1
|
Shi K, Wang L, Zheng XM, Zhang Z, Zhong F, Sun ZZ, Mohany M, Huang HH, Li J, Li S, Zhang L. Metronidazole-ursodeoxycholic acid bifunctional antibacterial: A promising strategy to combat Clostridium difficile infection and prevent recurrence. Eur J Med Chem 2025; 291:117631. [PMID: 40233427 DOI: 10.1016/j.ejmech.2025.117631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 03/12/2025] [Accepted: 04/11/2025] [Indexed: 04/17/2025]
Abstract
Current treatments against Clostridium difficile infection (CDI), such as vancomycin and metronidazole, frequently lead to severe recurrence due to disruption of gut microbiota balance, which results in a pressing need for new chemical entities to treat CDI. Bile acids, such as UDCA, have been demonstrated to inhibit the growth and spore germination of C. difficile, and regulate the structure of the intestinal flora. This study involved the synthesis of eight bile acid-metronidazole hybrids. Among them, the most promising hybrid, SCUT1-2, effectively killed the vegetative cells of C. difficile with a minimum inhibitory concentration (MIC) of 0.06-0.50 μg/mL, and inhibited spore germination in vitro. The absolute bioavailability of SCUT1-2 (F = 56.8 %) indicated that approximately half of SCUT1-2 was absorbed systemically, while a considerable portion remained in the gastrointestinal tract in its original form, laying a solid foundation for its effective action in vivo. SCUT1-2 could effectively alleviate the symptoms of weight loss and diarrhea in mice caused by CDI and effectively reduce the relevant expressions of inflammatory factors, outperforming metronidazole. Furthermore, SCUT1-2 demonstrated a favorable therapeutic effect in reducing mortality and disease symptoms in CDI mice by killing C. difficile cells and regulating the composition and structure of the intestinal flora. Notably, SCUT1-2 could effectively prevent recurrent CDI. This work provides a potential clinical lead for the development of CDI therapies and highlights hybrid medication as a new strategy.
Collapse
Affiliation(s)
- Kun Shi
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, PR China
| | - Li Wang
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
| | - Xiao-Min Zheng
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, PR China
| | - Zhe Zhang
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, PR China
| | - Fan Zhong
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, PR China
| | - Zhen-Zhu Sun
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, PR China
| | - Mohamed Mohany
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Hai-Hui Huang
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
| | - Jing Li
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, PR China
| | - Shan Li
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, PR China.
| | - Lei Zhang
- MOE International Joint Research Laboratory on Synthetic Biology and Medicines, School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, PR China.
| |
Collapse
|
2
|
Wang L, Xu T, Wu S, Zhao C, Huang H. The efficacy and underlying mechanisms of berberine in the treatment of recurrent Clostridioides difficile infection. Int J Antimicrob Agents 2025; 65:107468. [PMID: 39986400 DOI: 10.1016/j.ijantimicag.2025.107468] [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: 10/14/2024] [Revised: 01/05/2025] [Accepted: 02/14/2025] [Indexed: 02/24/2025]
Abstract
Recurrent Clostridioides difficile infection (rCDI) is a global health threat that has received considerable attention. Berberine (BBR), a natural pentacyclic isoquinoline alkaloid, has been used as a cost-effective treatment for intestinal infections in Asia for many years. However, the effect of BBR on rCDI is not clear. The efficacy and underlying mechanisms of BBR were evaluated in a vancomycin-dependent rCDI mouse model and an intestinal organoids model. The study findings showed that BBR treatment alleviated the severity of infection and increased survival rate in rCDI mice. Mechanistically, BBR alleviated intestinal epithelial damage with higher Occludin expression, suppressed some inflammatory pathways and reduced the level of inflammatory factors in both the caecum and serum. Moreover, 16S rRNA sequencing analysis indicated that BBR reshaped the gut microbiota by increasing the abundance of Firmicutes and reducing the abundance of Proteobacteria. At genus level, BBR treatment increased levels of Blautia and Bilophila, and reduced levels of Proteus. In addition, acetic acid, one of the short-chain fatty acids (SCFAs), was also increased after BBR treatment in rCDI mice. Collectively, BBR exerted a protective effect in rCDI via multiple underlying mechanisms and is a potential drug candidate for alleviating rCDI, but further research is needed in this area.
Collapse
Affiliation(s)
- Li Wang
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China; Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission of the PRC, Shanghai, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Teng Xu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China; Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission of the PRC, Shanghai, China
| | - Shi Wu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China; Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission of the PRC, Shanghai, China
| | - Chao Zhao
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China; MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| | - Haihui Huang
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, China; Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission of the PRC, Shanghai, China.
| |
Collapse
|
3
|
Shin JH, Jackson-Akers JY, Hoang SC, Behm BW, Warren CA. Fulminant Clostridioides difficile Infection: A Journey into the Unknown! Med Clin North Am 2025; 109:721-734. [PMID: 40185558 DOI: 10.1016/j.mcna.2025.01.001] [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: 04/07/2025]
Abstract
Clostridioides difficile is 1 of the 5 urgent antibiotic resistance threats in the United States as reported by the Centers for Disease Control and Prevention. Fulminant C difficile infection (CDI), characterized by hallmarks of critical illness such as hypotension, shock, or megacolon, has been difficult to define and treat. In this article, we describe the diagnostic criteria for fulminant CDI, clinical factors and inflammatory markers. We review the currently recommended treatment modalities including antibiotics and surgical interventions, colectomy, and diverting loop ileostomy. We also included treatment approaches that are still investigational such as intestinal microbiota transplant, tigecycline, and intravenous immunoglobulin.
Collapse
Affiliation(s)
- Jae H Shin
- Infectious Disease, Hoag Memorial Hospital Presbyterian, 1 Hoag Drive, Newport Beach, CA 92663, USA
| | | | - Sook C Hoang
- Colorectal Surgery, University of Virginia, 1215 Lee Street, Charlottesville, VA 22903, USA
| | - Brian W Behm
- Division of Gastroenterology, University of Virginia, 1300 Jefferson Park Avenue, Charlottesville, VA 22903, USA
| | - Cirle A Warren
- Division of Infectious Diseases and International Health, University of Virginia, 345 Crispell Drive, Charlottesville, VA 22903, USA.
| |
Collapse
|
4
|
Pascual M, Sogbe M, Yuste JR. Arrhythmic storm in a liver transplant recipient: Could bezlotoxumab be the trigger? Med Clin (Barc) 2025; 164:440-441. [PMID: 39701915 DOI: 10.1016/j.medcli.2024.10.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 12/21/2024]
Affiliation(s)
- Maria Pascual
- Liver Unit, Clínica Universidad de Navarra, Pamplona, Spain; Internal Medicine Department, Clínica Universidad de Navarra, Pamplona, Spain; Infectious Diseases Division, Clínica Universidad de Navarra, Pamplona, Spain
| | - Miguel Sogbe
- Liver Unit, Clínica Universidad de Navarra, Pamplona, Spain; Internal Medicine Department, Clínica Universidad de Navarra, Pamplona, Spain.
| | - José Ramón Yuste
- Internal Medicine Department, Clínica Universidad de Navarra, Pamplona, Spain; Infectious Diseases Division, Clínica Universidad de Navarra, Pamplona, Spain; IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| |
Collapse
|
5
|
Rahimić J, Alibegović E, Lekić L, Boras MM, Džidić-Krivić A, Farhat EK, Sher EK. Evaluating Vancomycin Monotherapy and Dual Therapy with Nifuroxazide for Medium-Severe Clostridioides Difficile Infection. Antibiotics (Basel) 2025; 14:400. [PMID: 40298556 PMCID: PMC12024110 DOI: 10.3390/antibiotics14040400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND All currently used therapeutic protocols and drugs for Clostridioides difficile infection (CDI) treatment do not have a satisfying success and usually cost a lot. OBJECTIVES To compare the efficacy of vancomycin monotherapy vs modified dual therapy with vancomycin + nifuroxazide as a therapeutic protocol for a medium-severe form of CDI. In addition, the effects of a modified therapeutic protocol with standard monotherapy on the number of stools and stool consistency in a medium-severe CDI will be compared. MATERIALS AND METHODS A prospective, randomized, controlled clinical trial that included 60 patients divided into two groups was conducted. One group of patients was treated with vancomycin monotherapy. The other group was treated with the modified therapeutic protocol (vancomycin + nifuroxazide). RESULTS The modified therapy with vancomycin + nifuroxazide demonstrated enhanced pharmacological efficacy in the management of CDI compared to the standard vancomycin monotherapy. Patients treated with dual therapy reported a significantly lower number of stools in first, second and third control; first control (4.47 ± 2.20 compared to 5.70 ± 1.91 in vancomycin group (p = 0.024)), second control (2.37 ± 0.85 compared to 3.13 ± 0.90 in vancomycin group (p = 0.001)), and third control (1.53 ± 0.51 compared to 1.80 ± 0.61 in vancomycin group (p = 0.035)). Also, the first and third controls noted significant improvements in stool consistency, measured as a decrease in the number of completely watery stools (p = 0.011 and p < 0.001, respectively). CONCLUSIONS Nifuroxazide and vancomycin have demonstrated accelerated improvement in patient status and hold promise as a novel dual therapeutic regimen for managing patients diagnosed with a medium-severe form of CDI.
Collapse
Affiliation(s)
- Jasna Rahimić
- Faculty of Medicine, European University Kallos, Tuzla, XVIII Hrvatske Brigade 8, 75000 Tuzla, Bosnia and Herzegovina;
- International Society of Engineering Science and Technology UK, Nottingham NG7 1GN, UK; (M.M.B.)
| | - Ervin Alibegović
- Department of Clinical Center, Tuzla University, 75000 Tuzla, Bosnia and Herzegovina
| | - Lana Lekić
- Faculty of Health Studies, University of Sarajevo, Stjepana Tomića 1, 71000 Sarajevo, Bosnia and Herzegovina
| | - Marijana Marković Boras
- International Society of Engineering Science and Technology UK, Nottingham NG7 1GN, UK; (M.M.B.)
- Department of Laboratory Diagnostic, University Clinical Hospital Mostar, 88000 Mostar, Bosnia and Herzegovina
| | - Amina Džidić-Krivić
- International Society of Engineering Science and Technology UK, Nottingham NG7 1GN, UK; (M.M.B.)
- Department of Neurology, Clinical Hospital Zenica, 72000 Zenica, Bosnia and Herzegovina
| | - Esma Karahmet Farhat
- Faculty of Food Technology, Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Emina Karahmet Sher
- International Society of Engineering Science and Technology UK, Nottingham NG7 1GN, UK; (M.M.B.)
- School of Science and Technology, Nottingham Trent University, Nottingham NG11 8NS, UK
| |
Collapse
|
6
|
Finn LM, Cummer R, Castagner B, Keller BG. Allosterically switchable network orients β-flap in Clostridioides difficile toxins. Proc Natl Acad Sci U S A 2025; 122:e2419263122. [PMID: 40172960 PMCID: PMC12002228 DOI: 10.1073/pnas.2419263122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 02/25/2025] [Indexed: 04/04/2025] Open
Abstract
Allosteric proteins exhibit a functional response upon ligand binding far from the active site. Clostridioides difficile toxins use allosteric binding by the endogenous cofactor myo-inositol hexakisphosphate to orchestrate self-cleavage from within the target cell. This binding event induces a conformational shift, primarily effecting a lever-like β-flap region, with two known orientations. We uncovered a mechanism for this allosteric transition using extensive atomistic molecular dynamics simulations and computational and experimental mutagenesis. The mechanism relies on a switchable interaction network. The most prominent interaction pair is K600-E743, with K600 interactions explaining ∼70% of the allosteric effect. Rather than gradually morphing between two end states, the interaction network adopts two mutually exclusive configurations in the active and inactive state. Similar switchable networks may explain allostery more broadly. This mechanism in particular could aid in drug development targeting the C. difficile toxins autoproteolysis.
Collapse
Affiliation(s)
- Lauren M. Finn
- Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, Berlin14195, Germany
| | - Rebecca Cummer
- Department of Pharmacology and Therapeutics, Faculty of Medicine and Health Sciences, McGill University, Montreal, QCH3G 1Y6, Canada
| | - Bastien Castagner
- Department of Pharmacology and Therapeutics, Faculty of Medicine and Health Sciences, McGill University, Montreal, QCH3G 1Y6, Canada
| | - Bettina G. Keller
- Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, Berlin14195, Germany
| |
Collapse
|
7
|
Feuerstadt P, Allegretti J, Khanna S. Treatment of Clostridioides difficile : The Times They Are Changing. Am J Gastroenterol 2025:00000434-990000000-01688. [PMID: 40192143 DOI: 10.14309/ajg.0000000000003445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 03/26/2025] [Indexed: 04/24/2025]
Affiliation(s)
- Paul Feuerstadt
- Section of Digestive Diseaese, Yale School of Medicine, Section of Digestive Disease, New Haven, Connecticut, USA
- PACT Gastroenterology Center, Hamden, Connecticut, USA
| | - Jessica Allegretti
- Fecal Microbiota Transplant Program, Crohn's and Colitis Center, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Division of Gastroenterology, Hepatology and Endoscopy, Harvard Medical School, Division of Gastroenterology, Hepatology and Endoscopy, Boston, Massachusetts, USA
| | - Sahil Khanna
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
8
|
Berry P, Khanna S. The evolving landscape of live biotherapeutics in the treatment of Clostridioides difficile infection. Indian J Gastroenterol 2025; 44:129-141. [PMID: 39821715 DOI: 10.1007/s12664-024-01717-9] [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] [Accepted: 11/18/2024] [Indexed: 01/19/2025]
Abstract
Clostridioides difficile (C. difficile) infection (CDI) is common after antibiotic exposure and presents significant morbidity, mortality and healthcare costs worldwide. The rising incidence of recurrent CDI, driven by hypervirulent strains, widespread antibiotic use and increased community transmission, has led to an urgent need for novel therapeutic strategies. Conventional antibiotic treatments, although effective, face limitations due to rising antibiotic resistance and high recurrence rates, which can reach up to 60% after multiple infections. This has prompted exploration of alternative therapies such as fecal microbiota-based therapies, including fecal microbiota transplantation (FMT) and live biotherapeutics (LBPs), which demonstrate superior efficacy in preventing recurrence. They are aimed at restoring the gut microbiota. Fecal microbiota, live-jslm and fecal microbiota spores, live-brpk have been approved by the U.S. Food and Drug Administration in individuals aged 18 years or older for recurrent CDI after standard antimicrobial treatment. They have demonstrated high efficacy and a favorable safety profile in clinical trials. Another LBP under study includes VE-303, which is not derived from human donor stool. This review provides a comprehensive overview of the current therapeutic landscape for CDI, including its epidemiology, pathophysiology, risk factors, diagnostic modalities and treatment strategies. The review delves into the emerging role of live biotherapeutics, with a particular focus on fecal microbiota-based therapies. We explore their development, mechanisms of action, clinical applications and potential to revolutionize CDI management.
