1
|
Bénard MV, de Bruijn CMA, Matamoros S, Wentink-Bonnema EMS, Benninga MA, Ponsioen CY, Zonneveld R. Transient colonization with Blastocystis spp. after transmission via fecal microbiota transplantation. Eur J Clin Microbiol Infect Dis 2025:10.1007/s10096-025-05124-6. [PMID: 40257672 DOI: 10.1007/s10096-025-05124-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 04/02/2025] [Indexed: 04/22/2025]
Abstract
BACKGROUND The pathogenicity of Blastocystis spp. is still debated. Guidelines for feces donor screening differ in their advice to screen for Blastocystis spp., but when tested, its presence is a common reason for exclusion. Blastocystis spp. are correlated to increased bacterial alpha-diversity and distinct bacterial groups and therefore its presence may indicate favorable efficacy of fecal microbiota transplantation (FMT). The latest European consensus report no longer advices rejecting feces donors testing positive for Blastocystis spp. Only one paper has been published on human transmission of Blastocystis spp. via frozen FMT. OBJECTIVE To investigate the transmission and long-term effects of Blastocystis-positive FMT, prepared with fresh (i.e., unfrozen) feces. METHODS In a trial (NCT03074227) on FMT for refractory Irritable Bowel Syndrome (IBS), adolescents (age 16-20 years) received two administrations - at baseline and after 6 weeks - of fresh allogeneic FMT from a Blastocystis-positive donor via nasoduodenal tube. The follow-up was 48 weeks. Blastocystis spp. presence, viability and subtyping were determined using microscopy, culture, PCR and sequencing. RESULTS Three recipients received FMT from one donor colonized with Blastocystis subtype 3 (ST3). At baseline, two recipients were negative for Blastocystis spp. and one recipient carried ST2. Culturing revealed viable Blastocystis spp. in fresh donor feces but not in frozen samples. After FMT with fresh feces, the two prior-negative recipients tested positive for the donor's ST3 at 12 weeks, but had lost this subtype by week 24 and 48. The recipient initially colonized with ST2 remained colonized with ST2 and did not acquire ST3. Transient adverse events occurred, but did not differ from patients treated with Blastocystis-negative FMT. No FMT-related serious adverse events emerged. CONCLUSION We present the first long-term data on viable Blastocystis spp. transmission via fresh FMT in three cases. Transient colonization with Blastocystis spp. was observed, without serious FMT-related adverse events.
Collapse
Affiliation(s)
- M V Bénard
- Department of Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM), Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Inflammatory Bowel Disease Center, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - C M A de Bruijn
- Emma Children's Hospital, Pediatric Gastroenterology, Hepatology and Nutrition, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development Research Institute, Amsterdam University Medical Center, Emma Children's Hospital, Amsterdam, The Netherlands
| | - S Matamoros
- Department of Medical Microbiology and Infection Prevention, Section Clinical Parasitology, Academic Medical Center, Amsterdam University Medical Center, University of Amsterdam, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
| | - E M S Wentink-Bonnema
- Department of Medical Microbiology and Infection Prevention, Section Clinical Parasitology, Academic Medical Center, Amsterdam University Medical Center, University of Amsterdam, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
| | - M A Benninga
- Emma Children's Hospital, Pediatric Gastroenterology, Hepatology and Nutrition, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Reproduction & Development Research Institute, Amsterdam University Medical Center, Emma Children's Hospital, Amsterdam, The Netherlands
| | - C Y Ponsioen
- Department of Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM), Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Inflammatory Bowel Disease Center, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - R Zonneveld
- Department of Medical Microbiology and Infection Prevention, Section Clinical Parasitology, Academic Medical Center, Amsterdam University Medical Center, University of Amsterdam, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands.
| |
Collapse
|
2
|
Randolph NK, Salerno M, Klein H, Diaz-Campos D, van Balen JC, Winston JA. Preparation of Fecal Microbiota Transplantation Products for Companion Animals. PLoS One 2025; 20:e0319161. [PMID: 40203217 PMCID: PMC11981653 DOI: 10.1371/journal.pone.0319161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 01/28/2025] [Indexed: 04/11/2025] Open
Abstract
Fecal microbiota transplantation (FMT) is increasingly utilized in small animal medicine for the treatment of a variety of gastrointestinal and non-gastrointestinal disorders. Despite proven clinical efficacy, there is no detailed protocol available for the preparation and storage of FMT products for veterinarians in a variety of clinical settings. Herein, the effect of processing technique on the microbial community structure was assessed with amplicon sequence analysis. Microbial viability was assessed with standard culture techniques using selective media. Given the fastidious nature of many intestinal microbes, colony forming units are considered surrogate viable microbes, representing a portion of potentially viable microbes. FMT products from four screened canine fecal donors and six screened feline fecal donors were processed aerobically according to a double centrifugation protocol adapted from the human medical literature. Fresh feces from an additional three screened canine fecal donors were used to evaluate the effect of cryopreservative, centrifugation, and short-term storage on microbial community structure and in vitro surrogate bacterial viability. Finally, fresh feces from a third group of three screened canine and three screened feline fecal donors were used to evaluate the long-term in vitro surrogate bacterial viability of three frozen and lyophilized FMT products. Microbiota analysis revealed that each canine fecal donor has a unique microbial profile. Processing of canine and feline feces for FMT does not significantly alter the overall microbial community structure. The addition of cryopreservatives and lyopreservatives significantly improved long-term viability, up to 6 months, for frozen and lyophilized FMT products compared to unprocessed raw feces with no cryopreservative. These results prove the practicality of this approach for FMT preparation in veterinary medicine and provide a detailed protocol for researchers and companion animal practitioners. Future in vivo research is needed to evaluate how the preparation and microbial viability of FMT impacts the recipient's microbial community and clinical outcomes across multiple disease phenotypes.
Collapse
Affiliation(s)
- Nina K. Randolph
- Department of Veterinary Clinical Sciences. College of Veterinary Medicine, The Ohio State University. Columbus, Ohio, United States of America
- Comparative Hepatobiliary and Intestinal Research Program, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Matthew Salerno
- Department of Veterinary Clinical Sciences. College of Veterinary Medicine, The Ohio State University. Columbus, Ohio, United States of America
- Comparative Hepatobiliary and Intestinal Research Program, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Hannah Klein
- Department of Veterinary Clinical Sciences. College of Veterinary Medicine, The Ohio State University. Columbus, Ohio, United States of America
- Comparative Hepatobiliary and Intestinal Research Program, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Dubraska Diaz-Campos
- Department of Veterinary Clinical Sciences. College of Veterinary Medicine, The Ohio State University. Columbus, Ohio, United States of America
| | - Joany C. van Balen
- Department of Veterinary Clinical Sciences. College of Veterinary Medicine, The Ohio State University. Columbus, Ohio, United States of America
| | - Jenessa A. Winston
- Department of Veterinary Clinical Sciences. College of Veterinary Medicine, The Ohio State University. Columbus, Ohio, United States of America
- Comparative Hepatobiliary and Intestinal Research Program, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, United States of America
| |
Collapse
|
3
|
Wang X, Zhao D, Bi D, Li L, Tian H, Yin F, Zuo T, Ianiro G, Li N, Chen Q, Qin H. Fecal microbiota transplantation: transitioning from chaos and controversial realm to scientific precision era. Sci Bull (Beijing) 2025; 70:970-985. [PMID: 39855927 DOI: 10.1016/j.scib.2025.01.029] [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: 09/21/2024] [Revised: 12/05/2024] [Accepted: 12/13/2024] [Indexed: 01/27/2025]
Abstract
With the popularization of modern lifestyles, the spectrum of intestinal diseases has become increasingly diverse, presenting significant challenges in its management. Traditional pharmaceutical interventions have struggled to keep pace with these changes, leaving many patients refractory to conventional pharmaceutical treatments. Fecal microbiota transplantation (FMT) has emerged as a promising therapeutic approach for enterogenic diseases. Still, controversies persist regarding its active constituents, mechanism of action, scheme of treatment evaluation, indications, and contraindications. In this review, we investigated the efficacy of FMT in addressing gastrointestinal and extraintestinal conditions, drawing from follow-up data on over 8000 patients. We systematically addressed the controversies surrounding FMT's clinical application. We delved into key issues such as its technical nature, evaluation methods, microbial restoration mechanisms, and impact on the host-microbiota interactions. Additionally, we explored the potential colonization patterns of FMT-engrafted new microbiota throughout the entire intestine and elucidated the specific pathways through which the new microbiota modulates host immunity, metabolism, and genome.
Collapse
Affiliation(s)
- Xinjun Wang
- Tenth People's Hospital of Tongji University, Shanghai 200072, China; Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215000, China; Department of Functional Intestinal Diseases, General Surgery of Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Shanghai Gastrointestinal Microecology Research Center, Shanghai 200072, China; Shanghai Institution of Gut Microbiota Research and Engineering Development, Shanghai 200072, China; Clinical Research Center for Digestive Diseases, Tongji University School of Medicine, Shanghai 200072, China.
| | - Di Zhao
- Tenth People's Hospital of Tongji University, Shanghai 200072, China; Department of Functional Intestinal Diseases, General Surgery of Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Shanghai Gastrointestinal Microecology Research Center, Shanghai 200072, China; Shanghai Institution of Gut Microbiota Research and Engineering Development, Shanghai 200072, China; Clinical Research Center for Digestive Diseases, Tongji University School of Medicine, Shanghai 200072, China
| | - Dexi Bi
- Department of Pathology, Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Long Li
- Tenth People's Hospital of Tongji University, Shanghai 200072, China; Department of Functional Intestinal Diseases, General Surgery of Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Shanghai Gastrointestinal Microecology Research Center, Shanghai 200072, China; Shanghai Institution of Gut Microbiota Research and Engineering Development, Shanghai 200072, China; Clinical Research Center for Digestive Diseases, Tongji University School of Medicine, Shanghai 200072, China
| | - Hongliang Tian
- Tenth People's Hospital of Tongji University, Shanghai 200072, China; Department of Functional Intestinal Diseases, General Surgery of Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Shanghai Gastrointestinal Microecology Research Center, Shanghai 200072, China; Shanghai Institution of Gut Microbiota Research and Engineering Development, Shanghai 200072, China; Clinical Research Center for Digestive Diseases, Tongji University School of Medicine, Shanghai 200072, China
| | - Fang Yin
- Tenth People's Hospital of Tongji University, Shanghai 200072, China; Department of Functional Intestinal Diseases, General Surgery of Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Shanghai Gastrointestinal Microecology Research Center, Shanghai 200072, China; Shanghai Institution of Gut Microbiota Research and Engineering Development, Shanghai 200072, China; Clinical Research Center for Digestive Diseases, Tongji University School of Medicine, Shanghai 200072, China
| | - Tao Zuo
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou 510655, China
| | - Gianluca Ianiro
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, 00168, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, 00168, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato, Rome, 00168, Italy
| | - Ning Li
- Tenth People's Hospital of Tongji University, Shanghai 200072, China; Department of Functional Intestinal Diseases, General Surgery of Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Shanghai Gastrointestinal Microecology Research Center, Shanghai 200072, China; Shanghai Institution of Gut Microbiota Research and Engineering Development, Shanghai 200072, China; Clinical Research Center for Digestive Diseases, Tongji University School of Medicine, Shanghai 200072, China
| | - Qiyi Chen
- Tenth People's Hospital of Tongji University, Shanghai 200072, China; Department of Functional Intestinal Diseases, General Surgery of Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Shanghai Gastrointestinal Microecology Research Center, Shanghai 200072, China; Shanghai Institution of Gut Microbiota Research and Engineering Development, Shanghai 200072, China; Clinical Research Center for Digestive Diseases, Tongji University School of Medicine, Shanghai 200072, China.
| | - Huanlong Qin
- Tenth People's Hospital of Tongji University, Shanghai 200072, China; Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215000, China.
| |
Collapse
|
4
|
Murgiano M, Bartocci B, Puca P, di Vincenzo F, Del Gaudio A, Papa A, Cammarota G, Gasbarrini A, Scaldaferri F, Lopetuso LR. Gut Microbiota Modulation in IBD: From the Old Paradigm to Revolutionary Tools. Int J Mol Sci 2025; 26:3059. [PMID: 40243712 PMCID: PMC11988433 DOI: 10.3390/ijms26073059] [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: 01/12/2025] [Revised: 03/18/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
Inflammatory bowel diseases (IBDs) are chronic inflammatory disorders primarily comprising two main conditions: ulcerative colitis and Crohn's disease. The gut microbiota's role in driving inflammation in IBD has garnered significant attention, yet the precise mechanisms through which the microbiota influences IBD pathogenesis remain largely unclear. Given the limited therapeutic options for IBD, alternative microbiota-targeted therapies-including prebiotics, probiotics, postbiotics, and symbiotics-have been proposed. While these approaches have shown promising results, microbiota modulation is still mainly considered an adjunct therapy to conventional treatments, with a demonstrated impact on patients' quality of life. Fecal microbiota transplantation (FMT), already approved for treating Clostridioides difficile infection, represents the first in a series of innovative microbiota-based therapies under investigation. Microbial biotherapeutics are emerging as personalized and cutting-edge tools for IBD management, encompassing next-generation probiotics, bacterial consortia, bacteriophages, engineered probiotics, direct metabolic pathway modulation, and nanotherapeutics. This review explores microbial modulation as a therapeutic strategy for IBDs, highlighting current approaches and examining promising tools under development to better understand their potential clinical applications in managing intestinal inflammatory disorders.
Collapse
Affiliation(s)
- Marco Murgiano
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
| | - Bianca Bartocci
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
| | - Pierluigi Puca
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
- Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy
| | - Federica di Vincenzo
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
| | - Angelo Del Gaudio
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
| | - Alfredo Papa
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
- Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy
| | - Giovanni Cammarota
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
- Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
- Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy
| | - Franco Scaldaferri
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
- Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, 00168 Rome, Italy
| | - Loris Riccardo Lopetuso
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (B.B.); (P.P.); (F.d.V.); (A.D.G.); (A.P.); (G.C.); (A.G.); (F.S.)
- Dipartimento di Scienze della Vita, della Salute e delle Professioni Sanitarie, Università degli Studi Link, 00165 Rome, Italy
| |
Collapse
|
5
|
Wang M, Liu Y, Zhong L, Wu F, Wang J. Advancements in the investigation of gut microbiota-based strategies for stroke prevention and treatment. Front Immunol 2025; 16:1533343. [PMID: 40103814 PMCID: PMC11914130 DOI: 10.3389/fimmu.2025.1533343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 02/11/2025] [Indexed: 03/20/2025] Open
Abstract
Stroke represents a predominant cause of mortality and disability on a global scale, impacting millions annually and exerting a considerable strain on healthcare systems. The incidence of stroke exhibits regional variability, with ischemic stroke accounting for the majority of occurrences. Post-stroke complications, such as cognitive impairment, motor dysfunction, and recurrent stroke, profoundly affect patients' quality of life. Recent advancements have elucidated the microbiota-gut-brain axis (MGBA), underscoring the complex interplay between gut health and brain function. Dysbiosis, characterized by an imbalance in gut microbiota, is significantly linked to an elevated risk of stroke and unfavorable outcomes. The MGBA plays a crucial role in modulating immune function, neurotransmitter levels, and metabolic byproducts, which may intensify neuroinflammation and impair cerebral health. This review elucidates the role of MGBA in stroke pathophysiology and explores potential gut-targeted therapeutic strategies to reduce stroke risk and promote recovery, including probiotics, prebiotics, pharmacological interventions, and dietary modifications. However, the current prevention and treatment strategies based on intestinal flora still face many problems, such as the large difference of individual intestinal flora, the stability of efficacy, and the long-term safety need to be considered. Further research needs to be strengthened to promote its better application in clinical practice.
Collapse
Affiliation(s)
- Min Wang
- Department of Gastroenterology, The First People's Hospital of Xiaoshan District, Hangzhou, Zhejiang, China
| | - Yan Liu
- Department of Gastroenterology, The First People's Hospital of Xiaoshan District, Hangzhou, Zhejiang, China
| | - Li Zhong
- Department of Gastroenterology, The First People's Hospital of Xiaoshan District, Hangzhou, Zhejiang, China
| | - Fang Wu
- Department of Gastroenterology, The First People's Hospital of Xiaoshan District, Hangzhou, Zhejiang, China
| | - Jinjin Wang
- Department of Gastroenterology, The First People's Hospital of Xiaoshan District, Hangzhou, Zhejiang, China
| |
Collapse
|
6
|
Facchin S, Cardin R, Patuzzi I, Carlotto C, Minotto M, Barberio B, Zingone F, Besutti VM, Castagliuolo I, Cattelan A, Savarino EV. Long-term stability and efficacy of frozen fecal microbiota transplant (FMT) product at 24 months. Dig Liver Dis 2025; 57:702-706. [PMID: 39672770 DOI: 10.1016/j.dld.2024.11.025] [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: 10/09/2024] [Revised: 11/27/2024] [Accepted: 11/28/2024] [Indexed: 12/15/2024]
Abstract
BACKGROUND Freezing donor fecal microbiota has improved fecal microbiota transplantation (FMT) for recurrent C. difficile infection (CDI), achieving short-term effectiveness similar to fresh-samples. Research shows frozen fecal matter remains effective for up to 12-months at -80 °C. OBJECTIVE To assess how long-term-freezing and thawing affect the viability, microbial composition, and clinical efficacy of frozen-stools for FMT. METHODS Stool samples from three donors were processed into 18 aliquots, thawed at intervals over two years, and analyzed for cell viability and microbial load. Microbiota composition was assessed through 16S-sequencing, with diversity evaluated using the Shannon-index and Principal-Coordinates-Analysis based on Bray-Curtis-distance (α/β-diversity). The same donors provided fecal material for a total of 23 FMT procedures, including 15 for CDI and 8 off-label. RESULTS We found that donor stools frozen for two years contained viable bacteria comparable to fresh samples, with anaerobic and aerobic species remaining viable for 24 months. Despite a reduction in colony-forming-units, FMT was successful in 71.4 % and 100 % of the cases at one year and at the end of follow-up, respectively. Most bacterial changes occurred among anaerobic species (Blautia producta and Bifidobacterium adolescentis), increasing post-thawing. Notably, specific taxa, (C. aerofaciens and Erysipelotrichaceae_Cc115), showed significant unexplained increase. CONCLUSION Long-term-stool-storage enhances FMT accessibility without compromising its success, despite taxonomic changes after 24 months.
Collapse
Affiliation(s)
- Sonia Facchin
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padua, Padua, Italy; Gastroenterology Unit, Azienda Ospedale Università di Padova, Padova, Italy
| | - Romilda Cardin
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padua, Padua, Italy; Gastroenterology Unit, Azienda Ospedale Università di Padova, Padova, Italy
| | - Ilaria Patuzzi
- Research & Development Division, EuBiome S.r.l., 35131 Padova, Italy
| | - Chiara Carlotto
- Gastroenterology Unit, Azienda Ospedale Università di Padova, Padova, Italy
| | - Milena Minotto
- Gastroenterology Unit, Azienda Ospedale Università di Padova, Padova, Italy
| | - Brigida Barberio
- Gastroenterology Unit, Azienda Ospedale Università di Padova, Padova, Italy
| | - Fabiana Zingone
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padua, Padua, Italy; Gastroenterology Unit, Azienda Ospedale Università di Padova, Padova, Italy
| | - Valeria M Besutti
- Microbiology Unit, Azienda Ospedale Università di Padova, Padova, Italy
| | | | - Annanaria Cattelan
- Infectious and Tropical Diseases Unit, Padova University Hospital, Padua, Italy
| | - Edoardo V Savarino
- Department of Surgery, Oncology and Gastroenterology (DISCOG), University of Padua, Padua, Italy; Gastroenterology Unit, Azienda Ospedale Università di Padova, Padova, Italy.
| |
Collapse
|
7
|
Xu J, Chen N, Li Z, Liu Y. Gut microbiome and liver diseases. FUNDAMENTAL RESEARCH 2025; 5:890-901. [PMID: 40242515 PMCID: PMC11997574 DOI: 10.1016/j.fmre.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 04/18/2025] Open
Abstract
Symbiotic microbiota plays a crucial role in the education, development, and maintenance of the host immune system, significantly contributing to overall health. Through the gut-liver axis, the gut microbiota and liver have a bidirectional relationship that is becoming increasingly evident as more research highlights the translocation of the gut microbiota and its metabolites. The focus of this narrative review is to examine and discuss the importance of the gut-liver axis and the enterohepatic barrier in maintaining overall health. Additionally, we emphasize the crucial role of the gut microbiome in liver diseases and explore potential therapeutic strategies for liver diseases by manipulating the microbiota.
