1
|
Groenewegen B, van Lingen E, Kovynev A, van den Berg AJ, Berssenbrugge EKL, Sanders IMJG, van Prehn J, van Nood E, Goorhuis A, Kuijper EJ, Smits WK, Wiese M, Keller JJ, Ducarmon QR, Terveer EM. The presence of Clostridioides difficile in faeces before and after faecal microbiota transplantation and its relation with recurrent C. difficile infection and the gut microbiota in a Dutch cohort. Clin Microbiol Infect 2025; 31:568-574. [PMID: 39662821 DOI: 10.1016/j.cmi.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 11/19/2024] [Accepted: 12/04/2024] [Indexed: 12/13/2024]
Abstract
OBJECTIVES The objectives of this study are to investigate the presence of Clostridioides difficile in faeces of patients with recurrent C. difficile infection (rCDI) before and after faecal microbiota transplantation (FMT) and to identify risk factors for faecal C. difficile and C. difficile infection (CDI) recurrence. METHODS n = 83 faecal sample triads (pre-FMT [∼1 day], post-FMT [∼3 weeks], and a corresponding FMT donor sample), and n = 22 long-term (∼1-3 years) follow-up faecal samples were collected from FMT-treated patients. The presence of C. difficile in faeces was assessed by enrichment broth culture and PCR (tcdB gene) and associated with patient characteristics, FMT outcome, duration of pre-FMT vancomycin, FMT donor, post-FMT antibiotic use, and faecal microbiota composition (shotgun metagenomics). RESULTS The FMT cure rate for rCDI was 92.8% (77/83), with six early CDI recurrences (<2 months post-FMT). Toxigenic C. difficile was cultured in 27.7% (23/83) of all patients post-FMT, 23.4% (18/77) of patients cured 2 months post-FMT, and 13.6% (3/22) at long-term follow-up. Early CDI recurrence (n = 6) was associated with positive C. difficile culture post-FMT (21.7% [5/23] vs. 1.7% [1/60], p 0.01), post-FMT antibiotics (30.0% [3/10] vs. 4.6% [3/65], p 0.03), and a short course of pre-FMT vancomycin (median 6.0 days, IQR [5-12] vs. 18 days, IQR [10.8-29], p < 0.05). Additionally, positive C. difficile culture directly pre-FMT was associated with a short course of pre-FMT vancomycin (median 9 days IQR [5-18] vs. 17 days, IQR [10-29.2], p 0.04). Gut microbiota analyses did not reveal signatures associated with C. difficile culture result, despite statistically non-significant trends in relative abundances of the Enterobacteriaceae family, and Dorea, Roseburia, and Clostridiales species. DISCUSSION Although eradication of C. difficile is not required for clinical cure of rCDI by FMT, it is associated with reduced prevalence of early CDI recurrence, as are the full completion of pre-FMT vancomycin (at least 10 days) and avoiding post-FMT antibiotics.
Collapse
Affiliation(s)
- Bas Groenewegen
- Netherlands Donor Feces Bank, Leiden University Center of Infectious Diseases Medical Microbiology and Infection Prevention, Leiden University Medical Center, Leiden, The Netherlands; Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Emilie van Lingen
- Netherlands Donor Feces Bank, Leiden University Center of Infectious Diseases Medical Microbiology and Infection Prevention, Leiden University Medical Center, Leiden, The Netherlands; Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Artemiy Kovynev
- Center for Microbiome Analyses and Therapeutics, Leiden University Center of Infectious Diseases Research, Leiden University Medical Center, Leiden, The Netherlands
| | - Alexander J van den Berg
- Netherlands Donor Feces Bank, Leiden University Center of Infectious Diseases Medical Microbiology and Infection Prevention, Leiden University Medical Center, Leiden, The Netherlands
| | - Eric K L Berssenbrugge
- Netherlands Donor Feces Bank, Leiden University Center of Infectious Diseases Medical Microbiology and Infection Prevention, Leiden University Medical Center, Leiden, The Netherlands
| | - Ingrid M J G Sanders
- Leiden University Center of Infectious Diseases Research, Leiden University Medical Center, Leiden, The Netherlands
| | - Joffrey van Prehn
- Netherlands Donor Feces Bank, Leiden University Center of Infectious Diseases Medical Microbiology and Infection Prevention, Leiden University Medical Center, Leiden, The Netherlands; Center for Microbiome Analyses and Therapeutics, Leiden University Center of Infectious Diseases Research, Leiden University Medical Center, Leiden, The Netherlands
| | - Els van Nood
- Department of Medical Microbiology and Infectious Diseases and Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Abraham Goorhuis
- Department of Internal Medicine, Amsterdam University Medical Centers, Location Academic Medical Center, Amsterdam, The Netherlands
| | - Ed J Kuijper
- Netherlands Donor Feces Bank, Leiden University Center of Infectious Diseases Medical Microbiology and Infection Prevention, Leiden University Medical Center, Leiden, The Netherlands; Center for Microbiome Analyses and Therapeutics, Leiden University Center of Infectious Diseases Research, Leiden University Medical Center, Leiden, The Netherlands
| | - Wiep Klaas Smits
- Center for Microbiome Analyses and Therapeutics, Leiden University Center of Infectious Diseases Research, Leiden University Medical Center, Leiden, The Netherlands
| | - Maria Wiese
- Center for Microbiome Analyses and Therapeutics, Leiden University Center of Infectious Diseases Research, Leiden University Medical Center, Leiden, The Netherlands; Department of Microbiology and Systems Biology, Netherlands Organization for Applied Scientific Research (TNO), Leiden, The Netherlands
| | - Josbert J Keller
- Netherlands Donor Feces Bank, Leiden University Center of Infectious Diseases Medical Microbiology and Infection Prevention, Leiden University Medical Center, Leiden, The Netherlands; Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands; Department of Gastroenterology and Hepatology, Haaglanden Medical Center, Den Haag, The Netherlands
| | - Quinten R Ducarmon
- Center for Microbiome Analyses and Therapeutics, Leiden University Center of Infectious Diseases Research, Leiden University Medical Center, Leiden, The Netherlands
| | - Elisabeth M Terveer
- Netherlands Donor Feces Bank, Leiden University Center of Infectious Diseases Medical Microbiology and Infection Prevention, Leiden University Medical Center, Leiden, The Netherlands; Center for Microbiome Analyses and Therapeutics, Leiden University Center of Infectious Diseases Research, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
2
|
Garrett EM, Pu M, Bobenchik AM. Evaluation of a Fluorescence Immunoassay for Detection of Clostridioides difficile Glutamate Dehydrogenase and Toxin Antigens. J Appl Lab Med 2025:jfaf010. [PMID: 40105901 DOI: 10.1093/jalm/jfaf010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 01/13/2025] [Indexed: 03/21/2025]
Abstract
BACKGROUND Clostridioides difficile infection (CDI) is a leading cause of nosocomial infections in the United States, causing longer hospital stays, significant morbidity, and increased healthcare costs. Accurate CDI diagnosis is essential for timely treatment and infection control. Laboratory diagnosis of CDI commonly involves the detection of glutamate dehydrogenase (GDH) and/or toxins A and B by immunoassays or the toxin genes by nucleic acid amplification. This study assesses the performance of a new commercial test, the Sofia® 2 C. difficile Fluorescent Immunoassay (Sofia 2; FIA; QuidelOrtho), for detecting C. difficile GDH and toxins. METHODS Sofia 2 was compared to enzyme immunoassays (EIAs) C. diff Quik Chek Complete (Techlab Inc.) and Immunocard (Meridian Bioscience) using remnant stool samples from 262 patients with suspected CDI. RESULTS Sofia 2 demonstrated high agreement with the EIA methods for GDH (positive percentage agreement (PPA): 100%, negative percentage agreement (NPA): 94%, overall percentage of agreement (OPA): 95%) and toxins (PPA: 100%, NPA: 99%, OPA: 99%) detection. Compared to standard-of-care (SOC) testing including toxin gene PCR with the following toxin antigen test, Sofia 2 demonstrates strong PPA (100%), NPA (98%), positive predictive value (71%), and negative predictive value (100%). CONCLUSIONS Sofia 2 C. difficile FIA generates rapid results that are comparable to other commercial immunoassays with a simple workflow, supporting its use for CDI diagnosis in clinical practice.
Collapse
Affiliation(s)
- Elizabeth M Garrett
- Department of Pathology and Laboratory Medicine, Penn State College of Medicine, Hershey, PA, United States
| | - Meng Pu
- Department of Pathology and Laboratory Medicine, Penn State College of Medicine, Hershey, PA, United States
| | - April M Bobenchik
- Department of Pathology and Laboratory Medicine, Penn State College of Medicine, Hershey, PA, United States
| |
Collapse
|
3
|
Vernon JJ, Eyre DW, Wilcox MH, Freeman J. Molecular clock complexities of Clostridioides difficile. Anaerobe 2025:102953. [PMID: 40118334 DOI: 10.1016/j.anaerobe.2025.102953] [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: 11/21/2024] [Revised: 03/07/2025] [Accepted: 03/09/2025] [Indexed: 03/23/2025]
Abstract
OBJECTIVES Reconstruct the phylogenetic status of a collection of historical Clostridioides difficile isolates and evaluate the congruence of their evolutionary trajectories with established molecular clock models. METHODS Phylogenetic analysis was performed on Illumina sequence reads from previously analysed historic C. difficile isolates (1980-86; n=75) demonstrating multiple antimicrobial resistances. Data was grouped by ribotype (RT), including comparators from European surveillance (2012-13) and phylogenetic studies (1985-2010). Reads were mapped to CD630/CD196 reference genomes and compared using recombination-adjusted maximum likelihood trees. Prediction intervals for expected SNP differences by age were calculated using a Poisson distribution and molecular clock estimates (0.74 SNPs per genome/per year). Root-to-tip analysis was performed to determine the date of most common recent ancestor of genomes sharing a ribotype. RESULTS Moxifloxacin-resistant (>16 mg/L) RT027 isolate JV67 (1986) was two SNPs distinct from a 2006 genome, fewer than the expected lower estimate (4.4 SNPs) under current molecular clock calculations; (p=3.93x10-5). For isolate JV02 (1981), the 13 SNP divergence from a 2008 isolate was consistent with expectations (5.9 SNPs; p=0.07). JV73 (1983) demonstrated an 8 SNP difference, which although above the expected lower limit (5.5 SNPs), was outside the 95% prediction interval; (p= 4.51x10-3). Only sixty-nine percent of historical genomes fit within the prediction interval for the number of SNPs expected compared to recent isolates, with fewer SNPs observed more frequently than expected. Root-to-tip analysis demonstrated only a weak linear correlation. CONCLUSIONS C. difficile molecular clock estimations may be more complex than previously considered, with periods of spore quiescence potentially complicating analyses.
Collapse
Affiliation(s)
- Jon J Vernon
- Healthcare-Associated Infections Research Group, Leeds Institute for Medical Research, University of Leeds, Wellcome Trust Brenner Building, St James University Hospital, Leeds, West Yorkshire, LS9 7TF; Division of Oral Biology, School of Dentistry, University of Leeds, Wellcome Trust Brenner Building, St James University Hospital, Leeds, West Yorkshire, LS9 7TF.
| | - David W Eyre
- Big Data Institute, Nuffield Department of Population Health, University of Oxford, Oxford, OX3 7LF; NIHR Oxford Biomedical Research Centre, University of Oxford, OX3 9DU; NIHR Health Protection Research Unit in Healthcare Associated Infections and Antimicrobial Resistance, University of Oxford, OX3 9DU
| | - Mark H Wilcox
- Healthcare-Associated Infections Research Group, Leeds Institute for Medical Research, University of Leeds, Wellcome Trust Brenner Building, St James University Hospital, Leeds, West Yorkshire, LS9 7TF; Microbiology, Leeds Teaching Hospitals Trust, Leeds, UK
| | - Jane Freeman
- Healthcare-Associated Infections Research Group, Leeds Institute for Medical Research, University of Leeds, Wellcome Trust Brenner Building, St James University Hospital, Leeds, West Yorkshire, LS9 7TF; Microbiology, Leeds Teaching Hospitals Trust, Leeds, UK
| |
Collapse
|
4
|
Glampedakis E, Snoussi MC, Sobgoui B, Battistella F, Cuiña Iglesias P, Riccio C, Qalla-Widmer L, Cassini A, Nahimana Tessemo MI. Bodily waste management and related hygiene practices in nursing homes of Vaud: findings from a multicentre cross-sectional survey as a basis for targeted interventions. Antimicrob Resist Infect Control 2025; 14:20. [PMID: 40069828 PMCID: PMC11895322 DOI: 10.1186/s13756-025-01535-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 02/23/2025] [Indexed: 03/15/2025] Open
Abstract
INTRODUCTION Bodily waste management is a critical yet frequently neglected domain of infection prevention and control. We conducted a survey to examine various aspects of bodily waste management and related hygiene practices in nursing homes (NH) based on existing recommendations. METHODS All NHs (n = 120) of canton Vaud in Switzerland were invited to participate in this cross-sectional survey between July 2022 and February 2023 using a questionnaire. RESULTS Eighty-seven NHs participated in the survey (72.5%). Of these, 33% had internal protocols on bodily waste management, 98% had at least a dirty utility room (median: 4 per NH) and all a bedpan washer-disinfector (WD), yet only 66% met the cantonal recommendation of bedpan WD density (1/15 beds). Separation of soiled and clean compartments was present in 51%, complete hand hygiene supplies in 73% and personal protective equipment (PPE) in 30% of utility rooms. Fifty-four percent of NHs reported having a lid for each bedpan. Systematic use of lids was reported in 33% of institutions and of gloves in 98%, for the transport of used bodily waste collection tools. All surveyed institutions reported performing automated reprocessing of bodily waste collection tools in bedpan WDs and use of manual pre-cleaning was anecdotal. Regular maintenance and validation of bedpan WDs was present in almost all participating NHs. CONCLUSION Identified actionable priorities include making bodily waste management protocols accessible to staff, delineation of clean and soiled compartments in utility rooms and equipping them with PPE and hand hygiene supplies, as well as educating healthcare workers on best practices for the transport and disposal of bodily waste.
