted: 02/14/2024] [Indexed: 03/02/2024]
Abstract
Huntington's disease (HD) is a currently incurable neurogenerative disorder and is typically characterized by progressive movement disorder (including chorea), cognitive deficits (culminating in dementia), psychiatric abnormalities (the most common of which is depression), and peripheral symptoms (including gastrointestinal dysfunction). There are currently no approved disease-modifying therapies available for HD, with death usually occurring approximately 10-25 years after onset, but some therapies hold promising potential. HD subjects are often burdened by chronic diarrhea, constipation, esophageal and gastric inflammation, and a susceptibility to diabetes. Our understanding of the microbiota-gut-brain axis in HD is in its infancy and growing evidence from preclinical and clinical studies suggests a role of gut microbial population imbalance (gut dysbiosis) in HD pathophysiology. The gut and the brain can communicate through the enteric nervous system, immune system, vagus nerve, and microbiota-derived-metabolites including short-chain fatty acids, bile acids, and branched-chain amino acids. This review summarizes supporting evidence demonstrating the alterations in bacterial and fungal composition that may be associated with HD. We focus on mechanisms through which gut dysbiosis may compromise brain and gut health, thus triggering neuroinflammatory responses, and further highlight outcomes of attempts to modulate the gut microbiota as promising therapeutic strategies for HD. Ultimately, we discuss the dearth of data and the need for more longitudinal and translational studies in this nascent field. We suggest future directions to improve our understanding of the association between gut microbes and the pathogenesis of HD, and other 'brain and body disorders'.
Collapse
Affiliation(s)
- Millicent N. Ekwudo
- Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkvilleAustralia
| | - Carolina Gubert
- Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkvilleAustralia
| | - Anthony J. Hannan
- Florey Institute of Neuroscience and Mental HealthUniversity of MelbourneParkvilleAustralia
- Department of Anatomy and PhysiologyUniversity of MelbourneParkvilleAustralia
| |
Collapse
|
13
|
Hensen ADO, Vehreschild MJGT, Gerding DN, Krut O, Chen W, Young VB, Tzipori S, Solbach P, Gibani MM, Chiu C, de Keersmaecker SCJ, Dasyam D, Morel S, Devaster JM, Corti N, Kuijper EJ, Roestenberg M, Smits WK. How to develop a controlled human infection model for Clostridioides difficile. Clin Microbiol Infect 2025; 31:373-379. [PMID: 39214188 DOI: 10.1016/j.cmi.2024.08.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Clostridioides difficile (C. difficile) remains the leading cause of healthcare-associated diarrhoea, posing treatment challenges because of antibiotic resistance and high relapse rates. Faecal microbiota transplantation is a novel treatment strategy to prevent relapses of C. difficile infection (CDI), however, the exact components conferring colonization resistance are unknown, hampering its translation to a medicinal product. The development of novel products independent of antibiotics, which increase colonization resistance or induce protective immune mechanisms is urgently needed. OBJECTIVES To establish a framework for a Controlled Human Infection Model (CHIM) of C. difficile, in which healthy volunteers are exposed to toxigenic C. difficile spores, offering the possibility to test novel approaches and identify microbiota and immunological targets. Whereas experimental exposure to non-toxigenic C. difficile has been done before, a toxigenic C. difficile CHIM faces ethical, scientific, logistical, and biosafety challenges. SOURCES Specific challenges in developing a C. difficile CHIM were discussed by a group of international experts during a workshop organized by Inno4Vac, an Innovative Health Initiative-funded consortium. CONTENT The experts agreed that the main challenges are: developing a clinically relevant CHIM that induces mild to moderate CDI symptoms but not severe CDI, determining the optimal C. difficile inoculum dose, and understanding the timing and duration of antibiotic pretreatment in inducing susceptibility to CDI in healthy volunteers. IMPLICATIONS Should these challenges be tackled, a C. difficile CHIM will not only provide a way forward for the testing of novel products but also offer a framework for a better understanding of the pathophysiology, pathogenesis, and immunology of C. difficile colonization and infection.
Collapse
Affiliation(s)
- Annefleur D O Hensen
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Maria J G T Vehreschild
- Department of Internal Medicine, Division of Infectious Diseases, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; German Center for Infection Research (DZIF), Cologne, Germany
| | - Dale N Gerding
- Department of Veterans Affairs, Edward Hines Jr VA Hospital, Hines, IL, United States
| | - Oleg Krut
- Paul-Ehrlich-Institut (PEI), Langen, Germany
| | - Wilbur Chen
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Vincent B Young
- Department of Internal Medicine/Infectious Diseases Division and the Department of Microbiology & Immunology, The University of Michigan, Ann Arbor, MI, United States
| | - Saul Tzipori
- Division of Infectious Disease and Global Health, Tufts University, Medford, MA, United States
| | - Philipp Solbach
- First Department of Medicine, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Malick Mahdi Gibani
- Department of Infectious Disease, Imperial College London (ICL), London, United Kingdom
| | - Christopher Chiu
- Department of Infectious Disease, Imperial College London (ICL), London, United Kingdom
| | | | | | | | | | | | - Ed J Kuijper
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Meta Roestenberg
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), Leiden, The Netherlands.
| | - Wiep Klaas Smits
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), Leiden, The Netherlands
| |
Collapse
|
14
|
Cheng Y, Hu G, Deng L, Zan Y, Chen X. Therapeutic role of gut microbiota in lung injury-related cognitive impairment. Front Nutr 2025; 11:1521214. [PMID: 40017811 PMCID: PMC11867030 DOI: 10.3389/fnut.2024.1521214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 12/16/2024] [Indexed: 03/01/2025] Open
Abstract
Lung injury can lead to specific neurocognitive dysfunction, and the "triple-hit" phenomenon may be the key theoretical mechanism for the progressive impairment of lung injury-related cognitive impairment. The lung and brain can communicate biologically through immune regulation pathway, hypoxic pathway, neural circuit, mitochondrial dysfunction, and microbial influence, which is called the "lung-brain axis." The gut microbiota is a highly complex community of microorganisms that reside in the gut and communicate with the lung via the "gut-lung axis." The dysregulation of gut microbiota may lead to the migration of pathogenic bacteria to the lung, and directly or indirectly regulate the lung immune response through their metabolites, which may cause or aggravate lung injury. The gut microbiota and the brain interact through the "gut-brain axis." The gut microbiota can influence and regulate cognitive function and behavior of the brain through neural pathway mechanisms, immune regulation pathway and hypothalamic-pituitary-adrenal (HPA) axis regulation. Based on the gut microbiota regulation mechanism of the "gut-lung axis" and "gut-brain axis," combined with the mechanisms of cognitive impairment caused by lung injury, we proposed the "triple-hit" hypothesis. It states that the pathophysiological changes of lung injury trigger a series of events such as immune disorder, inflammatory responses, and microbiota changes, which activate the "lung-gut axis," thus forming a "triple-hit" that leads to the development or deterioration of cognitive impairment. This hypothesis provides a more comprehensive framework for studying and understanding brain dysfunction in the context of lung injury. This review proposes the existence of an interactive tandem network for information exchange among the gut, lung, and brain, referred to as the "gut-lung-brain axis." It further explores the potential mechanism of lung injury-related cognitive impairment caused by multiple interactions of gut microbiota in the "gut-lung-brain axis." We found that there are many numerous pathophysiological factors that influence the interaction within the "gut-lung-brain axis." The impact of gut microbiota on cognitive functions related to lung injury may be mediated through mechanisms such as the "triple-hit" hypothesis, direct translocation of microbes and their metabolites, hypoxic pathway, immune modulation, vagal nerve activity, and the HPA axis regulation, among others. As the research deepens, based on the "triple-hit" hypothesis of lung injury, it is further discovered that gut microbial therapy can significantly change the pathogenesis of the inflammatory process on the "gut-lung-brain axis." It can also relieve lung injury and therapeutically modulate brain function and behavior. This perspective provides a new idea for the follow-up treatment of lung injury-related cognitive impairment caused by dysregulation of gut microbiota.
Collapse
Affiliation(s)
| | | | | | | | - Xia Chen
- Department of Pediatrics, Child and Adolescent Psychiatric Center of Jiangbei Campus, The First Affiliated Hospital of Army Medical University (Army 958th Hospital), Chongqing, China
| |
Collapse
|
15
|
Bland CM, Love BL, Jones BM. Human microbiome: Impact of newly approved treatments on C. difficile infection. Am J Health Syst Pharm 2025; 82:174-183. [PMID: 39230353 DOI: 10.1093/ajhp/zxae249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Indexed: 09/05/2024] Open
Abstract
PURPOSE The primary purposes of this review are to provide a brief overview of the microbiome, discuss the most relevant outcome data and key characteristics of each live microbiome agent, and pose questions for consideration going forward as these agents are integrated into clinical practice. SUMMARY The management of Clostridiodes difficile infection (CDI) remains a difficult clinical conundrum, with recurrent CDI occurring in 15% to 35% of patients and causing significant morbidity and decreased quality of life. For patients with frequent CDI recurrences, fecal microbiota transplantation (FMT) has been demonstrated to have significant benefit but also significant risks, and FMT is not approved by the US Food and Drug Administration (FDA) for that indication. FDA has established a new therapeutic class for agents known as live biotherapeutic products (LBPs) that offer significant advantages over FMT, including standardized screening, testing, and manufacturing as well as known quantities of organisms contained within. Two new live microbiome products within this class were recently approved by FDA for prevention of CDI recurrences in adult patients following treatment for recurrent CDI with standard antimicrobial therapy. Both agents had demonstrated efficacy in registry trials in preventing CDI recurrence but differ significantly in a number of characteristics, such as route of administration. Cost as well as logistics are current obstacles to use of these therapies. CONCLUSION Live microbiome therapy is a promising solution for patients with recurrent CDI. Future studies should provide further evidence within yet-to-be-evaluated populations not included in registry studies. This along with real-world evidence will inform future use and clinical guideline placement.