Collapse
Affiliation(s)
- Jun Xu
- Department of Gastroenterology, Peking University People's Hospital, Beijing 100044, China
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People's Hospital, Beijing 100044, China
| | - Ning Chen
- Department of Gastroenterology, Peking University People's Hospital, Beijing 100044, China
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People's Hospital, Beijing 100044, China
| | - Zhou Li
- Beijing Key Laboratory of Micro-nano Energy and Sensor, Beijing Institute of Nanoenergy and Nanosystems Chinese Academy of Sciences, Beijing 101400, China
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yulan Liu
- Department of Gastroenterology, Peking University People's Hospital, Beijing 100044, China
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People's Hospital, Beijing 100044, China
| |
Collapse
|
8
|
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
|
9
|
Lopetuso LR, Deleu S, Puca P, Abreu MT, Armuzzi A, Barbara G, Caprioli F, Chieng S, Costello SP, Damiani A, Danese S, Del Chierico F, D'Haens G, Dotan I, Facciotti F, Falony G, Fantini MC, Fiorino G, Gionchetti P, Godny L, Hart A, Kupčinskas J, Iqbal T, Laterza L, Lombardini L, Maharshak N, Marasco G, Masucci L, Papa A, Paramsothy S, Petito V, Piovani D, Pugliese D, Putignani L, Raes J, Ribaldone DG, Sanguinetti M, Savarino EV, Sokol H, Vetrano S, Ianiro G, Cammarota G, Cominelli F, Pizarro TT, Tilg H, Gasbarrini A, Vermeire S, Scaldaferri F. Guidance for Fecal Microbiota Transplantation Trials in Ulcerative Colitis: The Second ROME Consensus Conference. Inflamm Bowel Dis 2025:izaf013. [PMID: 39932857 DOI: 10.1093/ibd/izaf013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Indexed: 02/13/2025]
Abstract
BACKGROUND Fecal microbiota transplantation (FMT) is emerging as a potential treatment modality for individuals living with inflammatory bowel disease (IBD). Despite its promise, the effectiveness of FMT for treating IBD, particularly for ulcerative colitis (UC), still requires thorough clinical investigation. Notwithstanding differences in methodologies, current studies demonstrate its potential for inducing remission in UC patients. Therefore, standardized and robust randomized clinical trials (RCTs) are needed to further support its efficacy for managing UC. The aim of the second Rome Consensus Conference was to address gaps and uncertainties identified in previous research regarding FMT and to offer a robust framework for future studies applied to the treatment of UC. METHODS Global experts in the field of clinical IBD, mucosal immunology, and microbiology (N = 48) gathered to address the need for standardized clinical trials in FMT investigation. The group focused on key issues, such as stool donation, donor selection, characterization of fecal biomass, potential administration routes, as well as the process of induction, maintenance, and endpoint readouts. RESULTS AND CONCLUSIONS The consensus achieved during this conference established standardization of methods and protocols to enhance the current quality of research, with the aim of eventual implementation of FMT in managing UC and the ultimate goal of improving patient outcomes.
Collapse
Affiliation(s)
- Loris R Lopetuso
- IBD Unit, CEMAD, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Department of Life Science, Health, and Health Professions, Link Campus University, Rome, Italy
| | - Sara Deleu
- IBD Unit, CEMAD, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Department of Chronic Diseases, Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Pierluigi Puca
- IBD Unit, CEMAD, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Maria Teresa Abreu
- Division of Gastroenterology, Department of Medicine, Crohn's and Colitis Center, University of Miami - Miller School of Medicine, Miami, FL, USA
| | - Alessandro Armuzzi
- IBD Unit, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Giovanni Barbara
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Rome, Italy
- IRCCS Azienda Ospedaliero Universitaria Di Bologna, Bologna, Italy
| | - Flavio Caprioli
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Siew Chieng
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Samuel Paul Costello
- Department of Gastroenterology, The Queen Elizabeth Hospital, Adelaide, SA, Australia
| | - Andrea Damiani
- Real World Data Facility, Gemelli Generator, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Silvio Danese
- Department of Gastroenterology and Digestive Endoscopy, IRCCS San Raffaele Hospital and Vita-Salute San Raffaele University, Milan, Italy
| | - Federica Del Chierico
- Immunology, Rheumatology and Infectious Diseases Research Area, Unit of Human Microbiome, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Geert D'Haens
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Iris Dotan
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel
| | - Federica Facciotti
- Dipartimento di Biotecnologie e Bioscienze, Università di Milano-Bicocca, Milan, Italy
| | - Gwen Falony
- Institute of Medical Microbiology and Hygiene and Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Massimo Claudio Fantini
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
- Gastroenterology Unit, Azienda Ospedaliero-Universitaria di Cagliari, Cagliari, Italy
| | | | - Paolo Gionchetti
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- IBD Unit, IRCCS Azienda Ospedaliero Universitaria di Bologna, Bologna,, Italy
| | - Lihi Godny
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel
| | - Ailsa Hart
- IBD Unit, St Mark's Hospital, Harrow, Middlesex, UK
| | - Juozas Kupčinskas
- Department of Gastroenterology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Tariq Iqbal
- Microbiome Treatment Center, University of Birmingham, Birmingham, UK
| | - Lucrezia Laterza
- IBD Unit, CEMAD, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Letizia Lombardini
- Centro Nazionale Trapianti (CNT), Istituto Superiore di Sanità, Rome, Italy
| | - Nitsan Maharshak
- Division of Gastroenterology, Rabin Medical Center, Petah-Tikva, Israel
| | - Giovanni Marasco
- IRCCS Azienda Ospedaliero Universitaria Di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna,, Italy
| | - Luca Masucci
- Microbiology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Alfredo Papa
- IBD Unit, CEMAD, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Sudarshan Paramsothy
- Gastroenterology and Liver Services, Concord Repatriation General Hospital, Sydney, Australia
| | - Valentina Petito
- IBD Unit, CEMAD, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Daniele Piovani
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Daniela Pugliese
- IBD Unit, CEMAD, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Lorenza Putignani
- Unit of Microbiology and Diagnostic Immunology, Unit of Microbiomics and Research Area of Immunology, Rheumatology and Infectious Diseases, Unit of Human Microbiome, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Jeroen Raes
- Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
- Center for Microbiology, VIB, Gent, Belgium
| | | | - Maurizio Sanguinetti
- Microbiology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | | | - Harry Sokol
- INSERM, Centre de Recherche Saint-Antoine, CRSA, Sorbonne Université, Paris, France
- Department of Gastroenterology, Saint Antoine Hospital, Paris, France
| | - Stefania Vetrano
- Laboratory of Gastrointestinal Immunopathology, Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Gianluca Ianiro
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento di Scienze Mediche e Chirurgiche, UOC di Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Giovanni Cammarota
- Dipartimento di Scienze Mediche e Chirurgiche, UOC di Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Fabio Cominelli
- Division of Gastroenterology and Liver Diseases, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Theresa T Pizarro
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Herbert Tilg
- Department of Gastroenterology, Saint Antoine Hospital, Paris, France
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Antonio Gasbarrini
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento di Scienze Mediche e Chirurgiche, UOC di Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Severine Vermeire
- Department of Chronic Diseases, Metabolism (CHROMETA), KU Leuven, Leuven, Belgium
- Department of Gastroenterology and Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Franco Scaldaferri
- IBD Unit, CEMAD, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
10
|
Moutsoglou D, Syal A, Lopez S, Nelson EC, Chen L, Kabage AJ, Fischer M, Khoruts A, Vaughn BP, Staley C. Novel Microbial Engraftment Trajectories Following Microbiota Transplant Therapy in Ulcerative Colitis. J Crohns Colitis 2025; 19:jjae142. [PMID: 39240145 DOI: 10.1093/ecco-jcc/jjae142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND AND AIMS Microbiota transplant therapy (MTT) is an emerging treatment for ulcerative colitis (UC). One proposed mechanism for the benefit of MTT is through engraftment of donor microbiota; however, engraftment kinetics are unknown. We identified SourceTracker as an efficient method both to determine engraftment and for the kinetic study of engrafting donor taxa to aid in determining the mechanism of how this therapy may treat UC. METHODS Ulcerative colitis patients received either encapsulated (drug name MTP-101C) or placebo capsules daily for 8 weeks followed by a 4-week washout period. Amplicon sequence data from donors and patients were analyzed using the Bayesian algorithm SourceTracker. RESULTS Twenty-seven patients were enrolled, 14 to placebo and 13 to MTT. Baseline Shannon and Chao1 indices negatively correlated with week 12 donor engraftment for patients treated with active drug capsules but not for placebo patients. SourceTracker engraftment positively correlated with the week 12 distance from donors measured using the Bray-Curtis similarity metric in treated patients but not with placebo. Engraftment at week 12 was significantly higher in the MTT group than in the placebo group. We identified engrafting taxa from donors in our patients and quantified the proportion of donor similarity or engraftment during weeks 1 through 8 (active treatment) and week 12, 4 weeks after the last dose. CONCLUSION SourceTracker can be used as a simple and reliable method to quantify donor microbial community engraftment and donor taxa contribution in patients with UC and other inflammatory conditions treated with MTT.
Collapse
Affiliation(s)
- Daphne Moutsoglou
- Department of Gastroenterology, Minneapolis VA Health Care System, MN 55417, USA
- Department of Medicine, University of Minnesota, Minneapolis, MN 55455, USA
| | - Aneesh Syal
- Division of Basic and Translational Research, Department of Surgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Sharon Lopez
- Division of Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis, MN 55355, USA
| | - Elizabeth C Nelson
- Division of Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis, MN 55355, USA
| | - Lulu Chen
- Division of Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis, MN 55355, USA
| | - Amanda J Kabage
- Division of Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis, MN 55355, USA
| | - Monika Fischer
- Division of Gastroenterology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Alexander Khoruts
- Division of Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis, MN 55355, USA
| | - Byron P Vaughn
- Division of Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis, MN 55355, USA
| | - Christopher Staley
- Division of Basic and Translational Research, Department of Surgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| |
Collapse
|
11
|
Porcari S, Mullish BH, Asnicar F, Ng SC, Zhao L, Hansen R, O'Toole PW, Raes J, Hold G, Putignani L, Hvas CL, Zeller G, Koren O, Tun H, Valles-Colomer M, Collado MC, Fischer M, Allegretti J, Iqbal T, Chassaing B, Keller J, Baunwall SM, Abreu M, Barbara G, Zhang F, Ponziani FR, Costello SP, Paramsothy S, Kao D, Kelly C, Kupcinskas J, Youngster I, Franceschi F, Khanna S, Vehreschild M, Link A, De Maio F, Pasolli E, Miguez AB, Brigidi P, Posteraro B, Scaldaferri F, Stojanovic MR, Megraud F, Malfertheiner P, Masucci L, Arumugam M, Kaakoush N, Segal E, Bajaj J, Leong R, Cryan J, Weersma RK, Knight R, Guarner F, Shanahan F, Cani PD, Elinav E, Sanguinetti M, de Vos WM, El-Omar E, Dorè J, Marchesi J, Tilg H, Sokol H, Segata N, Cammarota G, Gasbarrini A, Ianiro G. International consensus statement on microbiome testing in clinical practice. Lancet Gastroenterol Hepatol 2025; 10:154-167. [PMID: 39647502 DOI: 10.1016/s2468-1253(24)00311-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/10/2024] [Accepted: 09/13/2024] [Indexed: 12/10/2024]
Abstract
There is growing interest in the potential exploitation of the gut microbiome as a diagnostic tool in medicine, but evidence supporting its clinical usefulness is scarce. An increasing number of commercial providers offer direct-to-consumer microbiome diagnostic tests without any consensus on their regulation or any proven value in clinical practice, which could result in considerable waste of individual and health-care resources and potential drawbacks in the clinical management of patients. We convened an international multidisciplinary expert panel to standardise best practices of microbiome testing for clinical implementation, including recommendations on general principles and minimum requirements for their provision, indications, pre-testing protocols, method of analyses, reporting of results, and potential clinical value. We also evaluated current knowledge gaps and future directions in this field. We aimed to establish a framework to regulate the provision of microbiome testing and minimise the use of inappropriate tests and pave the way for the evidence-based development and use of human microbiome diagnostics in clinical medicine.
Collapse
Affiliation(s)
- Serena Porcari
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Benjamin H Mullish
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, St Mary's Hospital Campus, Imperial College London, London, UK; Departments of Gastroenterology and Hepatology, St Mary's Hospital, Imperial College Healthcare NHS Trust, London, UK
| | | | - Siew C Ng
- Microbiota I-Center (MagIC), Hong Kong Special Administrative Region, China; Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Li Ka Shing Institute of Health Sciences, State Key Laboratory of Digestive Disease, Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Liping Zhao
- Department of Biochemistry and Microbiology, New Jersey Institute of Food, Nutrition and Health, Rutgers University, New Brunswick, NY, USA
| | - Richard Hansen
- Division of Molecular and Clinical Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - Paul W O'Toole
- APC Microbiome Ireland, Department of Medicine, University College Cork, Cork, Ireland; School of Microbiology, University College Cork, Cork, Ireland
| | - Jeroen Raes
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Leuven, Belgium; VIB, Center for Microbiology, Leuven, Belgium
| | - Georgina Hold
- Microbiome Research Centre, University of New South Wales, Sydney, Australia
| | - Lorenza Putignani
- Unit of Microbiomics and Unit of Human Microbiome, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Christian Lodberg Hvas
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus Denmark
| | - Georg Zeller
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center, Leiden, Netherlands; Center for Microbiome Analyses and Therapeutics, Leiden University Medical Center, Leiden, Netherlands; Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Omry Koren
- Azrieli Faculty of Medicine Bar-Ilan University, Safed, Israel
| | - Hein Tun
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Mireia Valles-Colomer
- Department CIBIO, University of Trento, Trento, Italy; MELIS Department, Pompeu Fabra University, Barcelona, Spain
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Valencia, Spain
| | - Monika Fischer
- Division of Gastroenterology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jessica Allegretti
- Division of Gastroenterology, Brigham and Women's Hospital, Boston, MA, USA
| | - Tariq Iqbal
- Department of Gastroenterology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK; Microbiome Treatment Centre, University of Birmingham, Edgbaston, UK
| | - Benoit Chassaing
- Microbiome-Host Interactions, Institut Pasteur, Université Paris Cité, INSERM, Paris, France
| | - Josbert Keller
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, Netherlands; Public Health Laboratory, Faculty of Medicine, University of Birmingham, Birmingham, UK
| | - Simon Mark Baunwall
- Department of Hepatology and Gastroenterology, Aarhus University Hospital, Aarhus Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Maria Abreu
- Division of Gastroenterology, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Giovanni Barbara
- IRCCS Azienda Ospedaliero, University of Bologna, Bologna, Italy; Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Faming Zhang
- Medical Center for Digestive Diseases, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China; Key Lab of Holistic Integrative Enterology, Nanjing Medical University, Nanjing, China
| | - Francesca Romana Ponziani
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Liver Unit-Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Sam P Costello
- Department of Gastroenterology, The Queen Elizabeth Hospital, Adelaide, South Australia, Australia; Faculty of Health and Medical Sciences, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia
| | - Sudarshan Paramsothy
- Department of Gastroenterology and Hepatology, Concord Repatriation General Hospital, Sydney, Australia; Concord Clinical School, University of Sydney, Sydney, Australia
| | - Dina Kao
- Edmonton FMT program, Division of Gastroenterology, University of Alberta, Edmonton, AB, Canada
| | - Colleen Kelly
- Division of Gastroenterology, Brigham and Women's Hospital, Boston, MA, USA
| | - Juozas Kupcinskas
- Gastroenterology Department and Institute for Digestive Research; Lithuanian University of Health Sciences Kaunas, Lithuania
| | - Ilan Youngster
- Division of Pediatrics and the Center for Microbiome Research, Shamir Medical Center, Israel, Faculty of Medical & Health Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Francesco Franceschi
- Department of Emergency Medicine, Fondazione Policlinico Universitario, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Sahil Khanna
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Maria Vehreschild
- Goethe University Frankfurt, University Hospital Frankfurt, Department II of Internal Medicine, Infectious Diseases, Frankfurt am Main, Germany
| | - Alexander Link
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University, Magdeburg, Germany
| | - Flavio De Maio
- Department of Laboratory and Infectious Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Edoardo Pasolli
- University of Naples Federico II, Department of Agricultural Sciences, Portici, Italy
| | | | - Patrizia Brigidi
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Brunella Posteraro
- Department of Basic Biotechnological Sciences, Intensive and Perioperative Clinics, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Franco Scaldaferri
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Mirjana Rajilic Stojanovic
- Department for Biochemical Engineering and Biotechnology, University of Belgrade, Belgrade, Serbia; Faculty of Technology and Metallurgy, University of Belgrade, Belgrade, Serbia
| | | | - Peter Malfertheiner
- Department of Gastroenterology, Hepatology and Infectious Diseases, Otto-von-Guericke University, Magdeburg, Germany; LMU, University Clinic, Medical Department II, Munich, Germany
| | - Luca Masucci
- Department of Laboratory and Infectious Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Manimozhiyan Arumugam
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nadeem Kaakoush
- School of Biomedical Sciences, University of New South Wales, Sydney, Australia
| | - Eran Segal
- Computer Science and Applied Mathematics Department, Weizmann Institute of Science, Rehovot, Israel
| | - Jasmohan Bajaj
- Division of Gastroenterology, Hepatology, and Nutrition, Virginia Commonwealth University and Richmond VA Medical Center, Richmond, VA, USA
| | - Rupert Leong
- Department of Gastroenterology, Concord Repatriation General Hospital, Sydney, Australia; MQ Health, Macquarie University Hospital, Sydney, Australia
| | - John Cryan
- APC Microbiome Ireland, Department of Medicine, University College Cork, Cork, Ireland
| | - Rinse K Weersma
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, Netherlands
| | - Robert Knight
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, USA
| | | | - Fergus Shanahan
- APC Microbiome Ireland, Department of Medicine, University College Cork, Cork, Ireland
| | - Patrice D Cani
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition Research Group, UCLouvain, Université Catholique de Louvain, Brussels, Belgium; Institute of Experimental and Clinical Research (IREC), UCLouvain, Université Catholique de Louvain, Brussels, Belgium; Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WELBIO department, WEL Research Institute, Wavre, Belgium
| | - Eran Elinav
- Systems Immunology Department, Weizmann Institute of Science, Rehovot, Israel; Cancer-Microbiome Division, Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| | - Maurizio Sanguinetti
- Department of Laboratory and Infectious Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Willem M de Vos
- Laboratory of Microbiology, Wageningen University, Netherlands; Human Microbiome Research Program, University of Helsinki, Finland
| | - Emad El-Omar
- Microbiome Research Centre, St George & Sutherland Clinical Campuses, School of Clinical Medicine, University of New South Wales, Sydney, Australia
| | - Joel Dorè
- MR Micalis Institut, INRA, Paris-Saclay University, Jouy-En-Josas, France
| | - Julian Marchesi
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, St Mary's Hospital Campus, Imperial College London, London, UK
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Harry Sokol
- MR Micalis Institut, INRA, Paris-Saclay University, Jouy-En-Josas, France; Gastroenterology Department, Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint-Antoine Hospital, Paris, France; Paris Center for Microbiome MedICIsne (PaCeMM) FHU, Paris, France
| | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy; Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | - Giovanni Cammarota
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Antonio Gasbarrini
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Gianluca Ianiro
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| |
Collapse
|
12
|
Lin X, Yu Z, Liu Y, Li C, Hu H, Hu J, Liu M, Yang Q, Gu P, Li J, Nandakumar KS, Hu G, Zhang Q, Chen X, Ma H, Huang W, Wang G, Wang Y, Huang L, Wu W, Liu N, Zhang C, Liu X, Zheng L, Chen P. Gut-X axis. IMETA 2025; 4:e270. [PMID: 40027477 PMCID: PMC11865426 DOI: 10.1002/imt2.270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 03/05/2025]
Abstract
Recent advances in understanding the modulatory functions of gut and gut microbiota on human diseases facilitated our focused attention on the contribution of the gut to the pathophysiological alterations of many extraintestinal organs, including the liver, heart, brain, lungs, kidneys, bone, skin, reproductive, and endocrine systems. In this review, we applied the "gut-X axis" concept to describe the linkages between the gut and other organs and discussed the latest findings related to the "gut-X axis," including the underlying modulatory mechanisms and potential clinical intervention strategies.