Collapse
Affiliation(s)
- Emmanouil Glampedakis
- Cantonal Unit for Infection Prevention and Control, Public Health Service, Lausanne, Switzerland.
- Unité Cantonale Hygiène, Prévention Et Contrôle de L'infection (HPCi Vaud), Office du Médecin Cantonal, Avenue de La Gare 43, 1003, Lausanne, Switzerland.
| | - Marie-Catherine Snoussi
- Cantonal Unit for Infection Prevention and Control, Public Health Service, Lausanne, Switzerland
| | - Béatrix Sobgoui
- Cantonal Unit for Infection Prevention and Control, Public Health Service, Lausanne, Switzerland
| | - Firmino Battistella
- Cantonal Unit for Infection Prevention and Control, Public Health Service, Lausanne, Switzerland
| | - Patricia Cuiña Iglesias
- Cantonal Unit for Infection Prevention and Control, Public Health Service, Lausanne, Switzerland
| | - Coralie Riccio
- Cantonal Unit for Infection Prevention and Control, Public Health Service, Lausanne, Switzerland
| | - Laetitia Qalla-Widmer
- La Source School of Nursing, HES-SO University of Applied Sciences and Arts Western Switzerland, Lausanne, Switzerland
| | - Alessandro Cassini
- Public Health Department, Canton of Vaud, Lausanne, Switzerland
- Infection Prevention and Control Unit, Infectious Diseases Service, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | | |
Collapse
|
5
|
Hensen ADO, Vehreschild MJGT, Gerding DN, Krut O, Chen W, Young VB, Tzipori S, Solbach P, Gibani MM, Chiu C, de Keersmaecker SCJ, Dasyam D, Morel S, Devaster JM, Corti N, Kuijper EJ, Roestenberg M, Smits WK. How to develop a controlled human infection model for Clostridioides difficile. Clin Microbiol Infect 2025; 31:373-379. [PMID: 39214188 DOI: 10.1016/j.cmi.2024.08.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Clostridioides difficile (C. difficile) remains the leading cause of healthcare-associated diarrhoea, posing treatment challenges because of antibiotic resistance and high relapse rates. Faecal microbiota transplantation is a novel treatment strategy to prevent relapses of C. difficile infection (CDI), however, the exact components conferring colonization resistance are unknown, hampering its translation to a medicinal product. The development of novel products independent of antibiotics, which increase colonization resistance or induce protective immune mechanisms is urgently needed. OBJECTIVES To establish a framework for a Controlled Human Infection Model (CHIM) of C. difficile, in which healthy volunteers are exposed to toxigenic C. difficile spores, offering the possibility to test novel approaches and identify microbiota and immunological targets. Whereas experimental exposure to non-toxigenic C. difficile has been done before, a toxigenic C. difficile CHIM faces ethical, scientific, logistical, and biosafety challenges. SOURCES Specific challenges in developing a C. difficile CHIM were discussed by a group of international experts during a workshop organized by Inno4Vac, an Innovative Health Initiative-funded consortium. CONTENT The experts agreed that the main challenges are: developing a clinically relevant CHIM that induces mild to moderate CDI symptoms but not severe CDI, determining the optimal C. difficile inoculum dose, and understanding the timing and duration of antibiotic pretreatment in inducing susceptibility to CDI in healthy volunteers. IMPLICATIONS Should these challenges be tackled, a C. difficile CHIM will not only provide a way forward for the testing of novel products but also offer a framework for a better understanding of the pathophysiology, pathogenesis, and immunology of C. difficile colonization and infection.
Collapse
Affiliation(s)
- Annefleur D O Hensen
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Maria J G T Vehreschild
- Department of Internal Medicine, Division of Infectious Diseases, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; German Center for Infection Research (DZIF), Cologne, Germany
| | - Dale N Gerding
- Department of Veterans Affairs, Edward Hines Jr VA Hospital, Hines, IL, United States
| | - Oleg Krut
- Paul-Ehrlich-Institut (PEI), Langen, Germany
| | - Wilbur Chen
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Vincent B Young
- Department of Internal Medicine/Infectious Diseases Division and the Department of Microbiology & Immunology, The University of Michigan, Ann Arbor, MI, United States
| | - Saul Tzipori
- Division of Infectious Disease and Global Health, Tufts University, Medford, MA, United States
| | - Philipp Solbach
- First Department of Medicine, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Malick Mahdi Gibani
- Department of Infectious Disease, Imperial College London (ICL), London, United Kingdom
| | - Christopher Chiu
- Department of Infectious Disease, Imperial College London (ICL), London, United Kingdom
| | | | | | | | | | | | - Ed J Kuijper
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Meta Roestenberg
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), Leiden, The Netherlands.
| | - Wiep Klaas Smits
- Leiden University Center for Infectious Diseases (LUCID), Leiden University Medical Center (LUMC), Leiden, The Netherlands
| |
Collapse
|
6
|
Mougiou D, Gioula G, Skoura L, Anastassopoulou C, Kachrimanidou M. Insights into the Interaction Between Clostridioides difficile and the Gut Microbiome. J Pers Med 2025; 15:94. [PMID: 40137411 PMCID: PMC11943401 DOI: 10.3390/jpm15030094] [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: 01/22/2025] [Revised: 02/19/2025] [Accepted: 02/27/2025] [Indexed: 03/27/2025] Open
Abstract
Clostridioides difficile (C. difficile) is a significant healthcare-associated pathogen that is predominantly caused by antibiotic-induced microbiota disturbance. Antibiotics decrease microbial diversity, resulting in C. difficile colonization and infection. Clostridium difficile infection (CDI) manifests through toxins A and B, causing diarrhea and colitis. Antibiotic usage, old age, and hospitalization are significant risk factors. A healthy gut microbiota, which is dominated by Firmicutes and Bacteroidetes, provides colonization resistance to C. difficile due to competition for nutrients, creating inhibitory substances and stimulating the immune response. Antibiotic-induced dysbiosis decreases resistance, allowing C. difficile spores to transform into vegetative forms. Patients with CDI have decreased gut microbiota diversity, with a decrease in beneficial bacteria, including Bacteroidetes, Prevotella, and Bifidobacterium, and a rise in harmful bacteria like Clostridioides and Lactobacillus. This disparity worsens the infection's symptoms and complicates therapy. Fecal Microbiota Transplantation (FMT) has emerged as a potential therapy for recurrent CDI by restoring gut microbiota diversity and function. Comprehending the connection between gut microbiota and CDI pathogenesis is critical for establishing effective preventive and treatment plans. Maintaining a healthy gut microbiota through careful antibiotic use and therapeutic options such as FMT can help in the management and prevention of CDI.
Collapse
Affiliation(s)
- Dimitra Mougiou
- Department of Microbiology, Medical School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (D.M.); (G.G.)
| | - Georgia Gioula
- Department of Microbiology, Medical School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (D.M.); (G.G.)
| | - Lemonia Skoura
- Department of Microbiology, AHEPA University Hospital, 54124 Thessaloniki, Greece;
| | - Cleo Anastassopoulou
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Melania Kachrimanidou
- Department of Microbiology, Medical School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (D.M.); (G.G.)
| |
Collapse
|
7
|
Clement J, Barlingay G, Addepalli S, Bang H, Donnelley MA, Cohen SH, Crabtree S. Risk factors for the development of Clostridioides difficile infection in patients colonized with toxigenic Clostridioides difficile. Infect Control Hosp Epidemiol 2025:1-7. [PMID: 39989316 DOI: 10.1017/ice.2025.4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
OBJECTIVE Asymptomatic patients colonized with toxigenic Clostridioides difficile are at risk of progressing to C. difficile infection (CDI), but risk factors associated with progression are poorly understood. The objectives of this study were to estimate the incidence and identify risk factors to progression of hospital-onset CDI (HO-CDI) among colonized patients. METHODS This was a nested case-control study at an academic medical center including adult patients colonized with toxigenic C. difficile, detected via polymerase chain reaction (PCR) on a rectal swab collected on admission from 2017 to 2020. Patients with prior CDI or symptoms on admission, neutropenia, prior rectal surgery, or hospitalization less than 24 hours were excluded. Colonized patients that developed HO-CDI were matched 1:3 to colonized patients who did not based on PCR test date. Bivariate and multivariable-adjusted Cox regression analyses were used to identify risk factors. RESULTS Of 2,150 colonized patients, 109 developed HO-CDI, with an incidence of 5.1%. After exclusions, 321 patients (69 with HO-CDI) were included, with an estimated incidence of 4.2%. Risk factors included cirrhosis (aHR 1.94), ICU admission (aHR 1.76), malignancy (aHR 1.88), and hospitalization within six months (aHR 1.6). Prior antibiotic exposure in the past three months (aHR 2.14) and receipt of at-risk antibiotics were also identified as potential risk factors (aHR 2.17). CONCLUSIONS Progression to HO-CDI among colonized patients was not uncommon. This study highlights key risk factors associated with progression, underscoring the importance of enhanced monitoring and prevention efforts tailored to high-risk populations to mitigate HO-CDI.
Collapse
Affiliation(s)
- Josh Clement
- Department of Pharmacy, University of California Davis Health, Sacramento, CA, USA
- Department of Pharmacy, Mount Sinai Hospital, New York, NY, USA
| | - Gauri Barlingay
- Division of Infectious Disease, University of California Davis Medical Center, Sacramento, CA, USA
| | - Sindhu Addepalli
- Department of Internal Medicine, University of California Davis, Sacramento, CA, USA
| | - Heejung Bang
- Division of Biostatistics, University of California Davis, Davis, CA, USA
| | - Monica A Donnelley
- Department of Pharmacy, University of California Davis Health, Sacramento, CA, USA
| | - Stuart H Cohen
- Division of Infectious Disease, University of California Davis Medical Center, Sacramento, CA, USA
| | - Scott Crabtree
- Division of Infectious Disease, University of California Davis Medical Center, Sacramento, CA, USA
| |
Collapse
|
8
|
Pappas MA, Herzig SJ, Auerbach AD, Deshpande A, Blanchard E, Rothberg MB. Impact of empiric antibiotics on risk of Clostridioides difficile-a causal inference observational analysis. ANTIMICROBIAL STEWARDSHIP & HEALTHCARE EPIDEMIOLOGY : ASHE 2025; 5:e40. [PMID: 39950004 PMCID: PMC11822578 DOI: 10.1017/ash.2025.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/14/2024] [Accepted: 12/17/2024] [Indexed: 02/16/2025]
Abstract
Background Clostridioides difficile infection (CDI) is a common and often nosocomial infection associated with increased mortality and morbidity. Antibiotic use is the most important modifiable risk factor, but many patients require empiric antibiotics. We estimated the increased risk of hospital-onset CDI with one daily dose-equivalent (DDE) of various empiric antibiotics compared to management without that daily dose-equivalent. Methods Using a multicenter retrospective cohort of adults admitted between March 2, 2020 and February 11, 2021 for the treatment of SARS-CoV-2, we used a series of three-level logistic regression models to estimate the probability of receiving each of several antibiotics of interest. For each antibiotic, we then limited our data set to patient-days at intermediate probability of receipt and used augmented inverse-probability weighted models to estimate the average treatment effect of one daily dose-equivalent, compared to management without that daily dose-equivalent, on the probability of hospital-onset CDI. Results In 24,406 patient-days at intermediate probability of receipt, parenteral vancomycin increased risk of hospital-onset CDI, with an average treatment effect of 0.0096 cases per daily dose-equivalent (95% CI: 0.0053-0.0138). In 38,003 patient-days at intermediate probability of receipt, cefepime also increased subsequent CDI risk, with an estimated effect of 0.0074 more cases per daily dose-equivalent (95% CI: 0.0022-0.0126). Conclusions Among common empiric antibiotics, parenteral vancomycin and cefepime appeared to increase risk of hospital-onset CDI. Causal inference observational study designs can be used to estimate patient-level harms of interventions such as empiric antimicrobials.