Collapse
Affiliation(s)
- Parul Berry
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA
| | - Sahil Khanna
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
9
|
Puerta-Alcalde P, O'Keefe J, Woolstencroft R, Kaul S, López N, Cronin K, Lim A, Garcia-Pouton N, Álvarez M, Chee L, Espasa M, Grafia I, Suárez-Lledó M, Smibert O, Garcia-Vidal C, Slavin MA, Yong MK, Soriano A, Worth LJ. Clostridioides difficile infection and recurrence in cancer patients (CIRCA): A multicentre, international study. Int J Infect Dis 2025; 153:107785. [PMID: 39818267 DOI: 10.1016/j.ijid.2025.107785] [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: 11/04/2024] [Revised: 12/21/2024] [Accepted: 01/12/2025] [Indexed: 01/18/2025] Open
Abstract
OBJECTIVES We aimed to describe the characteristics of Clostridioides difficile infection (CDI) in cancer patients, analysing risk factors for 90-day recurrence and attributable mortality. METHODS Retrospective analysis on all CDI episodes from 2020 to 2022 in three Australian hospitals and one Spanish hospital. Logistic regression analyses were performed. RESULTS A total of 547 CDI episodes in cancer patients were documented. Treatment predominantly involved vancomycin (81.5%), followed by metronidazole (15.0%) and fidaxomicin (9.1%). Combined antibiotics were used in 61 (11.2%) episodes. The 90-day recurrence rate was 15.6%. Independent risk factors for CDI recurrence were female sex (OR 2.26, 95% CI 1.13-4.52), age >75 years (OR 2.69, 95% CI 1.30-5.59), dialysis (OR 5.15, 95% CI 1.45-18.27), vomiting at presentation (OR 0.06, 95% CI 0.01-0.55), colonic wall thickening in the CT abdomen (OR 2.42, 95% CI 1.06-5.49) and vancomycin therapy (OR 4.60, 95% CI 1.34-15.84). Overall, 90-day mortality was 22.3%, but attributable mortality was 4.9%. Risk factors for mortality attributed to CDI were age >65 years (OR 15.91, 95% CI 2.64-95.80), previous cerebrovascular disease (OR 20.27, 95% CI 3.12-131.84), antibiotic therapy within the last 30 days (OR 0.17, 95% CI 0.05-0.54), high-output diarrhoea (OR 6.68, 95% CI 1.68-26.56), high CRP-levels (OR 11.60, 95% CI 1.90-70.81) and need for treatment change (OR 6.65, 95% CI 2.20-20.08). CONCLUSIONS CDI recurrence rates among cancer patients remain significant. Nonetheless, fidaxomicin and other preventive strategies are seldom used. We identified several factors that could inform the implementation of these strategies in cancer patients.
Collapse
Affiliation(s)
- Pedro Puerta-Alcalde
- Department of Infectious Diseases, Hospital Clínic-IDIBAPS, Barcelona, Spain; Universitat de Barcelona (UB), Barcelona, Spain.
| | - Jessica O'Keefe
- Victorian Infectious Diseases Service, Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Rachel Woolstencroft
- Department of Infectious Diseases and National Center for Infection, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Shipraa Kaul
- Department of Infectious Diseases and Microbiology, Austin Health, Melbourne, VIC, Australia
| | - Néstor López
- Department of Infectious Diseases, Hospital Clínic-IDIBAPS, Barcelona, Spain
| | - Katie Cronin
- Microbiology Department, Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Andrew Lim
- Haematology Department, Austin Health, Melbourne, VIC, Australia
| | | | - Míriam Álvarez
- Microbiology Department, Centre Diagnòstic Biomèdic, Hospital Clínic, Barcelona, Spain
| | - Lynette Chee
- Haematology Department, Royal Melbourne Hospital, Melbourne, and Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Mateu Espasa
- Microbiology Department, Centre Diagnòstic Biomèdic, Hospital Clínic, Barcelona, Spain
| | - Ignacio Grafia
- Medical Oncology Department, Hospital Clínic-IDIBAPS, Barcelona, Spain
| | | | - Olivia Smibert
- Department of Infectious Diseases and Microbiology, Austin Health, Melbourne, VIC, Australia
| | - Carolina Garcia-Vidal
- Department of Infectious Diseases, Hospital Clínic-IDIBAPS, Barcelona, Spain; Universitat de Barcelona (UB), Barcelona, Spain; CIBERINF, CIBER in Infectious Diseases, Spain
| | - Monica A Slavin
- Victorian Infectious Diseases Service, Royal Melbourne Hospital, Melbourne, VIC, Australia; Department of Infectious Diseases and National Center for Infection, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, VIC, Australia
| | - Michelle K Yong
- Victorian Infectious Diseases Service, Royal Melbourne Hospital, Melbourne, VIC, Australia; Department of Infectious Diseases and National Center for Infection, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Alex Soriano
- Department of Infectious Diseases, Hospital Clínic-IDIBAPS, Barcelona, Spain; Universitat de Barcelona (UB), Barcelona, Spain; CIBERINF, CIBER in Infectious Diseases, Spain
| | - Leon J Worth
- Victorian Infectious Diseases Service, Royal Melbourne Hospital, Melbourne, VIC, Australia; Department of Infectious Diseases and National Center for Infection, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, VIC, Australia
| |
Collapse
|
10
|
Leroux-Roels I, Alhatemi A, Caubet M, De Boever F, de Wergifosse B, El Idrissi M, Ferreira GS, Jacobs B, Lambert A, Morel S, Servais C, Yarzabal JP. Safety and Immunogenicity of an Adjuvanted Clostridioides difficile Vaccine Candidate in Healthy Adults: A Randomized Placebo-Controlled Phase 1 Study. J Infect Dis 2025; 231:e511-e520. [PMID: 39447053 PMCID: PMC11911797 DOI: 10.1093/infdis/jiae466] [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: 02/29/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND This study investigated the safety, reactogenicity, and immunogenicity in healthy subjects of a Clostridioides difficile vaccine candidate with/without adjuvant, targeting toxins A and B. METHODS In this first-in-human, phase 1, observer-blind study, subjects aged 18-45 years were randomized to receive F2 antigen (n = 10) or placebo (n = 10), and subjects aged 50-70 years to receive F2 antigen plus AS01 adjuvant (n = 45), F2 antigen (n = 45), or placebo (n = 30) in 2 doses 1 month apart. A subcohort (n = 40) received a third dose 15 months later. Solicited adverse events (AEs) were recorded for 7 days and unsolicited AEs for 30 days after each dose. Immunogenicity was assessed at baseline and after each dose. RESULTS Solicited AEs were transient and most frequent in subjects receiving F2 antigen plus AS01. No serious AEs were considered related to study vaccine. Immunogenicity was substantially higher in subjects receiving F2 antigen plus AS01 than subjects receiving F2 antigen alone. A third dose increased the immune response in subjects with baseline neutralization titers below the assay lower limit of quantitation. CONCLUSIONS The GSK C. difficile vaccine candidate was immunogenic, especially when given with AS01, and was well tolerated with an acceptable safety profile. CLINICAL TRIAL REGISTRATION NCT04026009.
Collapse
Affiliation(s)
- Isabel Leroux-Roels
- Center for Vaccinology, Ghent University, Ghent University Hospital, Ghent, Belgium
| | - Azhar Alhatemi
- Center for Vaccinology, Ghent University, Ghent University Hospital, Ghent, Belgium
| | | | - Fien De Boever
- Center for Vaccinology, Ghent University, Ghent University Hospital, Ghent, Belgium
| | | | | | | | - Bart Jacobs
- Center for Vaccinology, Ghent University, Ghent University Hospital, Ghent, Belgium
| | | | | | | | | |
Collapse
|
11
|
Magnusson C, Ölfvingsson E, Hjortswang H, Östholm Å, Serrander L. Improved health-related quality of life in patients with recurrent Clostridioides difficile infection after treatment with faecal microbiota transplantation. Infect Dis (Lond) 2025; 57:239-246. [PMID: 39460926 DOI: 10.1080/23744235.2024.2415694] [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: 03/28/2024] [Revised: 08/26/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Clostridioides difficile is a major burden for both healthcare systems and the patients. Faecal microbiota transplantation (FMT) is becoming more common as a treatment since it reduces the risk of recurrent Clostridioides difficile infection (rCDI). OBJECTIVES To evaluate how treatment with FMT is affecting the health-related quality of life (HRQoL) in patients with rCDI. METHODS A prospective observational cohort study was conducted where patients who were offered FMT as a treatment for rCDI were asked to fill in a questionnaire based on the Short Health Scale (SHS) and EuroQol 5-Dimensions 5-Levels (EQ-5D-5L) about their HRQoL before and after treatment. RESULTS Patients with rCDI had poor HRQoL, which improved following FMT. CONCLUSIONS Since FMT cures, reduces the risk of new recurrences of CDI and improves the HRQoL of the patients, it should be offered as a treatment for patients with rCDI. Also, SHS is a useful and reliable instrument for measuring HRQoL in patients with rCDI.
Collapse
Affiliation(s)
- Cecilia Magnusson
- Department of Infectious Diseases, Region Jönköping County, Jönköping, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Elis Ölfvingsson
- Department of Infectious Diseases, Linköping University Hospital, Linköping, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Henrik Hjortswang
- Department of Gastroenterology and Hepatology, Linköping and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
| | - Åse Östholm
- Department of Infectious Diseases, Linköping University Hospital, Linköping, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Lena Serrander
- Department of Clinical Microbiology, and Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
12
|
Hirsch W, Fischer M, Khoruts A, Allegretti JR, Kelly CR, Vaughn B. Risk Factors for Antibiotic Exposure Post-Fecal Microbiota Transplantation for Recurrent Clostridioides difficile Infection: A Prospective Multicenter Observational Study. Open Forum Infect Dis 2025; 12:ofaf130. [PMID: 40103733 PMCID: PMC11913780 DOI: 10.1093/ofid/ofaf130] [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: 10/01/2024] [Accepted: 03/05/2025] [Indexed: 03/20/2025] Open
Abstract
Background Recurrent Clostridioides difficile infection (CDI) is primarily driven by antibiotic-induced disruption of the indigenous intestinal microbiota. Restoration of microbiota through fecal microbiota transplantation (FMT) is effective in preventing subsequent CDI, although this effect is attenuated with additional antibiotic exposure. The aim of this study was to identify the risk factors for recurrent antibiotic administration after FMT. Methods This is a prospective cohort of patients who were administered FMT for recurrent CDI from 1 July 2019 through 23 November 2023 across 6 institutions in the United States. Providers collected de-identified data at the time of FMT administration and in the months post-FMT administration. Results The analysis included 448 patients. Risk factors for non-CDI antibiotic administration within 2 months of FMT included immunocompromised status (odds ratio [OR], 2.2 [95% confidence interval {CI}, 1.1-4.4]; P = .02), >3 non-CDI antibiotic courses pre-FMT (OR, 3.1 [95% CI, 1.4-6.8]; P = .006), and prior hospitalization for CDI (OR, 2.0 [95% CI, 1.1-3.8]; P = .02). The most common indications for non-CDI antibiotic administration post-FMT were urinary tract infections, respiratory infections, and procedure prophylaxis. Conclusions Non-CDI antibiotic exposure significantly increases the risk of CDI recurrence post-FMT. Risk factors for non-CDI antibiotic administration within 2 months of FMT include immunocompromised status, multiple prior non-CDI antibiotics, and prior hospitalization for CDI. These individuals may benefit from additional or modified recurrent CDI prevention strategies.
Collapse
Affiliation(s)
- William Hirsch
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Monika Fischer
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Alexander Khoruts
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
- Center for Immunology, University of Minnesota, Minneapolis, Minnesota, USA
- BioTechnology Institute, University of Minnesota, St Paul, Minnesota, USA
| | | | - Colleen R Kelly
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Byron Vaughn
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
13
|
L'Huillier JC, Guo WA. The always evolving diagnosis and management of Clostridioides difficile colitis: What you need to know. J Trauma Acute Care Surg 2025; 98:357-367. [PMID: 39509684 DOI: 10.1097/ta.0000000000004474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
ABSTRACT The diagnosis, pharmacologic management, and surgical options for Clostridioides difficile infection (CDI) are rapidly evolving, which presents a challenge for the busy surgeon to remain up to date on the latest clinical guidelines. This review provides an evidence-based practical guide for CDI management tailored to the needs of surgeons and surgical intensivists. Historically, the diagnosis of CDI relied on slow cell culture cytotoxicity neutralization assays, but now, the rapidly resulting nucleic acid amplification tests and enzyme immunoassays have become mainstream. In terms of antibiotic therapy, metronidazole and oral vancomycin were the main "workhorse" antibiotics in the early 2000s, but large randomized controlled trials have now demonstrated that fidaxomicin produces superior results. Regarding surgical intervention, total abdominal colectomy was once the only procedure of choice; however, diverting loop ileostomy with colonic lavage is emerging as a viable alternative. Finally, novel adjuncts such as fecal microbiota transplantation and targeted therapy against toxin B (bezlotoxumab) are playing an increasingly important role in the management of CDI.
Collapse
Affiliation(s)
- Joseph C L'Huillier
- From the Department of Surgery (J.C.L., W.A.G.), Jacobs School of Medicine and Biomedical Sciences, and Division of Health Services Policy and Practice, Department of Epidemiology and Environmental Health (J.C.L.), School of Public Health and Health Professions, University at Buffalo; and Division of Trauma, Critical Care, and Acute Care Surgery, Department of Surgery (J.C.L., W.A.G.), Erie County Medical Center, Buffalo, New York
| | | |
Collapse
|
14
|
Hirai J, Mori N, Hanai Y, Asai N, Hagihara M, Mikamo H. Evaluating Bezlotoxumab-Fidaxomicin Combination Therapy in Clostridioides Infection: A Single-Center Retrospective Study from Aichi Prefecture, Japan. Antibiotics (Basel) 2025; 14:228. [PMID: 40149040 PMCID: PMC11939304 DOI: 10.3390/antibiotics14030228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/13/2025] [Accepted: 02/20/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives:Clostridioides difficile infection (CDI) poses a significant healthcare challenge, with recurrence rates reaching 30%, leading to substantial morbidity and costs. Fidaxomicin (FDX) and bezlotoxumab (BEZ) have shown potential in reducing recurrence; however, real-world data on the efficacy of their combination in high-risk CDI patients remain limited. This study aimed to evaluate the efficacy and safety of FDX + BEZ compared with FDX alone in CDI patients with recurrence risk factors. Methods: CDI patients with ≥two recurrence risk factors treated with FDX alone or FDX + BEZ were analyzed. Sixteen factors were evaluated as risk factors for recurrent CDI based on findings from previous studies. Patients with FDX treatment duration <10 days or other CDI treatment prior to FDX were excluded. Outcomes included recurrence within 2 months, global and clinical cure rates, and adverse events. Univariate and multivariate analyses were performed to evaluate efficacy. Results: Among 82 patients, the FDX + BEZ group (n = 30) demonstrated significantly higher global (86.7% vs. 65.4%; p < 0.05) and clinical cure rates (90.0% vs. 69.2%; p < 0.05) compared with the FDX-alone group (n = 52), despite more severe cases in the combination group. Recurrence rates were non-significantly lower in the FDX + BEZ group (3.3% vs. 11.5%). Combination therapy also accelerated diarrhea resolution without additional adverse events. Multivariate analysis identified FDX + BEZ as significantly associated with improved clinical cure (adjusted odds ratio 4.167; 95% CI: 1.029-16.885). Conclusions: FDX + BEZ therapy offers superior efficacy and safety in CDI patients with recurrence risk factors, presenting a promising strategy for optimizing CDI management.