Collapse
Affiliation(s)
- Xu Lin
- Department of Endocrinology and MetabolismShunde Hospital of Southern Medical University (The First People's Hospital of Shunde)Foshan City528308China
| | - Zuxiang Yu
- State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, The Institute of Cardiovascular Sciences and Institute of Systems BiomedicinePeking UniversityBeijing100191China
| | - Yang Liu
- State Key Laboratory of Reproductive Medicine and Offsprings Health, Center for Global HealthNanjing Medical UniversityNanjing211166China
| | - Changzhou Li
- Department of Plastic and Aesthetic Surgery, Nanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Hui Hu
- Department of Laboratory Medicine, Shanghai East HospitalTongji University School of MedicineShanghai200123China
| | - Jia‐Chun Hu
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia Medica, Chinese Academy of Medical Sciences/Peking Union Medical CollegeBeijing100050China
| | - Mian Liu
- Department of Obstetrics and Gynecology, Nanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Qin Yang
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Peng Gu
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Jiaxin Li
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Kutty Selva Nandakumar
- Department of Medical Biochemistry and BiophysicsKarolinska InstituteStockholm17177Sweden
| | - Gaofei Hu
- State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, The Institute of Cardiovascular Sciences and Institute of Systems BiomedicinePeking UniversityBeijing100191China
| | - Qi Zhang
- State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, The Institute of Cardiovascular Sciences and Institute of Systems BiomedicinePeking UniversityBeijing100191China
| | - Xinyu Chen
- State Key Laboratory of Reproductive Medicine and Offsprings Health, Center for Global HealthNanjing Medical UniversityNanjing211166China
| | - Huihui Ma
- State Key Laboratory of Reproductive Medicine and Offsprings Health, Center for Global HealthNanjing Medical UniversityNanjing211166China
| | - Wenye Huang
- State Key Laboratory of Reproductive Medicine and Offsprings Health, Center for Global HealthNanjing Medical UniversityNanjing211166China
| | - Gaofeng Wang
- Department of Plastic and Aesthetic Surgery, Nanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Yan Wang
- State Key Laboratory of Bioactive Substance and Function of Natural MedicinesInstitute of Materia Medica, Chinese Academy of Medical Sciences/Peking Union Medical CollegeBeijing100050China
| | - Liping Huang
- Department of Obstetrics and Gynecology, Nanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Wenjuan Wu
- Department of Laboratory Medicine, Shanghai East HospitalTongji University School of MedicineShanghai200123China
| | - Ning‐Ning Liu
- State Key Laboratory of Systems Medicine for Cancer, Center for Single‐Cell Omics, School of Public HealthShanghai Jiao Tong University School of MedicineShanghai200025China
| | - Chenhong Zhang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and BiotechnologyShanghai Jiao Tong UniversityShanghai200240China
| | - Xingyin Liu
- State Key Laboratory of Reproductive Medicine and Offsprings Health, Center for Global HealthNanjing Medical UniversityNanjing211166China
- School of MedicineSouthern University of Science and TechnologyShenzhenChina
| | - Leming Zheng
- State Key Laboratory of Vascular Homeostasis and Remodeling, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Health Science Center, The Institute of Cardiovascular Sciences and Institute of Systems BiomedicinePeking UniversityBeijing100191China
| | - Peng Chen
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
| |
Collapse
|
13
|
Novelle MG, Naranjo-Martínez B, López-Cánovas JL, Díaz-Ruiz A. Fecal microbiota transplantation, a tool to transfer healthy longevity. Ageing Res Rev 2025; 103:102585. [PMID: 39586550 DOI: 10.1016/j.arr.2024.102585] [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: 06/14/2024] [Revised: 10/13/2024] [Accepted: 11/12/2024] [Indexed: 11/27/2024]
Abstract
The complex gut microbiome influences host aging and plays an important role in the manifestation of age-related diseases. Restoring a healthy gut microbiome via Fecal Microbiota Transplantation (FMT) is receiving extensive consideration to therapeutically transfer healthy longevity. Herein, we comprehensively review the benefits of gut microbial rejuvenation - via FMT - to promote healthy aging, with few studies documenting life length properties. This review explores how preconditioning donors via standard - lifestyle and pharmacological - antiaging interventions reshape gut microbiome, with the resulting benefits being also FMT-transferable. Finally, we expose the current clinical uses of FMT in the context of aging therapy and address FMT challenges - regulatory landscape, protocol standardization, and health risks - that require refinement to effectively utilize microbiome interventions in the elderly.
Collapse
Affiliation(s)
- Marta G Novelle
- Department of Genetics, Physiology and Microbiology (Unity of Animal Physiology), Faculty of Biology, Complutense University of Madrid (UCM), Madrid, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Spain
| | - Beatriz Naranjo-Martínez
- Laboratory of Cellular and Molecular Gerontology, Precision Nutrition and Aging, Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM+CSIC, Madrid, Spain
| | - Juan L López-Cánovas
- Laboratory of Cellular and Molecular Gerontology, Precision Nutrition and Aging, Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM+CSIC, Madrid, Spain
| | - Alberto Díaz-Ruiz
- Laboratory of Cellular and Molecular Gerontology, Precision Nutrition and Aging, Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM+CSIC, Madrid, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Spain.
| |
Collapse
|
14
|
Pérez‐Accino J, Salavati M, Glendinning L, Salavati Schmitz S. Effect of a single rectal fecal microbiota transplantation on clinical severity and fecal microbial communities in dogs with chronic inflammatory enteropathy. J Vet Intern Med 2025; 39:e17264. [PMID: 39778887 PMCID: PMC11710856 DOI: 10.1111/jvim.17264] [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: 07/30/2024] [Accepted: 11/14/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Fecal microbiota transplantation (FMT) has been advocated as a treatment for chronic enteropathy (CE) in dogs. However, so far only short-term clinical effects have been reported whereas the effect on the microbiota remains unexplored. HYPOTHESIS/OBJECTIVES Assess if a single FMT enema can lead to clinical improvement in dogs with CE when accompanied by presumed favorable microbiota changes. The effect of glycerol as a cryopreservative when storing FMT preparations also was assessed. ANIMALS Seven dogs with CE that received FMTs from 2 healthy donor dogs. MATERIALS AND METHODS Six dogs received a single FMT, 1 dog received 3 consecutive FMTs. Canine chronic enteropathy clinical activity index (CCECAI) and fecal samples were obtained before (Day 0), and 7, 30 and 90 days after FMT. Samples were stored with and without 10% glycerol. Sequencing of microbiota (16S rRNA, Illumina) was performed and compared by accepted analysis pipelines. RESULTS Median CCECAI before FMT was 8 (range, 5-14), decreased to a median of 3 (range, 1-12) within 1 week and a median of 1 (range, 0-12) by Day 30 (P < .01), with an average duration of response of approximately 10 weeks. Significant variation in the donors' microbiota composition was observed across different donations. Recipient microbiota composition or diversity did not change over time. Glycerol addition was associated with a difference in microbiota composition (P ≤ .001). CONCLUSIONS AND CLINICAL IMPORTANCE A single FMT can be considered an appropriate treatment in dogs with CE, but consistent microbiota changes were not observed.
Collapse
Affiliation(s)
- Jorge Pérez‐Accino
- College of Medicine and Veterinary Medicine, The Royal (Dick) School of Veterinary Studies, Hospital for Small Animals, Easter Bush CampusUniversity of EdinburghMidlothianUK
- Present address:
Hospital CanisGironaSpain
| | - Mazdak Salavati
- South and West Faculty, Dairy Research Innovation CentreScotland's Rural CollegeDumfriesUK
| | - Laura Glendinning
- College of Medicine and Veterinary Medicine, The Roslin Institute, Genetics and Genomics Department, Easter Bush CampusUniversity of EdinburghMidlothianUK
| | - Silke Salavati Schmitz
- College of Medicine and Veterinary Medicine, The Royal (Dick) School of Veterinary Studies, Hospital for Small Animals, Easter Bush CampusUniversity of EdinburghMidlothianUK
| |
Collapse
|
15
|
Kamath S, Sokolenko E, Collins K, Chan NSL, Mills N, Clark SR, Marques FZ, Joyce P. IUPHAR themed review: The gut microbiome in schizophrenia. Pharmacol Res 2025; 211:107561. [PMID: 39732352 DOI: 10.1016/j.phrs.2024.107561] [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: 11/25/2024] [Revised: 12/11/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024]
Abstract
Gut microbial dysbiosis or altered gut microbial consortium, in schizophrenia suggests a pathogenic role through the gut-brain axis, influencing neuroinflammatory and neurotransmitter pathways critical to psychotic, affective, and cognitive symptoms. Paradoxically, conventional psychotropic interventions may exacerbate this dysbiosis, with antipsychotics, particularly olanzapine, demonstrating profound effects on microbial architecture through disruption of bacterial phyla ratios, diminished taxonomic diversity, and attenuated short-chain fatty acid synthesis. To address these challenges, novel therapeutic strategies targeting the gut microbiome, encompassing probiotic supplementation, prebiotic compounds, faecal microbiota transplantation, and rationalised co-pharmacotherapy, show promise in attenuating antipsychotic-induced metabolic disruptions while enhancing therapeutic efficacy. Harnessing such insights, precision medicine approaches promise to transform antipsychotic prescribing practices by identifying patients at risk of metabolic side effects based on their microbial profiles. This IUPHAR review collates the current literature landscape of the gut-brain axis and its intricate relationship with schizophrenia while advocating for integrating microbiome assessments and therapeutic management. Such a fundamental shift in proposing microbiome-informed psychotropic prescriptions to optimise therapeutic efficacy and reduce adverse metabolic impacts would align antipsychotic treatments with microbiome safety, prioritising 'gut-neutral' or gut-favourable drugs to safeguard long-term patient outcomes in schizophrenia therapy.
Collapse
Affiliation(s)
- Srinivas Kamath
- UniSA Clinical & Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Elysia Sokolenko
- Discipline of Anatomy and Pathology, School of Biomedicine, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Kate Collins
- UniSA Clinical & Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Nicole S L Chan
- UniSA Clinical & Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Natalie Mills
- Discipline of Psychiatry, Adelaide Medical School, University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Scott R Clark
- Discipline of Psychiatry, Adelaide Medical School, University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Francine Z Marques
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; Hypertension Research Laboratory, School of Biological Sciences and Victorian Heart Institute, Monash University, Melbourne, VIC, Australia
| | - Paul Joyce
- UniSA Clinical & Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia.
| |
Collapse
|
16
|
Bloom PP, Chung RT. The future of clinical trials of gut microbiome therapeutics in cirrhosis. JHEP Rep 2025; 7:101234. [PMID: 39717506 PMCID: PMC11663965 DOI: 10.1016/j.jhepr.2024.101234] [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: 05/07/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 12/25/2024] Open
Abstract
The last two decades have witnessed an explosion of microbiome research, including in hepatology, with studies demonstrating altered microbial composition in liver disease. More recently, efforts have been made to understand the association of microbiome features with clinical outcomes and to develop therapeutics targeting the microbiome. While microbiome therapeutics hold much promise, their unique features pose certain challenges for the design and conduct of clinical trials. Herein, we will briefly review indications for microbiome therapeutics in cirrhosis, currently available microbiome therapeutics, and the biological pathways targeted by these therapies. We will then focus on the best practices and important considerations for clinical trials of gut microbiome therapeutics in cirrhosis.
Collapse
Affiliation(s)
- Patricia P. Bloom
- University of Michigan, Division of Gastroenterology, Ann Arbor, MI, USA
| | - Raymond T. Chung
- Massachusetts General Hospital, Division of Gastroenterology, Boston, MA, USA
| |
Collapse
|
17
|
Cibulkova I, Rehorova V, Soukupova H, Waldauf P, Cahova M, Manak J, Matejovic M, Duska F. Allogenic faecal microbiota transplantation for antibiotic-associated diarrhoea in critically ill patients (FEBATRICE)-Study protocol for a multi-centre randomised controlled trial (phase II). PLoS One 2024; 19:e0310180. [PMID: 39729440 DOI: 10.1371/journal.pone.0310180] [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: 02/13/2024] [Accepted: 08/25/2024] [Indexed: 12/29/2024] Open
Abstract
BACKGROUND Exposure of critically ill patients to antibiotics lead to intestinal dysbiosis, which often manifests as antibiotic-associated diarrhoea. Faecal microbiota transplantation restores gut microbiota and may lead to faster resolution of diarrhoea. METHODS Into this prospective, multi-centre, randomized controlled trial we will enrol 36 critically ill patients with antibiotic-associated diarrhoea. We will exclude patients with ongoing sepsis, need of systemic antibiotics, or those after recent bowel surgery or any other reason that prevents the FMT. Randomisation will be in 1:1 ratio. Patients in the control group will receive standard treatment based on oral diosmectite. In the intervention group, patients will receive, in addition to the standard of care, faecal microbiota transplantation via rectal tube, in the form of a preparation mixed from 7 thawed aliquots (50 mL) made from fresh stool of 7 healthy unrelated donors and quarantined deep frozen for 3 to 12 months. Primary outcome is treatment failure defined as intervention not delivered or diarrhoea persisting at day 7 after randomisation. Secondary outcomes include safety measures such as systemic inflammatory response, adverse events, and also diarrhoea recurrence within 28 days. Exploratory outcomes focus on gut barrier function and composition of intestinal microbiota. DISCUSSION Faecal microbiota transplantation has been effective for dysbiosis in non-critically ill patients with recurrent C. difficile infections and it is plausible to hypothesize that it will be equally effective for symptoms of dysbiosis in the critically ill patients. In addition, animal experiments and observational data suggest other benefits such as reduced colonization with multi-drug resistant bacteria and improved gut barrier and immune function. The frozen faeces from unrelated donors are immediately available when needed, unlike those from the relatives, who require lengthy investigation. Using multiple donors maximises graft microbiota diversity. Nonetheless, in vulnerable critically ill patients, Faecal microbiota transplantation might lead to bacterial translocation and unforeseen complications. From growing number of case series it is clear that its off label use in the critically ill patients is increasing and that there is a burning need to objectively assess its efficacy and safety, which this trial aims. TRIAL REGISTRATION www.clinicaltrials.gov (NCT05430269).
Collapse
Affiliation(s)
- Ivana Cibulkova
- Division of Gastroenterology, Department of Internal Medicine, Kralovske Vinohrady University Hospital, Prague, Czech Republic
- The Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Veronika Rehorova
- The Third Faculty of Medicine, Charles University, Prague, Czech Republic
- Department of Anaesthesia and Intensive Care Medicine, Kralovske Vinohrady University Hospital, Prague, Czech Republic
| | - Hana Soukupova
- The Third Faculty of Medicine, Charles University, Prague, Czech Republic
- Department of Microbiology, Kralovske Vinohrady University Hospital, Prague, Czech Republic
| | - Petr Waldauf
- The Third Faculty of Medicine, Charles University, Prague, Czech Republic
- Department of Anaesthesia and Intensive Care Medicine, Kralovske Vinohrady University Hospital, Prague, Czech Republic
| | - Monika Cahova
- Department of Experimental Medicine, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Jan Manak
- 3rd Department of Internal Medicine-Metabolism and Gerontology, Charles University Teaching Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Martin Matejovic
- 1st Department of Internal Medicine, Faculty of Medicine in Pilsen, Pilsen University Hospital, Pilsen, Czech Republic
| | - Frantisek Duska
- The Third Faculty of Medicine, Charles University, Prague, Czech Republic
- Department of Anaesthesia and Intensive Care Medicine, Kralovske Vinohrady University Hospital, Prague, Czech Republic
| |
Collapse
|
18
|
Bevilacqua A, Speranza B, Racioppo A, Santillo A, Albenzio M, Derossi A, Caporizzi R, Francavilla M, Racca D, Flagella Z, De Santis MA, Elia A, Conversa G, Luchetti L, Sinigaglia M, Corbo MR. Ultra-Processed Food and Gut Microbiota: Do Additives Affect Eubiosis? A Narrative Review. Nutrients 2024; 17:2. [PMID: 39796436 PMCID: PMC11723215 DOI: 10.3390/nu17010002] [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: 11/18/2024] [Revised: 12/19/2024] [Accepted: 12/22/2024] [Indexed: 01/13/2025] Open
Abstract
The gut microbiota plays a key role in health and disease, but it could be affected by various factors (diet, lifestyle, environment, genetics, etc.). Focusing on diet, while the role of the different styles and choices (Mediterranean vs. Western diet, vegan or vegetarian diets) has been extensively studied, there are a few comprehensive papers on the effects of additives and food processing. Therefore, the main goal of this manuscript is to propose an overview of the link between ultra-processed foods and the gut microbiota based on papers and data available in the literature. The literature search was performed on PubMed and Clinicaltrials.gov, and after the selection of the most relevant articles, the paper proposes a synopsis of the effects of some classes of additives (sweeteners, preservatives, emulsifiers, glutamate, etc.), as well as of some treatments, on the gut microbiota and some pathological conditions.