Collapse
Affiliation(s)
- Matthew A. Pappas
- Department of Hospital Medicine, Cleveland Clinic, Cleveland, OH, USA
- Center for Value-Based Care Research, Cleveland Clinic, Cleveland, OH, USA
- COVID-19 Consortium of HCA Healthcare and Academia for Research Generation, Nashville, TN, USA
| | - Shoshana J. Herzig
- COVID-19 Consortium of HCA Healthcare and Academia for Research Generation, Nashville, TN, USA
- Department of Medicine, Division of General Medicine, and Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Andrew D. Auerbach
- COVID-19 Consortium of HCA Healthcare and Academia for Research Generation, Nashville, TN, USA
- Department of Hospital Medicine, University of California, San Francisco, CA, USA
| | - Abhishek Deshpande
- Center for Value-Based Care Research, Cleveland Clinic, Cleveland, OH, USA
| | - Eunice Blanchard
- COVID-19 Consortium of HCA Healthcare and Academia for Research Generation, Nashville, TN, USA
- Infection Control and Hospital Epidemiology, HCA Healthcare, Nashville, TN, USA
| | | |
Collapse
|
9
|
Alshannaq AF, Kates AE, Keating JA, Mckinley LL, Dixon JW, Safdar N. Diverse Sources and Latent Reservoirs of Community-Associated Clostridioides difficile Infection. Clin Infect Dis 2025; 80:37-42. [PMID: 39215602 DOI: 10.1093/cid/ciae429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/30/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024] Open
Abstract
Clostridioides difficile is a spore-forming, toxin-producing, anaerobic bacterium that infects the human gastrointestinal tract, causing diarrhea and life-threatening colitis. Clostridioides difficile epidemiology continues to evolve, and it is recognized as a major community-associated (CA) pathogen in addition to its established role in causing healthcare-associated (HA) infection. While current surveillance and prevention measures mainly focus on healthcare-associated C. difficile infections (HA-CDI), much less is known about the factors that drive CA-CDI. This review highlights the potential contribution of reservoirs, including asymptomatic carriers, to CA C. difficile transmission. The reservoirs discussed in this review provide potential avenues for research to better understand and reduce CA transmission of C. difficile.
Collapse
Affiliation(s)
- Ahmad F Alshannaq
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Ashley E Kates
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Medicine, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Julie A Keating
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Medicine, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Linda L Mckinley
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Medicine, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Jonah W Dixon
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Medicine, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| | - Nasia Safdar
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Medicine, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, USA
| |
Collapse
|
10
|
Angelopoulos N, Staines J, Chamberlin M, Bates S, McGain F. A narrative review of personal protective equipment gowns: lessons from COVID-19. Br J Anaesth 2025; 134:368-381. [PMID: 39516124 DOI: 10.1016/j.bja.2024.09.014] [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: 07/17/2023] [Revised: 08/25/2024] [Accepted: 09/18/2024] [Indexed: 11/16/2024] Open
Abstract
This narrative review evaluates the evidence regarding the protection offered by isolation gowns, approaches to imparting antimicrobial activity to gowns, and the environmental impacts of gown use, particularly during the COVID-19 pandemic. We conducted a search of the Medline, PubMed, and Google Scholar databases for articles published between January 1, 2019 to February 20, 2024. We found that current standards pertaining to isolation gowns might be irrelevant to the protection of healthcare workers from pathogen transmission, as they focus primarily on fluid barrier resistance values that are not reflective of all transmission conditions in hospitals. Although most available isolation gowns are disposable, reusable gowns could offer greater barrier protection and are more environmentally sustainable. Several techniques have been studied for their ability to impart antimicrobial properties to isolation gowns, extending their lifespan and reducing environmental impacts. However, evidence of the effectiveness of such techniques in clinical settings is scarce. We advocate for standardised guidelines inclusive of common pathogen survival tests, comfort, and durability, which reflect the actual infection risks encountered by healthcare workers, to improve the safety and efficacy of isolation gowns in hospital settings. Further research into the clinical effectiveness of antimicrobial gowns and their long-term implications on the environment is also warranted.
Collapse
Affiliation(s)
- Nikolaos Angelopoulos
- Department of Anaesthesia and Intensive Care, Western Health, Melbourne, VIC, Australia.
| | - Jo Staines
- Department of Mechanical Engineering, The University of Melbourne, Melbourne, VIC, Australia
| | - Meriel Chamberlin
- Limebranch Pty Ltd trading as Full Circle Fibres, Brisbane, QLD, Australia
| | - Samantha Bates
- Department of Anaesthesia and Intensive Care, Western Health, Melbourne, VIC, Australia; Department of Critical Care, Faculty of Medicine, Dentistry & Health Sciences, The University of Melbourne, Melbourne, VIC, Australia
| | - Forbes McGain
- Department of Anaesthesia and Intensive Care, Western Health, Melbourne, VIC, Australia; Department of Critical Care, Faculty of Medicine, Dentistry & Health Sciences, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
11
|
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
|
12
|
Tansarli GS, Falagas ME, Fang FC. Clinical significance of toxin EIA positivity in patients with suspected Clostridioides difficile infection: systematic review and meta-analysis. J Clin Microbiol 2025; 63:e0097724. [PMID: 39665542 PMCID: PMC11784090 DOI: 10.1128/jcm.00977-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 11/14/2024] [Indexed: 12/13/2024] Open
Abstract
The laboratory diagnosis of Clostridioides difficile infection (CDI) is controversial. Nucleic acid amplification tests (NAAT) and toxin enzyme immunoassays (EIA) are most widely used, often in combination. However, the interpretation of a positive NAAT and negative toxin immunoassay (NAAT+/EIA-) is uncertain. PubMed and EMBASE were searched for studies reporting clinical outcomes in NAAT+/EIA- versus NAAT+/EIA+ patients. Forty-six studies comprising 33,959 patients were included in this meta-analysis. All-cause mortality (RR 0.96, 95% CI 0.80-1.15), attributable mortality (RR 0.61, 95% CI 0.20-1.91), fulminant CDI (RR 0.83, 95% CI 0.57-1.20), radiographic evidence of CDI (RR 0.87, 95% CI 0.65-1.16), total CDI complications (RR 0.95, 95% CI 0.59-1.53), colectomies (RR 0.78, 95% CI 0.34-1.79), and ICU admission (RR 1.04, 95% CI 0.84-1.30) did not significantly differ between NAAT+/EIA- and NAAT+/EIA+ patients. However, rates of recurrent (RR 0.62, 95% CI 0.50-0.77) or severe (RR 0.74, 95% CI 0.63-0.88) CDI were significantly lower in NAAT+/EIA- patients than in NAAT+/EIA+ patients. The pooled prevalence of NAAT+/EIA- patients who were treated with antibiotics for CDI was 73.4% (pooled proportion 0.72, 95% CI 0.52-0.88). NAAT+/EIA- patients have lower rates of recurrence and are at reduced risk for severe CDI compared with NAAT+/EIA+ patients but have a risk of CDI-related complications and mortality comparable to that of NAAT+/EIA+ patients. Toxin results cannot rule in or rule out CDI, and the decision whether to treat symptomatic NAAT+/EIA- patients for CDI should be based on clinical presentation and not on the toxin result.IMPORTANCEClostridioides difficile infection (CDI) is a common cause of healthcare-associated infections and the leading cause of antibiotic-associated diarrhea. However, the laboratory diagnosis of CDI, primarily done by nucleic acid amplification test (NAAT) and enzyme immunoassay (EIA), is controversial, especially in patients who test positive by NAAT but negative by EIA. In this systematic review, we compared the clinical outcomes of NAAT+/EIA- versus NAAT+/EIA+ patients and found that the two groups have similar risk of mortality and CDI-related complications. However, NAAT+/EIA- patients had significantly lower rates of recurrence and severe CDI than NAAT+/EIA+ patients, and most NAAT+/EIA- patients received CDI therapy. Toxin testing can help to predict the likelihood of CDI recurrence or severe infection, but the toxin result should not be a determining factor in the administration of CDI therapy. The decision on whether to treat NAAT+/EIA- patients should be based on clinical assessment.
Collapse
Affiliation(s)
- Giannoula S. Tansarli
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Matthew E. Falagas
- Department of Medicine, Alfa Institute of Biomedical Science, Athens, Greece
- Department of Medicine, School of Medicine, European University Cyprus, Nicosia, Cyprus
| | - Ferric C. Fang
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
- Clinical Microbiology Laboratory, Harborview Medical Center, Seattle, Washington, USA
- Department of Microbiology, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
13
|
De-la-Rosa-Martínez D, Villaseñor-Echavarri R, Vilar-Compte D, Mosqueda-Larrauri V, Zinser-Peniche P, Blumberg S. Heterogeneity of Clostridioides difficile asymptomatic colonization prevalence: a systematic review and meta-analysis. Gut Pathog 2025; 17:6. [PMID: 39871276 PMCID: PMC11773978 DOI: 10.1186/s13099-024-00674-0] [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: 10/14/2024] [Accepted: 12/24/2024] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND Asymptomatic carriers significantly influence the transmission dynamics of C. difficile. This study aimed to assess the prevalence of toxigenic C. difficile asymptomatic colonization (tCDAC) and investigate its heterogeneity across different populations. We searched MEDLINE, Web of Science, and Scopus for articles published between 2000 and 2023 on tCDAC. Studies including asymptomatic adults with laboratory-confirmed tCDAC were eligible. We performed a random-effects meta-analysis to estimate the pooled prevalence by clinical characteristics, settings, and geographic areas. In addition, we used outlier analyses and meta-regression to explore sources of prevalence variability. RESULTS Fifty-one studies involving 39,447 patients were included. The tCDAC prevalence ranged from 0.5 to 51.5%. Among pooled estimates, a high prevalence was observed in patients with cystic fibrosis, outbreak settings, and cancer patients, whereas the lowest rates were found in healthy individuals and healthcare workers. Similar colonization rates were observed between admitted and hospitalized patients. Our meta-regression analysis revealed lower rates in healthy individuals and higher rates in cystic fibrosis patients and studies from North America. Additionally, compared with that among healthy individuals, the prevalence significantly increased by 15-47% among different populations and settings. CONCLUSION Our study revealed that tCDAC is a common phenomenon. We found high prevalence estimates that showed significant variability across populations. This heterogeneity could be partially explained by population characteristics and settings, supporting their role in the pathogenesis and burden of this disease. This highlights the need to identify high-risk groups to improve infection control strategies, decrease transmission dynamics, and better understand the natural history of this disease.
Collapse
Affiliation(s)
- Daniel De-la-Rosa-Martínez
- Francis I Proctor Foundation, University of California San Francisco, 490 Illinois St, San Francisco, CA, 94158, USA.
| | | | - Diana Vilar-Compte
- Department of Infectious Diseases, Instituto Nacional de Cancerología, Mexico City, Mexico
| | | | - Paola Zinser-Peniche
- Department of Infectious Diseases, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Seth Blumberg
- Francis I Proctor Foundation, University of California San Francisco, 490 Illinois St, San Francisco, CA, 94158, USA
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
14
|
Meln I, Cnossen V, Corti N, Andeweg A, Baay M, Chiu C, Coia J, Cornely O, Cox RJ, Dasyam D, De Keersmaecker SCJ, Deming M, Waldock J, Engelhardt OG, Guo M, Haj-Ali Saflo O, Hensen A, Jeeninga R, Kolstoe S, Krut O, Kuijper EJ, Leal L, Mazur N, Mohn KGI, Morel S, Osterhaus A, Moreira AP, Smits WK, Sridhar S, Toomey D, van Gerven J, Vehreschild MJGT, Yarzabal JP, Zimmer-Harwood P, Neels P, Olesen OF, Roestenberg M, Kamerling IMC. Regulatory workshop on standardisation of clinical procedures, endpoints and data robustness of human challenge studies - A stakeholder meeting report. Biologicals 2025; 90:101818. [PMID: 39824043 DOI: 10.1016/j.biologicals.2025.101818] [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: 09/25/2024] [Revised: 01/10/2025] [Accepted: 01/10/2025] [Indexed: 01/20/2025] Open
Abstract
Inno4Vac, a public-private partnership funded by the IMI2/EU/EFPIA Joint Undertaking (IMI2 JU), brings together academic institutions, SMEs, and pharmaceutical companies to accelerate and de-risk vaccine development. The project has made significant strides in the selection and production of challenge agents for influenza, respiratory syncytial virus (RSV), and toxigenic Clostridioides difficile for controlled human infection model studies (CHIMs). A regulatory workshop held on March 20, 2024, addressed the standardisation of clinical procedures, ethical considerations, endpoints, and data integrity, highlighting the ongoing initiatives related to these CHIMs. Key discussions focused on refining trial protocols to balance statistical power with participant burden, overseen by a data safety monitoring board. The meeting emphasised the importance of harmonizing CHIM protocols to ensure robust, reproducible, and transparent research. Mandatory trial registration and adherence to the Findable, Accessible, Interoperable, and Reusable (FAIR) data principles were recommended to enhance data reuse and scientific value. This report consolidates efforts to standardise CHIM protocols, essential for accelerating therapeutic innovations and advancing global health research.