Collapse
Affiliation(s)
- Jun Hirai
- Department of Clinical Infectious Diseases, Aichi Medical University Hospital, Aichi 480-1195, Japan; (J.H.); (N.M.); (N.A.)
- Department of Infection Control and Prevention, Aichi Medical University Hospital, Aichi 480-1195, Japan
- Division of Infection Control and Prevention, Nippon Medical School Chiba Hokusoh Hospital, Chiba 270-1694, Japan
| | - Nobuaki Mori
- Department of Clinical Infectious Diseases, Aichi Medical University Hospital, Aichi 480-1195, Japan; (J.H.); (N.M.); (N.A.)
- Department of Infection Control and Prevention, Aichi Medical University Hospital, Aichi 480-1195, Japan
| | - Yuki Hanai
- Department of Clinical Pharmacy, Faculty of Pharmaceutical Sciences, Toho University, Chiba 274-8510, Japan;
| | - Nobuhiro Asai
- Department of Clinical Infectious Diseases, Aichi Medical University Hospital, Aichi 480-1195, Japan; (J.H.); (N.M.); (N.A.)
- Department of Infection Control and Prevention, Aichi Medical University Hospital, Aichi 480-1195, Japan
| | - Mao Hagihara
- Department of Molecular Epidemiology and Biomedical Sciences, Aichi Medical University Hospital, Aichi 480-1195, Japan;
| | - Hiroshige Mikamo
- Department of Clinical Infectious Diseases, Aichi Medical University Hospital, Aichi 480-1195, Japan; (J.H.); (N.M.); (N.A.)
- Department of Infection Control and Prevention, Aichi Medical University Hospital, Aichi 480-1195, Japan
| |
Collapse
|
15
|
Naz F, Hagspiel N, Xu F, Thompson B, Brett Moreau G, Young M, Herbein J, Fox CB, Petri WA, Abhyankar MM. Enhanced immunogenicity of a Clostridioides difficile TcdB vaccine adjuvanted with a synthetic dual-TLR ligand adjuvant. NPJ Vaccines 2025; 10:33. [PMID: 39966390 PMCID: PMC11836405 DOI: 10.1038/s41541-025-01075-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 01/19/2025] [Indexed: 02/20/2025] Open
Abstract
We report a comprehensive evaluation of the toxin B (TcdB) vaccine adjuvanted with a dual Toll-like receptor ligand liposome adjuvant for Clostridioides difficile infection (CDI). The vaccine completely protected mice from a lethal infection. Compared to alum adjuvanted TcdB, it generated functionally superior systemic antibodies and supported strong memory B cell and gut IgA responses. This pharmaceutically acceptable adjuvant platform holds promise for developing a next-generation CDI vaccine.
Collapse
Affiliation(s)
- Farha Naz
- University of Virginia, Division of Infectious Diseases and International Health, Charlottesville, VA, 22908, USA
| | - Nicholas Hagspiel
- University of Virginia, Division of Infectious Diseases and International Health, Charlottesville, VA, 22908, USA
| | - Feifan Xu
- University of Virginia, Division of Infectious Diseases and International Health, Charlottesville, VA, 22908, USA
| | - Brandon Thompson
- University of Virginia, Division of Infectious Diseases and International Health, Charlottesville, VA, 22908, USA
| | - G Brett Moreau
- University of Virginia, Division of Infectious Diseases and International Health, Charlottesville, VA, 22908, USA
| | - Mary Young
- University of Virginia, Division of Infectious Diseases and International Health, Charlottesville, VA, 22908, USA
| | | | | | - William A Petri
- University of Virginia, Division of Infectious Diseases and International Health, Charlottesville, VA, 22908, USA
| | - Mayuresh M Abhyankar
- University of Virginia, Division of Infectious Diseases and International Health, Charlottesville, VA, 22908, USA.
| |
Collapse
|
16
|
Mourabiti F, Jouga F, Sakoui S, El Hosayny O, Zouheir Y, Soukri A, El Khalfi B. Mechanisms, therapeutic strategies, and emerging therapeutic alternatives for carbapenem resistance in Gram-negative bacteria. Arch Microbiol 2025; 207:58. [PMID: 39948320 DOI: 10.1007/s00203-025-04252-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/15/2025] [Accepted: 01/17/2025] [Indexed: 03/04/2025]
Abstract
Carbapenem-resistant Gram-negative bacteria (CR-GNB) have experienced an alarming surge in prevalence in recent years, escalating into a critical global healthcare crisis. As carbapenems represent the last line of defense against such pathogens, infections caused by CR-GNB have become increasingly challenging to treat, given the restricted therapeutic options and heightened mortality risks. The discovery and development of alternative therapeutic strategies that present novel avenues against multi-drug-resistant organisms are gaining increased attention, presenting a pressing need for innovative solutions. Our comprehensive review delves into the multifaceted landscape of carbapenem resistance in Gram-negative bacteria in response to this urgent challenge. The scope of this review aims to provide an up-to-date and in-depth exploration regarding the mode of action of carbapenem and the resisting mechanisms of carbapenem in GNB. Additionally, it discusses the state of the art of some clinical therapies for the treatment of infections caused by CR-GNB. Moreover, it describes several combinational and alternative therapies to combat CR-GNB, including the computational approach of "molecular docking". In light of the conclusions of this review, we call for the implementation of these strategies to develop comprehensive approaches to mitigate carbapenem resistance in Gram-negative bacteria.
Collapse
Affiliation(s)
- Fatima Mourabiti
- Laboratory of Physiopathology, Molecular Genetics & Biotechnology, Faculty of Sciences Ain Chock, Research Center of Health & Biotechnology, Hassan II University of Casablanca, 20100, Casablanca, Morocco
| | - Fatimazahra Jouga
- Laboratory of Physiopathology, Molecular Genetics & Biotechnology, Faculty of Sciences Ain Chock, Research Center of Health & Biotechnology, Hassan II University of Casablanca, 20100, Casablanca, Morocco
| | - Souraya Sakoui
- Laboratory of Physiopathology, Molecular Genetics & Biotechnology, Faculty of Sciences Ain Chock, Research Center of Health & Biotechnology, Hassan II University of Casablanca, 20100, Casablanca, Morocco
| | - Otmane El Hosayny
- Applied Language and Culture Studies Laboratory, Faculty of Letters and Human Sciences, Chouaib Doukkali University, 24000, El Jadida, Morocco
| | - Yassine Zouheir
- Laboratory of Molecular Bacteriology, Pasteur Institute, Casablanca, Morocco
| | - Abdelaziz Soukri
- Laboratory of Physiopathology, Molecular Genetics & Biotechnology, Faculty of Sciences Ain Chock, Research Center of Health & Biotechnology, Hassan II University of Casablanca, 20100, Casablanca, Morocco
| | - Bouchra El Khalfi
- Laboratory of Physiopathology, Molecular Genetics & Biotechnology, Faculty of Sciences Ain Chock, Research Center of Health & Biotechnology, Hassan II University of Casablanca, 20100, Casablanca, Morocco.
| |
Collapse
|
17
|
Xie Y, Irwin S, Nelson B, van Daelen M, Fontenot L, Jacobs JP, Cappelletti M, Feng H, Li Y, Koon HW. Citrulline Inhibits Clostridioides difficile Infection With Anti-inflammatory Effects. Cell Mol Gastroenterol Hepatol 2025; 19:101474. [PMID: 39923847 PMCID: PMC11986985 DOI: 10.1016/j.jcmgh.2025.101474] [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: 07/24/2024] [Revised: 01/30/2025] [Accepted: 02/01/2025] [Indexed: 02/11/2025]
Abstract
BACKGROUND & AIMS Clostridioides difficile infection (CDI) causes colitis and diarrhea. C. difficile bacterium produces toxins A and B, which cause intestinal inflammation. A metabolomics analysis discovered fecal metabolites with anti-inflammatory effects in CDI. We aimed to identify an anti-CDI metabolite that can inhibit CDI-mediated colitis and prevent recurrence. METHODS Fresh human colonic tissues and primary human cells were used to determine metabolite effects. Humanized C. difficile-infected HuCD34-NCG mice and antibiotics-treated human gut microbiota-treated (ABX + HGM) hamsters were used to simulate the human environment. RESULTS High-throughput screening and fecal metabolomics analysis identified anti-inflammatory metabolites. Compared with other tested metabolites, citrulline preserved the mucosal integrity of toxin-exposed fresh human colonic tissues with reduced macrophage inflammatory protein 1 alpha (MIP-1a) and increased interleukin-10 (IL-10) expression. Oral citrulline treatment alleviated cecal inflammation in hamsters infected with C. difficile ribotype 027. This was accomplished by the augmented expression of cecal IL-10 and the diminished level of cecal MIP-1a. Citrulline and vancomycin synergistically prevented recurrence in the infected ABX + HGM hamsters. In C57BL/6J mice infected with C. difficile VPI10463, citrulline ameliorated colitis by reducing colonic Ccl3 mRNA expression. In immunologically humanized HuCD34-NCG mice infected with toxin B-expressing C. difficile ribotype 017, citrulline ameliorated colitis with increased human IL-10 expression in colonic macrophages. Citrulline suppressed MIP-1a secretion and GSK3a/b dephosphorylation in the toxin A-exposed human colonic epithelial cells and promoted IL-10 expression in toxin B-exposed human macrophages and heat shock protein 27 phosphorylation. CONCLUSION Citrulline exerts anti-inflammatory effects in the intestines against C. difficile toxins and inhibits CDI recurrence in mice and hamsters.
Collapse
Affiliation(s)
- Ying Xie
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California; Department of Gastroenterology and Endoscopy, The First Hospital of China Medical University, Shenyang City, Liaoning Province, China
| | - Sophie Irwin
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California
| | - Becca Nelson
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California
| | - Mieke van Daelen
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California
| | - Lindsey Fontenot
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California
| | - Jonathan P Jacobs
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California; Goodman-Luskin Microbiome Center, David Geffen School of Medicine at UCLA, Los Angeles, California; Division of Gastroenterology, Hepatology and Parenteral Nutrition, VA Greater Los Angeles Healthcare System, Los Angeles, California
| | - Monica Cappelletti
- Immunogenetics Division, Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California
| | - Hanping Feng
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, Maryland
| | - Yiling Li
- Department of Gastroenterology and Endoscopy, The First Hospital of China Medical University, Shenyang City, Liaoning Province, China.
| | - Hon Wai Koon
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California Los Angeles, Los Angeles, California.
| |
Collapse
|
18
|
Vaughn BP, Khoruts A, Fischer M. Diagnosis and Management of Clostridioides difficile in Inflammatory Bowel Disease. Am J Gastroenterol 2025; 120:313-319. [PMID: 39230037 DOI: 10.14309/ajg.0000000000003076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 08/30/2024] [Indexed: 09/05/2024]
Abstract
Patients with inflammatory bowel disease (IBD) have an increased risk of Clostridioides difficile infection (CDI), which can lead to worse IBD outcomes. The diagnosis of CDI in patients with IBD is complicated by higher C. difficile colonization rates and shared clinical symptoms of intestinal inflammation. Traditional risk factors for CDI, such as antibiotic exposure, may be lacking in patients with IBD because of underlying intestinal microbiota dysbiosis. Although CDI disproportionately affects people with IBD, patients with IBD are typically excluded from CDI clinical trials creating a knowledge gap in the diagnosis and management of these 2 diseases. This narrative review aims to provide a comprehensive overview of the diagnosis, treatment, and prevention of CDI in patients with IBD. Distinguishing CDI from C. difficile colonization in the setting of an IBD exacerbation is important to avoid treatment delays. When CDI is diagnosed, extended courses of anti- C. difficile antibiotics may lead to better CDI outcomes. Regardless of a diagnosis of CDI, the presence of C. difficile in a patient with IBD should prompt a disease assessment of the underlying IBD. Microbiota-based therapies and bezlotoxumab seem to be effective in preventing CDI recurrence in patients with IBD. Patients with IBD should be considered at high risk of CDI recurrence and evaluated for a preventative strategy when diagnosed with CDI. Ultimately, the comanagement of CDI in a patient with IBD requires a nuanced, patient-specific approach to distinguish CDI from C. difficile colonization, prevent CDI recurrence, and manage the underlying IBD.
Collapse
Affiliation(s)
- Byron P Vaughn
- Division of Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis, Minnesota, USA
| | - Alexander Khoruts
- Division of Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis, Minnesota, USA
| | - Monika Fischer
- Division of Gastroenterology and Hepatology, Indiana University, Indianapolis, Indiana, USA
| |
Collapse
|
19
|
Vinterberg JE, Oddsdottir J, Nye M, Pinton P. Management of Recurrent Clostridioides difficile Infection (rCDI): A Systematic Literature Review to Assess the Feasibility of Indirect Treatment Comparison (ITC). Infect Dis Ther 2025; 14:327-355. [PMID: 39821840 PMCID: PMC11829878 DOI: 10.1007/s40121-024-01105-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 12/18/2024] [Indexed: 01/19/2025] Open
Abstract
Recurrent Clostridioides difficile infection (rCDI) is a major cause of increased morbidity, mortality, and healthcare costs. Fecal-microbiota-based therapies are recommended for rCDI on completion of standard-of-care (SoC) antibiotics to prevent further recurrence: these therapies include conventional fecal-microbiota transplantation and the US Food and Drug Administration-approved therapies REBYOTA® (RBL) and VOWST Oral Spores™ (VOS). As an alternative to microbiota-based therapies, bezlotoxumab, a monoclonal antibody, is used as adjuvant to SoC antibiotics to prevent rCDI. There are no head-to-head clinical trials comparing different microbiota-based therapies or bezlotoxumab for rCDI. To address this gap, we conducted a systematic literature review to identify clinical trials on rCDI treatments and assess the feasibility of using them to conduct an indirect treatment comparison (ITC). The feasibility analysis determined that trial heterogeneity, particularly relating to inclusion criteria, may significantly compromise ITC and prevent cross-trial comparisons. Our analysis underlines the need to adopt standardized protocols to ensure comparability across trials.
Collapse
Affiliation(s)
| | | | - Maria Nye
- EMEA RW Methods and Evidence Generation, IQVIA, Athens, Greece
| | - Philippe Pinton
- Global Research and Medical, Ferring Pharmaceuticals, Kastrup, Denmark.