Collapse
Affiliation(s)
- Antonio Bevilacqua
- Department of Agriculture, Food, Natural Resources and Engineering (DAFNE), University of Foggia, 71122 Foggia, Italy; (B.S.); (A.R.); (A.S.); (M.A.); (A.D.); (R.C.); (M.F.); (D.R.); (Z.F.); (M.A.D.S.); (A.E.); (G.C.); (L.L.); (M.S.); (M.R.C.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Chasov V, Gilyazova E, Ganeeva I, Zmievskaya E, Davletshin D, Valiullina A, Bulatov E. Gut Microbiota Modulation: A Novel Strategy for Rheumatoid Arthritis Therapy. Biomolecules 2024; 14:1653. [PMID: 39766360 PMCID: PMC11674688 DOI: 10.3390/biom14121653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 12/14/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that leads to joint inflammation, progressive tissue damage and significant disability, severely impacting patients' quality of life. While the exact mechanisms underlying RA remain elusive, growing evidence suggests a strong link between intestinal microbiota dysbiosis and the disease's development and progression. Differences in microbial composition between healthy individuals and RA patients point to the role of gut microbiota in modulating immune responses and promoting inflammation. Therapies targeting microbiota restoration have demonstrated promise in improving treatment efficacy, enhancing patient outcomes and slowing disease progression. However, the complex interplay between gut microbiota and autoimmune pathways in RA requires further investigation to establish causative relationships and mechanisms. Here, we review the current understanding of the gut microbiota's role in RA pathogenesis and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Vitaly Chasov
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia (I.G.)
| | - Elvina Gilyazova
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia (I.G.)
| | - Irina Ganeeva
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia (I.G.)
| | - Ekaterina Zmievskaya
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia (I.G.)
| | - Damir Davletshin
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia (I.G.)
| | - Aygul Valiullina
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia (I.G.)
| | - Emil Bulatov
- Laboratory of Biomedical Technologies, Institute of Fundamental Medicine and Biology, Kazan Federal University, 18 Kremlyovskaya Street, Kazan 420008, Russia (I.G.)
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117997, Russia
| |
Collapse
|
20
|
Li Y, Xiao P, Ding H, Wang H, Xu Q, Wang R, Zheng L, Song X, Wang Y, Zhang T. Fecal Microbiota Transplantation in Children with Autism. Neuropsychiatr Dis Treat 2024; 20:2391-2400. [PMID: 39677507 PMCID: PMC11646457 DOI: 10.2147/ndt.s488001] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 12/01/2024] [Indexed: 12/17/2024] Open
Abstract
Purpose This research aimed to explore the clinical efficacy of fecal microbiota transplantation (FMT) in treating children with autism spectrum disorder (ASD). Methods In this single-arm prospective study, every participant received FMT therapy, followed by an 8-week follow-up. Children unable to swallow lyophilized capsules (Caps) received fecal solution through transendoscopic enteral tube (TET) or nasal jejunal tube (NJT) approaches. All participants underwent assessments of ASD core symptoms, gastrointestinal (GI) symptoms and sleep status initially, after treatment and during follow-up. The study outcomes included the changes in scores of the Autism Behavior Checklist (ABC), Childhood Autism Rating Scale (CARS), Social Responsiveness Scale (SRS), Gastrointestinal Symptoms Rating Scale (GSRS) and Sleep Disturbance Scale for Children (SDSC), as well as the adverse events (AEs). Results 98 participants were involved, consisting of 80 males and 18 females, with a median age of 7 years. 73 children received the FMT in the form of Caps, while 13 patients underwent the procedure through TET and 12 patients via NJT. Improvements were observed in all outcome measures for Caps and NJT groups at both the post-treatment and 8-week follow-up evaluations. Adjusted between-group analyses at post-treatment and follow-up showed that Caps and NJT group had greater reduction in ABC, CARS and SRS scores compared with TET group, while NJT group had greater reduction in SDSC scores compared with Caps and TET group. The incidence of AEs was 8.2% in the Caps group, 23.1% in the TET group, and 8.3% in the NJT group, with no serious AEs reported. Conclusion FMT treatment can improve the core symptoms, GI symptoms and sleep disturbances in children with ASD. The upper GI tract routes, including Caps and NJT, may be more effective and safe compared to the lower GI tract route of TET.
Collapse
Affiliation(s)
- Youran Li
- Department of Gastroenterology, Hepatology and Nutrition, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Pei Xiao
- Department of Gastroenterology, Hepatology and Nutrition, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Hongfang Ding
- Department of Pediatrics, Shengli Oilfield Central Hospital, Dongying, Shandong, People’s Republic of China
| | - Hui Wang
- Department of Pediatrics, Shengli Oilfield Central Hospital, Dongying, Shandong, People’s Republic of China
| | - Qiao Xu
- Department of Gastroenterology, Hepatology and Nutrition, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Ruixue Wang
- Department of Gastroenterology, Hepatology and Nutrition, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Lu Zheng
- Department of Gastroenterology, Hepatology and Nutrition, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Xue Song
- Department of Gastroenterology, Hepatology and Nutrition, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Yizhong Wang
- Department of Gastroenterology, Hepatology and Nutrition, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Ting Zhang
- Department of Gastroenterology, Hepatology and Nutrition, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
- Institute of Pediatric Infection, Immunity and Critical Care Medicine, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| |
Collapse
|
21
|
Mullish BH, Thursz MR. Alcohol-associated liver disease: Emerging therapeutic strategies. Hepatology 2024; 80:1372-1389. [PMID: 38922808 DOI: 10.1097/hep.0000000000000986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024]
Abstract
The large and growing burden of alcohol-associated liver disease-and the considerable burden of morbidity and mortality associated with it-has been a drive toward ongoing research into novel strategies for its treatment, with a particular focus upon alcohol-associated hepatitis (AH). Management of alcohol-use disorder forms the central pillar of alcohol-associated liver disease care, with evidence-based psychological and pharmacological approaches being well established, and certain models demonstrating improved clinical outcomes when hepatology and addiction services are co-located. Corticosteroids have previously been used somewhat indiscriminately in patients with severe AH, but effective tools now exist to assess early response (and limit futile ongoing exposure). Techniques to predict risk of corticosteroid-related infection are also available, although current clinical strategies to mitigate this risk are limited. A variety of novel therapeutic approaches to AH are at different phases of trials and evidence gathering, with some of the most promising signals related to cytokine manipulation, epigenetic modulation, and targeting of the gut microbiota (ie, by means of fecal microbiota transplant). While remaining an ongoing source of debate, early liver transplant in severe AH has grown in interest and acceptability over the past decade as evidence supporting its efficacy builds, in the process challenging paradigms about mandatory pretransplant sobriety periods. However, uncertainty remains regarding the optimal selection criteria, and whether liver transplant has a role for only a highly limited proportion of patients with AH or more widespread application. This review aims to provide an overview of this fast-moving field.
Collapse
Affiliation(s)
- Benjamin H Mullish
- Section of Hepatology and Gastroenterology, Department of Metabolism, Digestion and Reproduction, Division of Digestive Diseases, Faculty of Medicine, Imperial College London, London, United Kingdom
- Department of Hepatology, St Mary's Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Mark R Thursz
- Section of Hepatology and Gastroenterology, Department of Metabolism, Digestion and Reproduction, Division of Digestive Diseases, Faculty of Medicine, Imperial College London, London, United Kingdom
- Department of Hepatology, St Mary's Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom
| |
Collapse
|
22
|
Liu YH, Chen J, Chen X, Liu H. Factors of faecal microbiota transplantation applied to cancer management. J Drug Target 2024; 32:101-114. [PMID: 38174845 DOI: 10.1080/1061186x.2023.2299724] [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: 06/28/2023] [Accepted: 09/25/2023] [Indexed: 01/05/2024]
Abstract
The homeostasis of the microbiota is essential for human health. In particular, the gut microbiota plays a critical role in the regulation of the immune system. Thus, faecal microbiota transplantation (FMT), a technology that has rapidly developed in the last decade, has specifically been utilised for the treatment of intestinal inflammation and has recently been found to be able to treat tumours in combination with immunotherapy. FMT has become a breakthrough in enhancing the response rate to immunotherapy in cancer patients by altering the composition of the patient's gut microbiota. This review discusses the mechanisms of faecal microorganism effects on tumour development, drug treatment efficacy, and adverse effects and describes the recent clinical research trials on FMT. Moreover, the factors influencing the efficacy and safety of FMT are described. We summarise the possibilities of faecal transplantation in the treatment of tumours and its complications and propose directions to explore the development of FMT.
Collapse
Affiliation(s)
- Yi-Huang Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, China
- Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, Hunan, China
- Research Center of Molecular Metabolomics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Juan Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, China
- Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, Hunan, China
- Research Center of Molecular Metabolomics, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, China
- Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, Hunan, China
- Research Center of Molecular Metabolomics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hong Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Changsha, Hunan, China
- Hunan Engineering Research Center of Skin Health and Disease, Changsha, Hunan, China
- Xiangya Clinical Research Center for Cancer Immunotherapy, Central South University, Changsha, Hunan, China
- Research Center of Molecular Metabolomics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
23
|
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
|
24
|
Finnegan YE, Neill HR, Prpa EJ, Pot B. "Gut" to grips with the science of the microbiome - a symposium report. GUT MICROBIOME (CAMBRIDGE, ENGLAND) 2024; 5:e11. [PMID: 39703540 PMCID: PMC11658944 DOI: 10.1017/gmb.2024.8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 06/18/2024] [Indexed: 12/21/2024]
Abstract
The latest Yakult Science Study Day was held virtually on 2 November 2023. Aimed at healthcare professionals, researchers, and students, a variety of experts explored the latest gut microbiome research and what it means in practice. The morning sessions discussed the role of the microbiome in health and disease, the rapid advancements in DNA sequencing and implications for personalised nutrition, the current state of evidence on health benefits associated with fermented foods, prebiotics and probiotics and the challenges involved in interpreting research in this area. The afternoon session considered the emerging research on the microbiota-gut-brain axis in mediating effects of food on mood, the bidirectional impact of menopause on the gut microbiota, and the interplay between the gut and skin with implications for the treatment of rare and common skin disorders. The session ended with an update on the use of faecal microbiota transplant in both research and clinical practice. Undoubtedly, the gut microbiome is emerging as a key conductor of human health, both in relation to gastrointestinal and non-gastrointestinal outcomes. As research continues to elucidate mechanisms of action and confirm their effects in human trials, the gut microbiome should be a key consideration within a holistic approach to health moving forward.
Collapse
Affiliation(s)
- Yvonne E. Finnegan
- Yvonne Finnegan FINNE Nutrition & Regulatory Consultancy, Kilkenny, Ireland
| | | | | | - Bruno Pot
- Yakult Europe BV, Science Department, Almere, The Netherlands
| |
Collapse
|
25
|
Morales C, Ballestero L, Del Río P, Barbero-Herranz R, Olavarrieta L, Gómez-Artíguez L, Galeano J, Avendaño-Ortiz J, Basterra J, Del Campo R. Should the Faecal Microbiota Composition Be Determined to Certify a Faecal Donor? Diagnostics (Basel) 2024; 14:2635. [PMID: 39682542 DOI: 10.3390/diagnostics14232635] [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: 10/20/2024] [Revised: 11/10/2024] [Accepted: 11/15/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES Faecal microbiota transplantation (FMT) is considered a safe and effective therapy for recurrent Clostridioides difficile infection. It is the only current clinical indication for this technique, although numerous clinical research studies and trials propose its potential usefulness for treating other pathologies. Donor selection is a very rigorous process, based on a personal lifestyle interview and the absence of known pathogens in faeces and serum, leading to only a few volunteers finally achieving the corresponding certification. However, despite the high amount of data generated from the ongoing research studies relating microbiota and health, there is not yet a consensus defining what is a "healthy" microbiota. To date, knowledge of the composition of the microbiota is not a requirement to be a faecal donor. The aim of this work was to evaluate whether the analysis of the composition of the microbiota by massive sequencing of 16S rDNA could be useful in the selection of the faecal donors. METHODS Samples from 10 certified donors from Mikrobiomik Healthcare Company were collected and sequenced using 16S rDNA in a MiSeq (Illumina) platform. Alpha (Chao1 and Shannon indices) and beta diversity (Bray-Curtis) were performed using the bioinformatic web server Microbiome Analyst. The differences in microbial composition at the genera and phyla levels among the donors were evaluated. RESULTS The microbial diversity metric by alpha diversity indexes showed that most donors exhibited a similar microbial diversity and richness, whereas beta diversity by 16S rDNA sequencing revealed significant inter-donor differences, with a more stable microbial composition over time in some donors. The phyla Bacillota and Bacteroidota were predominant in all donors, while the density of other phyla, such as Actinomycota and Pseudomonota, varied among individuals. Each donor exhibited a characteristic genera distribution pattern; however, it was possible to define a microbiome core consisting of the genera Agathobacter, Eubacterium, Bacteroides, Clostridia UCG-014 and Akkermansia. Conclusions: The results suggest that donor certification does not need to rely exclusively on their microbiota composition, as it is unique to each donor. While one donor showed greater microbial diversity and richness, clear criteria for microbial normality and health have yet to be established. Therefore, donor certification should focus more on clinical and lifestyle aspects.
Collapse
Affiliation(s)
- Celia Morales
- Mikrobiomik Healthcare Company, 48160 Vizcaya, Spain
| | - Luna Ballestero
- Servicio de Microbiología, Instituto Ramón y Cajal de Investigaciones Sanitarias (IRYCIS), Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | | | - Raquel Barbero-Herranz
- Servicio de Microbiología, Instituto Ramón y Cajal de Investigaciones Sanitarias (IRYCIS), Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Leticia Olavarrieta
- Unidad Central de Apoyo (UCA-GT), Instituto Ramón y Cajal de Investigaciones Sanitarias (IRYCIS), Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | | | - Javier Galeano
- Grupo de Sistemas Complejos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - José Avendaño-Ortiz
- Servicio de Microbiología, Instituto Ramón y Cajal de Investigaciones Sanitarias (IRYCIS), Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
- Ciber en Enfermedades Infecciosas CIBERINFEC, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Juan Basterra
- Mikrobiomik Healthcare Company, 48160 Vizcaya, Spain
| | - Rosa Del Campo
- Servicio de Microbiología, Instituto Ramón y Cajal de Investigaciones Sanitarias (IRYCIS), Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
- Ciber en Enfermedades Infecciosas CIBERINFEC, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Facultad de Ciencias de la Salud, Universidad Alfonso X El Sabio, 28691 Villanueva de la Cañada, Spain
| |
Collapse
|
26
|
Kousgaard SJ, Cold F, Halkjær SI, Petersen AM, Kjeldsen J, Hansen JM, Dall SM, Albertsen M, Nielsen HL, Kirk KF, Duch K, Sønderkær M, Thorlacius-Ussing O. The Effect of Non-pooled Multidonor Faecal Microbiota Transplantation for Inducing Clinical Remission in Patients with Chronic Pouchitis: Results from a Multicentre, Randomised, Double-blinded, Placebo-controlled Trial [MicroPouch]. J Crohns Colitis 2024; 18:1753-1766. [PMID: 38708959 DOI: 10.1093/ecco-jcc/jjae066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/10/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
BACKGROUND AND AIMS To investigate if treatment with non-pooled, multidonor faecal microbiota transplantation [FMT] for 4 weeks was superior to placebo to induce clinical remission in patients with chronic pouchitis. METHODS The study was a randomised, double-blinded, placebo-controlled study with a 4-week intervention period and 12-month follow-up. Eligible patients with chronic pouchitis were recruited from five Danish hospitals. Participants were randomised to non-pooled, multidonor FMT derived from four faecal donors, or placebo. Treatment was delivered daily by enema for 2 weeks, followed by every second day for 2 weeks. Disease severity was accessed at inclusion and 30-day follow-up, using the Pouchitis Disease Activity Index [PDAI]; PDAI <7 was considered equivalent to clinical remission. Faecal samples from participants and donors were analysed by shotgun metagenomic sequencing. RESULTS Inclusion was stopped after inclusion of 30 participants who were randomised 1:1 for treatment with FMT or placebo. There was no difference in participants achieving clinical remission between the two groups at 30-day follow-up, relative risk 1.0 (95% CI [0.55; 1.81]). Treatment with FMT resulted in a clinically relevant increase in adverse events compared with placebo, incidence rate ratio 1.67 (95% CI [1.10; 2.52]); no serious adverse events within either group. Faecal microbiota transplantation statistically significantly increased the similarity of participant faecal microbiome to the faecal donor microbiome at 30-day follow-up [p = 0.01], which was not seen after placebo. CONCLUSIONS Non-pooled, multidonor FMT was comparable to placebo in inducing clinical remission in patients with chronic pouchitis, but showed a clinically relevant increase in adverse events compared with placebo. ClincialTrials.gov number, NCT04100291.
Collapse
Affiliation(s)
- Sabrina Just Kousgaard
- Department of Gastrointestinal Surgery, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Frederik Cold
- Gastrounit, Medical Division, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Sofie Ingdam Halkjær
- Gastrounit, Medical Division, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Andreas Munk Petersen
- Gastrounit, Medical Division, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
- Department of Clinical Microbiology, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Jens Kjeldsen
- Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | - Jane Møller Hansen
- Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | | | - Mads Albertsen
- Center for Microbial Communities, Aalborg University, Aalborg, Denmark
| | - Hans Linde Nielsen
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
- Department of Clinical Microbiology, Aalborg University Hospital, Aalborg, Denmark
| | - Karina Frahm Kirk
- Department of Infectious Disease, Aalborg University Hospital, Aalborg, Denmark
| | - Kirsten Duch
- Research Data and Biostatistics, Aalborg University Hospital, Aalborg, Denmark
| | - Mads Sønderkær
- Department of Molecular Diagnostics, Aalborg University Hospital, Aalborg, Denmark
| | - Ole Thorlacius-Ussing
- Department of Gastrointestinal Surgery, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
27
|
Han YJ, Kim S, Shin H, Kim HW, Park JD. Protective effect of gut microbiota restored by fecal microbiota transplantation in a sepsis model in juvenile mice. Front Immunol 2024; 15:1451356. [PMID: 39502702 PMCID: PMC11534669 DOI: 10.3389/fimmu.2024.1451356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 09/23/2024] [Indexed: 11/08/2024] Open
Abstract
Introduction Restoring a balanced, healthy gut microbiota through fecal microbiota transplantation (FMT) has the potential to be a treatment option for sepsis, despite the current lack of evidence. This study aimed to investigate the effect of FMT on sepsis in relation to the gut microbiota through a sepsis model in juvenile mice. Methods Three-week-old male mice were divided into three groups: the antibiotic treatment (ABX), ABX-FMT, and control groups. The ABX and ABX-FMT groups received antibiotics for seven days. FMT was performed through oral gavage in the ABX-FMT group over the subsequent seven days. On day 14, all mice underwent cecal ligation and puncture (CLP) to induce abdominal sepsis. Blood cytokine levels and the composition of fecal microbiota were analyzed, and survival was monitored for seven days post-CLP. Results Initially, the fecal microbiota was predominantly composed of the phyla Bacteroidetes and Firmicutes. After antibiotic intake, an extreme predominance of the class Bacilli emerged. FMT successfully restored antibiotic-induced fecal dysbiosis. After CLP, the phylum Bacteroidetes became extremely dominant in the ABX-FMT and control groups. Alpha diversity of the microbiota decreased after antibiotic intake, was restored after FMT, and decreased again following CLP. In the ABX group, the concentrations of interleukin-1β (IL-1β), IL-2, IL-6, IL-10, granulocyte macrophage colony-stimulating factor, tumor necrosis factor-α, and C-X-C motif chemokine ligand 1 increased more rapidly and to a higher degree compared to other groups. The survival rate in the ABX group was significantly lower (20.0%) compared to other groups (85.7%). Conclusion FMT-induced microbiota restoration demonstrated a protective effect against sepsis. This study uniquely validates the effectiveness of FMT in a juvenile mouse sepsis model, offering potential implications for clinical research in critically ill children.