Collapse
Affiliation(s)
- Irina Meln
- European Vaccine Initiative (EVI), Heidelberg, Germany.
| | - Victor Cnossen
- Centre for Human Drug Research (CHDR), Leiden, the Netherlands
| | | | - Arno Andeweg
- European Medicines Agency (EMA), Amsterdam, the Netherlands
| | - Marc Baay
- P95 Clinical & Epidemiology Services, Leuven, Belgium
| | | | - John Coia
- Institute of Regional Health Research, University of Southern Denmark, Denmark
| | - Oliver Cornely
- Department of Internal Medicine, Division of Infectious Diseases, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; German Centre for Infection Research (DZIF), Site Bonn-Cologne, Cologne, Germany
| | - Rebecca J Cox
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
| | | | | | - Meagan Deming
- University of Maryland School of Medicine, United States
| | - Joanna Waldock
- Vaccines Division, Science & Research Group, Medicines and Healthcare products Regulatory Agency (MHRA), Potters Bar, United Kingdom
| | - Othmar G Engelhardt
- Vaccines Division, Science & Research Group, Medicines and Healthcare products Regulatory Agency (MHRA), Potters Bar, United Kingdom
| | - Manman Guo
- University of Oxford, Oxford, United Kingdom
| | | | - Annefleur Hensen
- Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Rienk Jeeninga
- Viroclinics-DDL, a Cerba Research Company (VC), Rotterdam, the Netherlands
| | - Simon Kolstoe
- University of Portsmouth, Portsmouth, United Kingdom
| | - Oleg Krut
- Paul-Ehrlich-Institut (PEI), Langen, Germany
| | - Ed J Kuijper
- Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Lorna Leal
- European Medicines Agency (EMA), Amsterdam, the Netherlands
| | - Natalie Mazur
- Department of Pediatrics, Wilhelmina Children's Hospital, University Medical Center Utrecht (UMCU), Utrecht, the Netherlands
| | - Kristin G I Mohn
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
| | | | - Ab Osterhaus
- Stiftung Tierärztliche Hochschule Hannover (TIHO), Hannover, Germany
| | | | - Wiep Klaas Smits
- Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | | | | | - Joop van Gerven
- Central Committee on Research Involving Human Subjects (CCMO), the Hague, the Netherlands
| | - Maria J G T Vehreschild
- Department of Internal Medicine, Division of Infectious Diseases, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; German Centre for Infection Research (DZIF), Site Bonn-Cologne, Cologne, Germany
| | | | | | - Pieter Neels
- International Alliance of Biological Standardization (IABS-EU), Lyon, France
| | - Ole F Olesen
- European Vaccine Initiative (EVI), Heidelberg, Germany
| | - Meta Roestenberg
- Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Ingrid M C Kamerling
- Centre for Human Drug Research (CHDR), Leiden, the Netherlands; Leiden University Medical Center (LUMC), Leiden, the Netherlands
| |
Collapse
|
15
|
Sulaiman JE, Thompson J, Cheung PLK, Qian Y, Mill J, James I, Im H, Vivas EI, Simcox J, Venturelli OS. Phocaeicola vulgatus shapes the long-term growth dynamics and evolutionary adaptations of Clostridioides difficile. Cell Host Microbe 2025; 33:42-58.e10. [PMID: 39730002 PMCID: PMC11852276 DOI: 10.1016/j.chom.2024.12.001] [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] [Received: 07/10/2024] [Revised: 10/14/2024] [Accepted: 12/02/2024] [Indexed: 12/29/2024]
Abstract
Clostridioides difficile can transiently or persistently colonize the human gut, posing a risk for infections. This colonization is influenced by complex molecular and ecological interactions with the human gut microbiota. By investigating C. difficile dynamics in human gut communities over hundreds of generations, we show patterns of stable coexistence, instability, or competitive exclusion. Lowering carbohydrate concentrations shifted a community containing C. difficile and the prevalent human gut symbiont Phocaeicola vulgatus from competitive exclusion to coexistence, facilitated by increased cross-feeding. In this environment, two key mutations in C. difficile altered its metabolic niche from proline to glucose utilization. These metabolic changes in C. difficile substantially impacted gut microbiota inter-species interactions and reduced disease severity in mice. In sum, interactions with P. vulgatus are crucial in shaping the long-term growth dynamics and evolutionary adaptations of C. difficile, offering key insights for developing anti-C. difficile strategies.
Collapse
Affiliation(s)
- Jordy Evan Sulaiman
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Jaron Thompson
- Department of Chemical & Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Pak Lun Kevin Cheung
- Department of Electrical and Computer Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Yili Qian
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Jericha Mill
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Isabella James
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Hanhyeok Im
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Eugenio I Vivas
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA; Gnotobiotic Animal Core Facility, University of Wisconsin-Madison, Madison, WI, USA
| | - Judith Simcox
- Howard Hughes Medical Institute, Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Ophelia S Venturelli
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA; Department of Chemical & Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA; Department of Biomedical Engineering, Duke University, Durham, NC, USA; Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
16
|
Rong X, Shu Q. Modulating gut microbiota for treating antibiotic-associated diarrhea from Clostridium difficile infection: insights from Lizhong decoction and its polysaccharide component. Lett Appl Microbiol 2024; 77:ovae121. [PMID: 39611340 DOI: 10.1093/lambio/ovae121] [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: 07/17/2024] [Revised: 11/23/2024] [Accepted: 11/27/2024] [Indexed: 11/30/2024]
Abstract
To investigate whether the polysaccharide component of the Traditional Chinese Medicine (TCM) formula Lizhong decoction (LZD) has therapeutic effects and regulates gut microbiota in antibiotic-associated diarrhea (AAD) related to Clostridium difficile infection, a polysaccharide was extracted from LZD, consisting of rhamnose, five monosaccharides, arabinose, galactose, glucose, and galacturonic acid in a molar ratio of 1.12:8.49:4.06:80.67:5.66. An AAD model related to C. difficile infection was established using clindamycin gavage and oral colonization with C. difficile. Subsequently, oral treatment experiments with LZD polysaccharides and different doses of the decoction were conducted. The results indicated that the polysaccharide showed the best therapeutic effect on AAD related to C. difficile infection, with post-treatment gut microbiota clustering closest to the normal control group. The polysaccharide from LZD has therapeutic effects on AAD related to C. difficile infection, possibly achieved through the regulation of gut microbiota. It modulates the dominant gut microbiota, specifically increasing the abundance of beneficial bacteria by adjusting the ratio of Bacteroidetes and Firmicutes, thereby reversing the dysbiosis caused by C. difficile infection and antibiotics. Thus, LZD polysaccharide may be one of the important active components for gut microbiota regulation in this TCM formula.
Collapse
Affiliation(s)
- XinQian Rong
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, No. 1688 Meiling Road, Nanchang 330004, PR China
| | - QingLong Shu
- College of Traditional Chinese Medicine, Jiangxi University of Chinese Medicine, No. 1688 Meiling Road, Nanchang 330004, PR China
| |
Collapse
|
17
|
Ray MJ, Strnad LC, Tucker KJ, Furuno JP, Lofgren ET, McCracken CM, Park H, Gerber JS, McGregor JC. Influence of Antibiotic Exposure Intensity on the Risk of Clostridioides difficile Infection. Clin Infect Dis 2024; 79:1129-1135. [PMID: 38743579 PMCID: PMC11581687 DOI: 10.1093/cid/ciae259] [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: 02/22/2024] [Revised: 05/01/2024] [Accepted: 05/07/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Antibiotics are a strong risk factor for Clostridioides difficile infection (CDI), and CDI incidence is often measured as an important outcome metric for antimicrobial stewardship interventions aiming to reduce antibiotic use. However, risk of CDI from antibiotics varies by agent and dependent on the intensity (ie, spectrum and duration) of antibiotic therapy. Thus, the impact of stewardship interventions on CDI incidence is variable, and understanding this risk requires a more granular measure of intensity of therapy than traditionally used measures like days of therapy (DOT). METHODS We performed a retrospective cohort study to measure the independent association between intensity of antibiotic therapy, as measured by the antibiotic spectrum index (ASI), and hospital-associated CDI (HA-CDI) at a large academic medical center between January 2018 and March 2020. We constructed a marginal Poisson regression model to generate adjusted relative risks for a unit increase in ASI per antibiotic day. RESULTS We included 35 457 inpatient encounters in our cohort. Sixty-eight percent of patients received at least 1 antibiotic. We identified 128 HA-CDI cases, which corresponds to an incidence rate of 4.1 cases per 10 000 patient-days. After adjusting for known confounders, each additional unit increase in ASI per antibiotic day was associated with 1.09 times the risk of HA-CDI (relative risk = 1.09; 95% CI: 1.06-1.13). CONCLUSIONS The ASI was strongly associated with HA-CDI and could be a useful tool in evaluating the impact of antibiotic stewardship on HA-CDI rates, providing more granular information than the more commonly used DOT.
Collapse
Affiliation(s)
- Michael J Ray
- Department of Pharmacy Practice, Oregon State University College of Pharmacy, Portland, Oregon, USA
- Oregon Health & Science University–Portland State University School of Public Health, Portland, Oregon, USA
| | - Luke C Strnad
- Oregon Health & Science University–Portland State University School of Public Health, Portland, Oregon, USA
- Division of Infectious Diseases, Oregon Health & Science University School of Medicine, Portland, Oregon, USA
| | - Kendall J Tucker
- Department of Pharmacy Practice, Oregon State University College of Pharmacy, Portland, Oregon, USA
| | - Jon P Furuno
- Department of Pharmacy Practice, Oregon State University College of Pharmacy, Portland, Oregon, USA
| | - Eric T Lofgren
- Washington State University Allen School for Global Health, Pullman, Washington, USA
| | - Caitlin M McCracken
- Department of Pharmacy Practice, Oregon State University College of Pharmacy, Portland, Oregon, USA
| | - Hiro Park
- Department of Pharmacy Practice, Oregon State University College of Pharmacy, Portland, Oregon, USA
| | - Jeffrey S Gerber
- Division of Infectious Diseases, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jessina C McGregor
- Department of Pharmacy Practice, Oregon State University College of Pharmacy, Portland, Oregon, USA
- Oregon Health & Science University–Portland State University School of Public Health, Portland, Oregon, USA
| |
Collapse
|
18
|
Chen M, Wang R, Wang T. Gut microbiota and skin pathologies: Mechanism of the gut-skin axis in atopic dermatitis and psoriasis. Int Immunopharmacol 2024; 141:112658. [PMID: 39137625 DOI: 10.1016/j.intimp.2024.112658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 07/01/2024] [Accepted: 07/07/2024] [Indexed: 08/15/2024]
Abstract
Atopic dermatitis (AD) and psoriasis are chronic skin diseases with a global impact, posing significant challenges to public health systems and severely affecting patients' quality of life. This review delves into the key role of the gut microbiota in these diseases, emphasizing the importance of the gut-skin axis in inflammatory mediators and immune regulation and revealing a complex bidirectional communication system. We comprehensively assessed the pathogenesis, clinical manifestations, and treatment strategies for AD and psoriasis, with a particular focus on how the gut microbiota and their metabolites influence disease progression via the gut-skin axis. In addition, personalized treatment plans based on individual patient microbiome characteristics have been proposed, offering new perspectives for future treatment approaches. We call for enhanced interdisciplinary cooperation to further explore the interactions between gut microbiota and skin diseases and to assess the potential of drugs and natural products in modulating the gut-skin axis, aiming to advance the treatment of skin diseases.
Collapse
Affiliation(s)
- Meng Chen
- Department of Dermatology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou 318000, China
| | - Rui Wang
- Department of Dermatology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou 318000, China.
| | - Ting Wang
- Department of Dermatology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou 318000, China.
| |
Collapse
|
19
|
Williams MJ, Atienza S, Franzen E, Rathod H, Mejaki B, Graff J, Korman S, Zouine N, Gul Z, Sana S, Medlin S, Buggy BP. Evaluation of Primary Oral Vancomycin Prophylaxis Against Clostridioides difficile Infection During Autologous Stem Cell Transplantation. Open Forum Infect Dis 2024; 11:ofae622. [PMID: 39498172 PMCID: PMC11532790 DOI: 10.1093/ofid/ofae622] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 10/09/2024] [Indexed: 11/07/2024] Open
Abstract
Background Evaluations of oral vancomycin prophylaxis (OVP) against Clostridioides difficile have been reported in stem cell transplant populations with short follow-up periods. The longest known duration of standardized follow-up post-OVP is 90 days within an allogeneic stem cell transplant population. In 2017, we implemented OVP 125 mg twice daily in autologous stem cell transplant (ASCT) recipients beginning the day of admission and continued until the day of discharge. Methods Patients who received an ASCT within our institution between 1 January 2012 and 31 December 2021 were included and separated into 2 groups based on the receipt of OVP. The primary study aim was to measure the incidence of C difficile infection (CDI) during the ASCT admission. A secondary aim was to evaluate for delayed CDI 180 days post-discharge. Other factors evaluated were prior history of CDI, use of systemic antimicrobials, and length of stay. Results Overall, 254 patients were evaluated and 58% received OVP, predominantly as primary prophylaxis (95%). Of the 18 patients who developed in-hospital CDI, 6 were in the OVP group versus 12 in the non-OVP cohort (4% vs 11%, P = .03). In the 180-day follow-up period, OVP use did not increase risk of developing CDI after discontinuation while in-hospital length of stay was identified as a significant factor. Conclusions The use of OVP significantly reduced the incidence of CDI during the in-hospital ASCT course without increasing CDI post-OVP use. These encouraging results should promote further research into the use of OVP in ASCT.