- Global Research and Medical, International PharmaScience Center, Ferring Pharmaceuticals A/S, Amager Strandvej 405, 2770, Kastrup, Denmark.
| |
Collapse
|
20
|
Castro-Cordova P, Lopez-Garcia OK, Orozco J, Montes-Bravo N, Gil F, Pizarro-Guajardo M, Paredes-Sabja D. Clostridioides difficile major toxins remodel the intestinal epithelia, affecting spore adherence/internalization into intestinal tissue and their association with gut vitronectin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.29.635439. [PMID: 39974910 PMCID: PMC11838273 DOI: 10.1101/2025.01.29.635439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The most common cause of healthcare-associated diarrhea and colitis in the U.S., is Clostridioides difficile, a spore-forming pathogen. Two toxins, TcdA and TcdB, are major virulence factors essential for disease manifestations, while C. difficile spores are essential for disease transmission and recurrence. Both toxins cause major damage to the epithelial barrier, trigger massive inflammation, and reshape the microbiome and metabolic composition, facilitating C. difficile colonization. C. difficile spores, essential for transmission and recurrence of the disease, persist adhered and internalized in the intestinal epithelia. Studies have suggested that toxin-neutralization in combination with antibiotic during CDI treatment in humans significantly reduces disease recurrence, suggesting a link between toxin-mediated damage and spore persistence. Here, we show that TcdA/TcdB-intoxication of intestinal epithelial Caco-2 cells leads to remodeling of accessible levels of fibronectin (Fn) and vitronectin (Vn) and their cognate alpha-integrin subunits. While TcdB-intoxication of intestinal tissue had no impact in accessible levels of Fn and Vn, but significantly increased levels of intracellular Vn. We observed that Fn and Vn released to the supernatant readily bind to C. difficile spores in vitro, while TcdB-intoxication of intestinal tissue led to increased association of C. difficile spores with gut Vn. Toxin-intoxication of the intestinal tissue also contributes to increased adherence and internalization of C. difficile spores. However, TcdB-intoxicated ligated loops infected of mice treated with Bezlotoxumanb (monoclonal anti-TcdB antibodies) did not prevent TcdB-mediated increased spore adherence and internalization into intestinal tissue. This study highlights the importance of studying the impact of C. difficile toxins of host tissues has in C. difficile interaction with host surfaces that may contribute to increased persistence and disease recurrence.
Collapse
Affiliation(s)
- Pablo Castro-Cordova
- Millennium Nucleus in the Biology of Intestinal Microbiota, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Laboratory of Nano-Regenerative Medicine, Centro de Investigación e Innovación Biomédica (CiiB), Faculty of Medicine, Universidad de los Andes, Chile
| | - Osiris K. Lopez-Garcia
- Interdisciplinary Program in Genetics & Genomics, Texas A&M University, College Station, TX USA
- Department of Biology, Texas A&M University, College Station, TX USA
| | - Josué Orozco
- Millennium Nucleus in the Biology of Intestinal Microbiota, Santiago, Chile
| | | | - Fernando Gil
- Millennium Nucleus in the Biology of Intestinal Microbiota, Santiago, Chile
- Microbiota-Host Interactions & Clostridia Research Group, Universidad Andres Bello, Santiago, Chile
| | - Marjorie Pizarro-Guajardo
- Millennium Nucleus in the Biology of Intestinal Microbiota, Santiago, Chile
- Interdisciplinary Program in Genetics & Genomics, Texas A&M University, College Station, TX USA
| | - Daniel Paredes-Sabja
- Millennium Nucleus in the Biology of Intestinal Microbiota, Santiago, Chile
- Interdisciplinary Program in Genetics & Genomics, Texas A&M University, College Station, TX USA
- Department of Biology, Texas A&M University, College Station, TX USA
| |
Collapse
|
21
|
Morado F, Nanda N. A Review of Therapies for Clostridioides difficile Infection. Antibiotics (Basel) 2024; 14:17. [PMID: 39858303 PMCID: PMC11762378 DOI: 10.3390/antibiotics14010017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
Clostridioides difficile is an urgent public health threat that affects approximately half a million patients annually in the United States. Despite concerted efforts aimed at the prevention of Clostridioides difficile infection (CDI), it remains a leading cause of healthcare-associated infections. CDI is associated with significant clinical, social, and economic burdens. Therefore, it is imperative to provide optimal and timely therapy for CDI. We conducted a systematic literature review and offer treatment recommendations based on available evidence for the treatment and prevention of CDI.
Collapse
Affiliation(s)
- Faiza Morado
- Department of Pharmacy, Keck Medical Center, University of Southern California, Los Angeles, CA 90033, USA;
| | - Neha Nanda
- Division of Infectious Diseases, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
22
|
Kuijper EJ, Gerding DN. The End of Toxoid Vaccine Development for Preventing Clostridioides difficile Infections? Clin Infect Dis 2024; 79:1512-1514. [PMID: 39178347 DOI: 10.1093/cid/ciae412] [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: 06/27/2024] [Revised: 07/10/2024] [Accepted: 08/21/2024] [Indexed: 08/25/2024] Open
Abstract
Abstract
Collapse
Affiliation(s)
- Ed J Kuijper
- National Expertise Center for Clostridioides difficile Infections of Leiden University Center for Infectious Diseases, Leiden University Medical Centre (LUMC), Leiden, The Netherlands and Center for Infectious Disease Control (CIb), National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Dale N Gerding
- Edward Hines, Jr, Veterans Affairs Hospital, Hines, Illinois, USA
| |
Collapse
|
23
|
Pettit NN, Shaeer KM, Chahine EB. Live Biotherapeutic Products for the Prevention of Recurrent Clostridioides difficile Infection. Ann Pharmacother 2024; 58:1204-1217. [PMID: 38546138 DOI: 10.1177/10600280241239685] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2024] Open
Abstract
OBJECTIVE To review the efficacy, safety, and role of live biotherapeutic products (LBPs) in the prevention of recurrent Clostridioides difficile infection (rCDI). DATA SOURCES A literature search was performed using PubMed and Google Scholar (through February 2024) with search terms RBX2660, SER-109, and fecal microbiota. Other resources included abstracts presented at recent conferences, national clinical practice guidelines, and manufacturers' websites. STUDY SELECTION AND DATA EXTRACTION All relevant studies, trial updates, conference abstracts, and guidelines in the English language were included. DATA SYNTHESIS Two LBPs were recently approved by the Food and Drug Administration for the prevention of recurrence in adults following antibiotic treatment for rCDI. Fecal microbiota, live-jslm is administered rectally as a retention enema, whereas fecal microbiota spores, live-brpk is given orally after bowel preparation. Several phase 2 and phase 3 clinical trials have established the safety and efficacy of these LBPs in reducing rates of rCDI compared with placebo. Patients with severe immunosuppression and those with inflammatory bowel disease were largely excluded from these trials. RELEVANCE TO PATIENT CARE AND CLINICAL PRACTICE IN COMPARISON WITH EXISTING DRUGS Live biotherapeutic products offer a similar mechanism to conventional fecal microbiota transplant (FMT) in preventing rCDI through microbiota restoration. The primary advantages of LBPs over FMT are their standardized composition and donor stool screening processes for transmissible pathogens. Bezlotoxumab is also available for the prevention of Clostridioides difficile infection; however, there are no clinical data available to compare the efficacy of LBPs with bezlotoxumab, and the benefit of simultaneous use of these preventative therapies is unclear. CONCLUSIONS Live biotherapeutic products provide a safe and effective option for the prevention of rCDI and represent an improvement over conventional FMT. Additional studies are needed to further determine their place in therapy relative to bezlotoxumab and in the setting of immunosuppression and inflammatory bowel disease.
Collapse
Affiliation(s)
| | - Kristy M Shaeer
- Department of Pharmacotherapeutics & Clinical Research, University of South Florida Taneja College of Pharmacy, Tampa, FL, USA
| | - Elias B Chahine
- Department of Pharmacy Practice, Palm Beach Atlantic University Lloyd L. Gregory School of Pharmacy, West Palm Beach, FL, USA
| |
Collapse
|
24
|
Chanket W, Pipatthana M, Sangphukieo A, Harnvoravongchai P, Chankhamhaengdecha S, Janvilisri T, Phanchana M. The complete catalog of antimicrobial resistance secondary active transporters in Clostridioides difficile: evolution and drug resistance perspective. Comput Struct Biotechnol J 2024; 23:2358-2374. [PMID: 38873647 PMCID: PMC11170357 DOI: 10.1016/j.csbj.2024.05.027] [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: 02/08/2024] [Revised: 05/01/2024] [Accepted: 05/16/2024] [Indexed: 06/15/2024] Open
Abstract
Secondary active transporters shuttle substrates across eukaryotic and prokaryotic membranes, utilizing different electrochemical gradients. They are recognized as one of the antimicrobial efflux pumps among pathogens. While primary active transporters within the genome of C. difficile 630 have been completely cataloged, the systematical study of secondary active transporters remains incomplete. Here, we not only identify secondary active transporters but also disclose their evolution and role in drug resistance in C. difficile 630. Our analysis reveals that C. difficile 630 carries 147 secondary active transporters belonging to 27 (super)families. Notably, 50 (34%) of them potentially contribute to antimicrobial resistance (AMR). AMR-secondary active transporters are structurally classified into five (super)families: the p-aminobenzoyl-glutamate transporter (AbgT), drug/metabolite transporter (DMT) superfamily, major facilitator (MFS) superfamily, multidrug and toxic compound extrusion (MATE) family, and resistance-nodulation-division (RND) family. Surprisingly, complete RND genes found in C. difficile 630 are likely an evolutionary leftover from the common ancestor with the diderm. Through protein structure comparisons, we have potentially identified six novel AMR-secondary active transporters from DMT, MATE, and MFS (super)families. Pangenome analysis revealed that half of the AMR-secondary transporters are accessory genes, which indicates an important role in adaptive AMR function rather than innate physiological homeostasis. Gene expression profile firmly supports their ability to respond to a wide spectrum of antibiotics. Our findings highlight the evolution of AMR-secondary active transporters and their integral role in antibiotic responses. This marks AMR-secondary active transporters as interesting therapeutic targets to synergize with other antibiotic activity.
Collapse
Affiliation(s)
- Wannarat Chanket
- Graduate Program in Molecular Medicine, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Methinee Pipatthana
- Department of Microbiology, Faculty of Public Health, Mahidol University, Bangkok, Thailand
| | - Apiwat Sangphukieo
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | | | | | - Tavan Janvilisri
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Matthew Phanchana
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| |
Collapse
|
25
|
Herbin SR, Crum H, Gens K. Breaking the Cycle of Recurrent Clostridioides difficile Infections: A Narrative Review Exploring Current and Novel Therapeutic Strategies. J Pharm Pract 2024; 37:1361-1373. [PMID: 38739837 DOI: 10.1177/08971900241248883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Clostridioides difficile is a toxin-producing bacteria that is a main cause of antibiotic-associated diarrhea. Clostridioides difficile infections (CDI) are associated with disruptions within the gastrointestinal (GI) microbiota which can be further exacerbated by CDI-targeted antibiotic treatment thereby causing recurrent CDI (rCDI) and compounding the burden placed on patients and the healthcare system. Treatment of rCDI consists of antibiotics which can be paired with preventative therapeutics, such as bezlotoxumab or fecal microbiota transplants (FMTs), if sustained clinical response is not obtained. Newer preventative strategies have been recently approved to assist in restoring balance within the GI system with the goal of preventing recurrent infections.
Collapse
Affiliation(s)
- Shelbye R Herbin
- Antimicrobial Stewardship and Medication Safety, John D. Dingell VA Medical Center, Detroit, MI, USA
| | - Hannah Crum
- Mercy Hospital Southeast, Cape Girardeau, MO, USA
| | - Krista Gens
- Allina Health, Minneapolis, MN, USA
- Abbott Northwestern Hospital, Minneapolis, MN, USA
| |
Collapse
|
26
|
Khanna S. Microbiota restoration for recurrent Clostridioides difficile infection. Panminerva Med 2024; 66:417-426. [PMID: 39382853 DOI: 10.23736/s0031-0808.24.05111-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
Since the publication of the recent North American and European guidelines on management of Clostridioides difficile infection (CDI), new evidence describing the epidemiology, testing and treatment of CDI has emerged. Despite all advances in infection control and antibiotic stewardship, the incidence and burden of CDI in the hospitals and the community remains at a stable high. Coupled with the incidence of primary CDI, there is a stable high incidence of recurrent CDI. Testing for primary and recurrent CDI remains a clinical challenge owing to high sensitivity of the PCR (leading to false positives) and somewhat limited sensitivity of EIA for toxin. The pathophysiology of recurrent CDI involves an ongoing disruption of the microbiota owing to the infection and the treatment of CDI employed. Broad spectrum antibiotics such as vancomycin leads to further disruption of microbiota compared to fidaxomicin which has a lower disruption of the microbiota and leads to fewer recurrences. Owing to these data fidaxomicin is considered as the first line antibiotic for recurrent CDI. Intravenous bezlotoxumab is a monoclonal antibody that reduces the risk of recurrence in high-risk patients but does not restore the microbiota. Experimental fecal microbiota transplantation (FMT) has been available for more than a decade. Owing to the success of FMT, two new non-invasive donor dependent Food and Drug Administration (FDA) approved therapies have been available since late 2022. This review summarizes all these conundrums regarding CDI and provides clinical pearls to use in day-to-day practice.
Collapse
Affiliation(s)
- Sahil Khanna
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA -
| |
Collapse
|
27
|
Rubak T, Baunwall SMD, Gregersen M, Paaske SE, Asferg M, Barat I, Secher-Johnsen J, Riis MG, Rosenbæk JB, Hansen TK, Ørum M, Steves CJ, Veilbæk H, Hvas CL, Damsgaard EMS. Early geriatric assessment and management in older patients with Clostridioides difficile infection in Denmark (CLODIfrail): a randomised trial. THE LANCET. HEALTHY LONGEVITY 2024; 5:100648. [PMID: 39488230 DOI: 10.1016/j.lanhl.2024.100648] [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: 04/30/2024] [Revised: 09/21/2024] [Accepted: 09/26/2024] [Indexed: 11/04/2024] Open
Abstract
BACKGROUND Clostridioides difficile infection causes diarrhoea and colitis. Older patients with C difficile infection are often frail and have comorbidities, leading to high mortality rates. The frailty burden in older people might restrict access to treatments, such as C difficile infection-specific antibiotics and faecal microbiota transplantation. We aimed to investigate the clinical effects of early comprehensive geriatric assessment (CGA) and frailty evaluation, including home visits and assessment for faecal microbiota transplantation, in older patients with C difficile infection. METHODS In this randomised, quality improvement trial with a pragmatic design, patients from the Central Denmark Region aged 70 years or older with a positive PCR test for C difficile toxin were randomly assigned (1:1) to CGA or standard care, both with equal access to faecal microbiota transplantation. Patients and investigators were unmasked to treatment. The primary outcome was 90-day mortality, and was compared in the study groups according to the intention-to-treat principle. The study is registered with ClinicalTrials.gov, NCT05447533. FINDINGS Between Sept 1, 2022, and May 3, 2023, we randomly assigned 217 patients to CGA (n=109) or standard care (n=108). The median patient age was 78 years (IQR 74-84). 116 (53%) of 217 patients were female and 101 (47%) were male. 16 (15%; 95% CI 9-23) of 109 patients in the CGA group and 22 (20%; 14-29) of 108 patients in the standard-care group died within 90 days (odds ratio 0·66, 95% CI 0·32-1·38. No serious adverse events or deaths related to patient assessment or faecal microbiota transplantation were recorded in either group. Deaths directly attributable to C difficile infection were lower in the CGA group (seven [44%] of 16 deaths vs 18 [82%] of 22 deaths in the standard-care group; p=0·020). INTERPRETATION Older patients who received CGA had a 90-day mortality rate similar to that of patients who received standard care, but with fewer deaths directly attributable to C difficile infection. FUNDING Innovation Fund Denmark, Novo Nordisk Foundation, and Helsefonden.