Collapse
Affiliation(s)
- Young Joo Han
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - SungSu Kim
- Laboratory Animal Experiment Center, Bionsystems, Uiwang-si, Gyeonggi-do, Republic of Korea
| | - Haksup Shin
- Wide River Institute of Immunology, Seoul National University, Hongcheon-Gun, Gangwon-do, Republic of Korea
| | - Hyun Woo Kim
- Bio Convergence Team, Gangwon Techno Park Technology Innovation Support Center, Chuncheon-si, Gangwon-do, Republic of Korea
| | - June Dong Park
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
28
|
Bénard MV, de Goffau MC, Blonk J, Hugenholtz F, van Buuren J, Paramsothy S, Kaakoush NO, D'Haens GRAM, Borody TJ, Kamm MA, Ponsioen CY. Gut Microbiota Features in Relation to Fecal Microbiota Transplantation Outcome in Ulcerative Colitis: A Systematic Review and Meta-Analysis. Clin Gastroenterol Hepatol 2024:S1542-3565(24)00907-8. [PMID: 39442743 DOI: 10.1016/j.cgh.2024.10.001] [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: 07/03/2024] [Revised: 10/10/2024] [Accepted: 10/13/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND & AIMS Fecal microbiota transplantation (FMT) can induce remission in patients with ulcerative colitis, yet its efficacy needs improvement. We conducted a comprehensive evaluation of the current literature on microbial factors affecting outcome, as well as a meta-analysis on some of the largest datasets regarding composition. METHODS MEDLINE, Embase, and Cochrane were systematically searched through August 2024 for relevant studies. The quality of studies was analyzed with JBI tools and a composite critical appraisal score. Additionally, species-level data from 2 landmark FMT trials (the Transplantation of Feces in Ulcerative Colitis; Returning Nature's Homeostasis [TURN] and Fecal Microbiota Transplantation for Chronic Active Ulcerative Colitis [FOCUS] trials) were reanalyzed from a compositional perspective. RESULTS Out of 3755 citations identified, 56 met the inclusion criteria, of which 29 fulfilled quality standards. Higher microbial α-diversity, either in donors or recipients (at baseline or following FMT treatment), was associated with better clinical response rates. Engraftment of the donors' microbiota could not be clearly linked with clinical response, possibly because not every donor has an ideal microbiome. Butyrate-producing species from the Lachnospiraceae and Oscillospiraceae families were often related with response, whereas the reverse was true for Fusobacteria, many Proteobacteria, and Ruminococcus gnavus. Compositional analyses showed that clinical response is associated with a shift from a low-diversity, often Bacteroides-dominant composition to one with higher diversity, either dominated by various butyrate producers, the Christensenellaceae-Methanobrevibacter trophic network, or a moderate/high-diversity composition with abundant but not excessive levels of Prevotella copri. CONCLUSIONS This systematic review/meta-analysis yielded a coherent picture from a compositional perspective, which may help identify beneficial donor profiles and guide personalized FMT approaches.
Collapse
Affiliation(s)
- Mèlanie V Bénard
- Department of Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Inflammatory Bowel Disease Centre, Amsterdam UMC, Amsterdam, the Netherlands
| | | | - Justine Blonk
- Department of Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Floor Hugenholtz
- Center for Experimental and Molecular Medicine, Amsterdam UMC, Amsterdam Medical Center, University of Amsterdam, Amsterdam, the Netherlands
| | - Joep van Buuren
- Department of Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Sudarshan Paramsothy
- Faculty of Medicine and Health, Concord Clinical School, University of Sydney, Sydney, New South Wales, Australia; Department of Gastroenterology, Concord Repatriation General Hospital, Concord, New South Wales, Australia; Department of Gastroenterology, Macquarie University Hospital, Sydney, New South Wales, Australia
| | - Nadeem O Kaakoush
- School of Biomedical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Geert R A M D'Haens
- Department of Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Inflammatory Bowel Disease Centre, Amsterdam UMC, Amsterdam, the Netherlands
| | - Thomas J Borody
- Centre for Digestive Diseases, Sydney, New South Wales, Australia
| | - Michael A Kamm
- Department of Gastroenterology, St Vincent's Hospital, Melbourne, Victoria, Australia; Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Cyriel Y Ponsioen
- Department of Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Inflammatory Bowel Disease Centre, Amsterdam UMC, Amsterdam, the Netherlands.
| |
Collapse
|
29
|
Arcay R, Barceló-Nicolau M, Suárez L, Martín L, Reigada R, Höring M, Liebisch G, Garrido C, Cabot G, Vílchez H, Cortés-Lara S, González de Herrero E, López-Causapé C, Oliver A, Barceló-Coblijn G, Mena A. Gut microbiome and plasma lipidome analysis reveals a specific impact of Clostridioides difficile infection on intestinal bacterial communities and sterol metabolism. mBio 2024; 15:e0134724. [PMID: 39189787 PMCID: PMC11481895 DOI: 10.1128/mbio.01347-24] [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: 05/08/2024] [Accepted: 07/15/2024] [Indexed: 08/28/2024] Open
Abstract
Clostridioides difficile infection (CDI) causes alterations in the intestinal microbiota, frequently associated with changes in the gut metabolism of bile acids and cholesterol. In addition to the impact on microbiome composition and given the metabolic changes occurring during CDI, our work focuses on the importance to know the effects at the local and systemic levels, both during the infection and its treatment, by paying particular attention to plasma lipid metabolism due to its relationship with CDI pathogenesis. Specific changes, characterized by a loss of microbial richness and diversity and related to a reduction in short-chain acid-producing bacteria and an increase in bile salt hydrolase-producing bacteria, were observed in the gut microbiota of CDI patients, especially in those suffering from recurrent CDI (RCDI). However, gut microbiota showed its ability to restore itself after treatment, resembling healthy individuals, in those patients treated by fecal microbiome transfer (FMT), in contrast with those treated with antibiotics, and displaying increased levels of Eubacterium coprostanoligenes, a cholesterol-reducing anaerobe. Interestingly, changes in plasma lipidome revealed a global depletion in circulating lipids in CDI, with the largest impact on cholesteryl esters. CDI patients also showed a specific and consistent decrease in the levels of lipid species containing linoleic acid-an essential fatty acid-which were only partially recovered after antibiotic treatment. Analysis of the plasma lipidome reflects CDI impact on the gut microbiota and its metabolism, evidencing changes in sterol and fatty acid metabolism that are possibly related to specific alterations observed in gut microbial communities of CDI patients. IMPORTANCE There is increasing evidence about the influence the changes in microbiota and its metabolism has on numerous diseases and infections such as Clostridioides difficile infection (CDI). The knowledge of these changes at local and systemic levels can help us manage this infection to avoid recurrences and apply the best therapies, such as fecal microbiota transfer (FMT). This study shows a better restoration of the gut in FMT-treated patients than in antibiotic-treated patients, resembling healthy controls and showing increased levels of cholesterol-reducing bacteria. Furthermore, it evidences the CDI impact on plasma lipidome. We observed in CDI patients a global depletion in circulating lipids, particularly cholesteryl esters, and a specific decrease in linoleic acid-containing lipids, an essential fatty acid. Our observations could impact CDI management because the lipid content was only partially recovered after treatment, suggesting that continued nutritional support, aiming to restore healthy lipid levels, could be essential for a full recovery.
Collapse
Affiliation(s)
- Ricardo Arcay
- Microbiology Department, Hospital Universitari Son Espases, Palma, Balearic Islands, Spain
- Institut d’Investigació Sanitària Illes Balears (IdISBa, Health Research Institute of the Balearic Islands), Palma, Balearic Islands, Spain
| | - Maria Barceló-Nicolau
- Institut d’Investigació Sanitària Illes Balears (IdISBa, Health Research Institute of the Balearic Islands), Palma, Balearic Islands, Spain
- Research Unit, University Hospital Son Espases, Palma, Balearic Islands, Spain
| | - Loreto Suárez
- Microbiology Department, Hospital Universitari Son Espases, Palma, Balearic Islands, Spain
- Institut d’Investigació Sanitària Illes Balears (IdISBa, Health Research Institute of the Balearic Islands), Palma, Balearic Islands, Spain
| | - Luisa Martín
- Institut d’Investigació Sanitària Illes Balears (IdISBa, Health Research Institute of the Balearic Islands), Palma, Balearic Islands, Spain
- Internal Medicine Department, Hospital Universitari Son Espases, Palma, Balearic Islands, Spain
| | - Rebeca Reigada
- Institut d’Investigació Sanitària Illes Balears (IdISBa, Health Research Institute of the Balearic Islands), Palma, Balearic Islands, Spain
- Research Unit, University Hospital Son Espases, Palma, Balearic Islands, Spain
| | - Marcus Höring
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Bavaria, Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Bavaria, Germany
| | - Carmen Garrido
- Gastroenterology Department, Hospital Universitari Son Espases, Palma, Balearic Islands, Spain
| | - Gabriel Cabot
- Institut d’Investigació Sanitària Illes Balears (IdISBa, Health Research Institute of the Balearic Islands), Palma, Balearic Islands, Spain
- Research Unit, University Hospital Son Espases, Palma, Balearic Islands, Spain
| | - Helem Vílchez
- Institut d’Investigació Sanitària Illes Balears (IdISBa, Health Research Institute of the Balearic Islands), Palma, Balearic Islands, Spain
- Internal Medicine Department, Hospital Universitari Son Espases, Palma, Balearic Islands, Spain
| | - Sara Cortés-Lara
- Institut d’Investigació Sanitària Illes Balears (IdISBa, Health Research Institute of the Balearic Islands), Palma, Balearic Islands, Spain
- Research Unit, University Hospital Son Espases, Palma, Balearic Islands, Spain
| | - Elisa González de Herrero
- Microbiology Department, Hospital Universitari Son Espases, Palma, Balearic Islands, Spain
- Institut d’Investigació Sanitària Illes Balears (IdISBa, Health Research Institute of the Balearic Islands), Palma, Balearic Islands, Spain
| | - Carla López-Causapé
- Microbiology Department, Hospital Universitari Son Espases, Palma, Balearic Islands, Spain
- Institut d’Investigació Sanitària Illes Balears (IdISBa, Health Research Institute of the Balearic Islands), Palma, Balearic Islands, Spain
- Research Unit, University Hospital Son Espases, Palma, Balearic Islands, Spain
| | - Antonio Oliver
- Microbiology Department, Hospital Universitari Son Espases, Palma, Balearic Islands, Spain
- Institut d’Investigació Sanitària Illes Balears (IdISBa, Health Research Institute of the Balearic Islands), Palma, Balearic Islands, Spain
- Research Unit, University Hospital Son Espases, Palma, Balearic Islands, Spain
| | - Gwendolyn Barceló-Coblijn
- Institut d’Investigació Sanitària Illes Balears (IdISBa, Health Research Institute of the Balearic Islands), Palma, Balearic Islands, Spain
- Research Unit, University Hospital Son Espases, Palma, Balearic Islands, Spain
| | - Ana Mena
- Microbiology Department, Hospital Universitari Son Espases, Palma, Balearic Islands, Spain
- Institut d’Investigació Sanitària Illes Balears (IdISBa, Health Research Institute of the Balearic Islands), Palma, Balearic Islands, Spain
| |
Collapse
|
30
|
Marsiglia R, Pane S, Del Chierico F, Russo A, Vernocchi P, Romani L, Cardile S, Diamanti A, Galli L, Tamborino A, Terlizzi V, De Angelis P, Angelino G, Putignani L. Fecal Microbiota Transplantation for Recurrent Clostridioides difficile Infections in a Cystic Fibrosis Child Previously Screen Positive, Inconclusive Diagnosis (CFSPID): A Case Report. Microorganisms 2024; 12:2059. [PMID: 39458368 PMCID: PMC11509880 DOI: 10.3390/microorganisms12102059] [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: 09/25/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Clostridioides difficile infection (CDI) is generally treated with vancomycin, metronidazole or fidaxomicin, although fecal microbiota transplantation (FMT) represents a promising therapeutic option for antibiotic-resistant recurrent C. difficile infections (rCDIs) in adults. In pediatric cystic fibrosis (CF) patients, CDIs are generally asymptomatic and respond to treatment. Here, we present the case of an 8-year-old female, initially diagnosed as "CFTR-related metabolic syndrome/cystic fibrosis screen positive, inconclusive diagnosis" (CMRS/CFSPID), who then progressed to CF at 12 months. In the absence of CF-related symptoms, she presented multiple and disabling episodes of bloody diarrhoea with positive tests for C. difficile antigen and A/B toxin. After conventional treatments failed and several CDI relapses, FMT was proposed. Donor screening and GM donor-receiver matching identified her mother as a donor. Metataxonomy and targeted metabolomics provided, through a pre- and post-FMT time course, gut microbiota (GM) profiling to assess GM engraftment. At first, the GM map revealed severe dysbiosis, with a prevalence of Bacteroidetes and Proteobacteria (i.e., Klebsiella spp., Escherichia coli), a reduction in Firmicutes, a GM nearly entirely composed of Enterococcaceae (i.e., Enterococcus) and an almost complete depletion of Verrucomicrobia and Actinobacteria, mostly represented by Veillonella dispar. Post FMT, an increment in Bifidobacterium spp. and Collinsella spp. with a decrease in V. dispar restored intestinal eubiosis. Consistently, four weeks after FMT treatment, the child's gut symptoms cleared, without CDI recurrence.
Collapse
Affiliation(s)
- Riccardo Marsiglia
- Immunology, Rheumatology and Infectious Diseases Research Area, Unit of Research Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (R.M.); (F.D.C.); (P.V.)
| | - Stefania Pane
- Unit of Microbiomics, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (S.P.); (A.R.)
| | - Federica Del Chierico
- Immunology, Rheumatology and Infectious Diseases Research Area, Unit of Research Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (R.M.); (F.D.C.); (P.V.)
| | - Alessandra Russo
- Unit of Microbiomics, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (S.P.); (A.R.)
| | - Pamela Vernocchi
- Immunology, Rheumatology and Infectious Diseases Research Area, Unit of Research Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy; (R.M.); (F.D.C.); (P.V.)
| | - Lorenza Romani
- Infectious Diseases Unit, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy;
| | - Sabrina Cardile
- Unit of Gastroenterology and Nutrition, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (S.C.); (A.D.); (P.D.A.); (G.A.)
| | - Antonella Diamanti
- Unit of Gastroenterology and Nutrition, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (S.C.); (A.D.); (P.D.A.); (G.A.)
| | - Luisa Galli
- Department of Health Sciences, University of Florence, 50121 Florence, Italy;
- Infectious Disease Unit, Meyer Children’s Hospital IRCCS, 50121 Florence, Italy;
| | - Agnese Tamborino
- Infectious Disease Unit, Meyer Children’s Hospital IRCCS, 50121 Florence, Italy;
| | - Vito Terlizzi
- Department of Pediatric Medicine, Meyer Children’s Hospital IRCCS, Cystic Fibrosis Regional Reference Center, Viale Gaetano Pieraccini 24, 50139 Florence, Italy;
| | - Paola De Angelis
- Unit of Gastroenterology and Nutrition, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (S.C.); (A.D.); (P.D.A.); (G.A.)
| | - Giulia Angelino
- Unit of Gastroenterology and Nutrition, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (S.C.); (A.D.); (P.D.A.); (G.A.)
| | - Lorenza Putignani
- Unit of Microbiomics and Unit of Research of Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy
| |
Collapse
|
31
|
Sintes R, McLellan P, Navelli G, Landman C, Delage S, Truong S, Benech N, Kapel N, Moreino Sabater A, Schnuriger A, Eckert C, Bleibtreu A, Joly AC, Sokol H. Use of frozen native feces for fecal microbiota transplantation in recurrent Clostridioides difficile infection: a simple way to improve the efficiency of donor feces preparation. Antimicrob Agents Chemother 2024; 68:e0073424. [PMID: 39166867 PMCID: PMC11459919 DOI: 10.1128/aac.00734-24] [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: 05/16/2024] [Accepted: 07/29/2024] [Indexed: 08/23/2024] Open
Abstract
Preparing fecal microbiota transplants immediately after donation is resource-intensive, and a proportion are destroyed following abnormal screening results. We retrospectively compared two processes, frozen fecal preparation (FFP) and fresh native frozen preparation (FNFP), for clinical efficacy in the treatment of recurrent Clostridioides difficile infection (rCDI). FFP and FNFP were similarly effective with clinical success rates of 76.7% and 86.7% (P = 0.32), respectively. FNFP is an efficient procedure that saves resources while maintaining clinical efficacy in rCDI.
Collapse
Affiliation(s)
- Rachel Sintes
- Department of Pharmacy, Sorbonne Université, AP-HP, Saint-Antoine Hospital, Paris, France
- Assistance Publique-Hopitaux de Paris (AP-HP) FMT Center, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
- French Group for Fecal Microbiota Transplantation (GFTF), Paris, France
| | - Paul McLellan
- Assistance Publique-Hopitaux de Paris (AP-HP) FMT Center, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
- French Group for Fecal Microbiota Transplantation (GFTF), Paris, France
- Department of Gastroenterology, Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint-Antoine Hospital, Paris, France
| | - Gabriele Navelli
- Department of Pharmacy, Sorbonne Université, AP-HP, Saint-Antoine Hospital, Paris, France
- Assistance Publique-Hopitaux de Paris (AP-HP) FMT Center, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
- French Group for Fecal Microbiota Transplantation (GFTF), Paris, France
| | - Cécilia Landman
- Assistance Publique-Hopitaux de Paris (AP-HP) FMT Center, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
- French Group for Fecal Microbiota Transplantation (GFTF), Paris, France
- Department of Gastroenterology, Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint-Antoine Hospital, Paris, France
| | - Sandrine Delage
- Department of Pharmacy, Sorbonne Université, AP-HP, Saint-Antoine Hospital, Paris, France
- Assistance Publique-Hopitaux de Paris (AP-HP) FMT Center, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
- French Group for Fecal Microbiota Transplantation (GFTF), Paris, France
| | - Sandrine Truong
- Assistance Publique-Hopitaux de Paris (AP-HP) FMT Center, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
- French Group for Fecal Microbiota Transplantation (GFTF), Paris, France
- Department of Gastroenterology, Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint-Antoine Hospital, Paris, France
| | - Nicolas Benech
- French Group for Fecal Microbiota Transplantation (GFTF), Paris, France
- Department of Hepato-Gastroenterology, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| | - Nathalie Kapel
- Assistance Publique-Hopitaux de Paris (AP-HP) FMT Center, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
- French Group for Fecal Microbiota Transplantation (GFTF), Paris, France
- Functional Coprology Laboratory, APHP, Pitié Salpêtrière Hospital and Université Paris Cité, INSERM UMRS-1139, Centre d'Immunologie et des Maladies Infectieuses, (CIMI-PARIS), Inserm U1135, Sorbonne Université, Paris, France
| | - Alicia Moreino Sabater
- Assistance Publique-Hopitaux de Paris (AP-HP) FMT Center, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
- French Group for Fecal Microbiota Transplantation (GFTF), Paris, France
- Sorbonne Université, APHP, National Institute of Health and Medical Research, Center for Immunology and Infectious Diseases-Paris, Saint Antoine Hospital, Parasitology-Mycology, Paris, France
| | - Aurélie Schnuriger
- Assistance Publique-Hopitaux de Paris (AP-HP) FMT Center, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
- French Group for Fecal Microbiota Transplantation (GFTF), Paris, France
- Sorbonne Université, AP-HP, Saint-Antoine Hospital, Virology, Paris, France
| | - Catherine Eckert
- Assistance Publique-Hopitaux de Paris (AP-HP) FMT Center, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
- French Group for Fecal Microbiota Transplantation (GFTF), Paris, France
- Sorbonne Université, AP-HP, Saint-Antoine Hospital, Bacteriology et Centre d'Immunologie et des Maladies Infectieuses, INSERM, U1135, Sorbonne Université, Paris, France
| | - Alexandre Bleibtreu
- Assistance Publique-Hopitaux de Paris (AP-HP) FMT Center, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
- French Group for Fecal Microbiota Transplantation (GFTF), Paris, France
- Department of Infectious Disease, Sorbonne Université, AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Anne-Christine Joly
- Department of Pharmacy, Sorbonne Université, AP-HP, Saint-Antoine Hospital, Paris, France
- Assistance Publique-Hopitaux de Paris (AP-HP) FMT Center, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
- French Group for Fecal Microbiota Transplantation (GFTF), Paris, France
| | - Harry Sokol
- Assistance Publique-Hopitaux de Paris (AP-HP) FMT Center, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
- French Group for Fecal Microbiota Transplantation (GFTF), Paris, France
- Department of Gastroenterology, Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint-Antoine Hospital, Paris, France
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| |
Collapse
|
32
|
Cibulková I, Řehořová V, Wilhelm M, Soukupová H, Hajer J, Duška F, Daňková H, Cahová M. Evaluating Bacterial Viability in Faecal Microbiota Transplantation: A Comparative Analysis of In Vitro Cultivation and Membrane Integrity Methods. J Clin Lab Anal 2024; 38:e25105. [PMID: 39360586 PMCID: PMC11520942 DOI: 10.1002/jcla.25105] [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: 07/02/2024] [Revised: 09/03/2024] [Accepted: 09/08/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Faecal microbiota transplantation (FMT) is a developing therapy for disorders related to gut dysbiosis. Despite its growing application, standardised protocols for FMT filtrate preparation and quality assessment remain undeveloped. The viability of bacteria in the filtrate is crucial for FMT's efficacy and for validating protocol execution. We compared two methods-in vitro cultivation and membrane integrity assessment-for their accuracy, reproducibility and clinical applicability in measuring bacterial viability in frozen FMT stool filtrate. METHODS Bacterial viability in stool filtrate was evaluated using (i) membrane integrity through fluorescent DNA staining with SYTO9 and propidium iodide, followed by flow cytometry and (ii) culturable bacteria counts (colony-forming units, CFU) under aerobic or anaerobic conditions. RESULTS Using different types of samples (pure bacterial culture, stool of germ-free and conventionally bred mice, native and heat-treated human stool), we refined the bacterial DNA staining protocol integrated with flow cytometry for assessment of bacterial viability in frozen human stool samples. Both the membrane integrity-based and cultivation-based methods exhibited significant variability in bacterial viability across different FMT filtrates, without correlation. The cultivation-based method showed a mean coefficient of variance of 30.3%, ranging from 7.4% to 60.1%. Conversely, the membrane integrity approach yielded more reproducible results, with a mean coefficient of variance for viable cells of 6.4% ranging from 0.2% to 18.2%. CONCLUSION Bacterial viability assessment in stool filtrate using the membrane integrity method offers robust and precise data, making it a suitable option for faecal material evaluation in FMT. In contrast, the cultivation-dependent methods produce inconsistent outcomes.