Collapse
Affiliation(s)
- Michael J Williams
- Malignant Hematology and Stem Cell Transplant, Aurora St Luke's Medical Center, Milwaukee, Wisconsin, USA
- Department of Pharmacy, Aurora St Luke's Medical Center, Milwaukee, Wisconsin, USA
| | - Sol Atienza
- Department of Pharmacy, Aurora St Luke's Medical Center, Milwaukee, Wisconsin, USA
| | - Erin Franzen
- Department of Pharmacy, Aurora St Luke's Medical Center, Milwaukee, Wisconsin, USA
| | - Heena Rathod
- Department of Pharmacy, Aurora St Luke's Medical Center, Milwaukee, Wisconsin, USA
| | - Brittany Mejaki
- Malignant Hematology and Stem Cell Transplant, Aurora St Luke's Medical Center, Milwaukee, Wisconsin, USA
- Department of Pharmacy, Aurora St Luke's Medical Center, Milwaukee, Wisconsin, USA
| | - Justin Graff
- Malignant Hematology and Stem Cell Transplant, Aurora St Luke's Medical Center, Milwaukee, Wisconsin, USA
- Department of Pharmacy, Aurora St Luke's Medical Center, Milwaukee, Wisconsin, USA
| | - Sandra Korman
- Quality Management, Aurora St Luke's Medical Center, Milwaukee, Wisconsin, USA
| | - Noah Zouine
- Quality Management, Aurora St Luke's Medical Center, Milwaukee, Wisconsin, USA
| | - Zartash Gul
- Malignant Hematology and Stem Cell Transplant, Aurora St Luke's Medical Center, Milwaukee, Wisconsin, USA
| | - Sherjeel Sana
- Malignant Hematology and Stem Cell Transplant, Aurora St Luke's Medical Center, Milwaukee, Wisconsin, USA
| | - Stephen Medlin
- Malignant Hematology and Stem Cell Transplant, Aurora St Luke's Medical Center, Milwaukee, Wisconsin, USA
| | - Brian P Buggy
- Infectious Diseases Section, Aurora St Luke's Medical Center, Milwaukee, Wisconsin, USA
| |
Collapse
|
20
|
Zhou H, Wang X, She Z, Huang L, Wei H, Yang S, Wei Z, Chen H, Yang B, Hu Z, Feng X, Zhu P, Li Z, Shen J, Liu H, Dong H, Chen G, Zhang Q. Combining bioinformatics and multiomics strategies to investigate the key microbiota and active components of Liupao tea ameliorating hyperlipidemia. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118438. [PMID: 38848972 DOI: 10.1016/j.jep.2024.118438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/27/2024] [Accepted: 06/05/2024] [Indexed: 06/09/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Hyperlipidemia as a major health issue has attracted much public attention. As a geographical indication product of China, Liupao tea (LPT) is a typical representative of traditional Chinese dark tea that has shown good potential in regulating glucose and lipid metabolism. LPT has important medicinal value in hyperlipidemia prevention. However, the active ingredients and metabolic mechanisms by which LPT alleviates hyperlipidemia remain unclear. AIM OF THE STUDY This study aimed to systematically investigate the metabolic mechanisms and active ingredients of LPT extract in alleviating hyperlipidemia. MATERIALS AND METHODS Firstly, we developed a mouse model of hyperlipidemia to study the pharmacodynamics of LPT. Subsequently, network pharmacology and molecular docking were performed to predict the potential key active ingredients and core targets of LPT against hyperlipidemia. LC-MS/MS was used to validate the identity of key active ingredients in LPT with chemical standards. Finally, the effect and metabolic mechanisms of LPT extract in alleviating hyperlipidemia were investigated by integrating metabolomic, lipidomic, and gut microbiome analyses. RESULTS Results showed that LPT extract effectively improved hyperlipidemia by suppressing weight gain, remedying dysregulation of glucose and lipid metabolism, and reducing hepatic damage. Network pharmacology analysis and molecular docking suggested that four potential active ingredients and seven potential core targets were closely associated with roles for hyperlipidemia treatment. Ellagic acid, catechin, and naringenin were considered to be the key active ingredients of LPT alleviating hyperlipidemia. Additionally, LPT extract modulated the mRNA expression levels of Fxr, Cyp7a1, Cyp8b1, and Cyp27a1 associated with bile acid (BA) metabolism, mitigated the disturbances of BA and glycerophospholipid (GP) metabolism in hyperlipidemia mice. Combining fecal microbiota transplantation and correlation analysis, LPT extract effectively improved species diversity and abundance of gut microbiota, particularly the BA and GP metabolism-related gut microbiota, in the hyperlipidemia mice. CONCLUSIONS LPT extract ameliorated hyperlipidemia by modulating GP and BA metabolism by regulating Lactobacillus and Dubosiella, thereby alleviating hyperlipidemia. Three active ingredients of LPT served as the key factors in exerting an improvement on hyperlipidemia. These findings provide new insights into the active ingredients and metabolic mechanisms of LPT in improving hyperlipidemia, suggesting that LPT can be used to prevent and therapeutic hyperlipidemia.
Collapse
Affiliation(s)
- Hailin Zhou
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Guangxi, China.
| | - Xuancheng Wang
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Guangxi, China.
| | - Zhiyong She
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Guangxi, China.
| | - Li Huang
- College of Light Industry and Food Engineering, Guangxi University, Guangxi, China.
| | - Huijie Wei
- College of Light Industry and Food Engineering, Guangxi University, Guangxi, China.
| | - Shanyi Yang
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Guangxi, China.
| | - Zhijuan Wei
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Guangxi, China.
| | - Hongwei Chen
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Guangxi, China.
| | - Bao Yang
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Diseases, Hubei Minzu University, Hubei, China.
| | - Zehua Hu
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Diseases, Hubei Minzu University, Hubei, China.
| | - Xue Feng
- Center for Instrumental Analysis, Guangxi University, Guangxi, China.
| | - Pingchuan Zhu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Guangxi, China.
| | - Zijian Li
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Guangxi, China.
| | - Jiahui Shen
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Guangxi, China.
| | - Huan Liu
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Guangxi, China.
| | - Huanxiao Dong
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Guangxi, China.
| | - Guanghui Chen
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Guangxi University of Chinese Medicine, Guangxi, China.
| | - Qisong Zhang
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Guangxi, China; Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic Diseases, Hubei Minzu University, Hubei, China; Center for Instrumental Analysis, Guangxi University, Guangxi, China.
| |
Collapse
|
21
|
Ray MJ, Furuno JP, Strnad LC, Lofgren ET, McGregor JC. Examining the impact of the COVID-19 pandemic on hospital-associated Clostridioides difficile infection. Infect Control Hosp Epidemiol 2024:1-7. [PMID: 39429023 DOI: 10.1017/ice.2024.128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
OBJECTIVE To evaluate the impact of changes in the size and characteristics of the hospitalized patient population during the COVID-19 pandemic on the incidence of hospital-associated Clostridioides difficile infection (HA-CDI). DESIGN Interrupted time-series analysis. SETTING A 576-bed academic medical center in Portland, Oregon. METHODS We established March 23, 2020 as our pandemic onset and included 24 pre-pandemic and 24 pandemic-era 30-day intervals. We built an autoregressive segmented regression model to evaluate immediate and gradual changes in HA-CDI rate during the pandemic while controlling for changes in known CDI risk factors. RESULTS We observed 4.5 HA-CDI cases per 10,000 patient-days in the two years prior to the pandemic and 4.7 cases per 10,000 patient-days in the first two years of the pandemic. According to our adjusted segmented regression model, there were neither significant changes in HA-CDI rate at the onset of the pandemic (level-change coefficient = 0.70, P-value = 0.57) nor overtime during the pandemic (slope-change coefficient = 0.003, P-value = 0.97). We observed significant increases in frequency and intensity of antibiotic use, time at risk, comorbidities, and patient age before and after the pandemic onset. Frequency of C. difficile testing did not significantly change during the pandemic (P= 0.72). CONCLUSIONS Despite large increases in several CDI risk factors, we did not observe the expected corresponding changes in HA-CDI rate during the first two years of the COVID-19 pandemic. We hypothesize that infection prevention measures responding to COVID-19 played a role in CDI prevention.
Collapse
Affiliation(s)
- Michael J Ray
- Oregon State University College of Pharmacy, Department of Pharmacy Practice, Portland, OR, USA
- Oregon Health & Science University-Portland State University School of Public Health, Portland, OR, USA
| | - Jon P Furuno
- Oregon State University College of Pharmacy, Department of Pharmacy Practice, Portland, OR, USA
| | - Luke C Strnad
- Oregon Health & Science University-Portland State University School of Public Health, Portland, OR, USA
- Oregon Health & Science University School of Medicine, Division of Infectious Diseases, Portland, OR, USA
- Washington State University Allen School for Global Health, Pullman, WA, USA
| | - Eric T Lofgren
- Washington State University Allen School for Global Health, Pullman, WA, USA
| | - Jessina C McGregor
- Oregon State University College of Pharmacy, Department of Pharmacy Practice, Portland, OR, USA
- Oregon Health & Science University-Portland State University School of Public Health, Portland, OR, USA
| |
Collapse
|
22
|
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
|
23
|
Sansonetti PJ, Doré J. [The human microbiome proofed by the Anthropocene: from correlation to causality and intervention]. Med Sci (Paris) 2024; 40:757-765. [PMID: 39450961 DOI: 10.1051/medsci/2024121] [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: 10/26/2024] Open
Abstract
The deleterious effects of human activities on biodiversity in the vegetal and animal world, and on climate changes are now well-established facts. However, little is yet known on the impact of human activities on microbial diversity on the planet and more specifically on the human microbiota Large implementation of metagenomics allows exaustive microbial cataloguing with broad spatio-temporal resolution of human microbiota. A reduction in bacterial richness and diversity in the human microbiota, particularly in the intestinal tract, is now established and particularly obvious in the most industrialized regions of the planet. Massive, uncontrolled use of antibiotics, drastic changes in traditional food habits and some elements of the "global exposome" that remain to identify are usually considered as stressors accounting for this situation of "missing microbes". As a consequence, a dysbiotic situation develops, a "dysbiosis" being characterized by the erosion of the central core of shared bacterial species across individuals and the development of opportunistic "pathobionts" in response to a weaker barrier capacity of these impoverished microbiota. The current challenge is to establish a causality link between the extension of these dysbiotic situations and the steady emergence of epidemic, non-communicable diseases such as asthma, allergy, obesity, diabetes, autoimmune diseases and some cancers. Experimental animal models combined with controlled, prospective clinical interventions are in demand to consolidate causality links, with the understanding that in the deciphering of the mechanisms of alteration of the human-microbiome symbiosis resides a novel exciting chapter of medicine: "microbial medicine".
Collapse
Affiliation(s)
| | - Joël Doré
- Université Paris-Saclay, INRAE, MetaGenoPolis, AgroParisTech, MICALIS Jouy-en-Josas France
| |
Collapse
|
24
|
Ke F, Dong ZH, Bu F, Li CN, He QT, Liu ZC, Lu J, Yu K, Wang DG, Xu HN, Ye CT. Clostridium difficile infection following colon subtotal resection in a patient with gallstones: A case report and review of literature. World J Gastrointest Surg 2024; 16:3048-3056. [PMID: 39351567 PMCID: PMC11438826 DOI: 10.4240/wjgs.v16.i9.3048] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 07/02/2024] [Accepted: 07/10/2024] [Indexed: 09/18/2024] Open
Abstract
BACKGROUND Clostridium difficile (C. difficile) infection (CDI) is a rare clinical disease caused by changes in the intestinal microenvironment, which has a variety of causes and a poor prognosis, and for which there is no standardized clinical treatment. CASE SUMMARY A patient experienced recurrent difficulty in bowel movements over the past decade. Recently, symptoms worsened within the last ten days, leading to a clinic visit due to constipation. The patient was subsequently referred to our department. Preoperatively, the patient was diagnosed with obstructed colon accompanied by gallstones. Empirical antibiotics were administered both before and after surgery to prevent infection. On the fourth day post-surgery, symptoms of CDI emerged. Stool cultures confirmed the presence of C. difficile DNA. Treatment involved a combination of vancomycin and linezolid, resulting in the patient's successful recovery upon discharge. However, the patient failed to adhere to the prescribed medication after discharge and was discovered deceased during a follow-up two months later. CONCLUSION CDI is the leading cause of nosocomial post-operative care, with limited clinical cases and poor patient prognosis, and comprehensive clinical treatment guidelines are still lacking. This infection can be triggered by a variety of factors, including intestinal hypoxia, inappropriate antibiotic use, and bile acid circulation disorders. In patients with chronic bowel disease and related etiologies, prompt preoperative attention to possible CDI and preoperative bowel preparation is critical. Adequate and prolonged medication should be maintained in the treatment of CDI to prevent recurrence of the disease.
Collapse
Affiliation(s)
- Feng Ke
- Department of General Surgery, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun 130000, Jilin Province, China
| | - Zhen-Hua Dong
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Fan Bu
- Department of Plastic and Aesthetic Surgery, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Cheng-Nan Li
- Department of Encephalopathy Rehabilitation, Chaoyi Hospital, Yanbian Korean Autonomous Prefecture, Yanji 133000, Jilin Province, China
| | - Qi-Tong He
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Zhi-Cheng Liu
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Ji Lu
- Department of Urology, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Kai Yu
- Department of Urology, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Da-Guang Wang
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - He-Nan Xu
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Chang-Tao Ye
- Department of Urology, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| |
Collapse
|
25
|
Boyce JM. Hand and environmental hygiene: respective roles for MRSA, multi-resistant gram negatives, Clostridioides difficile, and Candida spp. Antimicrob Resist Infect Control 2024; 13:110. [PMID: 39334403 PMCID: PMC11437781 DOI: 10.1186/s13756-024-01461-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
Healthcare-associated infections (HAIs) caused by multidrug-resistant organisms (MDROs) represent a global threat to human health and well-being. Because transmission of MDROs to patients often occurs via transiently contaminated hands of healthcare personnel (HCP), hand hygiene is considered the most important measure for preventing HAIs. Environmental surfaces contaminated with MDROs from colonized or infected patients represent an important source of HCP hand contamination and contribute to transmission of pathogens. Accordingly, facilities are encouraged to adopt and implement recommendations included in the World Health Organization hand hygiene guidelines and those from the Society for Healthcare Epidemiology of America/Infectious Diseases Society of America/Association for Professionals in Infection Control and Epidemiology. Alcohol-based hand rubs are efficacious against MDROs with the exception of Clostridiodes difficile, for which soap and water handwashing is indicated. Monitoring hand hygiene adherence and providing HCP with feedback are of paramount importance. Environmental hygiene measures to curtail MDROs include disinfecting high-touch surfaces in rooms of patients with C. difficile infection daily with a sporicidal agent such as sodium hypochlorite. Some experts recommend also using a sporicidal agent in rooms of patients colonized with C. difficile, and for patients with multidrug-resistant Gram-negative bacteria. Sodium hypochlorite, hydrogen peroxide, or peracetic acid solutions are often used for daily and/or terminal disinfection of rooms housing patients with Candida auris or other MDROs. Products containing only a quaternary ammonium agent are not as effective as other agents against C. auris. Portable medical equipment should be cleaned and disinfected between use on different patients. Detergents are not recommended for cleaning high-touch surfaces in MDRO patient rooms, unless their use is followed by using a disinfectant. Facilities should consider using a disinfectant instead of detergents for terminal cleaning of floors in MDRO patient rooms. Education and training of environmental services employees is essential in assuring effective disinfection practices. Monitoring disinfection practices and providing personnel with performance feedback using fluorescent markers, adenosine triphosphate assays, or less commonly cultures of surfaces, can help reduce MDRO transmission. No-touch disinfection methods such as electrostatic spraying, hydrogen peroxide vapor, or ultraviolet light devices should be considered for terminal disinfection of MDRO patient rooms. Bundles with additional measures are usually necessary to reduce MDRO transmission.