Collapse
Affiliation(s)
| | - Bryan L Love
- University of South Carolina College of Pharmacy, Columbia, SC, USA
| | - Bruce M Jones
- St. Joseph's/Candler Health System, Inc., Savannah, GA, and University of Georgia College of Pharmacy, Savannah, GA
| |
Collapse
|
16
|
Feuerstadt P, Chopra T, Knapple W, Van Hise NW, Dubberke ER, Baggott B, Guthmueller B, Bancke L, Gamborg M, Steiner TS, Van Handel D, Khanna S. PUNCH CD3-OLS: A Phase 3 Prospective Observational Cohort Study to Evaluate the Safety and Efficacy of Fecal Microbiota, Live-jslm (REBYOTA) in Adults With Recurrent Clostridioides difficile Infection. Clin Infect Dis 2025; 80:43-51. [PMID: 39180326 PMCID: PMC11797394 DOI: 10.1093/cid/ciae437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/25/2024] [Accepted: 08/21/2024] [Indexed: 08/26/2024] Open
Abstract
BACKGROUND The aim of this study was to evaluate the safety and efficacy of fecal microbiota, live-jslm (RBL; REBYOTA)-the first single-dose, broad consortia microbiota-based live biotherapeutic approved by the US Food and Drug Administration for preventing recurrent Clostridioides difficile infection (rCDI) in adults following standard-of-care (SOC) antibiotic treatment. METHODS PUNCH CD3-OLS was a prospective, phase 3, open-label study, conducted across the US and Canada. Participants were aged ≥18 years with documented rCDI and confirmed use of SOC antibiotics. Participants with comorbidities including inflammatory bowel disease and mild-to-moderate immunocompromising conditions could be enrolled. A single dose of RBL was rectally administered within 24-72 hours of antibiotic completion. The primary endpoint was the number of participants with RBL- or administration-related treatment-emergent adverse events (TEAEs). Secondary endpoints included treatment success and sustained clinical response, at 8 weeks and 6 months after RBL administration, respectively. RESULTS Overall, 793 participants were enrolled, of whom 697 received RBL. TEAEs through 8 weeks after administration were reported by 47.3% of participants; most events were mild or moderate gastrointestinal disorders. Serious TEAEs were reported by 3.9% of participants. The treatment success rate at 8 weeks was 73.8%; in participants who achieved treatment success, the sustained clinical response rate at 6 months was 91.0%. Safety and efficacy rates were similar across demographic and baseline characteristic subgroups. CONCLUSIONS RBL was safe and efficacious in participants with rCDI and common comorbidities. This is the largest microbiota-based live biotherapeutic study to date, and findings support use of RBL to prevent rCDI in a broad patient population. CLINICAL TRIALS REGISTRATION NCT03931941.
Collapse
Affiliation(s)
- Paul Feuerstadt
- Digestive Diseases, Yale School of Medicine, New Haven, Connecticut, USA
| | - Teena Chopra
- Division of Infectious Diseases, Wayne State University, Detroit, Michigan, USA
| | | | | | - Erik R Dubberke
- Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, Missouri, USA
| | - Brian Baggott
- Department of Gastroenterology, Hepatology and Nutrition, Cleveland Clinic, Cleveland, Ohio, USA
| | - Beth Guthmueller
- Global Clinical and Translational Sciences, Ferring Pharmaceuticals, Roseville, Minnesota, USA
| | - Lindy Bancke
- Global Clinical and Translational Sciences, Ferring Pharmaceuticals, Roseville, Minnesota, USA
- Global Clinical Operations US, Ferring Pharmaceuticals, Roseville, Minnesota, USA
| | - Michael Gamborg
- Global Biometrics, Ferring Pharmaceuticals, Copenhagen, Denmark
| | - Theodore S Steiner
- Division of Infectious Diseases, University of British Columbia, Vancouver, Canada
| | | | - Sahil Khanna
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
17
|
Drekonja DM, Shaukat A, Huang Y, Zhang JH, Reinink AR, Nugent S, Dominitz JA, Davis-Karim A, Gerding DN, Kyriakides TC. A Randomized Controlled Trial of Efficacy and Safety of Fecal Microbiota Transplant for Preventing Recurrent Clostridioides difficile Infection. Clin Infect Dis 2025; 80:52-60. [PMID: 39271107 DOI: 10.1093/cid/ciae467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 06/16/2024] [Accepted: 09/11/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Clostridioides difficile infection (CDI) is the most common cause of healthcare-associated infections in US hospitals, with 15%-30% of patients experiencing recurrence. The aim of our randomized, double-blind clinical trial was to assess the efficacy of capsule-delivered fecal microbiota transplant (FMT) versus placebo in reducing recurrent diarrhea and CDI recurrence. The secondary aim was FMT safety assessment. METHODS Between 2018 and 2022, Veterans across the Veterans Health Administration system with recurrent CDI who responded to antibiotic treatment were randomized in a 1:1 ratio to oral FMT or placebo capsules. Randomization was stratified by number of prior CDI recurrences (1 or ≥2). The primary endpoint was clinical recurrence by day 56, defined as >3 unformed stools daily for ≥2 days with or without laboratory confirmation of C. difficile, or death within 56 days. RESULTS The study was stopped due to futility after meeting prespecified criteria. Of 153 participants (76 FMT, 77 placebo) with an average age of 66.5 years, 25 participants (32.9%) in the FMT arm and 23 (29.9%) in the placebo arm experienced the primary endpoint of diarrhea and possible or definite CDI recurrence or death within 56 days of capsule administration (absolute difference, 3.0% [95% confidence interval, -11.7% to 17.7%]). Stratification by number of recurrences revealed no statistically significant differences. There were no clinically important differences in adverse events. CONCLUSIONS FMT therapy versus placebo did not reduce CDI recurrence or death at 56 days. There were no meaningful differences in adverse events between treatment groups. CLINICAL TRIALS REGISTRATION NCT03005379.
Collapse
Affiliation(s)
- Dimitri M Drekonja
- Division of Infectious Diseases, Department of Medicine, Minneapolis Veterans Affairs Health Care System, Minnesota, USA
| | - Aasma Shaukat
- Division of Gastroenterology, Department of Medicine, New York Harbor Veterans Affairs Healthcare System, New York, USA
| | - Yuan Huang
- Veterans Affair Cooperative Studies Program Coordinating Center-West Haven, West Haven, Connecticut, USA
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut, USA
| | - Jane H Zhang
- Veterans Affair Cooperative Studies Program Coordinating Center-West Haven, West Haven, Connecticut, USA
| | - Andrew R Reinink
- Division of Infectious Diseases, Department of Medicine, Minneapolis Veterans Affairs Health Care System, Minnesota, USA
| | - Sean Nugent
- Division of Infectious Diseases, Department of Medicine, Minneapolis Veterans Affairs Health Care System, Minnesota, USA
| | - Jason A Dominitz
- Division of Gastroenterology, Department of Medicine, Veterans Affairs Puget Sound Health Care System, Seattle, Washington, USA
| | - Anne Davis-Karim
- Veterans Affairs Cooperative Studies Program Clinical Research Pharmacy Coordinating Center, Albuquerque, New Mexico, USA
| | - Dale N Gerding
- Division of Infectious Diseases, Department of Medicine, Edward Hines Jr Veterans Affairs Hospital, Hines, Illinois, USA
| | - Tassos C Kyriakides
- Veterans Affair Cooperative Studies Program Coordinating Center-West Haven, West Haven, Connecticut, USA
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut, USA
| |
Collapse
|
18
|
Stallmach A. [The gastrointestinal microbiome - vision and mission]. Dtsch Med Wochenschr 2025; 150:157-162. [PMID: 39879970 DOI: 10.1055/a-2303-3368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
The gastrointestinal microbiome influences physiological functions and is altered in a variety of diseases. The causality of "dysbiosis" in the pathogenesis is not always proven; association studies are often involved. Patients with IBD, bacteria, fungi, bacteriophages, and archaea show disease-typical patterns associated with metabolome disturbances. Fecal microbiome transfer (FMT) for treating various diseases is the subject of numerous clinical studies. Currently, recurrent Clostridioides difficile infection (rCDI) is the only confirmed indication recommended in medical guidelines. In Germany, the FMT is subject to the Medicines Act and may only be carried out as part of individual healing attempts or clinical studies. For patient safety, repeated donor screening, ideally with the construction of a chair bench, is necessary. This significantly limits the nationwide availability of the FMT in Germany. Microbiota-based therapeutics prepared from the stool of tested donors have recently been approved by the US Food and Drug Administration (FDA) for the prevention of rCDI. More microbiome-based medicines can be expected in the future.
Collapse
|
19
|
Bednárik DS, Földvári-Nagy KC, Simon V, Rancz A, Gede N, Veres DS, Paraskevopoulos P, Schnabel T, Erőss B, Hegyi P, Lenti K, Földvári-Nagy L. Comparative effectiveness of different therapies for Clostridioides difficile infection in adults: a systematic review and network meta-analysis of randomized controlled trials. THE LANCET REGIONAL HEALTH. EUROPE 2025; 49:101151. [PMID: 39989875 PMCID: PMC11846439 DOI: 10.1016/j.lanepe.2024.101151] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 02/25/2025]
Abstract
Background Clostridioides difficile infection (CDI) is a leading cause of healthcare-associated diarrhea, with substantial morbidity and mortality. CDI is a severe and growing problem with numerous treatment options. We evaluated the effectiveness of all therapies in recurrent and non-recurrent infections and their prevention. Methods This network meta-analysis and systematic review of randomized controlled trials (RCTs) compared all CDI therapies and preventions. We included RCTs published until 19 August 2024 and focused on adult population. We performed a systematic search in MEDLINE, EMBASE, and Cochrane Central Register of Controlled Trials. Inclusion criteria were patients: adults (>16) treated against CDI; study type: randomized controlled trial; outcome: cure rate, recurrence or effectiveness of prevention. Any publication not meeting all criteria was considered to be ineligible and excluded. We applied random-effects meta-analysis using frequentist methods. We reported our main results as odds ratios (as a symmetric effect size measure, OR) with 95% confidence interval (95% CI). We used the Cochrane risk-of-bias tool to assess the risk of bias. Our study protocol was preregistered in PROSPERO (CRD42022371210). Findings We assessed 73 RCTs with 28 interventions, involving 27,959 patients (49.2% female) in five networks. Fecal microbiota transplantation (FMT) was the most effective treatment in terms of the cure rate overall (P-score: 0.9952) and in recurrent cases (P-score: 0.9836). In recurrent cases, fidaxomicin (P-score: 0.6734) showed significantly greater effectiveness than vancomycin (P-score: 0.3677) and tolevamer (P-score: 0.0365). For non-recurrent CDI treatments ridinilazole, fidaxomicin, FMT and nitazoxanide were equally effective. Ridinilazole (P-score: 0.7671) and fidaxomicin (P-score: 0.7627) emerged as the most effective in preventing recurrence. Probiotics were not effective in preventing CDI, since network meta-analyses did not show significant differences between probiotics and placebo. In probiotics' subgroups pairwise meta-analyses Lactobacillaceae proved to be significantly more effective in prevention than placebo. Oral and colonoscopic FMT administration methods were equally effective. The study-level aggregated risk of bias of the publications included ranged from low to high. We observed relevant heterogeneity among studies in therapeutic doses, treatment durations, and follow-up times. Interpretation The superiority of FMT in the treatment of CDI highlights the potential for increased use of FMT in clinical settings. Further research on optimizing FMT protocols and exploring its long-term safety and efficacy in larger samples is needed. Our findings suggest that the preventive use of probiotics might be questioned. Funding None.