Collapse
Affiliation(s)
- Ivana Cibulková
- Division of Gastroenterology, Department of Internal MedicineKralovske Vinohrady University HospitalPragueCzech Republic
- The Third Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Veronika Řehořová
- The Third Faculty of MedicineCharles UniversityPragueCzech Republic
- Department of Anaesthesia and Intensive Care MedicineKralovske Vinohrady University HospitalPragueCzech Republic
| | - Marek Wilhelm
- The Third Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Hana Soukupová
- The Third Faculty of MedicineCharles UniversityPragueCzech Republic
- Department of MicrobiologyKralovske Vinohrady University HospitalPragueCzech Republic
| | - Jan Hajer
- Division of Gastroenterology, Department of Internal MedicineKralovske Vinohrady University HospitalPragueCzech Republic
- The Third Faculty of MedicineCharles UniversityPragueCzech Republic
| | - František Duška
- The Third Faculty of MedicineCharles UniversityPragueCzech Republic
- Department of Anaesthesia and Intensive Care MedicineKralovske Vinohrady University HospitalPragueCzech Republic
| | - Helena Daňková
- Department of Experimental MedicineInstitute for Clinical and Experimental MedicinePragueCzech Republic
| | - Monika Cahová
- Department of Experimental MedicineInstitute for Clinical and Experimental MedicinePragueCzech Republic
| |
Collapse
|
33
|
Ye ZN, Eslick GD, Huang SG, He XX. Faecal microbiota transplantation for eradicating Helicobacter pylori infection: clinical practice and theoretical postulation. EGASTROENTEROLOGY 2024; 2:e100099. [PMID: 39944265 PMCID: PMC11770466 DOI: 10.1136/egastro-2024-100099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025]
Abstract
The sustained increase in antibiotic resistance leads to a declining trend in the eradication rate of Helicobacter pylori (H. pylori) infection with antibiotic-based eradication regimens. Administration of a single probiotic shows limited efficacy in eradicating H. pylori infection. This review indicates that faecal microbiota transplantation (FMT), a novel therapeutic approach, either as a monotherapy or adjunctive therapy, exhibits beneficial effects in terms of the eradication of H. pylori infection and the prevention of adverse events. The role of FMT in H. pylori eradication may be associated directly or indirectly with some therapeutic constituents within the faecal suspension, including bacteria, viruses, antimicrobial peptides and metabolites. In addition, variations in donor selection, faecal suspension preparation and delivery methods are believed to be the main factors determining the effectiveness of FMT for the treatment of H. pylori infection. Future research should refine the operational procedures of FMT to achieve optimal efficacy for H. pylori infection and explore the mechanisms by which FMT acts against H. pylori.
Collapse
Affiliation(s)
- Zhi-Ning Ye
- The Affiliated Dongguan Hospital of Guangzhou University of Chinese Medicine, Dongguan, China
| | - Guy D Eslick
- The Australian Paediatric Surveillance Unit, The University of Sydney, The Children's Hospital, Sydney, New South Wale, Australia
| | - Shao-Gang Huang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xing-Xiang He
- The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
34
|
Karimi M, Shirsalimi N, Hashempour Z, Salehi Omran H, Sedighi E, Beigi F, Mortezazadeh M. Safety and efficacy of fecal microbiota transplantation (FMT) as a modern adjuvant therapy in various diseases and disorders: a comprehensive literature review. Front Immunol 2024; 15:1439176. [PMID: 39391303 PMCID: PMC11464302 DOI: 10.3389/fimmu.2024.1439176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 09/09/2024] [Indexed: 10/12/2024] Open
Abstract
The human gastrointestinal (GI) tract microbiome is a complex and all-encompassing ecological system of trillions of microorganisms. It plays a vital role in digestion, disease prevention, and overall health. When this delicate balance is disrupted, it can lead to various health issues. Fecal microbiota transplantation (FMT) is an emerging therapeutic intervention used as an adjuvant therapy for many diseases, particularly those with dysbiosis as their underlying cause. Its goal is to restore this balance by transferring fecal material from healthy donors to the recipients. FMT has an impressive reported cure rate between 80% and 90% and has become a favored treatment for many diseases. While FMT may have generally mild to moderate transient adverse effects, rare severe complications underscore the importance of rigorous donor screening and standardized administration. FMT has enormous potential as a practical therapeutic approach; however, additional research is required to further determine its potential for clinical utilization, as well as its safety and efficiency in different patient populations. This comprehensive literature review offers increased confidence in the safety and effectiveness of FMT for several diseases affecting the intestines and other systems, including diabetes, obesity, inflammatory and autoimmune illness, and other conditions.
Collapse
Affiliation(s)
- Mehdi Karimi
- Bogomolets National Medical University (NMU), Kyiv, Ukraine
| | - Niyousha Shirsalimi
- Faculty of Medicine, Hamadan University of Medical Science (UMSHA), Hamadan, Iran
| | - Zahra Hashempour
- School of Medicine, Shiraz University of Medical Sciences (SUMS), Shiraz, Iran
| | - Hossein Salehi Omran
- School of Medicine, Shahid Beheshti University of Medical Sciences (SBMUS), Tehran, Iran
| | - Eshagh Sedighi
- Department of Veterinary Medicine, Islamic Azad University Branch of Urmia, Urmia, Iran
| | - Farzan Beigi
- Students Research Committee, Arak University of Medical Sciences, Arak, Iran
| | - Masoud Mortezazadeh
- Department of Internal Medicine, Sina Hospital, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
35
|
Lo SW, Hung TH, Lin YT, Lee CS, Chen CY, Fang CJ, Lai PC. Clinical efficacy and safety of faecal microbiota transplantation in the treatment of irritable bowel syndrome: a systematic review, meta-analysis and trial sequential analysis. Eur J Med Res 2024; 29:464. [PMID: 39289768 PMCID: PMC11409544 DOI: 10.1186/s40001-024-02046-5] [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] [Accepted: 08/30/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND The aim of this study is to evaluate the efficacy and safety of faecal microbiota transplantation (FMT) for the treatment of irritable bowel syndrome (IBS). METHODS We searched four databases for randomised controlled trials (RCTs) that compared FMT with a control intervention in patients with IBS. The revised Cochrane risk-of-bias (RoB) tool was chosen for appraisal. Meta-analysis with trial sequential analysis (TSA) was conducted. Grading of Recommendations Assessment Development and Evaluation (GRADE) methodology was used to assess the certainty of evidence (CoE). RESULTS We included 12 RCTs with a total of 615 participants. Meta-analyses showed no significant difference between the FMT and control groups in terms of clinical responses (relative risk [RR] = 1.44, 95% confidence interval [CI] 0.88-2.33) and changes in IBS Severity Scoring System (IBS-SSS) scores (standardised mean difference [SMD] = - 0.31, 95% CI - 0.72 to 0.09) and IBS Quality of Life (IBS-QOL) scores (SMD = 0.30, 95% CI - 0.09 to 0.69). Subgroup analysis revealed that in studies with low RoB and using endoscopy, nasojejunal tube and rectal enema delivery, FMT led to a significant improvement in clinical responses and changes in IBS-SSS and IBS-QOL scores. TSA suggested that the current evidence is inconclusive and that the CoE is very low. CONCLUSION This study suggests that patients with IBS may benefit from FMT especially when it is administered via endoscopy, nasojejunal tube or rectal enema. However, the certainty of evidence is very low. Further research is needed to confirm the efficacy and safety of FMT for IBS treatment. TRIAL REGISTRATION PROSPERO registration number CRD42020211002.
Collapse
Affiliation(s)
- Shao-Wei Lo
- Education Centre, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, No.138, Sheng Li Road, Tainan, 704, Taiwan
| | - Tsung-Hsuan Hung
- Education Centre, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, No.138, Sheng Li Road, Tainan, 704, Taiwan
| | - Yen-Tsen Lin
- Education Centre, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, No.138, Sheng Li Road, Tainan, 704, Taiwan
| | - Chun-Shen Lee
- Education Centre, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, No.138, Sheng Li Road, Tainan, 704, Taiwan
| | - Chiung-Yu Chen
- Education Centre, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, No.138, Sheng Li Road, Tainan, 704, Taiwan
- Department of Internal Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Ching-Ju Fang
- Medical Library, National Cheng Kung University, Tainan, Taiwan
- Department of Secretariat, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Chun Lai
- Education Centre, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, No.138, Sheng Li Road, Tainan, 704, Taiwan.
- Department of Paediatrics, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
36
|
Dean NJ, d'Arienzo PD, Ibraheim H, Lee KA, Olsson-Brown AC, Pinato DJ, Powell N. The role of the gut microbiome in regulating the response to immune checkpoint inhibitor therapy. Best Pract Res Clin Gastroenterol 2024; 72:101944. [PMID: 39645284 DOI: 10.1016/j.bpg.2024.101944] [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: 06/04/2024] [Accepted: 08/05/2024] [Indexed: 12/09/2024]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionised cancer therapy, yet the proportion of patients who achieve long-term disease control remain suboptimal. Over the past decade, the gut microbiome has been shown to influence immune-mediated tumour suppression as well as responses to ICI therapies. Compositional differences in gut microbiome may account for the differences in outcomes from immune checkpoint blockade. Identifying microbiota species associated with favourable/unfavourable outcomes and modelling their dynamics throughout the course of ICI treatment could help develop predictive biomarkers of immunotherapy response, and manipulating the gut microbiome represent a novel approach to enhancing ICI effectiveness. Clinically, this raises the prospect of using gut microbiome-based therapies to overcome primary resistance to ICIs, mitigate the effects of microbiome-altering drugs such as antibiotics or proton pump inhibitors, and improve overall survival in patients across numerous different cancer types.
Collapse
Affiliation(s)
- Nathan J Dean
- Cancer Services Division, The Royal Marsden Hospital, London, United Kingdom
| | - Paolo D d'Arienzo
- Cancer Services Division, The Royal Marsden Hospital, London, United Kingdom
| | - Hajir Ibraheim
- Cancer Services Division, The Royal Marsden Hospital, London, United Kingdom; Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom
| | - Karla A Lee
- Cancer Services Division, The Royal Marsden Hospital, London, United Kingdom
| | - Anna C Olsson-Brown
- Sussex Cancer Centre, University Hospitals Sussex NHS Foundation Trust, Brighton, United Kingdom
| | - David J Pinato
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Nicholas Powell
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, United Kingdom.
| |
Collapse
|
37
|
Lauwers E, Sabino J, Hoffman I, van Hoeve K. Faecal microbiota transplantation in children: A systematic review. Acta Paediatr 2024; 113:1991-2002. [PMID: 38391047 DOI: 10.1111/apa.17167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 01/25/2024] [Accepted: 02/13/2024] [Indexed: 02/24/2024]
Abstract
AIM Novel technologies offer insights into the potential role of the intestinal microbiota in human health and disease. Dysbiosis has been associated with several diseases, and it is thought to play a role in the pathogenesis of different gastrointestinal diseases. Faecal microbiota transplantation (FMT) is emerging as a method to modulate the gastrointestinal microbial ecosystem. While recurrent Clostridioides difficile infection is the recognised FMT indication, exploration of other therapeutic uses is ongoing. METHODS Following PRISMA guidelines, we conducted a systematic review, extracting 583 articles from Embase and PubMed (index date to October 2022). RESULTS The search yielded 58 studies for full review, with 50 included in the systematic review. Articles were categorised by FMT indication, study design, efficacy, adverse events, donor selection and administration route. FMT appears safe and effective for recurrent Clostridioides difficile infection, although severe adverse events are reported in children. However, there are currently insufficient data to support the use of FMT for other potential therapeutic indications (such as irritable or inflammatory bowel disease or obesity), beside the potential to decolonise multi-drug resistant organisms. CONCLUSION This underscores the need for randomised, controlled, prospective cohort studies in children to assess FMT effectiveness in diverse conditions and counteract publication bias.
Collapse
Affiliation(s)
- Ella Lauwers
- Department of Paediatric Gastroenterology & Hepatology & Nutrition, University Hospitals Leuven, Leuven, Belgium
| | - João Sabino
- TARGID, Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
- Department of Gastroenterology & Hepatology, University Hospitals Leuven, Leuven, Belgium
| | - Ilse Hoffman
- Department of Paediatric Gastroenterology & Hepatology & Nutrition, University Hospitals Leuven, Leuven, Belgium
| | - Karen van Hoeve
- Department of Paediatric Gastroenterology & Hepatology & Nutrition, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
38
|
Piperni E, Nguyen LH, Manghi P, Kim H, Pasolli E, Andreu-Sánchez S, Arrè A, Bermingham KM, Blanco-Míguez A, Manara S, Valles-Colomer M, Bakker E, Busonero F, Davies R, Fiorillo E, Giordano F, Hadjigeorgiou G, Leeming ER, Lobina M, Masala M, Maschio A, McIver LJ, Pala M, Pitzalis M, Wolf J, Fu J, Zhernakova A, Cacciò SM, Cucca F, Berry SE, Ercolini D, Chan AT, Huttenhower C, Spector TD, Segata N, Asnicar F. Intestinal Blastocystis is linked to healthier diets and more favorable cardiometabolic outcomes in 56,989 individuals from 32 countries. Cell 2024; 187:4554-4570.e18. [PMID: 38981480 DOI: 10.1016/j.cell.2024.06.018] [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: 05/12/2023] [Revised: 02/23/2024] [Accepted: 06/11/2024] [Indexed: 07/11/2024]
Abstract
Diet impacts human health, influencing body adiposity and the risk of developing cardiometabolic diseases. The gut microbiome is a key player in the diet-health axis, but while its bacterial fraction is widely studied, the role of micro-eukaryotes, including Blastocystis, is underexplored. We performed a global-scale analysis on 56,989 metagenomes and showed that human Blastocystis exhibits distinct prevalence patterns linked to geography, lifestyle, and dietary habits. Blastocystis presence defined a specific bacterial signature and was positively associated with more favorable cardiometabolic profiles and negatively with obesity (p < 1e-16) and disorders linked to altered gut ecology (p < 1e-8). In a diet intervention study involving 1,124 individuals, improvements in dietary quality were linked to weight loss and increases in Blastocystis prevalence (p = 0.003) and abundance (p < 1e-7). Our findings suggest a potentially beneficial role for Blastocystis, which may help explain personalized host responses to diet and downstream disease etiopathogenesis.
Collapse
Affiliation(s)
- Elisa Piperni
- Department CIBIO, University of Trento, Trento, Italy; IEO, Istituto Europeo di Oncologia IRCSS, Milan, Italy
| | - Long H Nguyen
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA; Harvard Chan Microbiome in Public Health Center, Boston, MA, USA
| | - Paolo Manghi
- Department CIBIO, University of Trento, Trento, Italy
| | - Hanseul Kim
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Edoardo Pasolli
- Department of Agricultural Sciences, University of Naples Federico II, Portici, Italy
| | - Sergio Andreu-Sánchez
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Alberto Arrè
- Department CIBIO, University of Trento, Trento, Italy; Zoe Ltd, London, UK
| | - Kate M Bermingham
- Zoe Ltd, London, UK; Department of Nutritional Sciences, King's College London, London, UK
| | | | - Serena Manara
- Department CIBIO, University of Trento, Trento, Italy
| | | | | | - Fabio Busonero
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato, Cagliari, Italy
| | | | - Edoardo Fiorillo
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato, Cagliari, Italy
| | | | | | - Emily R Leeming
- Department of Twins Research and Genetic Epidemiology, King's College London, London, UK
| | - Monia Lobina
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato, Cagliari, Italy
| | - Marco Masala
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato, Cagliari, Italy
| | - Andrea Maschio
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato, Cagliari, Italy
| | | | - Mauro Pala
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato, Cagliari, Italy
| | - Maristella Pitzalis
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato, Cagliari, Italy
| | | | - Jingyuan Fu
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Alexandra Zhernakova
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Simone M Cacciò
- Department of Infectious Diseases, Istituto Superiore Di Sanità, Rome, Italy
| | - Francesco Cucca
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (CNR), Monserrato, Cagliari, Italy; Dipartimento di Scienze Biomediche, Università degli Studi di Sassari, Sassari, Italy
| | - Sarah E Berry
- Department of Nutritional Sciences, King's College London, London, UK
| | - Danilo Ercolini
- Department of Agricultural Sciences, University of Naples Federico II, Portici, Italy
| | - Andrew T Chan
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA; Harvard Chan Microbiome in Public Health Center, Boston, MA, USA
| | - Curtis Huttenhower
- Harvard T.H. Chan School of Public Health, Boston, MA, USA; The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Tim D Spector
- Department of Twins Research and Genetic Epidemiology, King's College London, London, UK
| | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy; IEO, Istituto Europeo di Oncologia IRCSS, Milan, Italy; Department of Twins Research and Genetic Epidemiology, King's College London, London, UK.
| | | |
Collapse
|
39
|
Yamada CH, Ortis GB, Buso GM, Martins TC, Zequinao T, Telles JP, Wollmann LC, Montenegro CDO, Dantas LR, Cruz JW, Tuon FF. Validation of Lyophilized Human Fecal Microbiota for the Treatment of Clostridioides difficile Infection: A Pilot Study with Pharmacoeconomic Analysis of a Middle-Income Country-Promicrobioma Project. Microorganisms 2024; 12:1741. [PMID: 39203583 PMCID: PMC11356882 DOI: 10.3390/microorganisms12081741] [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: 07/30/2024] [Revised: 08/09/2024] [Accepted: 08/21/2024] [Indexed: 09/03/2024] Open
Abstract
BACKGROUND Clostridioides difficile infection (CDI) represents a prevalent and potentially severe health concern linked to the usage of broad-spectrum antibiotics. The aim of this study was to evaluate a new lyophilized product based on human fecal microbiota for transplant, including cost-benefit analysis in the treatment of recurrent or refractory CDI. METHODS The product for fecal microbiota transplant was obtained from two donors. Microbiological, viability, and genomic analysis were evaluated. After validation, a clinical pilot study including recurrent or refractory CDI with 24 patients was performed. Clinical response and 4-week recurrence were the outcome. Cost-benefit analysis compared the fecal microbiota transplant with conventional retreatment with vancomycin or metronidazole. RESULTS The microbiota for transplant presented significant bacterial viability, with and adequate balance of Firmicutes and Bacteroidetes. The clinical response with the microbiota transplant was 92%. In financial terms, estimated expenditure for CDI solely related to recurrence, based on stochastic modeling, totals USD 222.8 million per year in Brazil. CONCLUSIONS The lyophilized human fecal microbiota for transplant is safe and can be an important step for a new product with low cost, even with genomic sequencing. Fecal microbiota transplantation emerges as a more cost-effective alternative compared to antimicrobials in the retreatment of CDI.