Collapse
Affiliation(s)
- John M Boyce
- J.M. Boyce Consulting, LLC, 214 Hudson View Terrace, Hyde Park, NY, USA.
| |
Collapse
|
26
|
Mizrahi A, Péan de Ponfilly G, Sapa D, Suau A, Mangin I, Baliarda A, Hoys S, Pilmis B, Lambert S, Brosse A, Le Monnier A. A Mouse Model of Mild Clostridioides difficile Infection for the Characterization of Natural Immune Responses. Microorganisms 2024; 12:1933. [PMID: 39458243 PMCID: PMC11509167 DOI: 10.3390/microorganisms12101933] [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: 08/02/2024] [Revised: 09/13/2024] [Accepted: 09/20/2024] [Indexed: 10/28/2024] Open
Abstract
(1) Background: We describe a model of primary mild-Clostridioides difficile infection (CDI) in a naïve host, including gut microbiota analysis, weight loss, mortality, length of colonization. This model was used in order to describe the kinetics of humoral (IgG, IgM) and mucosal (IgA) immune responses against toxins (TcdA/TcdB) and surface proteins (SlpA/FliC). (2) Methods: A total of 105 CFU vegetative forms of C. difficile 630Δerm were used for challenge by oral administration after dysbiosis, induced by a cocktail of antibiotics. Gut microbiota dysbiosis was confirmed and described by 16S rDNA sequencing. We sacrificed C57Bl/6 mice after different stages of infection (day 6, 2, 7, 14, 21, 28, and 56) to evaluate IgM, IgG against TcdA, TcdB, SlpA, FliC in blood samples, and IgA in the cecal contents collected. (3) Results: In our model, we observed a reproducible gut microbiota dysbiosis, allowing for C. difficile digestive colonization. CDI was objectivized by a mean weight loss of 13.1% and associated with a low mortality rate of 15.7% of mice. We observed an increase in IgM anti-toxins as early as D7 after challenge. IgG increased since D21, and IgA anti-toxins were secreted in cecal contents. Unexpectedly, neither anti-SlpA nor anti-FliC IgG or IgA were observed in our model. (4) Conclusions: In our model, we induced a gut microbiota dysbiosis, allowing a mild CDI to spontaneously resolve, with a digestive clearance observed since D14. After this primary CDI, we can study the development of specific immune responses in blood and cecal contents.
Collapse
Affiliation(s)
- Assaf Mizrahi
- Service de Microbiologie Clinique, Hôpitaux Saint-Joseph & Marie-Lannelongue, 75014 Paris, France; (G.P.d.P.); (B.P.); (A.L.M.)
- Institut Micalis UMR 1319, Université Paris-Saclay, INRAe, AgroParisTech, 91400 Orsay, France; (D.S.); (A.B.); (S.H.); (S.L.); (A.B.)
| | - Gauthier Péan de Ponfilly
- Service de Microbiologie Clinique, Hôpitaux Saint-Joseph & Marie-Lannelongue, 75014 Paris, France; (G.P.d.P.); (B.P.); (A.L.M.)
- Institut Micalis UMR 1319, Université Paris-Saclay, INRAe, AgroParisTech, 91400 Orsay, France; (D.S.); (A.B.); (S.H.); (S.L.); (A.B.)
| | - Diane Sapa
- Institut Micalis UMR 1319, Université Paris-Saclay, INRAe, AgroParisTech, 91400 Orsay, France; (D.S.); (A.B.); (S.H.); (S.L.); (A.B.)
| | - Antonia Suau
- USC ANSES-Cnam Metabiot, Conservatoire National des Arts et Métiers, 75003 Paris, France; (A.S.); (I.M.)
| | - Irène Mangin
- USC ANSES-Cnam Metabiot, Conservatoire National des Arts et Métiers, 75003 Paris, France; (A.S.); (I.M.)
| | - Aurélie Baliarda
- Institut Micalis UMR 1319, Université Paris-Saclay, INRAe, AgroParisTech, 91400 Orsay, France; (D.S.); (A.B.); (S.H.); (S.L.); (A.B.)
| | - Sandra Hoys
- Institut Micalis UMR 1319, Université Paris-Saclay, INRAe, AgroParisTech, 91400 Orsay, France; (D.S.); (A.B.); (S.H.); (S.L.); (A.B.)
| | - Benoît Pilmis
- Service de Microbiologie Clinique, Hôpitaux Saint-Joseph & Marie-Lannelongue, 75014 Paris, France; (G.P.d.P.); (B.P.); (A.L.M.)
- Institut Micalis UMR 1319, Université Paris-Saclay, INRAe, AgroParisTech, 91400 Orsay, France; (D.S.); (A.B.); (S.H.); (S.L.); (A.B.)
| | - Sylvie Lambert
- Institut Micalis UMR 1319, Université Paris-Saclay, INRAe, AgroParisTech, 91400 Orsay, France; (D.S.); (A.B.); (S.H.); (S.L.); (A.B.)
| | - Anaïs Brosse
- Institut Micalis UMR 1319, Université Paris-Saclay, INRAe, AgroParisTech, 91400 Orsay, France; (D.S.); (A.B.); (S.H.); (S.L.); (A.B.)
| | - Alban Le Monnier
- Service de Microbiologie Clinique, Hôpitaux Saint-Joseph & Marie-Lannelongue, 75014 Paris, France; (G.P.d.P.); (B.P.); (A.L.M.)
- Institut Micalis UMR 1319, Université Paris-Saclay, INRAe, AgroParisTech, 91400 Orsay, France; (D.S.); (A.B.); (S.H.); (S.L.); (A.B.)
| |
Collapse
|
27
|
Tao Y, Luo CJ, Zhang BH, Shen XY, Zhao RK, Ma BY, Shen N, Luo CY, Wang JM, Xia YJ, Xie L, Chen J, Mo X. Diagnostic performance of a multiplexed gastrointestinal PCR panel for identifying diarrheal pathogens in children undergoing hematopoietic stem cell transplant. World J Pediatr 2024; 20:966-975. [PMID: 38367140 PMCID: PMC11422252 DOI: 10.1007/s12519-023-00776-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 10/24/2023] [Indexed: 02/19/2024]
Abstract
BACKGROUND Diarrhea is a common complication of hematopoietic stem cell transplantation (HSCT) and is associated with substantial morbidity, but its etiology is often unknown. Etiologies of diarrhea in this population include infectious causes, chemotherapy- or medication-induced mucosal injury and graft-versus-host disease (GVHD). Distinguishing these potential causes of diarrhea is challenging since diarrheal symptoms are often multifactorial, and the etiologies often overlap in transplant patients. The objectives of this study were to evaluate whether the FilmArray gastrointestinal (GI) panel would increase diagnostic yield and the degree to which pre-transplantation colonization predicts post-transplantation infection. METHODS From November 2019 to February 2021, a total of 158 patients undergoing HSCT were prospectively included in the study. Stool specimens were obtained from all HSCT recipients prior to conditioning therapy, 28 ± 7 days after transplantation and at any new episode of diarrhea. All stool samples were tested by the FilmArray GI panel and other clinical microbiological assays. RESULTS The primary cause of post-transplantation diarrhea was infection (57/84, 67.86%), followed by medication (38/84, 45.24%) and GVHD (21/84, 25.00%). Ninety-five of 158 patients were colonized with at least one gastrointestinal pathogen before conditioning therapy, and the incidence of infectious diarrhea was significantly higher in colonized patients (47/95, 49.47%) than in non-colonized patients (10/63, 15.87%) (P < 0.001). Fourteen of 19 (73.68%) patients who were initially colonized with norovirus pre-transplantation developed a post-transplantation norovirus infection. Twenty-four of 62 (38.71%) patients colonized with Clostridium difficile developed a diarrheal infection. In addition, FilmArray GI panel testing improved the diagnostic yield by almost twofold in our study (55/92, 59.78% vs. 30/92, 32.61%). CONCLUSIONS Our data show that more than half of pediatric patients who were admitted for HSCT were colonized with various gastrointestinal pathogens, and more than one-third of these pathogens were associated with post-transplantation diarrhea. In addition, the FilmArray GI panel can increase the detection rate of diarrheal pathogens in pediatric HSCT patients, but the panel needs to be optimized for pathogen species, and further studies assessing its clinical impact and cost-effectiveness in this specific patient population are also needed.
Collapse
Affiliation(s)
- Yue Tao
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dongfang Rd., Shanghai, 200127, China.
| | - Cheng-Juan Luo
- Department of Hematology and Oncology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dongfang Rd., Shanghai, 200127, China
| | - Bing-Hua Zhang
- Department of Hematology and Oncology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dongfang Rd., Shanghai, 200127, China
| | - Xin-Yan Shen
- Department of Hematology and Oncology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dongfang Rd., Shanghai, 200127, China
| | - Rui-Ke Zhao
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dongfang Rd., Shanghai, 200127, China
| | - Bei-Ying Ma
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dongfang Rd., Shanghai, 200127, China
| | - Nan Shen
- Department of Infectious Diseases, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chang-Ying Luo
- Department of Hematology and Oncology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dongfang Rd., Shanghai, 200127, China
| | - Jian-Min Wang
- Department of Hematology and Oncology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dongfang Rd., Shanghai, 200127, China
| | - Yi-Jun Xia
- Medical Affairs, BioMérieux (Shanghai) Company, Limited, Shanghai, China
| | - Li Xie
- Clinical Research Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Chen
- Department of Hematology and Oncology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dongfang Rd., Shanghai, 200127, China.
| | - Xi Mo
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dongfang Rd., Shanghai, 200127, China.
| |
Collapse
|
28
|
Lam JC, Bourassa-Blanchette S. Ten Clinical Pearls in Microbiology: How Effective Collaboration Optimizes Patient Care. Am J Med 2024; 137:818-824. [PMID: 38782247 DOI: 10.1016/j.amjmed.2024.05.013] [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: 05/03/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/25/2024]
Abstract
Medical microbiology laboratories play an essential role in patient care-appertaining to infectious diseases diagnostics and treatment, infection prevention, and antimicrobial stewardship. Collaboration between clinicians and the microbiology laboratory can promote and enhance the safety, quality, and efficiency of patient care. We review practical, evidence-informed core concepts to explicate how effective partnership between clinicians and the microbiology laboratory improves patient outcomes.
Collapse
Affiliation(s)
- John C Lam
- Division of Infectious Diseases, Department of Medicine, University of California Los Angeles, Los Angeles, CA.
| | - Samuel Bourassa-Blanchette
- Division of Infectious Diseases, Department of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland, Canada; Division of Microbiology, Department of Pathology and Laboratory Medicine, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| |
Collapse
|
29
|
Dang Z, Yang B, Xia P, Huang J, Liao J, Li Y, Tang S, Han Q, Luo S, Xia Y. Antimicrobial susceptibilities, resistance mechanisms and molecular characteristics of toxigenic Clostridioides difficile isolates in a large teaching hospital in Chongqing, China. J Glob Antimicrob Resist 2024; 38:198-204. [PMID: 39048055 DOI: 10.1016/j.jgar.2024.07.006] [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: 08/21/2023] [Revised: 07/05/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024] Open
Abstract
OBJECTIVES Clostridioides difficile ranks among the primary sources of healthcare-related infections and diarrhoea in numerous nations. We evaluated the drug susceptibility and resistance mechanisms of C. difficile isolates from a hospital in Chongqing, China, and identified resistance rates and resistance mechanisms that differed from previous findings. METHODS The toxin genes and drug resistance genes of clinical strains were detected using Polymerase Chain Reaction (PCR), and these strains were subjected to Multilocus Sequence Typing (MLST). The agar dilution technique was employed for assessing susceptibility of antibiotics. Clinical data collection was completed through a review of electronic medical records. RESULTS A total of 67 strains of toxin-producing C. difficile were detected. All C. difficile isolates demonstrated susceptibility to both metronidazole and vancomycin. However, resistance was observed in 8.95%, 16.42%, 56.72%, 56.72%, 31.34% and 5.97% of the isolates for tigecycline, tetracycline, clindamycin, erythromycin, moxifloxacin and rifampin, respectively. Among the strains with toxin genotypes A + B + CDT - and belonging to the ST3, six strains exhibited reduced susceptibility to tigecycline (MIC=0.5mg/L) and tetracycline (MIC=8mg/L). The tetA(P) and tetB(P) genes were present in these six strains, but were absent in tetracycline-resistant strains. Resistance genes (ermB, tetM, tetA(P) and tetB(P)) and mutations (in gyrA, gyrB, and rpoB) were identified in resistant strains. CONCLUSIONS In contrast to prior studies, we found higher proportions of ST3 isolates with decreased tigecycline sensitivity, sharing similar resistance patterns and resistance genes. In the resistance process of tigecycline and tetracycline, the tetA(P) and tetB(P) genes may play a weak role.