Collapse
Affiliation(s)
- Tone Rubak
- Department of Geriatrics Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| | | | - Merete Gregersen
- Department of Geriatrics Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Sara Ellegaard Paaske
- Department of Hepatology and Gastroenterology Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Malene Asferg
- Geriatric Section, Department of Medicine, Silkeborg Regional Hospital, Silkeborg, Denmark
| | - Ishay Barat
- Geriatric Section, Department of Medicine, Horsens Regional Hospital, Horsens, Denmark
| | - Joanna Secher-Johnsen
- Geriatric Section, Department of Medicine, Viborg Regional Hospital, Viborg, Denmark
| | - Mikael Groth Riis
- Geriatric Section, Department of Medicine, Randers Regional Hospital, Randers, Denmark
| | | | - Troels Kjærskov Hansen
- Department of Geriatrics Aarhus University Hospital, Aarhus, Denmark; Medical department, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Marianne Ørum
- Department of Geriatrics Aarhus University Hospital, Aarhus, Denmark
| | - Claire J Steves
- Department of Ageing and Health, Guy's and St Thomas' NHS Foundation Trust and Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Hanne Veilbæk
- Department of Geriatrics Aarhus University Hospital, Aarhus, Denmark
| | - Christian Lodberg Hvas
- Department of Hepatology and Gastroenterology Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Else Marie Skjøde Damsgaard
- Department of Geriatrics Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
28
|
Naz F, Hagspiel N, Xu F, Thompson B, Moreau GB, Young M, Herbein J, Fox CB, Petri WA, Abhyankar MM. Enhanced immunogenicity of a Clostridioides difficile TcdB vaccine adjuvanted with a synthetic dual-TLR ligand adjuvant. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.25.625229. [PMID: 39651154 PMCID: PMC11623652 DOI: 10.1101/2024.11.25.625229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
We report a comprehensive evaluation of the toxin B (TcdB) vaccine adjuvanted with a dual Toll-like receptor ligand liposome adjuvant for Clostridioides difficile infection (CDI). The vaccine completely protected mice from a lethal infection. Compared to alum adjuvanted TcdB, it generated functionally superior systemic antibodies and supported strong memory B cell and gut IgA responses. This pharmaceutically acceptable adjuvant platform holds promise for developing a next-generation CDI vaccine.
Collapse
|
29
|
Luo F, Xu C, Zhang C, Tan A, Lu D, Luo P, Cheng P, Zhang W, Bai L, Yu C, Sun S, Zeng H, Zou Q. mRNA-based platform for preventing and treating Staphylococcus aureus by targeted staphylococcal enterotoxin B. Front Immunol 2024; 15:1490044. [PMID: 39640268 PMCID: PMC11617584 DOI: 10.3389/fimmu.2024.1490044] [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: 09/02/2024] [Accepted: 10/29/2024] [Indexed: 12/07/2024] Open
Abstract
Staphylococcus aureus (S. aureus) possesses numerous virulence factors, with the increasing prevalence of drug-resistant strains heightening the threat posed by this pathogen. Staphylococcal enterotoxin B (SEB), a highly conserved toxin secreted by S. aureus, is also recognized as a potential bioweapon with super-antigenic activity. SEB represents a promising target in efforts to combat infections caused by S. aureus. We developed mRNA-based vaccine and antibody targeting SEB for both prophylactic and therapeutic purposes in varying S. aureus infection conditions. The mSEB mRNA vaccine (10 μg per mouse) induces more robust and persistent immune responses, including higher antibody titers and specific cellular immune responses, compared to immunization with 30 μg of mSEB protein adjuvanted with aluminum phosphate. Additionally, the anti-SEB mRNA antibody maintains secretion of anti-SEB monoclonal antibody (mAb) with a dosage that is 10 times lower than purified protein administration. The mRNA-based antibody exhibits superior pharmacokinetic profiles compared to its protein counterparts, efficiently neutralizing SEB and clearing S. aureus from circulation. Both the mRNA vaccine and mRNA antibody demonstrate preventive and therapeutic effects by eliciting specific immune responses and generating high-affinity antibodies in mice. We have laid the groundwork for the development and evaluation of mRNA-based vaccines and antibodies targeting SEB produced by S. aureus. Our studies demonstrate that these approaches are more effective than traditional protein-based vaccines and antibodies in terms of inducing immune responses, pharmacokinetics, and their prophylactic or therapeutic efficacy against S. aureus infections.
Collapse
Affiliation(s)
- Fumei Luo
- School of Pharmacy, University of South China, Hunan, China
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| | - Chuanfei Xu
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| | - Chengwen Zhang
- Medical Research Institute, Southwest University, Chongqing, China
| | - Aomo Tan
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Dongshui Lu
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| | - Ping Luo
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| | - Ping Cheng
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| | - Weijun Zhang
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| | - Lijuan Bai
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Cuiyun Yu
- School of Pharmacy, University of South China, Hunan, China
| | - Si Sun
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| | - Hao Zeng
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| | - Quanming Zou
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing, China
| |
Collapse
|
30
|
Tsoumbris PR, Vincent RM, Jaschke PR. Designing a simple and efficient phage biocontainment system using the amber suppressor initiator tRNA. Arch Virol 2024; 169:248. [PMID: 39557717 DOI: 10.1007/s00705-024-06170-9] [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: 07/29/2024] [Accepted: 10/21/2024] [Indexed: 11/20/2024]
Abstract
Multidrug-resistant infections are becoming increasingly prevalent worldwide. One of the fastest-emerging alternative and adjuvant therapies being proposed is phage therapy. Naturally isolated phages are used in the vast majority of phage therapy treatments today. Engineered phages are being developed to enhance the effectiveness of phage therapy, but concerns over their potential escape remain a salient issue. To address this problem, we designed a biocontained phage system based on conditional replication using amber stop codon suppression. This system can be easily installed on any natural phage with a known genome sequence. To test the system, we individually mutated the start codons of three essential capsid genes in phage φX174 to the amber stop codon (UAG). These phages were able to efficiently infect host cells expressing the amber initiator tRNA, which suppresses the amber stop codon and initiates translation at TAG stop codons. The amber phage mutants were also able to successfully infect host cells and reduce their population on solid agar and liquid culture but could not produce infectious particles in the absence of the amber initiator tRNA or complementing capsid gene. We did not detect any growth-inhibiting effects on E. coli strains known to lack a receptor for φX174 and we showed that engineered phages have a limited propensity for reversion. The approach outlined here may be useful to control engineered phage replication in both the lab and clinic.
Collapse
Affiliation(s)
- Pamela R Tsoumbris
- School of Natural Sciences, Macquarie University, Sydney, New South Wales, 2109, Australia
- ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney, Australia
| | - Russel M Vincent
- School of Natural Sciences, Macquarie University, Sydney, New South Wales, 2109, Australia
- ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney, Australia
| | - Paul R Jaschke
- School of Natural Sciences, Macquarie University, Sydney, New South Wales, 2109, Australia.
- ARC Centre of Excellence in Synthetic Biology, Macquarie University, Sydney, Australia.
| |
Collapse
|
31
|
Naz F, Hagspiel N, Young MK, Uddin J, Tyus D, Boone R, Brown AC, Ramakrishnan G, Rigo I, Madden GR, Petri WA. IL-33 protects from recurrent C. difficile infection by restoration of humoral immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.16.623943. [PMID: 39605647 PMCID: PMC11601440 DOI: 10.1101/2024.11.16.623943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Clostridioides difficile infection (CDI) recurs in one of five patients. Monoclonal antibodies targeting the virulence factor TcdB reduce disease recurrence, suggesting that an inadequate anti-TcdB response to CDI leads to recurrence. In patients with CDI, we discovered that IL-33 measured at diagnosis predicts future recurrence, leading us to test the role of IL-33 signaling in the induction of humoral immunity during CDI. Using a mouse recurrence model, IL-33 was demonstrated to be integral for anti-TcdB antibody production. IL-33 acted via ST2+ ILC2 cells, facilitating germinal center T follicular helper (GC-Tfh) cell generation of antibodies. IL-33 protection from reinfection was antibody-dependent, as μMT KO mice and mice treated with anti-CD20 mAb were not protected. These findings demonstrate the critical role of IL-33 in generating humoral immunity to prevent recurrent CDI.
Collapse
Affiliation(s)
- Farha Naz
- Department of Medicine, Division of Infectious Diseases and International Health, Charlottesville, Virginia, USA
| | - Nicholas Hagspiel
- Department of Medicine, Division of Infectious Diseases and International Health, Charlottesville, Virginia, USA
| | - Mary K. Young
- Department of Medicine, Division of Infectious Diseases and International Health, Charlottesville, Virginia, USA
| | - Jashim Uddin
- Department of Medicine, Division of Infectious Diseases and International Health, Charlottesville, Virginia, USA
| | - David Tyus
- Department of Medicine, Division of Infectious Diseases and International Health, Charlottesville, Virginia, USA
| | - Rachel Boone
- Department of Microbiology, Immunology and Cancer Biology, Charlottesville, Virginia, USA
| | - Audrey C. Brown
- Department of Medicine, Division of Infectious Diseases and International Health, Charlottesville, Virginia, USA
| | - Girija Ramakrishnan
- Department of Medicine, Division of Infectious Diseases and International Health, Charlottesville, Virginia, USA
| | - Isaura Rigo
- Department of Medicine, Division of Infectious Diseases and International Health, Charlottesville, Virginia, USA
| | - Gregory R. Madden
- Department of Medicine, Division of Infectious Diseases and International Health, Charlottesville, Virginia, USA
| | - William A. Petri
- Department of Medicine, Division of Infectious Diseases and International Health, Charlottesville, Virginia, USA
- Department of Microbiology, Immunology and Cancer Biology, Charlottesville, Virginia, USA
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|
32
|
Kuchay RAH. Novel and emerging therapeutics for antimicrobial resistance: A brief review. Drug Discov Ther 2024; 18:269-276. [PMID: 39462601 DOI: 10.5582/ddt.2024.01063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
A pandemic known as anti-microbial resistance (AMR) poses a challenge to contemporary medicine. To stop AMR's rise and quick worldwide spread, urgent multisectoral intervention is needed. This review will provide insight on new and developing treatment approaches for AMR. Future therapy options may be made possible by the development of novel drugs that make use of developments in "omics" technology, artificial intelligence, and machine learning. Vaccines, immunoconjugates, antimicrobial peptides, monoclonal antibodies, and nanoparticles may also be intriguing options for treating AMR in the future. Combination therapy may potentially prove to be a successful strategy for combating AMR. To lessen the impact of AMR, ideas like drug repurposing, antibiotic stewardship, and the one health approach may be helpful.
Collapse
|
33
|
Zasheva A, Batcheva E, Ivanova KD, Yanakieva A. Differences in Patient Access to Newly Approved Antibacterial Drugs in EU/EEA Countries. Antibiotics (Basel) 2024; 13:1077. [PMID: 39596770 PMCID: PMC11591277 DOI: 10.3390/antibiotics13111077] [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: 09/30/2024] [Revised: 11/02/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Abstract
The introduction of antibiotics in the beginning of the 20th century was one of the most important scientific breakthroughs in history. However, in recent decades, the growing threat of antimicrobial resistance (AMR) has shown the limitations of the current research and development programs for new antimicrobial drugs. In the last decade, 20 antibiotics, 7 β-lactam/β-lactamase inhibitor (BL/BLI) combinations and 4 non-traditional antibacterial drugs have been launched worldwide. METHODS This study aimed to assess the time to patient access for new antibacterial drugs in countries in the European Union and the European Economic Area (EU/EEA). Time differences in marketing authorization from the U.S. Food and Drug Agency (FDA) and the European Medicines Agency (EMA) were also described, as well as the availability of each drug in the countries in the EU/EEA according to the national competent authorities. RESULTS Substantial differences between countries were observed, with no or only one new drug available in some countries. CONCLUSIONS Improving pricing and reimbursement timelines and fostering collaboration between national health authorities and market authorization holders can enhance timely and equitable patient access to new antibacterial treatments in Europe. Equitable and sustainable access to antibacterial drugs is a cornerstone in the battle against AMR.
Collapse
Affiliation(s)
- Anelia Zasheva
- Department of Infectious Diseases, Military Medical Academy, 1606 Sofia, Bulgaria
| | - Elina Batcheva
- Department of Pharmacology, Pharmacotherapy and Toxicology, Faculty of Pharmacy, Medical University—Sofia, 1000 Sofia, Bulgaria;
| | - Kremena Dimitrova Ivanova
- Department of Health Technology Assessment, Faculty of Public Health, Medical University—Sofia, 1527 Sofia, Bulgaria; (K.D.I.); (A.Y.)
| | - Antoniya Yanakieva
- Department of Health Technology Assessment, Faculty of Public Health, Medical University—Sofia, 1527 Sofia, Bulgaria; (K.D.I.); (A.Y.)
| |
Collapse
|
34
|
Doroudian M, Ardalan MA, Beheshti M, Soezi M. Novel approaches for bacterial toxin neutralization; current advances and future perspectives. QJM 2024; 117:763-767. [PMID: 38851872 DOI: 10.1093/qjmed/hcae113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 05/23/2024] [Indexed: 06/10/2024] Open
Abstract
This review outlines diverse strategies for neutralizing bacterial toxins which are a significant threat to human health. Effective toxin neutralization is crucial in preventing and treating bacterial infections, especially those caused by antibiotic-resistant strains. Promising approaches include using monoclonal antibodies that target toxins and combining them with agents that directly target bacteria. Aptamers, synthetic molecules that bind to specific targets, provide a rapid and tailored method for inhibiting toxin activity and detecting pathogens. Cell-membrane-coated nanoparticles mimic host cells and effectively neutralize toxins by diverting them and stimulating immune responses. These advancements have the potential to combat bacterial infections and alleviate the associated public health burden.
Collapse
Affiliation(s)
- M Doroudian
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - M A Ardalan
- Internal Medicine Department, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - M Beheshti
- Department of Microbiology, Faculty of Veterinari Medicine, University of Tehran, Iran, Tehran, Iran
| | - M Soezi
- Infection Disease Research Center, AJA University of Medical Sciences, Tehran, Iran
- Medical Biotechnology Research Center, AJA University of Medical Sciences, Tehran, Iran
| |
Collapse
|
35
|
Hassoun-Kheir N, Guedes M, Arieti F, Pezzani MD, Gladstone BP, Robotham JV, Pouwels KB, Kingston R, Carmeli Y, Cassini A, Cecchini M, Drobniewski F, Frost I, Geurtsen J, Kronenberg A, Htay MNN, Paul M, Rocha-Pereira N, Rodríguez-Baño J, Scudeller L, Stewardson AJ, Tacconelli E, Harbarth S, Vella V, de Kraker ME. Expert consensus on antimicrobial resistance research priorities to focus development and implementation of antibacterial vaccines and monoclonal antibodies. Euro Surveill 2024; 29:2400212. [PMID: 39574390 PMCID: PMC11583311 DOI: 10.2807/1560-7917.es.2024.29.47.2400212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 10/07/2024] [Indexed: 11/24/2024] Open
Abstract
To reduce antimicrobial resistance (AMR), pathogen-specific AMR burden data are crucial to guide target selection for research and development of vaccines and monoclonal antibodies (mAbs). We identified knowledge gaps through previously conducted systematic reviews, which informed a Delphi expert consultation on future AMR research priorities and harmonisation strategies to support data-driven decision-making. Consensus (≥80% agreement) on importance and feasibility of research topics was achieved in two rounds, involving 24 of 39 and 19 of 24 invited experts, respectively. Priority pathogens and resistance profiles for future research were identified: third generation cephalosporin-resistant Klebsiella pneumoniae and Escherichia coli, for bloodstream and urinary tract infections, respectively, and meticillin-resistant Staphylococcus aureus for surgical-site infections. Prioritised high-risk populations included surgical, haemato-oncological and transplant patients. Mortality and resource use were prioritised as health-economic outcomes. The importance of age-stratified data and inclusion of a non-infected comparator group were highlighted. This agenda provides guidance for future research to fill knowledge gaps and support data-driven selection of target pathogens and populations for new preventive and treatment strategies, specifically vaccines and mAbs, to effectively address the AMR burden in Europe. These research priorities are also relevant to improve the evidence base for future AMR burden estimates.