Collapse
Affiliation(s)
- Carolina Hikari Yamada
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, PR, Brazil; (C.H.Y.); (G.B.O.); (T.C.M.); (T.Z.); (L.R.D.)
| | - Gabriel Burato Ortis
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, PR, Brazil; (C.H.Y.); (G.B.O.); (T.C.M.); (T.Z.); (L.R.D.)
| | - Gustavo Martini Buso
- School of Business, Pontifical Catholic University of Paraná, Rua Imaculada Conceição 1155, Curitiba 80215-901, PR, Brazil; (G.M.B.); (J.W.C.)
| | - Thalissa Colodiano Martins
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, PR, Brazil; (C.H.Y.); (G.B.O.); (T.C.M.); (T.Z.); (L.R.D.)
| | - Tiago Zequinao
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, PR, Brazil; (C.H.Y.); (G.B.O.); (T.C.M.); (T.Z.); (L.R.D.)
| | - Joao Paulo Telles
- Hospital Universitário Evangélico Mackenzie, Curitiba 80730-150, PR, Brazil; (J.P.T.); (L.C.W.)
| | | | - Carolina de Oliveira Montenegro
- School of Business, Pontifical Catholic University of Paraná, Rua Imaculada Conceição 1155, Curitiba 80215-901, PR, Brazil; (G.M.B.); (J.W.C.)
| | - Leticia Ramos Dantas
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, PR, Brazil; (C.H.Y.); (G.B.O.); (T.C.M.); (T.Z.); (L.R.D.)
| | - June Westarb Cruz
- School of Business, Pontifical Catholic University of Paraná, Rua Imaculada Conceição 1155, Curitiba 80215-901, PR, Brazil; (G.M.B.); (J.W.C.)
| | - Felipe Francisco Tuon
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, PR, Brazil; (C.H.Y.); (G.B.O.); (T.C.M.); (T.Z.); (L.R.D.)
| |
Collapse
|
40
|
Goloshchapov OV, Chukhlovin AB, Bug DS, Polev DE, Kosarev OV, Klementeva RV, Izmailova EA, Kazantsev IV, Khalipskaia MS, Goloshchapova МО, Yudintseva OS, Barkhatov IM, Petukhova NV, Zubarovskaya LS, Kulagin AD, Moiseev IS. Safety, Feasibility, and Advantages of Oral Microbiota Transplantation: The First Clinical Case. J Pediatr Hematol Oncol 2024; 46:287-296. [PMID: 38875447 PMCID: PMC11268550 DOI: 10.1097/mph.0000000000002896] [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: 12/24/2023] [Accepted: 04/30/2024] [Indexed: 06/16/2024]
Abstract
The pilot clinical study presented demonstrates the possibility, safety, and effectiveness of oral microbiota transplantation from a healthy donor to a patient with neuroblastoma to prevent chemotherapy-induced oral mucositis. A 6-month-old patient with a diagnosis of retroperitoneal neuroblastoma was treated according to the NB 2004 protocol. Due to the development of severe oral mucositis, it was decided to perform oral microbiota transplantation. During the next 3 chemotherapy cycles and conditioning regimen before autologous hematopoietic cell transplantation (auto-HCT), the patient was repeatedly injected per os with donor saliva from her healthy mother. Oral microbiota transplantation was shown to effectively prevent the development of oral mucositis after chemotherapy, and only grade 1 oral mucositis developed after auto-HCT. In all loci of the oral cavity, there was a decreased abundance of bacteria from the Staphylococcaceae, Micrococcaceae, and Xanthomonadaceae families. Conversely, there was an increase in the relative abundance of Streptococcaceae and certain other bacterial taxa. In conclusion, the transplantation of maternal saliva in this patient prevented severe mucositis and was accompanied by a compositional change of the patient's oral microbiota. No adverse events due to the transplantation of maternal saliva were noted.
Collapse
Affiliation(s)
| | | | | | | | - Oleg V. Kosarev
- Saint Petersburg Mining University, Saint Petersburg, Russian Federation
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Wang Q, He M, Liang J, Tan X, Wu Q, Wang J, Li X, Qiao M, Huang Z, Xie Q, Liu Z, Ren H, Wang L, Zhou H, Shao L, Shu R, Wu W, Yang W, Wang H, Sun Z, Xu X, Zhang X, Li Z, Zhang Y, Meng J, Zhu Y, Chen F, Qu R, Chen P, Li S, Shi Y, Mao X, Hu B, Zhang Y, Cao YJ, Guo Z. Chinese guidelines for integrated diagnosis and treatment of intestinal microecology technologies in tumor application (2024 Edition). J Cancer Res Ther 2024; 20:1130-1140. [PMID: 39206974 DOI: 10.4103/jcrt.jcrt_32_24] [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: 01/04/2024] [Accepted: 07/01/2024] [Indexed: 09/04/2024]
Abstract
ABSTRACT Intestinal microecology (IM) is the largest and most important microecological system of the human body. Furthermore, it is the key factor for activating and maintaining the physiological functions of the intestine. Numerous studies have investigated the effects of the gut microbiota on the different tissues and organs of the human body as well as their association with various diseases, and the findings are gradually being translated into clinical practice. The gut microbiota affects the occurrence, progression, treatment response, and toxic side effects of tumors. The deepening of research related to IM and tumors has opened a new chapter in IM research driven by methods and technologies such as second-generation sequencing and bioinformatics. The IM maintains the function of the host immune system and plays a pivotal role in tumor-control drug therapy. Increasing evidence has proven that the efficacy of tumor-control drugs largely depends on the IM balance, and strategies based on the IM technology show promising application prospects in the diagnosis and treatment of tumor. The Tumor and Microecology Professional Committee of the Chinese Anti-cancer Association gathered relevant experts to discuss and propose the "Chinese guidelines for integrated diagnosis and treatment of IM technologies in tumor application (2024 Edition)," which was established based on the research progress of the application of the IM technology in tumor to provide a basis for the standardization of the diagnosis and treatment of the IM technology in the tumor.
Collapse
Affiliation(s)
- Qiang Wang
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Mingxin He
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Jing Liang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, Jinan, China
| | - Xiaohua Tan
- Department of Oncology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China
| | - Qingming Wu
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Jun Wang
- The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Xiaoan Li
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Mingqiang Qiao
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin, China
| | - Ziming Huang
- Hubei Maternal and Child Health Care Hospital, Wuhan, China
| | - Qi Xie
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Lung Cancer Institute, Jinan, China
| | - Zhe Liu
- Medical College, Tianjin University, Tianjin, China
| | - Hua Ren
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong Province, China
| | - Liang Wang
- Department of Hematology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Hao Zhou
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liang Shao
- Department of Hematology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Rong Shu
- The Third People's Hospital of Hubei Province, Wuhan, China
| | - Wei Wu
- Guangdong Provincial Institute of Public Health, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou, China
| | - Wenyan Yang
- Shangdong First Medical University and Shangdong Academy of Medical Sciences, Jinan, China
| | - Hua Wang
- Department of Hematology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou, China
| | - Zhiqiang Sun
- Department of Hematology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Xiaojun Xu
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xingding Zhang
- The School of Medicine of Sun Yat-Sen University, Shenzhen, China
| | - Zhiming Li
- Medical Department, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou, China
| | - Yu Zhang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Shenzhen, China
| | - Jingye Meng
- Department of Hematology and Oncology, National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, Second Hospital Affiliated to Southern University of Science and Technology, Shenzhen, China
| | - Yanli Zhu
- The First Affliated Hosptial of Xinxiang Medical University, Xinxiang, China
| | - Feng Chen
- The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rong Qu
- Department of Critical Care Medicine, Huizhou Municipal Central Hospital, Huizhou, China
| | - Peng Chen
- Department of Hematology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Shuluan Li
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Yuanyuan Shi
- Shenzhen Cell Valley Biomedicine Co. LTD, Shenzhen, China
| | - Xin Mao
- Primary Health Care Foundation of China, Xiangyang, China
| | - Bichuan Hu
- Xiangyang Hospital of Integrated Traditional Chinese and Western Medicine, Xiangyang, China
| | - Yukui Zhang
- Xiangyang Hospital of Traditional Chinese Medicine, Xiangyang, China
| | - Yu J Cao
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Zhi Guo
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, China
| |
Collapse
|
42
|
Spigaglia P. Clostridioides difficile and Gut Microbiota: From Colonization to Infection and Treatment. Pathogens 2024; 13:646. [PMID: 39204246 PMCID: PMC11357127 DOI: 10.3390/pathogens13080646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/23/2024] [Accepted: 07/29/2024] [Indexed: 09/03/2024] Open
Abstract
Clostridioides difficile is the main causative agent of antibiotic-associated diarrhea (AAD) in hospitals in the developed world. Both infected patients and asymptomatic colonized individuals represent important transmission sources of C. difficile. C. difficile infection (CDI) shows a large range of symptoms, from mild diarrhea to severe manifestations such as pseudomembranous colitis. Epidemiological changes in CDIs have been observed in the last two decades, with the emergence of highly virulent types and more numerous and severe CDI cases in the community. C. difficile interacts with the gut microbiota throughout its entire life cycle, and the C. difficile's role as colonizer or invader largely depends on alterations in the gut microbiota, which C. difficile itself can promote and maintain. The restoration of the gut microbiota to a healthy state is considered potentially effective for the prevention and treatment of CDI. Besides a fecal microbiota transplantation (FMT), many other approaches to re-establishing intestinal eubiosis are currently under investigation. This review aims to explore current data on C. difficile and gut microbiota changes in colonized individuals and infected patients with a consideration of the recent emergence of highly virulent C. difficile types, with an overview of the microbial interventions used to restore the human gut microbiota.
Collapse
Affiliation(s)
- Patrizia Spigaglia
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Roma, Italy
| |
Collapse
|
43
|
Long AE, Pitta D, Hennessy M, Indugu N, Vecchiarelli B, Luethy D, Aceto H, Hurcombe S. Assessment of fecal bacterial viability and diversity in fresh and frozen fecal microbiota transplant (FMT) product in horses. BMC Vet Res 2024; 20:306. [PMID: 38987780 PMCID: PMC11234551 DOI: 10.1186/s12917-024-04166-w] [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: 03/13/2023] [Accepted: 07/01/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND Currently, lack of standardization for fecal microbiota transplantation (FMT) in equine practice has resulted in highly variable techniques, and there is no data on the bacterial metabolic activity or viability of the administered product. The objectives of this study were to compare the total and potentially metabolically active bacterial populations in equine FMT, and assess the effect of different frozen storage times, buffers, and temperatures on an equine FMT product. Fresh feces collected from three healthy adult horses was subjected to different storage methods. This included different preservation solutions (saline plus glycerol or saline only), temperature (-20 °C or -80 °C), and time (fresh, 30, 60, or 90 days). Samples underwent DNA extraction to assess total bacterial populations (both live and dead combined) and RNA extraction followed by reverse transcription to cDNA as a proxy to assess viable bacteria, then 16s rRNA gene amplicon sequencing using the V1-V2 region. RESULTS The largest difference in population indices and taxonomic composition at the genus level was seen when evaluating the results of DNA-based (total) and cDNA-based (potentially metabolically active) extraction method. At the community level, alpha diversity (observed species, Shannon diversity) was significantly decreased in frozen samples for DNA-based analysis (P < 0.05), with less difference seen for cDNA-based sequencing. Using DNA-based analysis, length of storage had a significant impact (P < 0.05) on the bacterial community profiles. For potentially metabolically active populations, storage overall had less of an effect on the bacterial community composition, with a significant effect of buffer (P < 0.05). Individual horse had the most significant effect within both DNA and cDNA bacterial communities. CONCLUSIONS Frozen storage of equine FMT material can preserve potentially metabolically active bacteria of the equine fecal microbiome, with saline plus glycerol preservation more effective than saline alone. Larger studies are needed to determine if these findings apply to other individual horses. The ability to freeze FMT material for use in equine patients could allow for easier clinical use of fecal transplant in horses with disturbances in their intestinal microbiome.
Collapse
Affiliation(s)
- Alicia E Long
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania, Kennett Square, PA, USA.
| | - Dipti Pitta
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania, Kennett Square, PA, USA
| | - Meagan Hennessy
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania, Kennett Square, PA, USA
| | - Nagaraju Indugu
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania, Kennett Square, PA, USA
| | - Bonnie Vecchiarelli
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania, Kennett Square, PA, USA
| | - Daniela Luethy
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania, Kennett Square, PA, USA
| | - Helen Aceto
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania, Kennett Square, PA, USA
| | - Samuel Hurcombe
- Department of Clinical Studies, New Bolton Center, University of Pennsylvania, Kennett Square, PA, USA
- Veterinary Innovative Partners, New York, NY, USA
| |
Collapse
|
44
|
Zhao JT, Zhang Y, Wang XW, Zou PY, Zhao Z, Mei H, Liu YX, Su NY, Zhu YJ, Wang B, Wei YL, Chen DF, Lan CH. Long-term effects of fecal microbiota transplantation on gut microbiota after Helicobacter pylori eradication with bismuth quadruple therapy: A randomized controlled trial. Helicobacter 2024; 29:e13079. [PMID: 38984661 DOI: 10.1111/hel.13079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 03/29/2024] [Accepted: 04/09/2024] [Indexed: 07/11/2024]
Abstract
BACKGROUND Eradicating Helicobacter pylori infection by bismuth quadruple therapy (BQT) is effective. However, the effect of BQT and subsequent fecal microbiota transplant (FMT) on the gut microbiota is less known. MATERIALS AND METHODS This prospective randomized controlled trial was conducted at a tertiary hospital in China from January 2019 to October 2020, with the primary endpoints the effect of BQT on the gut microbiota and the effect of FMT on the gut microbiota after bismuth quadruple therapy eradication therapy. A 14-day BQT with amoxicillin and clarithromycin was administered to H. pylori-positive subjects, and after eradication therapy, patients received a one-time FMT or placebo treatment. We then collected stool samples to assess the effects of 14-day BQT and FMT on the gut microbiota. 16 s rDNA and metagenomic sequencing were used to analyze the structure and function of intestinal flora. We also used Gastrointestinal Symptom Rating Scale (GSRS) to evaluate gastrointestinal symptom during treatment. RESULTS A total of 30 patients were recruited and 15 were assigned to either FMT or placebo groups. After eradication therapy, alpha-diversity was decreased in both groups. At the phylum level, the abundance of Bacteroidetes and Firmicutes decreased, while Proteobacteria increased. At the genus level, the abundance of beneficial bacteria decreased, while pathogenic bacteria increased. Eradication therapy reduced some resistance genes abundance while increased the resistance genes abundance linked to Escherichia coli. While they all returned to baseline by Week 10. Besides, the difference was observed in Week 10 by the diarrhea score between two groups. Compared to Week 2, the GSRS total score and diarrhea score decreased in Week 3 only in FMT group. CONCLUSIONS The balance of intestinal flora in patients can be considerably impacted by BQT in the short term, but it has reverted back to baseline by Week 10. FMT can alleviate gastrointestinal symptoms even if there was no evidence it promoted restoration of intestinal flora.
Collapse
Affiliation(s)
- Jing-Tao Zhao
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Yi Zhang
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Xing-Wei Wang
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Pei-Ying Zou
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Zhe Zhao
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Hao Mei
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Yu-Xiang Liu
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Na-Yun Su
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Yang-Jie Zhu
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Bin Wang
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Yan-Ling Wei
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Dong-Feng Chen
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| | - Chun-Hui Lan
- Department of Gastroenterology, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
45
|
Rondinella D, Quaranta G, Rozera T, Dargenio P, Fancello G, Venturini I, Guarnaccia A, Porcari S, Bibbò S, Sanguinetti M, Gasbarrini A, Masucci L, Cammarota G, Ianiro G. Donor screening for fecal microbiota transplantation with a direct stool testing-based strategy: a prospective cohort study. Microbes Infect 2024; 26:105341. [PMID: 38679228 DOI: 10.1016/j.micinf.2024.105341] [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: 02/19/2024] [Revised: 03/31/2024] [Accepted: 04/18/2024] [Indexed: 05/01/2024]
Abstract
Fecal microbiota transplantation (FMT) is effective against recurrent Clostridioides difficile infection (rCDI), but its safety is jeopardized by the potential transmission of pathogens, so international guidelines recommend either a quarantine or a direct stool testing. Whereas reports of the quarantine-based approach are emerging, data on the direct testing-based approach are not available. Our aim is to report outcomes of a donor screening framework for FMT including direct stool testing. In this prospective cohort study, all donor candidates recruited at our FMT centre underwent a four-step screening process to be enrolled as actual donors. Each collected stool donation was then evaluated with a direct stool testing including a molecular assay for gut pathogens and a culture assay for multi-drug resistant organisms (MDRO). From January 2019 to June 2023, 72 of 227 candidates (32%) were considered eligible and provided 277 stool donations. Ninety-nine donations (36%) were discarded for positivity to intestinal pathogens, most commonly enteropathogenic Escherichia coli (n = 37) and Blastocystis hominis (n = 20). Overall, 337 stool aliquots were obtained from 165 approved donations. All suspensions were used for patients with rCDI, and no serious adverse events or clinically evident infections were observed at 12 weeks after procedures. In our study, screening of donor faeces including direct stool testing led to the discard of a considerable rate of stool donations but was also extremely safe. This approach may represent a reliable strategy to guarantee the safety of FMT programs, especially in countries with high prevalence of MDRO.