Collapse
Affiliation(s)
- Dániel Steve Bednárik
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Heim Pál National Pediatric Institute, Budapest, Hungary
| | - Kincső Csepke Földvári-Nagy
- School of Life Sciences, University of Warwick, Coventry, United Kingdom
- Faculty of Health Sciences, Semmelweis University, Budapest, Hungary
| | - Viktor Simon
- Faculty of Health Sciences, Semmelweis University, Budapest, Hungary
| | - Anett Rancz
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Noémi Gede
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Dániel Sándor Veres
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | | | - Tamás Schnabel
- Department of Gastroenterology, Skien Hospital, Telemark Hospital Trust, Skien, Norway
| | - Bálint Erőss
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Institute of Pancreatic Diseases, Semmelweis University, Budapest, Hungary
- Institute for Translational Medicine, University of Pécs, Medical School, Pécs, Hungary
| | - Péter Hegyi
- Centre for Translational Medicine, Semmelweis University, Budapest, Hungary
- Institute of Pancreatic Diseases, Semmelweis University, Budapest, Hungary
- Institute for Translational Medicine, University of Pécs, Medical School, Pécs, Hungary
| | - Katalin Lenti
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, Budapest, Hungary
| | - László Földvári-Nagy
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, Budapest, Hungary
| |
Collapse
|
20
|
Kuijper EJ, Garey KW. The New Horizon of Clostridioides difficile Infection Treatment. Gastroenterology 2025; 168:219-222. [PMID: 39505220 DOI: 10.1053/j.gastro.2024.10.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/08/2024]
Affiliation(s)
- Ed J Kuijper
- National Expertise Center for Clostridioides difficile Infections, from Leiden University Center for Infectious Diseases, Leiden, The Netherlands, and National Institute for Public Health and the Environment, Bilthoven, The Netherlands.
| | - Kevin W Garey
- University of Houston College of Pharmacy, Houston, Texas
| |
Collapse
|
21
|
Pitashny M, Kesten I, Shlon D, Hur DB, Bar-Yoseph H. The Future of Microbiome Therapeutics. Drugs 2025; 85:117-125. [PMID: 39843757 PMCID: PMC11802617 DOI: 10.1007/s40265-024-02107-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2024] [Indexed: 01/24/2025]
Abstract
The human microbiome exerts profound influence over various biological processes within the body. Unlike many host determinants, it represents a readily accessible target for manipulation to promote health benefits. However, existing commercial microbiome-directed products often exhibit low efficacy. Advancements in technology are paving the way for the development of novel microbiome therapeutics, across a wide range of indications. In this narrative review, we provide an overview of state-of-the-art technologies in late-stage development, examining their advantages and limitations. By covering a spectrum, from fecal-derived products to live biotherapeutics, phage therapy, and synthetic biology, we illuminate the path toward the future of microbiome therapeutics.
Collapse
Affiliation(s)
- Milena Pitashny
- Bruce Rappaport Faculty of Medicine, Technion-Israel institute of technology, Haifa, Israel
- Clinical and Research Microbiome Center, Rambam Health Care Campus, Haifa, Israel
| | - Inbar Kesten
- Clinical and Research Microbiome Center, Rambam Health Care Campus, Haifa, Israel
| | - Dima Shlon
- Internal Medicine D, Rambam Health Care Campus, Haifa, Israel
| | - Dana Ben Hur
- Department of Gastroenterology, Rambam Health Care Campus, Haifa, Israel
- Internal Medicine H, Rambam Health Care Campus, Haifa, Israel
| | - Haggai Bar-Yoseph
- Bruce Rappaport Faculty of Medicine, Technion-Israel institute of technology, Haifa, Israel.
- Department of Gastroenterology, Rambam Health Care Campus, Haifa, Israel.
| |
Collapse
|
22
|
Vaughn BP, Khoruts A, Fischer M. Diagnosis and Management of Clostridioides difficile in Inflammatory Bowel Disease. Am J Gastroenterol 2025; 120:313-319. [PMID: 39230037 DOI: 10.14309/ajg.0000000000003076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 08/30/2024] [Indexed: 09/05/2024]
Abstract
Patients with inflammatory bowel disease (IBD) have an increased risk of Clostridioides difficile infection (CDI), which can lead to worse IBD outcomes. The diagnosis of CDI in patients with IBD is complicated by higher C. difficile colonization rates and shared clinical symptoms of intestinal inflammation. Traditional risk factors for CDI, such as antibiotic exposure, may be lacking in patients with IBD because of underlying intestinal microbiota dysbiosis. Although CDI disproportionately affects people with IBD, patients with IBD are typically excluded from CDI clinical trials creating a knowledge gap in the diagnosis and management of these 2 diseases. This narrative review aims to provide a comprehensive overview of the diagnosis, treatment, and prevention of CDI in patients with IBD. Distinguishing CDI from C. difficile colonization in the setting of an IBD exacerbation is important to avoid treatment delays. When CDI is diagnosed, extended courses of anti- C. difficile antibiotics may lead to better CDI outcomes. Regardless of a diagnosis of CDI, the presence of C. difficile in a patient with IBD should prompt a disease assessment of the underlying IBD. Microbiota-based therapies and bezlotoxumab seem to be effective in preventing CDI recurrence in patients with IBD. Patients with IBD should be considered at high risk of CDI recurrence and evaluated for a preventative strategy when diagnosed with CDI. Ultimately, the comanagement of CDI in a patient with IBD requires a nuanced, patient-specific approach to distinguish CDI from C. difficile colonization, prevent CDI recurrence, and manage the underlying IBD.
Collapse
Affiliation(s)
- Byron P Vaughn
- Division of Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis, Minnesota, USA
| | - Alexander Khoruts
- Division of Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis, Minnesota, USA
| | - Monika Fischer
- Division of Gastroenterology and Hepatology, Indiana University, Indianapolis, Indiana, USA
| |
Collapse
|
23
|
Vinterberg JE, Oddsdottir J, Nye M, Pinton P. Management of Recurrent Clostridioides difficile Infection (rCDI): A Systematic Literature Review to Assess the Feasibility of Indirect Treatment Comparison (ITC). Infect Dis Ther 2025; 14:327-355. [PMID: 39821840 PMCID: PMC11829878 DOI: 10.1007/s40121-024-01105-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 12/18/2024] [Indexed: 01/19/2025] Open
Abstract
Recurrent Clostridioides difficile infection (rCDI) is a major cause of increased morbidity, mortality, and healthcare costs. Fecal-microbiota-based therapies are recommended for rCDI on completion of standard-of-care (SoC) antibiotics to prevent further recurrence: these therapies include conventional fecal-microbiota transplantation and the US Food and Drug Administration-approved therapies REBYOTA® (RBL) and VOWST Oral Spores™ (VOS). As an alternative to microbiota-based therapies, bezlotoxumab, a monoclonal antibody, is used as adjuvant to SoC antibiotics to prevent rCDI. There are no head-to-head clinical trials comparing different microbiota-based therapies or bezlotoxumab for rCDI. To address this gap, we conducted a systematic literature review to identify clinical trials on rCDI treatments and assess the feasibility of using them to conduct an indirect treatment comparison (ITC). The feasibility analysis determined that trial heterogeneity, particularly relating to inclusion criteria, may significantly compromise ITC and prevent cross-trial comparisons. Our analysis underlines the need to adopt standardized protocols to ensure comparability across trials.
Collapse
Affiliation(s)
| | | | - Maria Nye
- EMEA RW Methods and Evidence Generation, IQVIA, Athens, Greece
| | - Philippe Pinton
- Global Research and Medical, Ferring Pharmaceuticals, Kastrup, Denmark.
- Global Research and Medical, International PharmaScience Center, Ferring Pharmaceuticals A/S, Amager Strandvej 405, 2770, Kastrup, Denmark.
| |
Collapse
|
24
|
Allegretti JR, Feuerstadt P, Knapple WL, Orenstein R, Pinton P, Sheh A, Khanna S. Safety and Efficacy of Fecal Microbiota, Live-jslm (REBYOTA®), for the Prevention of Recurrent Clostridioides difficile Infection in Participants With Inflammatory Bowel Disease in PUNCH CD3-OLS. Inflamm Bowel Dis 2025:izae291. [PMID: 39862395 DOI: 10.1093/ibd/izae291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Indexed: 01/27/2025]
Abstract
BACKGROUND Fecal microbiota, live-jslm (RBL; REBYOTA®), is the first single-dose, broad consortia, microbiota-based live biotherapeutic approved by the US Food and Drug Administration to prevent recurrent Clostridioides difficile infection (rCDI) in adults following standard-of-care antimicrobials. Inflammatory bowel disease (IBD) is a common risk factor for rCDI, yet patients with IBD are often excluded from prospective trials. This subgroup analysis of PUNCH CD3-OLS (NCT03931941) evaluated the safety and efficacy of RBL in participants with rCDI and IBD. METHODS Participants with IBD (ulcerative colitis [UC], Crohn's disease [CD], or unspecified) who had rCDI were included. Treatment-emergent adverse event (TEAE) data were collected for up to 6 months following RBL administration. Efficacy outcomes included treatment success at 8 weeks and sustained clinical response at 6 months. RESULTS Overall, 793 participants were enrolled, and 697 received RBL; 74 had IBD (UC: n = 45; CD: n = 25; unspecified IBD: n = 4). TEAEs within 8 weeks of administration were reported by 45.9% and 47.5% of participants with and without IBD, respectively; most were mild or moderate gastrointestinal symptoms. Serious TEAEs within 8 weeks of administration were reported by 1.4% and 4.2% of participants with and without IBD, respectively. The treatment success rate at 8 weeks was 78.9%, and the sustained clinical response rate at 6 months was 91.1% in participants with IBD, similar to rates in participants without IBD (73.2% and 91.0%, respectively). CONCLUSIONS The results of this subgroup analysis of PUNCH CD3-OLS suggest RBL is safe and efficacious in patients with IBD.