Collapse
Affiliation(s)
- Nasreen Hassoun-Kheir
- Infection Control Program, Geneva University Hospitals and Faculty of Medicine, WHO Collaborating Center, Geneva, Switzerland
| | - Mariana Guedes
- Instituto de Biomedicina de Sevilla (IBiS), Infectious Diseases and Microbiology Division, Hospital Universitario Virgen Macarena, Department of Medicine, University of Sevilla/CSIC, Sevilla, Spain
- Infection and Antimicrobial Resistance Control and Prevention Unit, Hospital Epidemiology Centre, Centro Hospitalar Universitário São João, Porto, Portugal
| | - Fabiana Arieti
- Infectious Diseases, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Maria Diletta Pezzani
- Infectious Diseases, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Beryl Primrose Gladstone
- DZIF-Clinical Research Unit, Infectious Diseases, Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Julie V Robotham
- HCAI, Fungal, AMR, AMU & Sepsis Division, UK Health Security Agency, London, United Kingdom
| | - Koen B Pouwels
- Health Economics Research Centre, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Rhys Kingston
- HCAI, Fungal, AMR, AMU & Sepsis Division, UK Health Security Agency, London, United Kingdom
| | - Yehuda Carmeli
- National Institute for Antibiotic Resistance and Infection Control, Ministry of Health, Tel Aviv; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Alessandro Cassini
- Infectious Diseases Service, Lausanne University Hospital, Lausanne, Switzerland and Public Health Department, Canton of Vaud, Lausanne, Switzerland
| | - Michele Cecchini
- Head of Public Health, Health Division, OECD (Organisation for Economic Co-operation and Development), Paris, France
| | - Francis Drobniewski
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Isabel Frost
- AstraZeneca, Eastbrook House, Cambridge, United Kingdom
| | - Jeroen Geurtsen
- Bacterial Vaccines Research & Early Development, Janssen Vaccines & Prevention B.V., Leiden, the Netherlands
| | - Andreas Kronenberg
- Swiss Centre for Antibiotic Resistance, Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Mila Nu Nu Htay
- Department of Community Medicine, Faculty of Medicine, Manipal University College Malaysia, Melaka, Malaysia
| | - Mical Paul
- Infectious Diseases Institute, Rambam Health Care Campus; Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Nuno Rocha-Pereira
- Department of Medicine, Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- Infection and Antimicrobial Resistance Control and Prevention Unit, Hospital Epidemiology Centre, Centro Hospitalar Universitário São João, Porto, Portugal
| | - Jesús Rodríguez-Baño
- CIBERINFEC, Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Biomedicina de Sevilla (IBiS), Infectious Diseases and Microbiology Division, Hospital Universitario Virgen Macarena, Department of Medicine, University of Sevilla/CSIC, Sevilla, Spain
| | - Luigia Scudeller
- Research and Innovation Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Andrew J Stewardson
- Department of Infectious Diseases, The Alfred Hospital and School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Evelina Tacconelli
- Infectious Diseases, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Stephan Harbarth
- Infection Control Program, Geneva University Hospitals and Faculty of Medicine, WHO Collaborating Center, Geneva, Switzerland
| | | | - Marlieke Ea de Kraker
- Infection Control Program, Geneva University Hospitals and Faculty of Medicine, WHO Collaborating Center, Geneva, Switzerland
| |
Collapse
|
36
|
Edgar JE, Bournazos S. Fc-FcγR interactions during infections: From neutralizing antibodies to antibody-dependent enhancement. Immunol Rev 2024; 328:221-242. [PMID: 39268652 PMCID: PMC11659939 DOI: 10.1111/imr.13393] [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: 09/17/2024]
Abstract
Advances in antibody technologies have resulted in the development of potent antibody-based therapeutics with proven clinical efficacy against infectious diseases. Several monoclonal antibodies (mAbs), mainly against viruses such as SARS-CoV-2, HIV-1, Ebola virus, influenza virus, and hepatitis B virus, are currently undergoing clinical testing or are already in use. Although these mAbs exhibit potent neutralizing activity that effectively blocks host cell infection, their antiviral activity results not only from Fab-mediated virus neutralization, but also from the protective effector functions mediated through the interaction of their Fc domains with Fcγ receptors (FcγRs) on effector leukocytes. Fc-FcγR interactions confer pleiotropic protective activities, including the clearance of opsonized virions and infected cells, as well as the induction of antiviral T-cell responses. However, excessive or inappropriate activation of specific FcγR pathways can lead to disease enhancement and exacerbated pathology, as seen in the context of dengue virus infections. A comprehensive understanding of the diversity of Fc effector functions during infection has guided the development of engineered antiviral antibodies optimized for maximal effector activity, as well as the design of targeted therapeutic approaches to prevent antibody-dependent enhancement of disease.
Collapse
Affiliation(s)
- Julia E. Edgar
- The London School of Hygiene and Tropical MedicineLondonUK
| | - Stylianos Bournazos
- The Laboratory of Molecular Genetics and ImmunologyThe Rockefeller UniversityNew YorkNew YorkUSA
| |
Collapse
|
37
|
Mekala JR, Nalluri HP, Reddy PN, S B S, N S SK, G V S D SK, Dhiman R, Chamarthy S, Komaragiri RR, Manyam RR, Dirisala VR. Emerging trends and therapeutic applications of monoclonal antibodies. Gene 2024; 925:148607. [PMID: 38797505 DOI: 10.1016/j.gene.2024.148607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 04/02/2024] [Accepted: 05/21/2024] [Indexed: 05/29/2024]
Abstract
Monoclonal antibodies (mAbs) are being used to prevent, detect, and treat a broad spectrum of malignancies and infectious and autoimmune diseases. Over the past few years, the market for mAbs has grown exponentially. They have become a significant part of many pharmaceutical product lines, and more than 250 therapeutic mAbs are undergoing clinical trials. Ever since the advent of hybridoma technology, antibody-based therapeutics were realized using murine antibodies which further progressed into humanized and fully human antibodies, reducing the risk of immunogenicity. Some of the benefits of using mAbs over conventional drugs include a drastic reduction in the chances of adverse reactions, interactions between drugs, and targeting specific proteins. While antibodies are very efficient, their higher production costs impede the process of commercialization. However, their cost factor has been improved by developing biosimilar antibodies, which are affordable versions of therapeutic antibodies. Along with biosimilars, innovations in antibody engineering have helped to design bio-better antibodies with improved efficacy than the conventional ones. These novel mAb-based therapeutics are set to revolutionize existing drug therapies targeting a wide spectrum of diseases, thereby meeting several unmet medical needs. In the future, mAbs generated by applying next-generation sequencing (NGS) are expected to become a powerful tool in clinical therapeutics. This article describes the methods of mAb production, pre-clinical and clinical development of mAbs, approved indications targeted by mAbs, and novel developments in the field of mAb research.
Collapse
Affiliation(s)
- Janaki Ramaiah Mekala
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation (KLEF), Vaddeswaram 522502, Guntur, Andhra Pradesh, INDIA.
| | - Hari P Nalluri
- Department of Biotechnology, Vignan's (Deemed to be) University, Guntur 522213, AP, India
| | - Prakash Narayana Reddy
- Department of Microbiology, Dr. V.S. Krishna Government College, Visakhapatnam 530013, India
| | - Sainath S B
- Department of Biotechnology, Vikrama Simhapuri University, Nellore 524320, AP, India
| | - Sampath Kumar N S
- Department of Biotechnology, Vignan's (Deemed to be) University, Guntur 522213, AP, India
| | - Sai Kiran G V S D
- Santhiram Medical College and General Hospital, Nandyal, Kurnool 518501, AP, India
| | - Rohan Dhiman
- Laboratory of Mycobacterial Immunology, Department of Life Sciences, National Institute of Technology Rourkela-769008, India
| | - Sahiti Chamarthy
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation (KLEF), Vaddeswaram 522502, Guntur, Andhra Pradesh, INDIA
| | - Raghava Rao Komaragiri
- Department of CSE, Koneru Lakshmaiah Education Foundation (KLEF), Vaddeswaram 522302, Andhra Pradesh, INDIA
| | - Rajasekhar Reddy Manyam
- Amrita School of Computing, Amrita Vishwa Vidyapeetham, Amaravati Campus, Amaravati, Andhra Pradesh, India
| | - Vijaya R Dirisala
- Department of Biotechnology, Vignan's (Deemed to be) University, Guntur 522213, AP, India.
| |
Collapse
|
38
|
Duo H, Yang Y, Zhang G, Chen Y, Cao Y, Luo L, Pan H, Ye Q. Comparative effectiveness of treatments for recurrent Clostridioides difficile infection: a network meta-analysis of randomized controlled trials. Front Pharmacol 2024; 15:1430724. [PMID: 39484168 PMCID: PMC11525118 DOI: 10.3389/fphar.2024.1430724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 10/07/2024] [Indexed: 11/03/2024] Open
Abstract
Background Clostridioides difficile infection (CDI) is the most common cause of healthcare-associated infectious diarrhea. A major clinical challenge is recurrent CDI (rCDI) without effective standard drug-based therapy. Additionally, a comprehensive comparison of various therapy effectiveness in rCDI patients is still under investigation. Methods A Bayesian network meta-analysis (NMA) of randomized control trials up to March 2024 was performed to investigate the efficacy of rCDI interventions. Results Seventeen trials were included, comprising 4,148 CDI patients with ten interventions, including fecal microbiota transplantation (FMT) by lower gastrointestinal (LGI), FMT by upper gastrointestinal (UGI), Autologous FMT (AFMT), vancomycin + FMT, vancomycin, placebo, fidaxomicin, Vowst (SER109), Rebyota (RBX2660), and monoclonal antibody. NMA showed that FMT by LGI had the highest efficacy in treating rCDIs with an odds ratio (95% confidence interval) of 32.33 (4.03, 248.69) compared with placebo. FMT by UGI also showed high efficacy, whereas the efficacy comparison between FMT by LGI and UGI was not statistically significant (ORs) (95% CI), 1.72 (0.65, 5.21). The rankogram and surface under the cumulative ranking curve (SUCRA) also showed FMT by LGI ranked at the top and FMT by UGI ranked second in the curative effect. Conclusion NMA demonstrates FMT's significant efficacy in rCDI management, regardless of administration route (lower or upper gastrointestinal). Despite its significant benefits, FMT's safety is a concern due to the lack of standardized FDAcompliant manufacturing and oversight. Microbiota-based therapies also exhibit potential. However, limited research mandates further clinical exploration. Antibiotics, in contrast, display comparatively reduced efficacy in rCDI, potentially linked to disruptions in native gut microflora balance. Systematic Review https://www.crd.york.ac.uk/PROSPERO/display_record.php?RecordID=368435, Identifier CRD42022368435.
Collapse
Affiliation(s)
- Hong Duo
- Hubei Key Laboratory of Medical Technology on Transplantation, National Quality Control Center for Donated Organ Procurement, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-Based Medical Materials, Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan, Hubei, China
| | - Yanwei Yang
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Clinical Research Center of Hubei Critical Care Medicine, Wuhan, China
| | - Guqing Zhang
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yingxin Chen
- Global Health Institute, School of Public Health, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Yumeng Cao
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Linjie Luo
- Department of Experimental Radiation Oncology and Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, United States
| | - Huaqin Pan
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplantation Intensive Care Unit, Transplant Center of Wuhan University, Hubei Key Laboratory of Medical Technology on Transplantation, Wuhan, China
| | - Qifa Ye
- Hubei Key Laboratory of Medical Technology on Transplantation, National Quality Control Center for Donated Organ Procurement, Hubei Clinical Research Center for Natural Polymer Biological Liver, Hubei Engineering Center of Natural Polymer-Based Medical Materials, Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan, Hubei, China
| |
Collapse
|
39
|
Moore SE, Song M, Swingler EA, Furmanek S, Chandler T, Smith D, Brenneman MT, Wilde AM. Comparing rates of recurrent infection for first occurrence of Clostridioides difficile between tapered oral vancomycin and standard vancomycin: a retrospective, propensity matched cohort study. Infect Control Hosp Epidemiol 2024:1-7. [PMID: 39400010 DOI: 10.1017/ice.2024.117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
OBJECTIVE To compare rates of Clostridioides difficile infection (CDI) recurrence following initial occurrence treated with tapered enteral vancomycin compared to standard vancomycin. DESIGN Retrospective cohort study. SETTING Community health system. PATIENTS Adults ≥18 years of age hospitalized with positive C. difficile polymerase chain reaction or toxin enzyme immunoassay who were prescribed either standard 10-14 days of enteral vancomycin four times daily or a 12-week tapered vancomycin regimen. METHODS Retrospective propensity score pair matched cohort study. Groups were matched based on age < or ≥ 65 years and receipt of non-C. difficile antibiotics during hospitalization or within 6 months post-discharge. Recurrence rates were analyzed via logistic regression conditioned on matched pairs and reported as conditional odds ratios. The primary outcome was recurrence rates compared between standard vancomycin versus tapered vancomycin for treatment of initial CDI. RESULTS The CDI recurrence rate at 6 months was 5.3% (4/75) in the taper cohort versus 28% (21/75) in the standard vancomycin cohort. The median time to CDI recurrence was 115 days versus 20 days in the taper and standard vancomycin cohorts, respectively. When adjusted for matching, patients in the taper arm were less likely to experience CDI recurrence at 6 months when compared to standard vancomycin (cOR = 0.19, 95% CI 0.07-0.56, p < 0.002). CONCLUSIONS Larger prospective trials are needed to elucidate the clinical utility of tapered oral vancomycin as a treatment option to achieve sustained clinical cure in first occurrences of CDI.