Collapse
Affiliation(s)
- Debora Rondinella
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Gianluca Quaranta
- Microbiology Unit, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Tommaso Rozera
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Pasquale Dargenio
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Giovanni Fancello
- Microbiology Unit, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Irene Venturini
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Alessandra Guarnaccia
- Microbiology Unit, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Serena Porcari
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Stefano Bibbò
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Maurizio Sanguinetti
- Microbiology Unit, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Antonio Gasbarrini
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Luca Masucci
- Microbiology Unit, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giovanni Cammarota
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Gianluca Ianiro
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy.
| |
Collapse
|
46
|
Bottino P, Vay D, Leli C, Ferrara L, Pizzo V, Gotta F, Raiteri A, Rapallo F, Roveta A, Maconi A, Rocchetti A. Evaluation of Bacterial Viability for Fecal Microbiota Transplantation: Impact of Thawing Temperature and Storage Time. Microorganisms 2024; 12:1294. [PMID: 39065063 PMCID: PMC11278783 DOI: 10.3390/microorganisms12071294] [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: 05/18/2024] [Revised: 06/17/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
Fecal Microbiota Transplantation (FMT) represents a promising therapeutic tool under study for several purposes and is currently applied to the treatment of recurrent Clostridioides difficile infection. However, since the use of fresh stool was affected by several issues linked to donor screening, the development of a frozen stool bank is a reliable option to standardize FMT procedures. Nevertheless, different environmental factors impact microbial viability. Herein, we report the effect of different thawing temperatures and storage conditions on bacterial suspensions in the FMT procedure. In total, 20 stool samples were divided into aliquots and tested across a combination of different storing periods (15, 30; 90 days) and thawing procedures (4 °C overnight, room temperature for 1 h; 37 °C for 5 min). Focusing on storage time, our data showed a significant reduction in viability for aerobic and anaerobic bacteria after thawing for 15 days, while no further reductions were observed until after 90 days. Instead, among the different thawing procedures, no significant differences were observed for aerobic bacteria, while for anaerobes, thawing at 37 °C for 5 min was more effective in preserving the bacterial viability. In conclusion, the frozen fecal microbiota remained viable for at least three months, with an excellent recovery rate in all three thawing conditions.
Collapse
Affiliation(s)
- Paolo Bottino
- Microbiology and Virology Laboratory, Azienda Ospedaliera Universitaria “SS. Antonio e Biagio e C. Arrigo”, 15121 Alessandria, Italy; (D.V.); (C.L.); (L.F.); (V.P.); (F.G.); (A.R.)
| | - Daria Vay
- Microbiology and Virology Laboratory, Azienda Ospedaliera Universitaria “SS. Antonio e Biagio e C. Arrigo”, 15121 Alessandria, Italy; (D.V.); (C.L.); (L.F.); (V.P.); (F.G.); (A.R.)
| | - Christian Leli
- Microbiology and Virology Laboratory, Azienda Ospedaliera Universitaria “SS. Antonio e Biagio e C. Arrigo”, 15121 Alessandria, Italy; (D.V.); (C.L.); (L.F.); (V.P.); (F.G.); (A.R.)
| | - Lidia Ferrara
- Microbiology and Virology Laboratory, Azienda Ospedaliera Universitaria “SS. Antonio e Biagio e C. Arrigo”, 15121 Alessandria, Italy; (D.V.); (C.L.); (L.F.); (V.P.); (F.G.); (A.R.)
| | - Valentina Pizzo
- Microbiology and Virology Laboratory, Azienda Ospedaliera Universitaria “SS. Antonio e Biagio e C. Arrigo”, 15121 Alessandria, Italy; (D.V.); (C.L.); (L.F.); (V.P.); (F.G.); (A.R.)
| | - Franca Gotta
- Microbiology and Virology Laboratory, Azienda Ospedaliera Universitaria “SS. Antonio e Biagio e C. Arrigo”, 15121 Alessandria, Italy; (D.V.); (C.L.); (L.F.); (V.P.); (F.G.); (A.R.)
| | - Alessio Raiteri
- Department of Science and Technological Innovation, University of Eastern Piedmont, 15121 Alessandria, Italy;
| | - Fabio Rapallo
- Department of Economics, University of Genova, 16126 Genova, Italy;
| | - Annalisa Roveta
- Research Training Innovation Infrastructure, Research and Innovation Department (DAIRI), Azienda Ospedaliera Universitaria “SS. Antonio e Biagio e C. Arrigo”, 15121 Alessandria, Italy; (A.R.); (A.M.)
| | - Antonio Maconi
- Research Training Innovation Infrastructure, Research and Innovation Department (DAIRI), Azienda Ospedaliera Universitaria “SS. Antonio e Biagio e C. Arrigo”, 15121 Alessandria, Italy; (A.R.); (A.M.)
| | - Andrea Rocchetti
- Microbiology and Virology Laboratory, Azienda Ospedaliera Universitaria “SS. Antonio e Biagio e C. Arrigo”, 15121 Alessandria, Italy; (D.V.); (C.L.); (L.F.); (V.P.); (F.G.); (A.R.)
| |
Collapse
|
47
|
Chen JH, Chiu CH, Chen CC, Chen YC, Yeh PJ, Kuo CJ, Chiu CT, Cheng HT, Pan YB, Le PH. Comparative Efficacy of Fecal Microbiota Transplantation in Treating Refractory or Recurrent Clostridioides difficile Infection among Patients with and without Inflammatory Bowel Disease: A Retrospective Cohort Study. Biomedicines 2024; 12:1396. [PMID: 39061970 PMCID: PMC11274107 DOI: 10.3390/biomedicines12071396] [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: 06/03/2024] [Revised: 06/17/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
Clostridioides difficile infection (CDI) worsens inflammatory bowel disease (IBD) prognosis. While fecal microbiota transplantation (FMT) is effective for refractory or recurrent CDI (rrCDI), comparative success rates between IBD and non-IBD patients are scarce. This study addresses this gap. A retrospective cohort study was conducted at Chang Gung Memorial Hospital from April 2019 to October 2023. Patients receiving FMT for rrCDI were categorized into IBD and non-IBD groups. Baseline characteristics and outcomes were compared at one month and one year, with successful FMT defined as the resolution of diarrhea without CDI recurrence. The study included 88 patients: 30 with IBD and 58 without IBD. The IBD group was younger, with fewer comorbidities. Success rates at one month were similar between groups (IBD: 80.0% vs. non-IBD: 78.9%, p = 0.908), as were negative toxin tests (IBD: 83.3% vs. non-IBD: 63.8%, p = 0.174). One-year success rates (IBD: 70.0% vs. non-IBD: 67.6%, p = 0.857) and eradication rates (IBD: 94.4% vs. non-IBD: 73.9%, p = 0.112) were also similar. Poor bowel preparation predicted FMT failure at one month (OR = 0.23, p = 0.019). No safety issues were reported. FMT is a safe, effective treatment for rrCDI, demonstrating similar success rates in patients with and without IBD.
Collapse
Affiliation(s)
- Jing-Han Chen
- School of Medicine, Chang Gung University, Taoyuan 333, Taiwan;
| | - Cheng-Hsun Chiu
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan; (C.-H.C.); (Y.-C.C.)
- Chang Gung Microbiota Therapy Center, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan; (C.-C.C.); (P.-J.Y.); (C.-J.K.); (C.-T.C.)
| | - Chien-Chang Chen
- Chang Gung Microbiota Therapy Center, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan; (C.-C.C.); (P.-J.Y.); (C.-J.K.); (C.-T.C.)
- Department of Pediatric Gastroenterology, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan
- Chang Gung Inflammatory Bowel Disease Center, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan
| | - Yi-Ching Chen
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan; (C.-H.C.); (Y.-C.C.)
- Chang Gung Microbiota Therapy Center, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan; (C.-C.C.); (P.-J.Y.); (C.-J.K.); (C.-T.C.)
| | - Pai-Jui Yeh
- Chang Gung Microbiota Therapy Center, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan; (C.-C.C.); (P.-J.Y.); (C.-J.K.); (C.-T.C.)
- Department of Pediatric Gastroenterology, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan
- Chang Gung Inflammatory Bowel Disease Center, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan
| | - Chia-Jung Kuo
- Chang Gung Microbiota Therapy Center, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan; (C.-C.C.); (P.-J.Y.); (C.-J.K.); (C.-T.C.)
- Chang Gung Inflammatory Bowel Disease Center, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan
- Taiwan Association of the Study of Intestinal Disease (TASID), Taoyuan 333, Taiwan
| | - Cheng-Tang Chiu
- Chang Gung Microbiota Therapy Center, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan; (C.-C.C.); (P.-J.Y.); (C.-J.K.); (C.-T.C.)
- Chang Gung Inflammatory Bowel Disease Center, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan
- Taiwan Association of the Study of Intestinal Disease (TASID), Taoyuan 333, Taiwan
| | - Hao-Tsai Cheng
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, New Taipei Municipal Tucheng Hospital, Tucheng, New Taipei City 236, Taiwan;
| | - Yu-Bin Pan
- Biostatistical Section, Clinical Trial Center, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan;
| | - Puo-Hsien Le
- Chang Gung Microbiota Therapy Center, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan; (C.-C.C.); (P.-J.Y.); (C.-J.K.); (C.-T.C.)
- Chang Gung Inflammatory Bowel Disease Center, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan
- Taiwan Association of the Study of Intestinal Disease (TASID), Taoyuan 333, Taiwan
| |
Collapse
|
48
|
Hassall J, Coxon C, Patel VC, Goldenberg SD, Sergaki C. Limitations of current techniques in clinical antimicrobial resistance diagnosis: examples and future prospects. NPJ ANTIMICROBIALS AND RESISTANCE 2024; 2:16. [PMID: 39843577 PMCID: PMC11721362 DOI: 10.1038/s44259-024-00033-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 05/07/2024] [Indexed: 01/24/2025]
Abstract
Antimicrobial resistance is a global threat to public health. Without proactive intervention, common infections may become untreatable, restricting the types of clinical intervention that can be undertaken and reversing improvements in mortality rates. Effective antimicrobial stewardship represents one approach to restrict the spread of antimicrobial resistance but relies on rapid and accurate diagnostics that minimise the unnecessary use of antibiotics. This is increasingly a key unmet clinical need. In this paper, we describe existing techniques for the detection of antimicrobial resistance, while examining their drawbacks and limitations. We also discuss emerging diagnostic technologies in the field, and the need for standardisation to allow for swifter and more widespread clinical adoption.
Collapse
Affiliation(s)
- Jack Hassall
- Science Research and Innovation, Medicines and Healthcare products Regulatory Agency, Blanche Lane, South Mimms, Potters Bar, Hertfordshire, EN6 3QG, UK
| | - Carmen Coxon
- Science Research and Innovation, Medicines and Healthcare products Regulatory Agency, Blanche Lane, South Mimms, Potters Bar, Hertfordshire, EN6 3QG, UK
| | - Vishal C Patel
- The Roger Williams Institute of Hepatology London, Foundation for Liver Research, 111 Coldharbour Lane, London, SE5 9NT, UK
- Institute of Liver Studies, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, 125 Coldharbour Lane, London, SE5 9NU, UK
- Institute of Liver Studies, King's College Hospital NHS Foundation Trust, Denmark Hill, London, SE5 9RS, UK
| | - Simon D Goldenberg
- Centre for Clinical Infection and Diagnostics Research, Guy's and St Thomas' NHS Foundation Trust and King's College, London, UK
| | - Chrysi Sergaki
- Science Research and Innovation, Medicines and Healthcare products Regulatory Agency, Blanche Lane, South Mimms, Potters Bar, Hertfordshire, EN6 3QG, UK.
| |
Collapse
|
49
|
Yadegar A, Bar-Yoseph H, Monaghan TM, Pakpour S, Severino A, Kuijper EJ, Smits WK, Terveer EM, Neupane S, Nabavi-Rad A, Sadeghi J, Cammarota G, Ianiro G, Nap-Hill E, Leung D, Wong K, Kao D. Fecal microbiota transplantation: current challenges and future landscapes. Clin Microbiol Rev 2024; 37:e0006022. [PMID: 38717124 PMCID: PMC11325845 DOI: 10.1128/cmr.00060-22] [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] [Indexed: 06/14/2024] Open
Abstract
SUMMARYGiven the importance of gut microbial homeostasis in maintaining health, there has been considerable interest in developing innovative therapeutic strategies for restoring gut microbiota. One such approach, fecal microbiota transplantation (FMT), is the main "whole gut microbiome replacement" strategy and has been integrated into clinical practice guidelines for treating recurrent Clostridioides difficile infection (rCDI). Furthermore, the potential application of FMT in other indications such as inflammatory bowel disease (IBD), metabolic syndrome, and solid tumor malignancies is an area of intense interest and active research. However, the complex and variable nature of FMT makes it challenging to address its precise functionality and to assess clinical efficacy and safety in different disease contexts. In this review, we outline clinical applications, efficacy, durability, and safety of FMT and provide a comprehensive assessment of its procedural and administration aspects. The clinical applications of FMT in children and cancer immunotherapy are also described. We focus on data from human studies in IBD in contrast with rCDI to delineate the putative mechanisms of this treatment in IBD as a model, including colonization resistance and functional restoration through bacterial engraftment, modulating effects of virome/phageome, gut metabolome and host interactions, and immunoregulatory actions of FMT. Furthermore, we comprehensively review omics technologies, metagenomic approaches, and bioinformatics pipelines to characterize complex microbial communities and discuss their limitations. FMT regulatory challenges, ethical considerations, and pharmacomicrobiomics are also highlighted to shed light on future development of tailored microbiome-based therapeutics.
Collapse
Affiliation(s)
- Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Haggai Bar-Yoseph
- Department of Gastroenterology, Rambam Health Care Campus, Haifa, Israel
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Tanya Marie Monaghan
- National Institute for Health Research Nottingham Biomedical Research Centre, University of Nottingham, Nottingham, United Kingdom
- Nottingham Digestive Diseases Centre, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Sepideh Pakpour
- School of Engineering, Faculty of Applied Sciences, UBC, Okanagan Campus, Kelowna, British Columbia, Canada
| | - Andrea Severino
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Ed J Kuijper
- Center for Microbiota Analysis and Therapeutics (CMAT), Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Wiep Klaas Smits
- Center for Microbiota Analysis and Therapeutics (CMAT), Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Elisabeth M Terveer
- Center for Microbiota Analysis and Therapeutics (CMAT), Leiden University Center for Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Sukanya Neupane
- Division of Gastroenterology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Ali Nabavi-Rad
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Javad Sadeghi
- School of Engineering, Faculty of Applied Sciences, UBC, Okanagan Campus, Kelowna, British Columbia, Canada
| | - Giovanni Cammarota
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Gianluca Ianiro
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
- Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Estello Nap-Hill
- Department of Medicine, Division of Gastroenterology, St Paul's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Dickson Leung
- Division of Gastroenterology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Karen Wong
- Division of Gastroenterology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Dina Kao
- Division of Gastroenterology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
50
|
Mullish BH, Merrick B, Quraishi MN, Bak A, Green CA, Moore DJ, Porter RJ, Elumogo NT, Segal JP, Sharma N, Marsh B, Kontkowski G, Manzoor SE, Hart AL, Settle C, Keller JJ, Hawkey P, Iqbal TH, Goldenberg SD, Williams HRT. The use of faecal microbiota transplant as treatment for recurrent or refractory Clostridioides difficile infection and other potential indications: second edition of joint British Society of Gastroenterology (BSG) and Healthcare Infection Society (HIS) guidelines. Gut 2024; 73:1052-1075. [PMID: 38609165 DOI: 10.1136/gutjnl-2023-331550] [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: 11/15/2023] [Accepted: 02/03/2024] [Indexed: 04/14/2024]
Abstract
The first British Society of Gastroenterology (BSG) and Healthcare Infection Society (HIS)-endorsed faecal microbiota transplant (FMT) guidelines were published in 2018. Over the past 5 years, there has been considerable growth in the evidence base (including publication of outcomes from large national FMT registries), necessitating an updated critical review of the literature and a second edition of the BSG/HIS FMT guidelines. These have been produced in accordance with National Institute for Health and Care Excellence-accredited methodology, thus have particular relevance for UK-based clinicians, but are intended to be of pertinence internationally. This second edition of the guidelines have been divided into recommendations, good practice points and recommendations against certain practices. With respect to FMT for Clostridioides difficile infection (CDI), key focus areas centred around timing of administration, increasing clinical experience of encapsulated FMT preparations and optimising donor screening. The latter topic is of particular relevance given the COVID-19 pandemic, and cases of patient morbidity and mortality resulting from FMT-related pathogen transmission. The guidelines also considered emergent literature on the use of FMT in non-CDI settings (including both gastrointestinal and non-gastrointestinal indications), reviewing relevant randomised controlled trials. Recommendations are provided regarding special areas (including compassionate FMT use), and considerations regarding the evolving landscape of FMT and microbiome therapeutics.
Collapse
Affiliation(s)
- Benjamin H Mullish
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
- Departments of Gastroenterology and Hepatology, St Mary's Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Blair Merrick
- Centre for Clinical Infection and Diagnostics Research, Guy's and St Thomas' NHS Foundation Trust, King's College London, London, UK
| | - Mohammed Nabil Quraishi
- Department of Gastroenterology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Microbiome Treatment Centre, University of Birmingham, Edgbaston, UK
- Institute of Cancer and Genomic Sciences, University of Birmingham, London, UK
| | - Aggie Bak
- Healthcare Infection Society, London, UK
| | - Christopher A Green
- Department of Infectious Diseases & Tropical Medicine, University Hospitals NHS Foundation Trust, Birmingham Heartlands Hospital, Birmingham, UK
- School of Chemical Engineering, University of Birmingham, Birmingham, UK
| | - David J Moore
- Institute of Applied Health Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Robert J Porter
- Department of Microbiology, Royal Devon and Exeter Hospitals, Barrack Road, UK
| | - Ngozi T Elumogo
- Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- Norfolk and Norwich University Hospital, Norwich, UK
| | - Jonathan P Segal
- Department of Gastroenterology, Royal Melbourne Hospital, Melbourne, Victoria, Australia
- Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Naveen Sharma
- Department of Gastroenterology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Microbiome Treatment Centre, University of Birmingham, Edgbaston, UK
- Institute of Cancer and Genomic Sciences, University of Birmingham, London, UK
| | - Belinda Marsh
- Lay representative for FMT Working Party, Healthcare Infection Society, London, UK
| | - Graziella Kontkowski
- Lay representative for FMT Working Party, Healthcare Infection Society, London, UK
- C.diff support, London, UK
| | - Susan E Manzoor
- Microbiome Treatment Centre, University of Birmingham, Edgbaston, UK
| | - Ailsa L Hart
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
- Department of Gastroenterology and Inflammatory Bowel Disease Unit, St Mark's Hospital and Academic Institute, Middlesex, UK
| | | | - Josbert J Keller
- Department of Gastroenterology, Haaglanden Medisch Centrum, The Hague, The Netherlands
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter Hawkey
- Microbiome Treatment Centre, University of Birmingham, Edgbaston, UK
- Public Health Laboratory, Faculty of Medicine, University of Birmingham, Birmingham, UK
| | - Tariq H Iqbal
- Department of Gastroenterology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Microbiome Treatment Centre, University of Birmingham, Edgbaston, UK
- Institute of Cancer and Genomic Sciences, University of Birmingham, London, UK
| | - Simon D Goldenberg
- Centre for Clinical Infection and Diagnostics Research, Guy's and St Thomas' NHS Foundation Trust, King's College London, London, UK
| | - Horace R T Williams
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
- Departments of Gastroenterology and Hepatology, St Mary's Hospital, Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|