Collapse
Affiliation(s)
- Jessica R Allegretti
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham & Women's Hospital, Boston, MA, USA
| | - Paul Feuerstadt
- Division of Digestive Disease, Yale University School of Medicine, New Haven, CT, USA
- PACT-Gastroenterology Center, Hamden, CT, USA
| | | | - Robert Orenstein
- Division of Infectious Diseases, Department of Internal Medicine, Mayo Clinic, Phoenix, AZ, USA
| | - Philippe Pinton
- Global Research and Medical, Ferring Pharmaceuticals A/S, Kastrup, Denmark
| | | | - Sahil Khanna
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
25
|
Takano T, Aiba H, Kaku M, Kunishima H. Gastroenterological Surgery and Management of Clostridioides difficile Infection: A Review. J Anus Rectum Colon 2025; 9:25-32. [PMID: 39882219 PMCID: PMC11772788 DOI: 10.23922/jarc.2024-078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 08/08/2024] [Indexed: 01/31/2025] Open
Abstract
Fever and diarrhea are the common symptoms of Clostridioides difficile infection (CDI); however, pseudomembranous enteritis, megacolonization, and paralytic ileus have been observed in severe cases. C. difficile spores are resistant to several types of disinfectants. Thus, they are often the causative pathogens of healthcare-associated infections. Rapid diagnostic tests based on glutamate dehydrogenase and toxins are the mainstay of CDI laboratory diagnosis owing to their simplicity. CDI can be diagnosed with high specificity using the nucleic acid amplification test, a genetic test for C. difficile toxins. The risk factors for CDI include age ≥65 years; history of antimicrobial use; previous hospitalization; history of gastrointestinal surgery, chronic kidney disease, or inflammatory bowel disease; nasal tube feeding; and use of proton pump inhibitors and histamine H2 receptor antagonists. The risk of CDI development persists even 1 year after discontinuation of proton pump inhibitor use. Furthermore, colorectal surgery and radical cystectomy with urinary diversion are associated with high incidences of postoperative CDI. The choice of therapeutic agent depends on the severity of the disease and recurrence. However, a combination of oral or nasogastric vancomycin, intracolonic vancomycin, and intravenous metronidazole can be considered in patients with toxic megacolonization and paralytic ileus. In January 2024, the European Committee on Antimicrobial Susceptibility Testing established a breakpoint for fidaxomicin (minimum inhibitory concentration breakpoint > 2 mg/L) against C. difficile. Rapid progress has been achieved in CDI treatment. Thus, multidisciplinary teams must collaborate to diagnose, treat, and control CDI.
Collapse
Affiliation(s)
- Tomonori Takano
- Department of Infectious Diseases, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Hiroyuki Aiba
- Department of Infectious Diseases, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Mitsuo Kaku
- Department of Infectious Diseases, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Hiroyuki Kunishima
- Department of Infectious Diseases, St. Marianna University School of Medicine, Kawasaki, Japan
| |
Collapse
|
26
|
Siguenza N, Bailey S, Sadegi M, Gootin H, Tiu M, Price JD, Ramer-Tait A, Zarrinpar A. Gut Competition Dynamics of Live Bacterial Therapeutics Are Shaped by Microbiome Complexity, Diet, and Therapeutic Transgenes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.21.634159. [PMID: 39896492 PMCID: PMC11785071 DOI: 10.1101/2025.01.21.634159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Competitive exclusion is conventionally believed to prevent the establishment of a secondary strain of the same bacterial species in the gut microbiome, raising concerns for the deployment of live bacterial therapeutics (LBTs), especially if the bacterial chassis is a strain native to the gut. In this study, we investigated factors influencing competition dynamics in the murine gut using isogenic native Escherichia coli strains. We found that competition outcomes are context-dependent, modulated by microbiome complexity, LBT transgene expression, intestinal inflammation, and host diet. Furthermore, we demonstrated that native LBTs can establish long-term engraftment in the gut alongside a parental strain, with transgene-associated fitness effects influencing competition. We identified various interventions, including strategic dosing and dietary modulation, that significantly enhanced LBT colonization levels by 2 to 3 orders of magnitude. These insights provide a framework for optimizing LBT engraftment and efficacy, supporting their potential translation for human therapeutic applications.
Collapse
Affiliation(s)
- Nicole Siguenza
- Division of Gastroenterology, University of California, San Diego, La Jolla, CA, USA
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA, USA
| | - Sharyl Bailey
- Division of Gastroenterology, University of California, San Diego, La Jolla, CA, USA
| | - Mohammad Sadegi
- Division of Gastroenterology, University of California, San Diego, La Jolla, CA, USA
| | - Hanna Gootin
- Division of Gastroenterology, University of California, San Diego, La Jolla, CA, USA
| | - Maria Tiu
- Division of Gastroenterology, University of California, San Diego, La Jolla, CA, USA
| | - Jeffrey D. Price
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Amanda Ramer-Tait
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, NE, USA
- Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Amir Zarrinpar
- Division of Gastroenterology, University of California, San Diego, La Jolla, CA, USA
- Division of Gastroenterology, Jennifer Moreno Department of Veterans Affairs Medical Center, La Jolla, CA, USA
- The Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Synthetic Biology Institute, University of California San Diego, La Jolla, CA, USA
- Institute of Diabetes and Metabolic Health, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
27
|
Dong Q, Harper S, McSpadden E, Son SS, Allen MM, Lin H, Smith RC, Metcalfe C, Burgo V, Woodson C, Sundararajan A, Rose A, McMillin M, Moran D, Little J, Mullowney MW, Sidebottom AM, Fortier LC, Shen A, Pamer EG. Protection against Clostridioides difficile disease by a naturally avirulent strain. Cell Host Microbe 2025; 33:59-70.e4. [PMID: 39610252 PMCID: PMC11731898 DOI: 10.1016/j.chom.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/24/2024] [Accepted: 11/01/2024] [Indexed: 11/30/2024]
Abstract
Clostridioides difficile is a leading cause of healthcare infections. Gut dysbiosis promotes C. difficile infection (CDI) and CDIs promote gut dysbiosis, leading to frequent CDI recurrence. Although therapies preventing recurrent CDI have been developed, including live biotherapeutic products, existing therapies are costly and do not prevent primary infections. Here, we show that an avirulent C. difficile isolate, ST1-75, protects mice from developing colitis induced by a virulent R20291 strain when coinfected at a 1:1 ratio. In metabolic analyses, avirulent ST1-75 depletes amino acids more rapidly than virulent R20291 and supplementation with amino acids ablates this competitive advantage, indicating that ST1-75 limits the growth of virulent R20291 through amino acid depletion. Overall, our study identifies inter-strain nutrient depletion as a potentially exploitable mechanism to reduce the incidence of CDI and reveals that the ST1-75 strain may be a biotherapeutic agent that can prevent CDI in high-risk patients.
Collapse
Affiliation(s)
- Qiwen Dong
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA; Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA.
| | - Stephen Harper
- Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA
| | - Emma McSpadden
- Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA
| | - Sophie S Son
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA; Interdisciplinary Scientist Training Program, University of Chicago, Chicago, IL 60637, USA
| | - Marie-Maude Allen
- Department of Microbiology and Infectious Diseases, Université de Sherbrooke, Sherbrooke, QC J1E 4K8, Canada
| | - Huaiying Lin
- Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA
| | - Rita C Smith
- Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA
| | - Carolyn Metcalfe
- Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA
| | - Victoria Burgo
- Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA
| | - Che Woodson
- Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA
| | | | - Amber Rose
- Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA
| | - Mary McMillin
- Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA
| | - David Moran
- Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA
| | - Jessica Little
- Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA
| | | | | | - Louis-Charles Fortier
- Department of Microbiology and Infectious Diseases, Université de Sherbrooke, Sherbrooke, QC J1E 4K8, Canada
| | - Aimee Shen
- Department of Molecular Biology and Microbiology, Tufts University, Boston, MA 02111, USA
| | - Eric G Pamer
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA; Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
28
|
Menon R, Bhattarai SK, Crossette E, Prince AL, Olle B, Silber JL, Bucci V, Faith J, Norman JM. Multi-omic profiling a defined bacterial consortium for treatment of recurrent Clostridioides difficile infection. Nat Med 2025; 31:223-234. [PMID: 39747680 DOI: 10.1038/s41591-024-03337-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 10/02/2024] [Indexed: 01/04/2025]
Abstract
Donor-derived fecal microbiota treatments are efficacious in preventing recurrent Clostridioides difficile infection (rCDI), but they have inherently variable quality attributes, are difficult to scale and harbor the risk of pathogen transfer. In contrast, VE303 is a defined consortium of eight purified, clonal bacterial strains developed for prevention of rCDI. In the phase 2 CONSORTIUM study, high-dose VE303 was well tolerated and reduced the odds of rCDI by more than 80% compared to placebo. VE303 organisms robustly colonized the gut in the high-dose group and were among the top taxa associated with non-recurrence. Multi-omic modeling identified antibiotic history, baseline stool metabolites and serum cytokines as predictors of both on-study CDI recurrence and VE303 colonization. VE303 potentiated early recovery of the host microbiome and metabolites with increases in short-chain fatty acids, secondary bile acids and bile salt hydrolase genes after antibiotic treatment for CDI, which is considered important to prevent CDI recurrences. These results support the idea that VE303 promotes efficacy in rCDI through multiple mechanisms.