Collapse
Affiliation(s)
- Sarah E Moore
- Norton Healthcare, Norton Infectious Diseases Institute, Louisville, KY, USA
| | - Matthew Song
- Norton Healthcare, Norton Infectious Diseases Institute, Louisville, KY, USA
| | - Elena A Swingler
- Norton Healthcare, Norton Infectious Diseases Institute, Louisville, KY, USA
| | - Stephen Furmanek
- Norton Healthcare, Norton Infectious Diseases Institute, Louisville, KY, USA
| | - Thomas Chandler
- Norton Healthcare, Norton Infectious Diseases Institute, Louisville, KY, USA
| | - Dakota Smith
- Norton Healthcare, Department of Pharmacy, Louisville, KY, USA
| | | | - Ashley M Wilde
- Norton Healthcare, Norton Infectious Diseases Institute, Louisville, KY, USA
| |
Collapse
|
40
|
Theodorakis N, Feretzakis G, Hitas C, Kreouzi M, Kalantzi S, Spyridaki A, Kollia Z, Verykios VS, Nikolaou M. Immunosenescence: How Aging Increases Susceptibility to Bacterial Infections and Virulence Factors. Microorganisms 2024; 12:2052. [PMID: 39458361 PMCID: PMC11510421 DOI: 10.3390/microorganisms12102052] [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: 09/18/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
The process of aging leads to a progressive decline in the immune system function, known as immunosenescence, which compromises both innate and adaptive responses. This includes impairments in phagocytosis and decreased production, activation, and function of T- and B-lymphocytes, among other effects. Bacteria exploit immunosenescence by using various virulence factors to evade the host's defenses, leading to severe and often life-threatening infections. This manuscript explores the complex relationship between immunosenescence and bacterial virulence, focusing on the underlying mechanisms that increase vulnerability to bacterial infections in the elderly. Additionally, it discusses how machine learning methods can provide accurate modeling of interactions between the weakened immune system and bacterial virulence mechanisms, guiding the development of personalized interventions. The development of vaccines, novel antibiotics, and antivirulence therapies for multidrug-resistant bacteria, as well as the investigation of potential immune-boosting therapies, are promising strategies in this field. Future research should focus on how machine learning approaches can be integrated with immunological, microbiological, and clinical data to develop personalized interventions that improve outcomes for bacterial infections in the growing elderly population.
Collapse
Affiliation(s)
- Nikolaos Theodorakis
- Department of Cardiology, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (N.T.); (C.H.); (M.N.)
- 65+ Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (M.K.); (S.K.); (A.S.); (Z.K.)
- School of Medicine, National and Kapodistrian University of Athens, 75 Mikras Asias, 11527 Athens, Greece
| | - Georgios Feretzakis
- School of Science and Technology, Hellenic Open University, 18 Aristotelous Str., 26335 Patras, Greece;
| | - Christos Hitas
- Department of Cardiology, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (N.T.); (C.H.); (M.N.)
- 65+ Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (M.K.); (S.K.); (A.S.); (Z.K.)
| | - Magdalini Kreouzi
- 65+ Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (M.K.); (S.K.); (A.S.); (Z.K.)
- Department of Internal Medicine, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece
| | - Sofia Kalantzi
- 65+ Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (M.K.); (S.K.); (A.S.); (Z.K.)
- Department of Internal Medicine, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece
| | - Aikaterini Spyridaki
- 65+ Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (M.K.); (S.K.); (A.S.); (Z.K.)
- Department of Internal Medicine, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece
| | - Zoi Kollia
- 65+ Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (M.K.); (S.K.); (A.S.); (Z.K.)
| | - Vassilios S. Verykios
- School of Science and Technology, Hellenic Open University, 18 Aristotelous Str., 26335 Patras, Greece;
| | - Maria Nikolaou
- Department of Cardiology, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (N.T.); (C.H.); (M.N.)
- 65+ Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (M.K.); (S.K.); (A.S.); (Z.K.)
| |
Collapse
|
41
|
Carter K, Kuznetsova O, Anisimov V, Krisam J, Scherer C, Ryeznik Y, Sverdlov O. Forced randomization: the what, why, and how. BMC Med Res Methodol 2024; 24:234. [PMID: 39379810 PMCID: PMC11459895 DOI: 10.1186/s12874-024-02340-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 09/13/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND When running a randomized controlled trial (RCT), a clinical site may face a situation when an eligible trial participant is to be randomized to the treatment that is not available at the site. In this case, there are two options: not to enroll the participant, or, without disclosing to the site, allocate the participant to a treatment arm with drug available at the site using a built-in feature of the interactive response technology (IRT). In the latter case, one has employed a "forced randomization" (FR). There seems to be an industry-wide consensus that using FR can be acceptable in confirmatory trials provided there are "not too many" instances of forcing. A better understanding of statistical properties of FR is warranted. METHODS We described four different IRT configurations with or without FR and illustrated them using a simple example. We discussed potential merits of FR and outlined some relevant theoretical risks and risk mitigation strategies. We performed a search using Cortellis Regulatory Intelligence database (IDRAC) ( www.cortellis.com ) to understand the prevalence of FR in clinical trial practice. We also proposed a structured template for development and evaluation of randomization designs featuring FR and showcased an application of this template for a hypothetical multi-center 1:1 RCT under three experimental settings ("base case", "slower recruitment", and "faster recruitment") to explore the effect of four different IRT configurations in combination with three different drug supply/re-supply strategies on some important operating characteristics of the trial. We also supplied the Julia code that can be used to reproduce our simulation results and generate additional results under user-specified experimental scenarios. RESULTS FR can eliminate refusals to randomize patients, which can cause frustration for patients and study site personnel, improve the study logistics, drug supply management, cost-efficiency, and recruitment time. Nevertheless, FR carries some potential risks that should be reviewed at the study planning stage and, ideally, prospectively addressed through risk mitigation planning. The Cortellis search identified only 9 submissions that have reported the use of FR; typically, the FR option was documented in IRT specifications. Our simulation evidence showed that under the considered realistic experimental settings, the percentage of FR is expected to be low. When FR with backfilling was used in combination with high re-supply strategy, the final treatment imbalance was negligibly small, the proportion of patients not randomized due to the lack of drug supply was close to zero, and the time to complete recruitment was shortened compared to the case when FR was not allowed. The drug overage was primarily determined by the intensity of the re-supply strategy and to a smaller extent by the presence or absence of the FR feature in IRT. CONCLUSION FR with a carefully chosen drug supply/re-supply strategy can result in quantifiable improvements in the patients' and site personnel experience, trial logistics and efficiency while preventing an undesirable refusal to randomize a patient and a consequential unblinding at the site. FR is a useful design feature of multi-center RCTs provided it is properly planned for and carefully implemented.
Collapse
Affiliation(s)
- Kerstine Carter
- Boehringer-Ingelheim Pharmaceuticals Inc, Ridgefield, CT, USA
| | | | | | - Johannes Krisam
- Boehringer-Ingelheim Pharma GmbH & Co. KG, Ingelheim, Germany
| | | | - Yevgen Ryeznik
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | | |
Collapse
|
42
|
Lv X, Zhang Y, Sun K, Yang Q, Luo J, Tao L, Lu P. De novo design of mini-protein binders broadly neutralizing Clostridioides difficile toxin B variants. Nat Commun 2024; 15:8521. [PMID: 39358329 PMCID: PMC11447207 DOI: 10.1038/s41467-024-52582-1] [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: 02/27/2024] [Accepted: 09/12/2024] [Indexed: 10/04/2024] Open
Abstract
Clostridioides difficile toxin B (TcdB) is the key virulence factor accounting for C. difficile infection-associated symptoms. Effectively neutralizing different TcdB variants with a universal solution poses a significant challenge. Here we present the de novo design and characterization of pan-specific mini-protein binders against major TcdB subtypes. Our design successfully binds to the first receptor binding interface (RBI-1) of the varied TcdB subtypes, exhibiting affinities ranging from 20 pM to 10 nM. The cryo-electron microscopy (cryo-EM) structures of the mini protein binder in complex with TcdB1 and TcdB4 are consistent with the computational design models. The engineered and evolved variants of the mini-protein binder and chondroitin sulfate proteoglycan 4 (CSPG4), another natural receptor that binds to the second RBI (RBI-2) of TcdB, better neutralize major TcdB variants both in cells and in vivo, as demonstrated by the colon-loop assay using female mice. Our findings provide valuable starting points for the development of therapeutics targeting C. difficile infections (CDI).
Collapse
Affiliation(s)
- Xinchen Lv
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, 310024, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, 310024, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310024, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, 310024, China
| | - Yuanyuan Zhang
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310024, China
- Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou, Zhejiang, 310024, China
- Center for General Practice Medicine, Department of Infectious Diseases, Zhejiang Provincial People's Hospital, Hangzhou Medical College Affiliated People's Hospital, Hangzhou, Zhejiang, 310014, China
| | - Ke Sun
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310024, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, 310024, China
| | - Qi Yang
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310024, China
- Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou, Zhejiang, 310024, China
| | - Jianhua Luo
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, 310024, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, 310024, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310024, China
- Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou, Zhejiang, 310024, China
| | - Liang Tao
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, 310024, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, 310024, China.
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310024, China.
- Center for Infectious Disease Research, School of Medicine, Westlake University, Hangzhou, Zhejiang, 310024, China.
| | - Peilong Lu
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, 310024, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, 310024, China.
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310024, China.
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, 310024, China.
| |
Collapse
|
43
|
Kraft CS, Sims M, Silverman M, Louie TJ, Feuerstadt P, Huang ES, Khanna S, Berenson CS, Wang EEL, Cohen SH, Korman L, Lee C, Kelly CR, Odio A, Cook PP, Lashner B, Ramesh M, Kumar P, De A, Memisoglu A, Lombardi DA, Hasson BR, McGovern BH, von Moltke L, Pardi DS. Integrated Safety and Efficacy Analyses of Phase 3 Trials of a Microbiome Therapeutic for Recurrent CDI. Infect Dis Ther 2024; 13:2105-2121. [PMID: 38941068 PMCID: PMC11416444 DOI: 10.1007/s40121-024-01007-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 04/29/2024] [Indexed: 06/29/2024] Open
Abstract
INTRODUCTION Recurrent Clostridioides difficile infection (rCDI) often occurs after standard-of-care antibiotics. VOWST oral spores (VOS, previously SER-109), an FDA-approved orally administered microbiome therapeutic, is indicated to prevent rCDI following antibiotics for rCDI. OBJECTIVE, DESIGN, AND PATIENTS To evaluate safety and efficacy of VOS from two phase 3 trials, (randomized, placebo-controlled [ECOSPOR III: NCT03183128] and open-label, single arm [ECOSPOR IV: NCT03183141]) of 349 adults with rCDI and prevalent comorbidities. METHODS VOS or placebo [ECOSPOR III only] (4 capsules once daily for 3 days). Integrated analysis of treatment-emergent adverse events (TEAEs) collected through week 8; serious TEAEs and TEAEs of special interest collected through week 24; and rates of rCDI (toxin-positive diarrhea requiring treatment) evaluated through weeks 8 and 24. RESULTS TEAEs were mostly mild or moderate and gastrointestinal. Most common treatment-related TEAEs were flatulence, abdominal pain and distension, fatigue, and diarrhea. There were 11 deaths (3.2%) and 48 patients (13.8%) with serious TEAEs, none treatment-related. The rCDI rate through week 8 was 9.5% (95% CI 6.6-13.0) and remained low through 24 weeks (15.2%; 95% CI 11.6-19.4). Safety and rCDI rates were consistent across subgroups including age, renal impairment/failure, diabetes, and immunocompromise/immunosuppression. CONCLUSIONS VOS was well tolerated and rates of rCDI remained low through week 24 including in those with comorbidities. These data support the potential benefit of VOS following antibiotics to prevent recurrence in high-risk patients. TRIAL REGISTRATION ClinicalTrials.gov identifier, NCT03183128 and NCT03183141.
Collapse
Affiliation(s)
- Colleen S Kraft
- Department of Pathology and Laboratory Medicine, Division of Infectious Diseases, Emory University, Atlanta, GA, USA
| | - Matthew Sims
- Section of Infectious Diseases and International Medicine, Department of Internal Medicine, Beaumont Royal Oak, Royal Oak, MI, USA
- Departments of Internal Medicine and Foundational Medical Studies, Oakland University William Beaumont School of Medicine, Rochester, MI, USA
| | | | - Thomas J Louie
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Paul Feuerstadt
- Division of Digestive Disease, Yale University School of Medicine, New Haven, CT, USA
- PACT-Gastroenterology Center, Hamden, CT, USA
| | - Edward S Huang
- Department of Gastroenterology, Palo Alto Medical Foundation, Sutter Health, Mountain View, CA, USA
| | - Sahil Khanna
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Charles S Berenson
- University at Buffalo, VA Western New York Healthcare System, Buffalo, NY, USA
| | - Elaine E L Wang
- Seres Therapeutics, 200 Sidney Street, Cambridge, MA, 02139, USA
| | - Stuart H Cohen
- University of California Davis Health, Sacramento, CA, USA
| | - Louis Korman
- Gastroenterology and Hepatology, Chevy Chase Clinical Research, Chevy Chase, MD, USA
| | - Christine Lee
- Island Medical Program, University of British Columbia and University of Victoria, Vancouver, BC, Canada
| | - Colleen R Kelly
- Division of Gastroenterology, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Paul P Cook
- Brody School of Medicine at East, Carolina University, Greenville, NC, USA
| | | | - Mayur Ramesh
- Division of Infectious Diseases, Henry Ford Health, Detroit, MI, USA
| | - Princy Kumar
- Division of Infectious Diseases and Tropical Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Ananya De
- Seres Therapeutics, 200 Sidney Street, Cambridge, MA, 02139, USA
| | - Asli Memisoglu
- Seres Therapeutics, 200 Sidney Street, Cambridge, MA, 02139, USA
| | - David A Lombardi
- Seres Therapeutics, 200 Sidney Street, Cambridge, MA, 02139, USA
| | - Brooke R Hasson
- Seres Therapeutics, 200 Sidney Street, Cambridge, MA, 02139, USA.
| | | | - Lisa von Moltke
- Seres Therapeutics, 200 Sidney Street, Cambridge, MA, 02139, USA
| | - Darrell S Pardi
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
44
|
Cocker D, Birgand G, Zhu N, Rodriguez-Manzano J, Ahmad R, Jambo K, Levin AS, Holmes A. Healthcare as a driver, reservoir and amplifier of antimicrobial resistance: opportunities for interventions. Nat Rev Microbiol 2024; 22:636-649. [PMID: 39048837 DOI: 10.1038/s41579-024-01076-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/25/2024] [Indexed: 07/27/2024]
Abstract
Antimicrobial resistance (AMR) is a global health challenge that threatens humans, animals and the environment. Evidence is emerging for a role of healthcare infrastructure, environments and patient pathways in promoting and maintaining AMR via direct and indirect mechanisms. Advances in vaccination and monoclonal antibody therapies together with integrated surveillance, rapid diagnostics, targeted antimicrobial therapy and infection control measures offer opportunities to address healthcare-associated AMR risks more effectively. Additionally, innovations in artificial intelligence, data linkage and intelligent systems can be used to better predict and reduce AMR and improve healthcare resilience. In this Review, we examine the mechanisms by which healthcare functions as a driver, reservoir and amplifier of AMR, contextualized within a One Health framework. We also explore the opportunities and innovative solutions that can be used to combat AMR throughout the patient journey. We provide a perspective on the current evidence for the effectiveness of interventions designed to mitigate healthcare-associated AMR and promote healthcare resilience within high-income and resource-limited settings, as well as the challenges associated with their implementation.