Collapse
Affiliation(s)
- Zijun Dang
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bingxue Yang
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Peiwen Xia
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jinzhu Huang
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiajia Liao
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yuqiong Li
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shiyu Tang
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qi Han
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shengli Luo
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yun Xia
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
30
|
Kunishima H, Ichiki K, Ohge H, Sakamoto F, Sato Y, Suzuki H, Nakamura A, Fujimura S, Matsumoto K, Mikamo H, Mizutani T, Morinaga Y, Mori M, Yamagishi Y, Yoshizawa S. Japanese Society for infection prevention and control guide to Clostridioides difficile infection prevention and control. J Infect Chemother 2024; 30:673-715. [PMID: 38714273 DOI: 10.1016/j.jiac.2024.03.018] [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: 03/04/2024] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 05/09/2024]
Affiliation(s)
- Hiroyuki Kunishima
- Department of Infectious Diseases. St. Marianna University School of Medicine, Japan.
| | - Kaoru Ichiki
- Department of Infection Control and Prevention, Hyogo Medical University Hospital, Japan
| | - Hiroki Ohge
- Department of Infectious Diseases, Hiroshima University Hospital, Japan
| | - Fumie Sakamoto
- Quality Improvement and Safety Center, Itabashi Chuo Medical Center, Japan
| | - Yuka Sato
- Department of Infection Control and Nursing, Graduate School of Nursing, Aichi Medical University, Japan
| | - Hiromichi Suzuki
- Department of Infectious Diseases, University of Tsukuba School of Medicine and Health Sciences, Japan
| | - Atsushi Nakamura
- Department of Infection Prevention and Control, Graduate School of Medical Sciences, Nagoya City University, Japan
| | - Shigeru Fujimura
- Division of Clinical Infectious Diseases and Chemotherapy, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Japan
| | - Kazuaki Matsumoto
- Division of Pharmacodynamics, Faculty of Pharmacy, Keio University, Japan
| | - Hiroshige Mikamo
- Department of Clinical Infectious Diseases, Aichi Medical University, Japan
| | | | - Yoshitomo Morinaga
- Department of Microbiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Japan
| | - Minako Mori
- Department of Infection Control, Hiroshima University Hospital, Japan
| | - Yuka Yamagishi
- Department of Clinical Infectious Diseases, Kochi Medical School, Kochi University, Japan
| | - Sadako Yoshizawa
- Department of Laboratory Medicine/Department of Microbiology and Infectious Diseases, Faculty of Medicine, Toho University, Japan
| |
Collapse
|
31
|
Spigaglia P. Clostridioides difficile and Gut Microbiota: From Colonization to Infection and Treatment. Pathogens 2024; 13:646. [PMID: 39204246 PMCID: PMC11357127 DOI: 10.3390/pathogens13080646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/23/2024] [Accepted: 07/29/2024] [Indexed: 09/03/2024] Open
Abstract
Clostridioides difficile is the main causative agent of antibiotic-associated diarrhea (AAD) in hospitals in the developed world. Both infected patients and asymptomatic colonized individuals represent important transmission sources of C. difficile. C. difficile infection (CDI) shows a large range of symptoms, from mild diarrhea to severe manifestations such as pseudomembranous colitis. Epidemiological changes in CDIs have been observed in the last two decades, with the emergence of highly virulent types and more numerous and severe CDI cases in the community. C. difficile interacts with the gut microbiota throughout its entire life cycle, and the C. difficile's role as colonizer or invader largely depends on alterations in the gut microbiota, which C. difficile itself can promote and maintain. The restoration of the gut microbiota to a healthy state is considered potentially effective for the prevention and treatment of CDI. Besides a fecal microbiota transplantation (FMT), many other approaches to re-establishing intestinal eubiosis are currently under investigation. This review aims to explore current data on C. difficile and gut microbiota changes in colonized individuals and infected patients with a consideration of the recent emergence of highly virulent C. difficile types, with an overview of the microbial interventions used to restore the human gut microbiota.
Collapse
Affiliation(s)
- Patrizia Spigaglia
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Roma, Italy
| |
Collapse
|
32
|
Di Bella S, Sanson G, Monticelli J, Zerbato V, Principe L, Giuffrè M, Pipitone G, Luzzati R. Clostridioides difficile infection: history, epidemiology, risk factors, prevention, clinical manifestations, treatment, and future options. Clin Microbiol Rev 2024; 37:e0013523. [PMID: 38421181 PMCID: PMC11324037 DOI: 10.1128/cmr.00135-23] [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: 03/02/2024] Open
Abstract
SUMMARYClostridioides difficile infection (CDI) is one of the major issues in nosocomial infections. This bacterium is constantly evolving and poses complex challenges for clinicians, often encountered in real-life scenarios. In the face of CDI, we are increasingly equipped with new therapeutic strategies, such as monoclonal antibodies and live biotherapeutic products, which need to be thoroughly understood to fully harness their benefits. Moreover, interesting options are currently under study for the future, including bacteriophages, vaccines, and antibiotic inhibitors. Surveillance and prevention strategies continue to play a pivotal role in limiting the spread of the infection. In this review, we aim to provide the reader with a comprehensive overview of epidemiological aspects, predisposing factors, clinical manifestations, diagnostic tools, and current and future prophylactic and therapeutic options for C. difficile infection.
Collapse
Affiliation(s)
- Stefano Di Bella
- Clinical Department of
Medical, Surgical and Health Sciences, Trieste
University, Trieste,
Italy
| | - Gianfranco Sanson
- Clinical Department of
Medical, Surgical and Health Sciences, Trieste
University, Trieste,
Italy
| | - Jacopo Monticelli
- Infectious Diseases
Unit, Trieste University Hospital
(ASUGI), Trieste,
Italy
| | - Verena Zerbato
- Infectious Diseases
Unit, Trieste University Hospital
(ASUGI), Trieste,
Italy
| | - Luigi Principe
- Microbiology and
Virology Unit, Great Metropolitan Hospital
“Bianchi-Melacrino-Morelli”,
Reggio Calabria, Italy
| | - Mauro Giuffrè
- Clinical Department of
Medical, Surgical and Health Sciences, Trieste
University, Trieste,
Italy
- Department of Internal
Medicine (Digestive Diseases), Yale School of Medicine, Yale
University, New Haven,
Connecticut, USA
| | - Giuseppe Pipitone
- Infectious Diseases
Unit, ARNAS Civico-Di Cristina
Hospital, Palermo,
Italy
| | - Roberto Luzzati
- Clinical Department of
Medical, Surgical and Health Sciences, Trieste
University, Trieste,
Italy
| |
Collapse
|
33
|
Huletsky A, Loo VG, Longtin Y, Longtin J, Trottier S, Tremblay CL, Gilca R, Lavallée C, Brochu É, Bérubé È, Bastien M, Bernier M, Gagnon M, Frenette J, Bestman-Smith J, Deschênes L, Bergeron MG. Comparison of rectal swabs and fecal samples for the detection of Clostridioides difficile infections with a new in-house PCR assay. Microbiol Spectr 2024; 12:e0022524. [PMID: 38687067 PMCID: PMC11237655 DOI: 10.1128/spectrum.00225-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/29/2024] [Indexed: 05/02/2024] Open
Abstract
The detection of Clostridioides difficile infections (CDI) relies on testing the stool of patients by toxin antigen detection or PCR methods. Although PCR and antigenic methods have significantly reduced the time to results, delays in stool collection can significantly add to the turnaround time. The use of rectal swabs to detect C. difficile could considerably reduce the time to diagnosis of CDI. We developed a new rapid PCR assay for the detection of C. difficile and evaluated this PCR assay on both stool and rectal swab specimens. We recruited a total of 623 patients suspected of C. difficile infection. Stool samples and rectal swabs were collected from each patient and tested by our PCR assay. Stool samples were also tested by the cell cytotoxicity neutralization assay (CCNA) as a reference. The PCR assay detected C. difficile in 60 stool specimens and 61 rectal swabs for the 64 patients whose stool samples were positive for C. difficile by CCNA. The PCR assay detected an additional 35 and 36 stool and rectal swab specimens positive for C. difficile, respectively, for sensitivity with stools and rectal swabs of 93.8% and 95.3%, specificity of 93.7% and 93.6%, positive predictive values of 63.2% and 62.9%, and negative predictive values of 99.2% and 99.4%. Detection of C. difficile using PCR on stools or rectal swabs yielded reliable and similar results. The use of PCR tests on rectal swabs could reduce turnaround time for CDI detection, thus improving CDI management and control of C. difficile transmission. IMPORTANCE Clostridioides difficile infection (CDI) is the leading cause of healthcare-associated diarrhea, resulting in high morbidity, mortality, and economic burden. In clinical laboratories, CDI testing is currently performed on stool samples collected from patients with diarrhea. However, the diagnosis of CDI can be delayed by the time required to collect stool samples. Barriers to sample collection could be overcome by using a rectal swab instead of a stool sample. Our study showed that CDI can be identified rapidly and reliably by a new PCR assay developed in our laboratory on both stool and rectal swab specimens. The use of PCR tests on rectal swabs could reduce the time for the detection of CDI and improve the management of this infection. It should also provide a useful alternative for infection-control practitioners to better control the spread of C. difficile.
Collapse
Affiliation(s)
- Ann Huletsky
- Centre de recherche en infectiologie de l’Université Laval, Québec City, Canada
- Centre de recherche du Centre hospitalier universitaire de Québec-Université Laval, Québec City, Canada
- Axe maladies infectieuses et immunitaires, Centre de recherche du Centre hospitalier universitaire de Québec-Université Laval, Québec City, Canada
| | - Vivian G. Loo
- Division of Infectious Diseases, Department of Medical Microbiology, McGill University Health Centre, Montréal, Canada
- Faculty of Medicine, McGill University, Montréal, Canada
| | - Yves Longtin
- Faculty of Medicine, McGill University, Montréal, Canada
- Sir Mortimer B. Davis Jewish General Hospital, Montréal, Canada
| | - Jean Longtin
- Centre hospitalier universitaire de Québec-Université Laval, Québec City, Canada
| | - Sylvie Trottier
- Centre de recherche en infectiologie de l’Université Laval, Québec City, Canada
- Centre de recherche du Centre hospitalier universitaire de Québec-Université Laval, Québec City, Canada
- Axe maladies infectieuses et immunitaires, Centre de recherche du Centre hospitalier universitaire de Québec-Université Laval, Québec City, Canada
| | - Cécile L. Tremblay
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Montréal, Canada
- Département de microbiologie, infectiologie et immunologie, Université de Montréal, Montréal, Canada
| | - Rodica Gilca
- Axe maladies infectieuses et immunitaires, Centre de recherche du Centre hospitalier universitaire de Québec-Université Laval, Québec City, Canada
- Département de médecine sociale et préventive, Faculté de médecine, Université Laval, Québec City, Canada
- Département de risque biologique et de la santé au travail, Institut national de santé publique du Québec, Québec City, Canada
| | - Christian Lavallée
- Département de microbiologie, infectiologie et immunologie, Université de Montréal, Montréal, Canada
- Service de maladies infectieuses et de microbiologie, Département de médecine spécialisée, Hôpital Maisonneuve-Rosemont - CIUSSS de l'Est-de-l'Ile-de-Montréal, Montréal, Canada
- Département clinique de médecine de laboratoire, Centre hospitalier de l'Université de Montréal, Montréal, Canada
| | - Éliel Brochu
- Centre de recherche en infectiologie de l’Université Laval, Québec City, Canada
- Centre de recherche du Centre hospitalier universitaire de Québec-Université Laval, Québec City, Canada
- Axe maladies infectieuses et immunitaires, Centre de recherche du Centre hospitalier universitaire de Québec-Université Laval, Québec City, Canada
| | - Ève Bérubé
- Centre de recherche en infectiologie de l’Université Laval, Québec City, Canada
- Centre de recherche du Centre hospitalier universitaire de Québec-Université Laval, Québec City, Canada
- Axe maladies infectieuses et immunitaires, Centre de recherche du Centre hospitalier universitaire de Québec-Université Laval, Québec City, Canada
| | - Martine Bastien
- Centre de recherche en infectiologie de l’Université Laval, Québec City, Canada
- Centre de recherche du Centre hospitalier universitaire de Québec-Université Laval, Québec City, Canada
- Axe maladies infectieuses et immunitaires, Centre de recherche du Centre hospitalier universitaire de Québec-Université Laval, Québec City, Canada
| | - Marthe Bernier
- Centre de recherche en infectiologie de l’Université Laval, Québec City, Canada
- Centre de recherche du Centre hospitalier universitaire de Québec-Université Laval, Québec City, Canada
- Axe maladies infectieuses et immunitaires, Centre de recherche du Centre hospitalier universitaire de Québec-Université Laval, Québec City, Canada
| | - Martin Gagnon
- Centre de recherche en infectiologie de l’Université Laval, Québec City, Canada
- Centre de recherche du Centre hospitalier universitaire de Québec-Université Laval, Québec City, Canada
- Axe maladies infectieuses et immunitaires, Centre de recherche du Centre hospitalier universitaire de Québec-Université Laval, Québec City, Canada
| | - Johanne Frenette
- Centre de recherche en infectiologie de l’Université Laval, Québec City, Canada
- Centre de recherche du Centre hospitalier universitaire de Québec-Université Laval, Québec City, Canada
- Axe maladies infectieuses et immunitaires, Centre de recherche du Centre hospitalier universitaire de Québec-Université Laval, Québec City, Canada
| | - Julie Bestman-Smith
- Axe maladies infectieuses et immunitaires, Centre de recherche du Centre hospitalier universitaire de Québec-Université Laval, Québec City, Canada
- Service de microbiologie-infectiologie, Centre hospitalier universitaire de Québec-Université Laval, Québec City, Canada
| | - Louise Deschênes
- Axe maladies infectieuses et immunitaires, Centre de recherche du Centre hospitalier universitaire de Québec-Université Laval, Québec City, Canada
- Service de microbiologie-infectiologie, Centre hospitalier universitaire de Québec-Université Laval, Québec City, Canada
| | - Michel G. Bergeron
- Centre de recherche en infectiologie de l’Université Laval, Québec City, Canada
- Centre de recherche du Centre hospitalier universitaire de Québec-Université Laval, Québec City, Canada
- Axe maladies infectieuses et immunitaires, Centre de recherche du Centre hospitalier universitaire de Québec-Université Laval, Québec City, Canada
| |
Collapse
|
34
|
Wang L, Villafuerte Gálvez JA, Lee C, Wu S, Kelly CP, Chen X, Cao Y. Understanding host immune responses in Clostridioides difficile infection: Implications for pathogenesis and immunotherapy. IMETA 2024; 3:e200. [PMID: 38898983 PMCID: PMC11183162 DOI: 10.1002/imt2.200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 06/21/2024]
Abstract
Clostridioides difficile (C. difficile) is the predominant causative agent of nosocomial diarrhea worldwide. Infection with C. difficile occurs due to the secretion of large glycosylating toxin proteins, which can lead to toxic megacolon or mortality in susceptible hosts. A critical aspect of C. difficile's biology is its ability to persist asymptomatically within the human host. Individuals harboring asymptomatic colonization or experiencing a single episode of C. difficile infection (CDI) without recurrence exhibit heightened immune responses compared to symptomatic counterparts. The significance of these immune responses cannot be overstated, as they play critical roles in the development, progression, prognosis, and outcomes of CDI. Nonetheless, our current comprehension of the immune responses implicated in CDI remains limited. Therefore, further investigation is imperative to elucidate their underlying mechanisms. This review explores recent advancements in comprehending CDI pathogenesis and how the host immune system response influences disease progression and severity, aiming to enhance our capacity to develop immunotherapy-based treatments for CDI.