Collapse
Affiliation(s)
| | - Shakti K Bhattarai
- Program in Microbiome Dynamics, Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | | | | | - Bernat Olle
- Vedanta Biosciences, Inc., Cambridge, MA, USA
| | | | - Vanni Bucci
- Program in Microbiome Dynamics, Department of Microbiology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jeremiah Faith
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | |
Collapse
|
29
|
Grilc NK, Kristl J, Zupančič Š. Can polymeric nanofibers effectively preserve and deliver live therapeutic bacteria? Colloids Surf B Biointerfaces 2025; 245:114329. [PMID: 39486375 DOI: 10.1016/j.colsurfb.2024.114329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/18/2024] [Accepted: 10/19/2024] [Indexed: 11/04/2024]
Abstract
Probiotics and live therapeutic bacteria (LTB), their strictly regulated therapeutic counterpart, are increasingly important in treating and preventing biofilm-related diseases. This necessitates new approaches to (i) preserve bacterial viability during manufacturing and storage and (ii) incorporate LTB into delivery systems for enhanced therapeutic efficacy. This review explores advances in probiotic and LTB product development, focusing on preservation, protection, and improved delivery. Preservation of bacteria can be achieved by drying methods that decelerate metabolism. These methods introduce stresses affecting viability which can be mitigated with suitable excipients like polymeric or low molecular weight stabilizers. The review emphasizes the incorporation of LTB into polymer-based nanofibers via electrospinning, enabling simultaneous drying, encapsulation, and delivery system production. Optimization of bacterial survival during electrospinning and storage is discussed, as well as controlled LTB release achievable through formulation design using gel-forming, gastroprotective, mucoadhesive, and pH-responsive polymers. Evaluation of the presence of the actual therapeutic strains, bacterial viability and activity by CFU enumeration or alternative analytical techniques is presented as a key aspect of developing effective and safe formulations with LTB. This review offers insights into designing delivery systems, especially polymeric nanofibers, for preservation and delivery of LTB, guiding readers in developing innovative biotherapeutic delivery systems.
Collapse
Affiliation(s)
- Nina Katarina Grilc
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana 1000, Slovenia
| | - Julijana Kristl
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana 1000, Slovenia
| | - Špela Zupančič
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, Ljubljana 1000, Slovenia.
| |
Collapse
|
30
|
Jin K, Huang Y, Che H, Wu Y. Engineered Bacteria for Disease Diagnosis and Treatment Using Synthetic Biology. Microb Biotechnol 2025; 18:e70080. [PMID: 39801378 PMCID: PMC11725985 DOI: 10.1111/1751-7915.70080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 12/18/2024] [Accepted: 12/26/2024] [Indexed: 01/16/2025] Open
Abstract
Using synthetic biology techniques, bacteria have been engineered to serve as microrobots for diagnosing diseases and delivering treatments. These engineered bacteria can be used individually or in combination as microbial consortia. The components within these consortia complement each other, enhancing diagnostic accuracy and providing synergistic effects that improve treatment efficacy. The application of microbial therapies in cancer, intestinal diseases, and metabolic disorders underscores their significant potential. The impact of these therapies on the host's native microbiota is crucial, as engineered microbes can modulate and interact with the host's microbial environment, influencing treatment outcomes and overall health. Despite numerous advancements, challenges remain. These include ensuring the long-term survival and safety of bacteria, developing new chassis microbes and gene editing techniques for non-model strains, minimising potential toxicity, and understanding bacterial interactions with the host microbiota. This mini-review examines the current state of engineered bacteria and microbial consortia in disease diagnosis and treatment, highlighting advancements, challenges, and future directions in this promising field.
Collapse
Affiliation(s)
- Kai Jin
- Department of Environmental and Chemical EngineeringShanghai UniversityShanghaiChina
| | - Yi Huang
- Department of Environmental and Chemical EngineeringShanghai UniversityShanghaiChina
| | - Hailong Che
- Department of Environmental and Chemical EngineeringShanghai UniversityShanghaiChina
| | - Yihan Wu
- Department of Environmental and Chemical EngineeringShanghai UniversityShanghaiChina
| |
Collapse
|
31
|
Morado F, Nanda N. A Review of Therapies for Clostridioides difficile Infection. Antibiotics (Basel) 2024; 14:17. [PMID: 39858303 PMCID: PMC11762378 DOI: 10.3390/antibiotics14010017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
Clostridioides difficile is an urgent public health threat that affects approximately half a million patients annually in the United States. Despite concerted efforts aimed at the prevention of Clostridioides difficile infection (CDI), it remains a leading cause of healthcare-associated infections. CDI is associated with significant clinical, social, and economic burdens. Therefore, it is imperative to provide optimal and timely therapy for CDI. We conducted a systematic literature review and offer treatment recommendations based on available evidence for the treatment and prevention of CDI.
Collapse
Affiliation(s)
- Faiza Morado
- Department of Pharmacy, Keck Medical Center, University of Southern California, Los Angeles, CA 90033, USA;
| | - Neha Nanda
- Division of Infectious Diseases, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
32
|
Zhang L, Bai J, Guo Q, Li L, Jia Y, Qiu X, Zhou D, Zhang Z, Niu H. Gut Microbial Composition and Antibiotic Resistance Profiles in Dairy Calves with Diarrhea. Life (Basel) 2024; 15:10. [PMID: 39859950 PMCID: PMC11766533 DOI: 10.3390/life15010010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 12/19/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025] Open
Abstract
Calf diarrhea is a prevalent and significant health issue in dairy farming, severely impacting feed intake, weight gain, and survival rates in young calves. This study aimed to investigate the microbial composition and antibiotic resistance profiles of diarrheic calves to provide insights into the epidemiology and management of the condition. The prevalence of diarrhea in 1685 calves was analyzed. Rectal fecal samples were collected from five healthy and five diarrheic Holstein calves on a large dairy farm in Shaanxi Province, China. High-throughput 16S-rRNA sequencing and PCR were utilized for microbial and resistance gene analysis. In 2023, the overall diarrhea rate among 1685 calves was 9.08%, with a significantly higher diarrhea rate during the suckling period (8.13%) compared to the post-weaning period (0.95%) (p < 0.001). No differences in species diversity and richness were detected among the different groups. However, LEfSe analysis identified six genera (Eubacterium, Eubacteriaceae, Prevotella, Comamonadaceae, Comamonas, and Firmicutes) significantly enriched in diarrheic calves compared to healthy ones (LDA scores > 2, p < 0.05). Additionally, antibiotic resistance genes for quinolones, β-lactams, chloramphenicol, tetracyclines, and aminoglycosides were detected, with significantly higher prevalence in diarrheic calves. These findings demonstrate distinct microbial and antibiotic resistance profiles between healthy and diarrheic calves, emphasizing the importance of microbial management in controlling calf diarrhea.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Animal Engineering, Yangling Vocational & Technical College, Yangling 712100, China; (L.Z.); (J.B.); (L.L.); (Y.J.); (X.Q.)
- Shaanxi Engineering Research Center of the Prevention and Control for Animal Disease, Yangling Vocational & Technical College, Yangling 712100, China
- Key Laboratory for Efficient Ruminant Breeding Technology of Higher Education Institutions in Shanxi Province, Yangling Vocational and Technical College, Yangling 712100, China
- The Youth Innovation Team of Shaanxi Universities, Yangling Vocational and Technical College, Yangling 712100, China
| | - Jun Bai
- Department of Animal Engineering, Yangling Vocational & Technical College, Yangling 712100, China; (L.Z.); (J.B.); (L.L.); (Y.J.); (X.Q.)
- Shaanxi Engineering Research Center of the Prevention and Control for Animal Disease, Yangling Vocational & Technical College, Yangling 712100, China
- Key Laboratory for Efficient Ruminant Breeding Technology of Higher Education Institutions in Shanxi Province, Yangling Vocational and Technical College, Yangling 712100, China
| | - Qian Guo
- ShaanXi Province Management Station of Animal Health and Slaughter, Xi’an 710000, China;
| | - Long Li
- Department of Animal Engineering, Yangling Vocational & Technical College, Yangling 712100, China; (L.Z.); (J.B.); (L.L.); (Y.J.); (X.Q.)
- Key Laboratory for Efficient Ruminant Breeding Technology of Higher Education Institutions in Shanxi Province, Yangling Vocational and Technical College, Yangling 712100, China
- The Youth Innovation Team of Shaanxi Universities, Yangling Vocational and Technical College, Yangling 712100, China
| | - Yanqing Jia
- Department of Animal Engineering, Yangling Vocational & Technical College, Yangling 712100, China; (L.Z.); (J.B.); (L.L.); (Y.J.); (X.Q.)
- Shaanxi Engineering Research Center of the Prevention and Control for Animal Disease, Yangling Vocational & Technical College, Yangling 712100, China
- Key Laboratory for Efficient Ruminant Breeding Technology of Higher Education Institutions in Shanxi Province, Yangling Vocational and Technical College, Yangling 712100, China
- The Youth Innovation Team of Shaanxi Universities, Yangling Vocational and Technical College, Yangling 712100, China
| | - Xinxin Qiu
- Department of Animal Engineering, Yangling Vocational & Technical College, Yangling 712100, China; (L.Z.); (J.B.); (L.L.); (Y.J.); (X.Q.)
- Shaanxi Engineering Research Center of the Prevention and Control for Animal Disease, Yangling Vocational & Technical College, Yangling 712100, China
- Key Laboratory for Efficient Ruminant Breeding Technology of Higher Education Institutions in Shanxi Province, Yangling Vocational and Technical College, Yangling 712100, China
| | - Dong Zhou
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China;
| | - Zhencang Zhang
- Department of Animal Engineering, Yangling Vocational & Technical College, Yangling 712100, China; (L.Z.); (J.B.); (L.L.); (Y.J.); (X.Q.)
- Shaanxi Engineering Research Center of the Prevention and Control for Animal Disease, Yangling Vocational & Technical College, Yangling 712100, China
- Key Laboratory for Efficient Ruminant Breeding Technology of Higher Education Institutions in Shanxi Province, Yangling Vocational and Technical College, Yangling 712100, China
| | - Huafeng Niu
- Department of Animal Engineering, Yangling Vocational & Technical College, Yangling 712100, China; (L.Z.); (J.B.); (L.L.); (Y.J.); (X.Q.)