Collapse
Affiliation(s)
- Derek Cocker
- David Price Evans Infectious Diseases & Global Health Group, University of Liverpool, Liverpool, UK
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
| | - Gabriel Birgand
- Centre d'appui pour la Prévention des Infections Associées aux Soins, Nantes, France
- National Institute for Health and Care Research (NIHR) Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at Imperial College London, London, UK
- Cibles et medicaments des infections et de l'immunitée, IICiMed, Nantes Universite, Nantes, France
| | - Nina Zhu
- National Institute for Health and Care Research (NIHR) Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at Imperial College London, London, UK
- Department of Infectious Disease, Imperial College London, London, UK
| | - Jesus Rodriguez-Manzano
- National Institute for Health and Care Research (NIHR) Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at Imperial College London, London, UK
- Department of Infectious Disease, Imperial College London, London, UK
| | - Raheelah Ahmad
- National Institute for Health and Care Research (NIHR) Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at Imperial College London, London, UK
- Department of Health Services Research & Management, City University of London, London, UK
- Dow University of Health Sciences, Karachi, Pakistan
| | - Kondwani Jambo
- Malawi-Liverpool-Wellcome Research Programme, Blantyre, Malawi
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Anna S Levin
- Department of Infectious Disease, School of Medicine & Institute of Tropical Medicine, University of São Paulo, São Paulo, Brazil
| | - Alison Holmes
- David Price Evans Infectious Diseases & Global Health Group, University of Liverpool, Liverpool, UK.
- National Institute for Health and Care Research (NIHR) Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance at Imperial College London, London, UK.
- Department of Infectious Disease, Imperial College London, London, UK.
| |
Collapse
|
45
|
Theodorakis N, Feretzakis G, Hitas C, Kreouzi M, Kalantzi S, Spyridaki A, Boufeas IZ, Sakagianni A, Paxinou E, Verykios VS, Nikolaou M. Antibiotic Resistance in the Elderly: Mechanisms, Risk Factors, and Solutions. Microorganisms 2024; 12:1978. [PMID: 39458286 PMCID: PMC11509523 DOI: 10.3390/microorganisms12101978] [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: 09/22/2024] [Revised: 09/27/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
Antibiotic resistance presents a critical challenge in healthcare, particularly among the elderly, where multidrug-resistant organisms (MDROs) contribute to increased morbidity, mortality, and healthcare costs. This review focuses on the mechanisms underlying resistance in key bacterial pathogens and highlights how aging-related factors like immunosenescence, frailty, and multimorbidity increase the burden of infections from MDROs in this population. Novel strategies to mitigate resistance include the development of next-generation antibiotics like teixobactin and cefiderocol, innovative therapies such as bacteriophage therapy and antivirulence treatments, and the implementation of antimicrobial stewardship programs to optimize antibiotic use. Furthermore, advanced molecular diagnostic techniques, including nucleic acid amplification tests and next-generation sequencing, allow for faster and more precise identification of resistant pathogens. Vaccine development, particularly through innovative approaches like multi-epitope vaccines and nanoparticle-based platforms, holds promise in preventing MDRO infections among the elderly. The role of machine learning (ML) in predicting resistance patterns and aiding in vaccine and antibiotic development is also explored, offering promising solutions for personalized treatment and prevention strategies in the elderly. By integrating cutting-edge diagnostics, therapeutic innovations, and ML-based approaches, this review underscores the importance of multidisciplinary efforts to address the global challenge of antibiotic resistance in aging populations.
Collapse
Affiliation(s)
- Nikolaos Theodorakis
- Department of Cardiology, 65+ Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (N.T.); (C.H.); (M.N.)
- School of Medicine, National and Kapodistrian University of Athens, 75 Mikras Asias, 11527 Athens, Greece
| | - Georgios Feretzakis
- School of Science and Technology, Hellenic Open University, 18 Aristotelous Str., 26335 Patras, Greece; (G.F.); (E.P.)
| | - Christos Hitas
- Department of Cardiology, 65+ Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (N.T.); (C.H.); (M.N.)
| | - Magdalini Kreouzi
- Department of Internal Medicine, 65+ Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (M.K.); (S.K.); (A.S.)
| | - Sofia Kalantzi
- Department of Internal Medicine, 65+ Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (M.K.); (S.K.); (A.S.)
| | - Aikaterini Spyridaki
- Department of Internal Medicine, 65+ Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (M.K.); (S.K.); (A.S.)
| | - Iris Zoe Boufeas
- Barts and The London School of Medicine and Dentistry, Queen Mary University of London, 64 Turner Street, London E1 2AD, UK;
| | - Aikaterini Sakagianni
- Intensive Care Unit, Sismanogelio General Hospital, 37 Sismanogleiou Str., 15126 Marousi, Greece;
| | - Evgenia Paxinou
- School of Science and Technology, Hellenic Open University, 18 Aristotelous Str., 26335 Patras, Greece; (G.F.); (E.P.)
| | - Vassilios S. Verykios
- School of Science and Technology, Hellenic Open University, 18 Aristotelous Str., 26335 Patras, Greece; (G.F.); (E.P.)
| | - Maria Nikolaou
- Department of Cardiology, 65+ Clinic, Amalia Fleming General Hospital, 14, 25th Martiou Str., 15127 Melissia, Greece; (N.T.); (C.H.); (M.N.)
| |
Collapse
|
46
|
Cummer R, Grosjean F, Bolteau R, Vasegh SE, Veyron S, Keogh L, Trempe JF, Castagner B. Structure-Activity Relationship of Inositol Thiophosphate Analogs as Allosteric Activators of Clostridioides difficile Toxin B. J Med Chem 2024; 67:16576-16597. [PMID: 39254660 DOI: 10.1021/acs.jmedchem.4c01408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Clostridioides difficile is a bacterium that causes life-threatening intestinal infections. Infection symptoms are mediated by a toxin secreted by the bacterium. Toxin pathogenesis is modulated by the intracellular molecule, inositol-hexakisphosphate (IP6). IP6 binds to a cysteine protease domain (CPD) on the toxin, inducing autoproteolysis, which liberates a virulence factor in the cell cytosol. We developed second-generation IP6 analogs designed to induce autoproteolysis in the gut lumen, prior to toxin uptake, circumventing pathogenesis. We synthesized a panel of thiophosphate-/sulfate-containing IP6 analogs and characterized their toxin binding affinity, autoproteolysis induction, and cation interactions. Our top candidate was soluble in extracellular cation concentrations, unlike IP6. The IP6 analogs were more negatively charged than IP6, which improved affinity and stabilization of the CPD, enhancing toxin autoproteolysis. Our data illustrate the optimization of IP6 with thiophosphate biomimetic which are more capable of inducing toxin autoproteolysis than the native ligand, warranting further studies in vivo.
Collapse
Affiliation(s)
- Rebecca Cummer
- Department of Pharmacology and Therapeutics, McGill University, Québec H3G 1Y6, Canada
| | - Félix Grosjean
- Department of Pharmacology and Therapeutics, McGill University, Québec H3G 1Y6, Canada
| | - Raphaël Bolteau
- Department of Pharmacology and Therapeutics, McGill University, Québec H3G 1Y6, Canada
| | - Seyed Ehsan Vasegh
- Department of Pharmacology and Therapeutics, McGill University, Québec H3G 1Y6, Canada
| | - Simon Veyron
- Department of Pharmacology and Therapeutics, McGill University, Québec H3G 1Y6, Canada
| | - Liam Keogh
- Department of Pharmacology and Therapeutics, McGill University, Québec H3G 1Y6, Canada
| | - Jean-François Trempe
- Department of Pharmacology and Therapeutics, McGill University, Québec H3G 1Y6, Canada
| | - Bastien Castagner
- Department of Pharmacology and Therapeutics, McGill University, Québec H3G 1Y6, Canada
| |
Collapse
|
47
|
Berry P, Khanna S. Fecal microbiota spores, live-brpk (VOWST™/VOS) for prevention of recurrent Clostridioides difficile infection. Future Microbiol 2024; 19:1519-1528. [PMID: 39320321 DOI: 10.1080/17460913.2024.2403892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 09/10/2024] [Indexed: 09/26/2024] Open
Abstract
Clostridioides difficile infection (CDI) is a health crisis comprising a majority of healthcare-associated infections and is now being seen in the community. Persistent dysbiosis despite treatment with standard-of-care antibiotics increases risk of recurrent infections. Fecal microbiota transplantation has been an effective way of addressing dysbiosis, but the studies have lacked standardization, which makes outcome and safety data difficult to interpret. Standardized microbiome therapies have demonstrated efficacy and safety for recurrent CDI and have been approved to prevent recurrent infection. In this review, we discuss the data behind and the practice use of fecal microbiota spores, live-brpk (VOWST™ / VOS), a US FDA approved live biotherapeutic for the prevention of recurrent CDI.
Collapse
Affiliation(s)
- Parul Berry
- C. difficile Clinic & Microbiome Restoration Program, Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Sahil Khanna
- C. difficile Clinic & Microbiome Restoration Program, Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
48
|
Villafuerte Gálvez JA, Kelly CP. An Anti-inflammatory Approach to Drug Repurposing for Clostridioides difficile Infection. J Infect Dis 2024; 230:527-528. [PMID: 38243873 DOI: 10.1093/infdis/jiae022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 01/17/2024] [Indexed: 01/22/2024] Open
Affiliation(s)
- Javier A Villafuerte Gálvez
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Ciarán P Kelly
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
49
|
Xie Y, Irwin S, Chupina Estrada A, Nelson B, Bullock A, Fontenot L, Feng H, Sun M, Koon HW. Loratadine as an Anti-inflammatory Agent Against Clostridium difficile Toxin B. J Infect Dis 2024; 230:545-557. [PMID: 38243838 PMCID: PMC11420802 DOI: 10.1093/infdis/jiae021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 11/30/2023] [Accepted: 01/17/2024] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND Clostridium difficile infection (CDI) is a debilitating nosocomial infection. C. difficile produces toxins A and B, which cause inflammation. Existing therapies have issues with recurrence, cost, and safety. We aim to discover a safe, effective, and economical nonmicrobiological therapeutic approach against CDI. METHODS We included human primary peripheral blood mononuclear cells (PBMCs), fresh human colonic explants, and humanized HuCD34-NCG mice. Toxin A+B+ VPI 10463 and A-B+ ribotype 017 C. difficile strains were used. We used single-cell RNA profiling and high-throughput screening to find actionable toxin B-dependent pathways in PBMCs. RESULTS Histamine 1 receptor-related drugs were found among the hit compounds that reversed toxin-mediated macrophage inflammatory protein (MIP) 1α expression in PBMCs. We identified loratadine as the safest representative antihistamine for therapeutic development. Loratadine inhibited toxin B-induced MIP-1α secretion in fresh human colonic tissues. Oral loratadine (10 mg/kg/d) maintained survival, inhibited intestinal CCl3 messenger RNA expression, and prevented vancomycin-associated recurrence in the VPI 10463-infected mice and ribotype 017-infected hamsters. Splenocytes from loratadine-treated mice conferred anti-inflammatory effects to the VPI 10463-infected T/B-cell--deficient Rag-/- mice. Oral loratadine suppressed human MIP-1α expression in monocytes/macrophages in toxin B-expressing ribotype 017-infected humanized HuCD34-NCG mice. CONCLUSIONS Loratadine may be repurposed to optimize existing therapies against CDI.
Collapse
Affiliation(s)
- Ying Xie
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, California, USA
- Department of Gastroenterology, The First Hospital of China Medical University, Shenyang City, China
| | - Sophie Irwin
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, California, USA
| | - Andrea Chupina Estrada
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, California, USA
| | - Becca Nelson
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, California, USA
| | - Ashlen Bullock
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, California, USA
| | - Lindsey Fontenot
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, California, USA
| | - Hanping Feng
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland, Baltimore, Maryland, USA
| | - Mingjun Sun
- Department of Gastroenterology, The First Hospital of China Medical University, Shenyang City, China
| | - Hon Wai Koon
- Vatche and Tamar Manoukian Division of Digestive Diseases, David Geffen School of Medicine at the University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
50
|
Anjou C, Royer M, Bertrand É, Bredon M, Le Bris J, Salgueiro IA, Caulat LC, Dupuy B, Barbut F, Morvan C, Rolhion N, Martin-Verstraete I. Adaptation mechanisms of Clostridioides difficile to auranofin and its impact on human gut microbiota. NPJ Biofilms Microbiomes 2024; 10:86. [PMID: 39284817 PMCID: PMC11405772 DOI: 10.1038/s41522-024-00551-3] [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: 04/02/2024] [Accepted: 08/20/2024] [Indexed: 09/19/2024] Open
Abstract
Auranofin (AF), a former rheumatoid polyarthritis treatment, gained renewed interest for its use as an antimicrobial. AF is an inhibitor of thioredoxin reductase (TrxB), a thiol and protein repair enzyme, with an antibacterial activity against several bacteria including C. difficile, an enteropathogen causing post-antibiotic diarrhea. Several studies demonstrated the effect of AF on C. difficile physiology, but the crucial questions of resistance mechanisms and impact on microbiota remain unaddressed. We explored potential resistance mechanisms by studying the impact of TrxB multiplicity and by generating and characterizing adaptive mutations. We showed that if mutants inactivated for trxB genes have a lower MIC of AF, the number of TrxBs naturally present in clinical strains does not impact the MIC. All stable mutations isolated after AF long-term exposure were in the anti-sigma factor of σB and strongly affect physiology. Finally, we showed that AF has less impact on human gut microbiota than vancomycin.
Collapse
Affiliation(s)
- Cyril Anjou
- Institut Pasteur, Université Paris Cité, UMR CNRS 6047, Laboratoire Pathogenèse des Bactéries Anaérobies, F-75015, Paris, France
| | - Marie Royer
- Institut Pasteur, Université Paris Cité, UMR CNRS 6047, Laboratoire Pathogenèse des Bactéries Anaérobies, F-75015, Paris, France
- Institut Pasteur, Université Paris Cité, UMR CNRS 6047, Unité Écologie et Évolution de la Résistance aux Antibiotiques, Paris, France
| | - Émilie Bertrand
- Institut Pasteur, Université Paris Cité, UMR CNRS 6047, Laboratoire Pathogenèse des Bactéries Anaérobies, F-75015, Paris, France
| | - Marius Bredon
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Julie Le Bris
- Microbial Evolutionary Genomics, Institut Pasteur, CNRS UMR3525, Université Paris Cité, Paris, France
- Sorbonne Université, Collège Doctoral, École Doctorale Complexité du Vivant, 75005, Paris, France
| | - Iria Alonso Salgueiro
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Léo C Caulat
- Institut Pasteur, Université Paris Cité, UMR CNRS 6047, Laboratoire Pathogenèse des Bactéries Anaérobies, F-75015, Paris, France
| | - Bruno Dupuy
- Institut Pasteur, Université Paris Cité, UMR CNRS 6047, Laboratoire Pathogenèse des Bactéries Anaérobies, F-75015, Paris, France
| | - Frédéric Barbut
- Université Paris Cité, INSERM, UMR-1139, Paris, France
- National Reference Laboratory for C. difficile, Assistance Publique Hôpitaux de Paris, Hôpital Saint-Antoine, 75012, Paris, France
| | - Claire Morvan
- Institut Pasteur, Université Paris Cité, UMR CNRS 6047, Laboratoire Pathogenèse des Bactéries Anaérobies, F-75015, Paris, France
| | - Nathalie Rolhion
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, Paris, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Isabelle Martin-Verstraete
- Institut Pasteur, Université Paris Cité, UMR CNRS 6047, Laboratoire Pathogenèse des Bactéries Anaérobies, F-75015, Paris, France.
- Institut Universitaire de France, Paris, France.
| |
Collapse
|