- Key Laboratory for Efficient Ruminant Breeding Technology of Higher Education Institutions in Shanxi Province, Yangling Vocational and Technical College, Yangling 712100, China
| |
Collapse
|
33
|
Pettit NN, Shaeer KM, Chahine EB. Live Biotherapeutic Products for the Prevention of Recurrent Clostridioides difficile Infection. Ann Pharmacother 2024; 58:1204-1217. [PMID: 38546138 DOI: 10.1177/10600280241239685] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2024] Open
Abstract
OBJECTIVE To review the efficacy, safety, and role of live biotherapeutic products (LBPs) in the prevention of recurrent Clostridioides difficile infection (rCDI). DATA SOURCES A literature search was performed using PubMed and Google Scholar (through February 2024) with search terms RBX2660, SER-109, and fecal microbiota. Other resources included abstracts presented at recent conferences, national clinical practice guidelines, and manufacturers' websites. STUDY SELECTION AND DATA EXTRACTION All relevant studies, trial updates, conference abstracts, and guidelines in the English language were included. DATA SYNTHESIS Two LBPs were recently approved by the Food and Drug Administration for the prevention of recurrence in adults following antibiotic treatment for rCDI. Fecal microbiota, live-jslm is administered rectally as a retention enema, whereas fecal microbiota spores, live-brpk is given orally after bowel preparation. Several phase 2 and phase 3 clinical trials have established the safety and efficacy of these LBPs in reducing rates of rCDI compared with placebo. Patients with severe immunosuppression and those with inflammatory bowel disease were largely excluded from these trials. RELEVANCE TO PATIENT CARE AND CLINICAL PRACTICE IN COMPARISON WITH EXISTING DRUGS Live biotherapeutic products offer a similar mechanism to conventional fecal microbiota transplant (FMT) in preventing rCDI through microbiota restoration. The primary advantages of LBPs over FMT are their standardized composition and donor stool screening processes for transmissible pathogens. Bezlotoxumab is also available for the prevention of Clostridioides difficile infection; however, there are no clinical data available to compare the efficacy of LBPs with bezlotoxumab, and the benefit of simultaneous use of these preventative therapies is unclear. CONCLUSIONS Live biotherapeutic products provide a safe and effective option for the prevention of rCDI and represent an improvement over conventional FMT. Additional studies are needed to further determine their place in therapy relative to bezlotoxumab and in the setting of immunosuppression and inflammatory bowel disease.
Collapse
Affiliation(s)
| | - Kristy M Shaeer
- Department of Pharmacotherapeutics & Clinical Research, University of South Florida Taneja College of Pharmacy, Tampa, FL, USA
| | - Elias B Chahine
- Department of Pharmacy Practice, Palm Beach Atlantic University Lloyd L. Gregory School of Pharmacy, West Palm Beach, FL, USA
| |
Collapse
|
34
|
Frutos-Grilo E, Ana Y, Gonzalez-de Miguel J, Cardona-I-Collado M, Rodriguez-Arce I, Serrano L. Bacterial live therapeutics for human diseases. Mol Syst Biol 2024; 20:1261-1281. [PMID: 39443745 PMCID: PMC11612307 DOI: 10.1038/s44320-024-00067-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/19/2024] [Accepted: 09/12/2024] [Indexed: 10/25/2024] Open
Abstract
The genomic revolution has fueled rapid progress in synthetic and systems biology, opening up new possibilities for using live biotherapeutic products (LBP) to treat, attenuate or prevent human diseases. Among LBP, bacteria-based therapies are particularly promising due to their ability to colonize diverse human tissues, modulate the immune system and secrete or deliver complex biological products. These bacterial LBP include engineered pathogenic species designed to target specific diseases, and microbiota species that promote microbial balance and immune system homeostasis, either through local administration or the gut-body axes. This review focuses on recent advancements in preclinical and clinical trials of bacteria-based LBP, highlighting both on-site and long-reaching strategies.
Collapse
Affiliation(s)
- Elisabet Frutos-Grilo
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Yamile Ana
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Javier Gonzalez-de Miguel
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Marcel Cardona-I-Collado
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Irene Rodriguez-Arce
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain.
| | - Luis Serrano
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain.
- ICREA, Pg. Lluis Companys 23, Barcelona, Spain.
| |
Collapse
|
35
|
Komeya M, Lundy SD. Microbiome-based Therapeutics: Cutting-edge Innovation. Eur Urol Focus 2024; 10:875-876. [PMID: 39609243 DOI: 10.1016/j.euf.2024.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 10/30/2024] [Indexed: 11/30/2024]
Abstract
From bacillus Calmette-Guérin to cutting-edge research, the microbiome has played an integral role in urology treatments and will continue to do so for many generations to come.
Collapse
Affiliation(s)
- Mitsuru Komeya
- Glickman Urological Institute, Cleveland Clinic, Cleveland OH 44195, USA.
| | - Scott D Lundy
- Glickman Urological Institute, Cleveland Clinic, Cleveland OH 44195, USA
| |
Collapse
|
36
|
Herbin SR, Crum H, Gens K. Breaking the Cycle of Recurrent Clostridioides difficile Infections: A Narrative Review Exploring Current and Novel Therapeutic Strategies. J Pharm Pract 2024; 37:1361-1373. [PMID: 38739837 DOI: 10.1177/08971900241248883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Clostridioides difficile is a toxin-producing bacteria that is a main cause of antibiotic-associated diarrhea. Clostridioides difficile infections (CDI) are associated with disruptions within the gastrointestinal (GI) microbiota which can be further exacerbated by CDI-targeted antibiotic treatment thereby causing recurrent CDI (rCDI) and compounding the burden placed on patients and the healthcare system. Treatment of rCDI consists of antibiotics which can be paired with preventative therapeutics, such as bezlotoxumab or fecal microbiota transplants (FMTs), if sustained clinical response is not obtained. Newer preventative strategies have been recently approved to assist in restoring balance within the GI system with the goal of preventing recurrent infections.
Collapse
Affiliation(s)
- Shelbye R Herbin
- Antimicrobial Stewardship and Medication Safety, John D. Dingell VA Medical Center, Detroit, MI, USA
| | - Hannah Crum
- Mercy Hospital Southeast, Cape Girardeau, MO, USA
| | - Krista Gens
- Allina Health, Minneapolis, MN, USA
- Abbott Northwestern Hospital, Minneapolis, MN, USA
| |
Collapse
|
37
|
Khanna S. Microbiota restoration for recurrent Clostridioides difficile infection. Panminerva Med 2024; 66:417-426. [PMID: 39382853 DOI: 10.23736/s0031-0808.24.05111-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
Since the publication of the recent North American and European guidelines on management of Clostridioides difficile infection (CDI), new evidence describing the epidemiology, testing and treatment of CDI has emerged. Despite all advances in infection control and antibiotic stewardship, the incidence and burden of CDI in the hospitals and the community remains at a stable high. Coupled with the incidence of primary CDI, there is a stable high incidence of recurrent CDI. Testing for primary and recurrent CDI remains a clinical challenge owing to high sensitivity of the PCR (leading to false positives) and somewhat limited sensitivity of EIA for toxin. The pathophysiology of recurrent CDI involves an ongoing disruption of the microbiota owing to the infection and the treatment of CDI employed. Broad spectrum antibiotics such as vancomycin leads to further disruption of microbiota compared to fidaxomicin which has a lower disruption of the microbiota and leads to fewer recurrences. Owing to these data fidaxomicin is considered as the first line antibiotic for recurrent CDI. Intravenous bezlotoxumab is a monoclonal antibody that reduces the risk of recurrence in high-risk patients but does not restore the microbiota. Experimental fecal microbiota transplantation (FMT) has been available for more than a decade. Owing to the success of FMT, two new non-invasive donor dependent Food and Drug Administration (FDA) approved therapies have been available since late 2022. This review summarizes all these conundrums regarding CDI and provides clinical pearls to use in day-to-day practice.
Collapse
Affiliation(s)
- Sahil Khanna
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA -
| |
Collapse
|
38
|
Lee JY, Kim Y, Kim J, Kim JK. Fecal Microbiota Transplantation: Indications, Methods, and Challenges. J Microbiol 2024; 62:1057-1074. [PMID: 39557804 DOI: 10.1007/s12275-024-00184-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/25/2024] [Accepted: 10/16/2024] [Indexed: 11/20/2024]
Abstract
Over the past two decades, as the importance of gut microbiota to human health has become widely known, attempts have been made to treat diseases by correcting dysbiosis of gut microbiota through fecal microbiota transplantation (FMT). Apart from current knowledge of gut microbiota, FMT to treat disease has a long history, from the treatment of food poisoning in the fourth century to the treatment of Clostridioides difficile infections in the twentieth century. In 2013, FMT was recognized as a standard treatment for recurrent C. difficile because it consistently showed high efficacy. Though recurrent C. difficile is the only disease internationally recognized for FMT efficacy, FMT has been tested for other diseases and shown some promising preliminary results. Different FMT methods have been developed using various formulations and administration routes. Despite advances in FMT, some issues remain to be resolved, such as donor screening, manufacturing protocols, and unknown components in the fecal microbiota. In this review, we discuss the mechanisms, clinical indications, methods, and challenges of current FMT. We also discuss the development of alternative therapies to overcome the challenges of FMT.
Collapse
Affiliation(s)
- Jee Young Lee
- Department of Microbiology, Kosin University College of Medicine, Busan, 49267, Republic of Korea
| | - Yehwon Kim
- Department of Medicine, Kosin University College of Medicine, Busan, 49267, Republic of Korea
| | - Jiyoun Kim
- Department of Medicine, Kosin University College of Medicine, Busan, 49267, Republic of Korea
| | - Jiyeun Kate Kim
- Department of Microbiology, Kosin University College of Medicine, Busan, 49267, Republic of Korea.
| |
Collapse
|
39
|
Tafader A, Bajaj JS. Present and future of fecal microbiome transplantation in cirrhosis. Liver Transpl 2024:01445473-990000000-00519. [PMID: 39591377 DOI: 10.1097/lvt.0000000000000542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024]
Abstract
Over the last few decades, there have been tremendous advances in our understanding of the role of the gut microbiome in cirrhosis and the clinical sequelae that follow. Progressive dysbiosis and immune dysregulation occur in patients with cirrhosis. In fact, alterations in the gut microbiome occur long before a diagnosis of cirrhosis is made. Understandably, our attention has recently been diverted toward potential modulators of the gut microbiome and the gut-liver axis as targets for treatment. The goal of this review is to highlight the utility of manipulating the gut microbiome with a focus on fecal microbiome transplantation (FMT) in patients with cirrhosis. In addition, we will provide an overview of disease-specific microbial alterations and the resultant impact this has on cirrhosis-related complications.
Collapse
Affiliation(s)
- Asiya Tafader
- Department of Medicine, Virginia Commonwealth University and Richmond VA Medical Center, Richmond, Virginia, USA
| | | |
Collapse
|
40
|
Boustany A, Feuerstadt P, Tillotson G. The 3 Ds: Depression, Dysbiosis, and Clostridiodes difficile. Adv Ther 2024; 41:3982-3995. [PMID: 39276186 PMCID: PMC11480130 DOI: 10.1007/s12325-024-02972-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 08/12/2024] [Indexed: 09/16/2024]
Abstract
This paper explores the intricate relationship between depression, gut dysbiosis, and Clostridioides difficile infections, collectively termed "The 3 Ds". Depression is a widespread mental disorder increasing in prevalence. It is recognized for its societal burden and complex pathophysiology, encompassing genetic, environmental, and microbiome-related factors. The consequent increased use of antidepressants has led to growing concerns about their effects on the gut microbiome. Various classes of antidepressants and antipsychotics show antimicrobial activity, potentially leading to shifts in the gut microbiome and contributing to the development of dysbiosis. Dysbiosis, in turn, can predispose individuals to opportunistic infections like C. difficile, a significant healthcare concern due to its high recurrence rates and severe impact on patients' quality of life. Further, the link between antidepressant use and an increased risk of C. difficile infection (CDI) is explored and, finally, the emergence of live biotherapeutic products as novel treatment options for recurrent CDI is discussed.
Collapse
Affiliation(s)
- Antoine Boustany
- Department of Internal Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Paul Feuerstadt
- Yale University School of Medicine, New Haven, CT, USA
- PACT-Gastroenterology Center, Hamden, CT, USA
| | | |
Collapse
|
41
|
Lee A, Yoo JS, Yoon EJ. Gut Microbiota and New Microbiome-Targeted Drugs for Clostridioides difficile Infections. Antibiotics (Basel) 2024; 13:995. [PMID: 39452261 PMCID: PMC11505460 DOI: 10.3390/antibiotics13100995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 10/26/2024] Open
Abstract
Clostridioides difficile is a major causative pathogen for antibiotic-associated diarrhea and C. difficile infections (CDIs) may lead to life-threatening diseases in clinical settings. Most of the risk factors for the incidence of CDIs, i.e., antibiotic use, treatment by proton pump inhibitors, old age, and hospitalization, are associated with dysbiosis of gut microbiota and associated metabolites and, consequently, treatment options for CDIs include normalizing the composition of the intestinal microbiome. In this review, with an introduction to the CDI and its global epidemiology, CDI-associated traits of the gut microbiome and its metabolites were reviewed, and microbiome-targeting treatment options were introduced, which was approved recently as a new drug by the United States Food and Drug Administration (U.S. FDA), rather than a medical practice.
Collapse
Affiliation(s)
| | | | - Eun-Jeong Yoon
- Division of Antimicrobial Resistance Research, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju-si 28159, Republic of Korea
| |
Collapse
|
42
|
Duo H, Yang Y, Zhang G, Chen Y, Cao Y, Luo L, Pan H, Ye Q. Comparative effectiveness of treatments for recurrent Clostridioides difficile infection: a network meta-analysis of randomized controlled trials. Front Pharmacol 2024; 15:1430724. [PMID: 39484168 PMCID: PMC11525118 DOI: 10.3389/fphar.2024.1430724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 10/07/2024] [Indexed: 11/03/2024] Open
Abstract
Background Clostridioides difficile infection (CDI) is the most common cause of healthcare-associated infectious diarrhea. A major clinical challenge is recurrent CDI (rCDI) without effective standard drug-based therapy. Additionally, a comprehensive comparison of various therapy effectiveness in rCDI patients is still under investigation. Methods A Bayesian network meta-analysis (NMA) of randomized control trials up to March 2024 was performed to investigate the efficacy of rCDI interventions. Results Seventeen trials were included, comprising 4,148 CDI patients with ten interventions, including fecal microbiota transplantation (FMT) by lower gastrointestinal (LGI), FMT by upper gastrointestinal (UGI), Autologous FMT (AFMT), vancomycin + FMT, vancomycin, placebo, fidaxomicin, Vowst (SER109), Rebyota (RBX2660), and monoclonal antibody. NMA showed that FMT by LGI had the highest efficacy in treating rCDIs with an odds ratio (95% confidence interval) of 32.33 (4.03, 248.69) compared with placebo. FMT by UGI also showed high efficacy, whereas the efficacy comparison between FMT by LGI and UGI was not statistically significant (ORs) (95% CI), 1.72 (0.65, 5.21). The rankogram and surface under the cumulative ranking curve (SUCRA) also showed FMT by LGI ranked at the top and FMT by UGI ranked second in the curative effect. Conclusion NMA demonstrates FMT's significant efficacy in rCDI management, regardless of administration route (lower or upper gastrointestinal). Despite its significant benefits, FMT's safety is a concern due to the lack of standardized FDAcompliant manufacturing and oversight. Microbiota-based therapies also exhibit potential. However, limited research mandates further clinical exploration. Antibiotics, in contrast, display comparatively reduced efficacy in rCDI, potentially linked to disruptions in native gut microflora balance. Systematic Review https://www.crd.york.ac.uk/PROSPERO/display_record.php?RecordID=368435, Identifier CRD42022368435.
Collapse
Affiliation(s)
- Hong Duo
- Hubei Key Laboratory of Medical Technology on Transplantation, National Quality Control Center for Donated Organ Procurement, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-Based Medical Materials, Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan, Hubei, China
| | - Yanwei Yang
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| | - Guqing Zhang
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yingxin Chen
- Global Health Institute, School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Yumeng Cao
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Linjie Luo
- Department of Experimental Radiation Oncology and Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, United States
| | - Huaqin Pan
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplantation Intensive Care Unit, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, China
| | - Qifa Ye
- Hubei Key Laboratory of Medical Technology on Transplantation, National Quality Control Center for Donated Organ Procurement, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-Based Medical Materials, Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
43
|
Allegretti JR, Khanna S, Mullish BH, Feuerstadt P. The Progression of Microbiome Therapeutics for the Management of Gastrointestinal Diseases and Beyond. Gastroenterology 2024; 167:885-902. [PMID: 38754739 DOI: 10.1053/j.gastro.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 05/02/2024] [Accepted: 05/09/2024] [Indexed: 05/18/2024]
Abstract
There has been an increased ability to investigate the human microbiota through next-generation sequencing and functional assessment. This advancement has rapidly expanded our ability to study and manipulate the gastrointestinal microbiome to mitigate disease. Fecal microbiota transplantation, a therapy that broadly transfers the entire intestinal ecosystem, has been explored as a potential therapeutic in a variety of gastrointestinal, hepatic, and extraintestinal conditions. The field, however, continues to evolve, with a movement toward precision microbiome therapeutics, individualizing care for various disorders. This review will describe the use of fecal microbiota transplantation, microbiota restoration, and precision microbiome therapeutics, focusing on gastrointestinal and hepatic diseases.
Collapse
Affiliation(s)
- Jessica R Allegretti
- Division of Gastroenterology, Hepatology, and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts.
| | - Sahil Khanna
- Division of Gastroenterology, Mayo Clinic, Rochester, Minnesota
| | - Benjamin H Mullish
- Division of Digestive Diseases, Imperial College London, London, United Kingdom; Departments of Gastroenterology and Hepatology, St Mary's Hospital, Imperial College Healthcare National Health Service Trust, London, United Kingdom
| | - Paul Feuerstadt
- Division of Gastroenterology, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
44
|
Hamilton AM, Krout IN, White AC, Sampson TR. Microbiome-based therapeutics for Parkinson's disease. Neurotherapeutics 2024; 21:e00462. [PMID: 39393983 PMCID: PMC11585879 DOI: 10.1016/j.neurot.2024.e00462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/18/2024] [Accepted: 09/26/2024] [Indexed: 10/13/2024] Open
Abstract
Recent experimental and clinical data demonstrate a significant dysregulation of the gut microbiome in individuals with Parkinson's disease (PD). With an immense influence on all aspects of physiology, this dysregulation has potential to directly or indirectly contribute to disease pathology. Experimental models have bridged these associations toward defined contributions, identifying various microbiome-dependent impacts to PD pathology. These studies have laid the foundation for human translation, examining whether certain members of the microbiome and/or whole restoration of the gut microbiome community can provide therapeutic benefit for people living with PD. Here, we review recent and ongoing clinically-focused studies that use microbiome-targeted therapies to limit the severity and progression of PD. Fecal microbiome transplants, prebiotic interventions, and probiotic supplementation are each emerging as viable methodologies to augment the gut microbiome and potentially limit PD symptoms. While still early, the data in the field to date support continued cross-talk between experimental systems and human studies to identify key microbial factors that contribute to PD pathologies.
Collapse
Affiliation(s)
- Adam M Hamilton
- Department of Cell Biology, Emory University School of Medicine, Atlanta GA 30322, USA
| | - Ian N Krout
- Department of Cell Biology, Emory University School of Medicine, Atlanta GA 30322, USA
| | - Alexandria C White
- Department of Cell Biology, Emory University School of Medicine, Atlanta GA 30322, USA
| | - Timothy R Sampson
- Department of Cell Biology, Emory University School of Medicine, Atlanta GA 30322, USA.
| |
Collapse
|
45
|
Kraft CS, Sims M, Silverman M, Louie TJ, Feuerstadt P, Huang ES, Khanna S, Berenson CS, Wang EEL, Cohen SH, Korman L, Lee C, Kelly CR, Odio A, Cook PP, Lashner B, Ramesh M, Kumar P, De A, Memisoglu A, Lombardi DA, Hasson BR, McGovern BH, von Moltke L, Pardi DS. Integrated Safety and Efficacy Analyses of Phase 3 Trials of a Microbiome Therapeutic for Recurrent CDI. Infect Dis Ther 2024; 13:2105-2121. [PMID: 38941068 PMCID: PMC11416444 DOI: 10.1007/s40121-024-01007-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 04/29/2024] [Indexed: 06/29/2024] Open
Abstract
INTRODUCTION Recurrent Clostridioides difficile infection (rCDI) often occurs after standard-of-care antibiotics. VOWST oral spores (VOS, previously SER-109), an FDA-approved orally administered microbiome therapeutic, is indicated to prevent rCDI following antibiotics for rCDI. OBJECTIVE, DESIGN, AND PATIENTS To evaluate safety and efficacy of VOS from two phase 3 trials, (randomized, placebo-controlled [ECOSPOR III: NCT03183128] and open-label, single arm [ECOSPOR IV: NCT03183141]) of 349 adults with rCDI and prevalent comorbidities. METHODS VOS or placebo [ECOSPOR III only] (4 capsules once daily for 3 days). Integrated analysis of treatment-emergent adverse events (TEAEs) collected through week 8; serious TEAEs and TEAEs of special interest collected through week 24; and rates of rCDI (toxin-positive diarrhea requiring treatment) evaluated through weeks 8 and 24. RESULTS TEAEs were mostly mild or moderate and gastrointestinal. Most common treatment-related TEAEs were flatulence, abdominal pain and distension, fatigue, and diarrhea. There were 11 deaths (3.2%) and 48 patients (13.8%) with serious TEAEs, none treatment-related. The rCDI rate through week 8 was 9.5% (95% CI 6.6-13.0) and remained low through 24 weeks (15.2%; 95% CI 11.6-19.4). Safety and rCDI rates were consistent across subgroups including age, renal impairment/failure, diabetes, and immunocompromise/immunosuppression. CONCLUSIONS VOS was well tolerated and rates of rCDI remained low through week 24 including in those with comorbidities. These data support the potential benefit of VOS following antibiotics to prevent recurrence in high-risk patients. TRIAL REGISTRATION ClinicalTrials.gov identifier, NCT03183128 and NCT03183141.
Collapse
Affiliation(s)
- Colleen S Kraft
- Department of Pathology and Laboratory Medicine, Division of Infectious Diseases, Emory University, Atlanta, GA, USA
| | - Matthew Sims
- Section of Infectious Diseases and International Medicine, Department of Internal Medicine, Beaumont Royal Oak, Royal Oak, MI, USA
- Departments of Internal Medicine and Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI, USA
| | | | - Thomas J Louie
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Paul Feuerstadt
- Division of Digestive Disease, Yale University School of Medicine, New Haven, CT, USA
- PACT-Gastroenterology Center, Hamden, CT, USA
| | - Edward S Huang
- Department of Gastroenterology, Palo Alto Medical Foundation, Sutter Health, Mountain View, CA, USA
| | - Sahil Khanna
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Charles S Berenson
- University at Buffalo, VA Western New York Healthcare System, Buffalo, NY, USA
| | - Elaine E L Wang
- Seres Therapeutics, 200 Sidney Street, Cambridge, MA, 02139, USA
| | - Stuart H Cohen
- University of California Davis Health, Sacramento, CA, USA
| | - Louis Korman
- Gastroenterology and Hepatology, Chevy Chase Clinical Research, Chevy Chase, MD, USA
| | - Christine Lee
- Island Medical Program, University of British Columbia and University of Victoria, Vancouver, BC, Canada
| | - Colleen R Kelly
- Division of Gastroenterology, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Paul P Cook
- Brody School of Medicine at East, Carolina University, Greenville, NC, USA
| | | | - Mayur Ramesh
- Division of Infectious Diseases, Henry Ford Health, Detroit, MI, USA
| | - Princy Kumar
- Division of Infectious Diseases and Tropical Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Ananya De
- Seres Therapeutics, 200 Sidney Street, Cambridge, MA, 02139, USA
| | - Asli Memisoglu
- Seres Therapeutics, 200 Sidney Street, Cambridge, MA, 02139, USA
| | - David A Lombardi
- Seres Therapeutics, 200 Sidney Street, Cambridge, MA, 02139, USA
| | - Brooke R Hasson
- Seres Therapeutics, 200 Sidney Street, Cambridge, MA, 02139, USA.
| | | | - Lisa von Moltke
- Seres Therapeutics, 200 Sidney Street, Cambridge, MA, 02139, USA
| | - Darrell S Pardi
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
46
|
Saeed A, Batra N, Rezgui R, Alshaghdali K, Alkhalaf I, Yadav DK, Dey P. Gut microbiota-centered risk factors and altered immunometabolism in the pathogenesis and prophylaxis of Clostridium difficile infection. JOURNAL OF KING SAUD UNIVERSITY - SCIENCE 2024; 36:103374. [DOI: 10.1016/j.jksus.2024.103374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2025]
|
47
|
Berry P, Khanna S. Fecal microbiota spores, live-brpk (VOWST™/VOS) for prevention of recurrent Clostridioides difficile infection. Future Microbiol 2024; 19:1519-1528. [PMID: 39320321 DOI: 10.1080/17460913.2024.2403892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 09/10/2024] [Indexed: 09/26/2024] Open
Abstract
Clostridioides difficile infection (CDI) is a health crisis comprising a majority of healthcare-associated infections and is now being seen in the community. Persistent dysbiosis despite treatment with standard-of-care antibiotics increases risk of recurrent infections. Fecal microbiota transplantation has been an effective way of addressing dysbiosis, but the studies have lacked standardization, which makes outcome and safety data difficult to interpret. Standardized microbiome therapies have demonstrated efficacy and safety for recurrent CDI and have been approved to prevent recurrent infection. In this review, we discuss the data behind and the practice use of fecal microbiota spores, live-brpk (VOWST™ / VOS), a US FDA approved live biotherapeutic for the prevention of recurrent CDI.
Collapse
Affiliation(s)
- Parul Berry
- C. difficile Clinic & Microbiome Restoration Program, Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Sahil Khanna
- C. difficile Clinic & Microbiome Restoration Program, Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
48
|
Villafuerte Gálvez JA, Kelly CP. An Anti-inflammatory Approach to Drug Repurposing for Clostridioides difficile Infection. J Infect Dis 2024; 230:527-528. [PMID: 38243873 DOI: 10.1093/infdis/jiae022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 01/17/2024] [Indexed: 01/22/2024] Open
Affiliation(s)
- Javier A Villafuerte Gálvez
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Ciarán P Kelly
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
49
|
Xie Y, Irwin S, Chupina Estrada A, Nelson B, Bullock A, Fontenot L, Feng H, Sun M, Koon HW. Loratadine as an Anti-inflammatory Agent Against Clostridium difficile Toxin B. J Infect Dis 2024; 230:545-557. [PMID: 38243838 PMCID: PMC11420802 DOI: 10.1093/infdis/jiae021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 11/30/2023] [Accepted: 01/17/2024] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND Clostridium difficile infection (CDI) is a debilitating nosocomial infection. C. difficile produces toxins A and B, which cause inflammation. Existing therapies have issues with recurrence, cost, and safety. We aim to discover a safe, effective, and economical nonmicrobiological therapeutic approach against CDI. METHODS We included human primary peripheral blood mononuclear cells (PBMCs), fresh human colonic explants, and humanized HuCD34-NCG mice. Toxin A+B+ VPI 10463 and A-B+ ribotype 017 C. difficile strains were used. We used single-cell RNA profiling and high-throughput screening to find actionable toxin B-dependent pathways in PBMCs. RESULTS Histamine 1 receptor-related drugs were found among the hit compounds that reversed toxin-mediated macrophage inflammatory protein (MIP) 1α expression in PBMCs. We identified loratadine as the safest representative antihistamine for therapeutic development. Loratadine inhibited toxin B-induced MIP-1α secretion in fresh human colonic tissues. Oral loratadine (10 mg/kg/d) maintained survival, inhibited intestinal CCl3 messenger RNA expression, and prevented vancomycin-associated recurrence in the VPI 10463-infected mice and ribotype 017-infected hamsters. Splenocytes from loratadine-treated mice conferred anti-inflammatory effects to the VPI 10463-infected T/B-cell--deficient Rag-/- mice. Oral loratadine suppressed human MIP-1α expression in monocytes/macrophages in toxin B-expressing ribotype 017-infected humanized HuCD34-NCG mice. CONCLUSIONS Loratadine may be repurposed to optimize existing therapies against CDI.
Collapse
Affiliation(s)
- Ying Xie
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, California, USA
- Department of Gastroenterology, The First Hospital of China Medical University, Shenyang City, China
| | - Sophie Irwin
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, California, USA
| | - Andrea Chupina Estrada
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, California, USA
| | - Becca Nelson
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, California, USA
| | - Ashlen Bullock
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, California, USA
| | - Lindsey Fontenot
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, California, USA
| | - Hanping Feng
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, Maryland, USA
| | - Mingjun Sun
- Department of Gastroenterology, The First Hospital of China Medical University, Shenyang City, China
| | - Hon Wai Koon
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
50
|
Acevedo-Román A, Pagán-Zayas N, Velázquez-Rivera LI, Torres-Ventura AC, Godoy-Vitorino F. Insights into Gut Dysbiosis: Inflammatory Diseases, Obesity, and Restoration Approaches. Int J Mol Sci 2024; 25:9715. [PMID: 39273662 PMCID: PMC11396321 DOI: 10.3390/ijms25179715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/30/2024] [Accepted: 09/07/2024] [Indexed: 09/15/2024] Open
Abstract
The gut microbiota is one of the most critical factors in human health. It involves numerous physiological processes impacting host health, mainly via immune system modulation. A balanced microbiome contributes to the gut's barrier function, preventing the invasion of pathogens and maintaining the integrity of the gut lining. Dysbiosis, or an imbalance in the gut microbiome's composition and function, disrupts essential processes and contributes to various diseases. This narrative review summarizes key findings related to the gut microbiota in modern multifactorial inflammatory conditions such as ulcerative colitis or Crohn's disease. It addresses the challenges posed by antibiotic-driven dysbiosis, particularly in the context of C. difficile infections, and the development of novel therapies like fecal microbiota transplantation and biotherapeutic drugs to combat these infections. An emphasis is given to restoration of the healthy gut microbiome through dietary interventions, probiotics, prebiotics, and novel approaches for managing gut-related diseases.
Collapse
Affiliation(s)
- Andy Acevedo-Román
- Microbiology Department, University of Puerto Rico Medical Sciences Campus, San Juan 00936, Puerto Rico
| | - Natalia Pagán-Zayas
- Microbiology Department, University of Puerto Rico Medical Sciences Campus, San Juan 00936, Puerto Rico
| | - Liz I Velázquez-Rivera
- Microbiology Department, University of Puerto Rico Medical Sciences Campus, San Juan 00936, Puerto Rico
| | - Aryanne C Torres-Ventura
- Microbiology Department, University of Puerto Rico Medical Sciences Campus, San Juan 00936, Puerto Rico
| | - Filipa Godoy-Vitorino
- Microbiology Department, University of Puerto Rico Medical Sciences Campus, San Juan 00936, Puerto Rico
| |
Collapse
|