Collapse
Affiliation(s)
- Lamei Wang
- College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Javier A. Villafuerte Gálvez
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Christina Lee
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Shengru Wu
- College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
| | - Ciaran P. Kelly
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Xinhua Chen
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Yangchun Cao
- College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
35
|
Cersosimo LM, Worley JN, Bry L. Approaching toxigenic Clostridia from a One Health perspective. Anaerobe 2024; 87:102839. [PMID: 38552896 PMCID: PMC11180571 DOI: 10.1016/j.anaerobe.2024.102839] [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: 12/31/2023] [Revised: 02/29/2024] [Accepted: 03/17/2024] [Indexed: 04/08/2024]
Abstract
Spore-forming pathogens have a unique capacity to thrive in diverse environments, and with temporal persistence afforded through their ability to sporulate. Their prevalence in diverse ecosystems requires a One Health approach to identify critical reservoirs and outbreak-associated transmission chains, given their capacity to freely move across soils, waterways, foodstuffs and as commensals or infecting pathogens in human and animal populations. Among anaerobic spore-formers, genomic resources for pathogens including C. botulinum, C. difficile, and C. perfringens enable our capacity to identify common and unique factors that support their persistence in diverse reservoirs and capacity to cause disease. Publicly available genomic resources for spore-forming pathogens at NCBI's Pathogen Detection program aid outbreak investigations and longitudinal monitoring in national and international programs in public health and food safety, as well as for local healthcare systems. These tools also enable research to derive new knowledge regarding disease pathogenesis, and to inform strategies in disease prevention and treatment. As global community resources, the continued sharing of strain genomic data and phenotypes further enhances international resources and means to develop impactful applications. We present examples showing use of these resources in surveillance, including capacity to assess linkages among clinical, environmental, and foodborne reservoirs and to further research investigations into factors promoting their persistence and virulence in different settings.
Collapse
Affiliation(s)
- Laura M Cersosimo
- Massachusetts Host-Microbiome Center, Dept. Pathology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jay N Worley
- Massachusetts Host-Microbiome Center, Dept. Pathology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA; National Center for Biotechnology Information, NIH, Bethesda, MD, USA
| | - Lynn Bry
- Massachusetts Host-Microbiome Center, Dept. Pathology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
36
|
Rigo I, Young MK, Abhyankar MM, Xu F, Ramakrishnan G, Naz F, Madden GR, Petri WA. The impact of existing total anti-toxin B IgG immunity in outcomes of recurrent Clostridioides difficile infection. Anaerobe 2024; 87:102842. [PMID: 38552897 PMCID: PMC11180572 DOI: 10.1016/j.anaerobe.2024.102842] [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: 12/30/2023] [Revised: 02/22/2024] [Accepted: 03/19/2024] [Indexed: 04/15/2024]
Abstract
Late anti-toxin-B humoral immunity acquired after treatment is important for preventing recurrent Clostridioides difficile infection. We prospectively-measured anti-toxin-B IgG and neutralization titers at diagnosis as potential early predictors of recurrence. High anti-toxin-B-IgG/neutralizing antibodies were associated with short-lasting protection within 6-weeks, however, no difference in recurrence risk was observed by 90-days post-infection.
Collapse
Affiliation(s)
- Isaura Rigo
- University of Virginia, Department of Medicine/ Infectious Disease and International Health, USA
| | - Mary K Young
- University of Virginia, Department of Medicine/ Infectious Disease and International Health, USA
| | - Mayuresh M Abhyankar
- University of Virginia, Department of Medicine/ Infectious Disease and International Health, USA
| | - Feifan Xu
- University of Virginia, Department of Medicine/ Infectious Disease and International Health, USA
| | - Girija Ramakrishnan
- University of Virginia, Department of Medicine/ Infectious Disease and International Health, USA
| | - Farha Naz
- University of Virginia, Department of Medicine/ Infectious Disease and International Health, USA
| | - Gregory R Madden
- University of Virginia, Department of Medicine/ Infectious Disease and International Health, USA
| | - William A Petri
- University of Virginia, Department of Medicine/ Infectious Disease and International Health, USA.
| |
Collapse
|
37
|
Zhang S, Ma C, Zhang H, Zhao C, Guo R, Liu J, Wang J, Yuan J, Jia K, Wu A, Chen Y, Lei J. Toxin genotypes, antibiotic resistance and their correlations in Clostridioides difficile isolated from hospitals in Xi'an, China. BMC Microbiol 2024; 24:177. [PMID: 38783194 PMCID: PMC11112860 DOI: 10.1186/s12866-024-03327-z] [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: 04/13/2023] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Clostridioides difficile is the main pathogen of antimicrobial-associated diarrhoea and health care facility-associated infectious diarrhoea. This study aimed to investigate the prevalence, toxin genotypes, and antibiotic resistance of C. difficile among hospitalized patients in Xi'an, China. RESULTS We isolated and cultured 156 strains of C. difficile, representing 12.67% of the 1231 inpatient stool samples collected. Among the isolates, tcdA + B + strains were predominant, accounting for 78.2% (122/156), followed by 27 tcdA-B + strains (27/156, 17.3%) and 6 binary toxin gene-positive strains. The positive rates of three regulatory genes, tcdC, tcdR, and tcdE, were 89.1% (139/156), 96.8% (151/156), and 100%, respectively. All isolates were sensitive to metronidazole, and the resistance rates to clindamycin and cephalosporins were also high. Six strains were found to be resistant to vancomycin. CONCLUSION Currently, the prevalence rate of C. difficile infection (CDI) in Xi'an is 12.67% (156/1231), with the major toxin genotype of the isolates being tcdA + tcdB + cdtA-/B-. Metronidazole and vancomycin were still effective drugs for the treatment of CDI, but we should pay attention to antibiotic management and epidemiological surveillance of CDI.
Collapse
Affiliation(s)
- Sukai Zhang
- Clinical Medicine Class of 2019, Xi'an Jiaotong University, Xi'an, China
| | - Chen Ma
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Haiyue Zhang
- Clinical Medicine Class of 2019, Xi'an Jiaotong University, Xi'an, China
| | - Congcong Zhao
- Clinical Medicine Class of 2019, Xi'an Jiaotong University, Xi'an, China
| | - Ruibing Guo
- Clinical Medicine Class of 2019, Xi'an Jiaotong University, Xi'an, China
| | - Jiahao Liu
- Clinical Medicine Class of 2019, Xi'an Jiaotong University, Xi'an, China
| | - Jing Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jing Yuan
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Kai Jia
- Xi'an Children's Hospital, Xi'an, China
| | | | - Yanjiong Chen
- Department of Immunology and Pathogenic Biology, College of Basic Medicine, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Jin'e Lei
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
38
|
Abstract
Colorectal cancer (CRC) is a substantial source of global morbidity and mortality in dire need of improved prevention and treatment strategies. As our understanding of CRC grows, it is becoming increasingly evident that the gut microbiota, consisting of trillions of microorganisms in direct interface with the colon, plays a substantial role in CRC development and progression. Understanding the roles that individual microorganisms and complex microbial communities play in CRC pathogenesis, along with their attendant mechanisms, will help yield novel preventive and therapeutic interventions for CRC. In this Review, we discuss recent evidence concerning global perturbations of the gut microbiota in CRC, associations of specific microorganisms with CRC, the underlying mechanisms by which microorganisms potentially drive CRC development and the roles of complex microbial communities in CRC pathogenesis. While our understanding of the relationship between the microbiota and CRC has improved in recent years, our findings highlight substantial gaps in current research that need to be filled before this knowledge can be used to the benefit of patients.
Collapse
Affiliation(s)
- Maxwell T White
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cynthia L Sears
- Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
39
|
Leal J, Shen Y, Faris P, Dalton B, Sabuda D, Ocampo W, Bresee L, Chow B, Fletcher JR, Henderson E, Kaufman J, Kim J, Raman M, Kraft S, Lamont NC, Larios O, Missaghi B, Holroyd-Leduc J, Louie T, Conly J. Effectiveness of Bio-K+ for the prevention of Clostridioides difficile infection: Stepped-wedge cluster-randomized controlled trial. Infect Control Hosp Epidemiol 2024; 45:443-451. [PMID: 38073551 PMCID: PMC11007362 DOI: 10.1017/ice.2023.169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 05/20/2023] [Accepted: 07/08/2023] [Indexed: 04/10/2024]
Abstract
OBJECTIVE To evaluate the impact of administering probiotics to prevent Clostridioides difficile infection (CDI) among patients receiving therapeutic antibiotics. DESIGN Stepped-wedge cluster-randomized trial between September 1, 2016, and August 31, 2019. SETTING This study was conducted in 4 acute-care hospitals across an integrated health region. PATIENTS Hospitalized patients, aged ≥55 years. METHODS Patients were given 2 probiotic capsules daily (Bio-K+, Laval, Quebec, Canada), containing 50 billion colony-forming units of Lactobacillus acidophilus CL1285, L. casei LBC80R, and L. rhamnosus CLR2. We measured hospital-acquired CDI (HA-CDI) and the number of positive C. difficile tests per 10,000 patient days as well as adherence to administration of Bio-K+ within 48 and 72 hours of antibiotic administration. Mixed-effects generalized linear models, adjusted for influenza admissions and facility characteristics, were used to evaluate the impact of the intervention on outcomes. RESULTS Overall adherence of Bio-K+ administration ranged from 76.9% to 84.6% when stratified by facility and periods. Rates of adherence to administration within 48 and 72 hours of antibiotic treatment were 60.2% -71.4% and 66.7%-75.8%, respectively. In the adjusted analysis, there was no change in HA-CDI (incidence rate ratio [IRR], 0.92; 95% confidence interval [CI], 0.68-1.23) or C. difficile positivity rate (IRR, 1.05; 95% CI, 0.89-1.24). Discharged patients may not have received a complete course of Bio-K+. Our hospitals had a low baseline incidence of HA-CDI. Patients who did not receive Bio-K+ may have differential risks of acquiring CDI, introducing selection bias. CONCLUSIONS Hospitals considering probiotics as a primary prevention strategy should consider the baseline incidence of HA-CDI in their population and timing of probiotics relative to the start of antimicrobial administration.
Collapse
Affiliation(s)
- Jenine Leal
- Infection Prevention and Control, Alberta Health Services, Alberta, Canada
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- O’Brien Institute for Public Health, University of Calgary, Calgary, Alberta, Canada
| | - Ye Shen
- Infection Prevention and Control, Alberta Health Services, Alberta, Canada
| | - Peter Faris
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Analytics, Alberta Health Services, Alberta, Canada
| | - Bruce Dalton
- Pharmacy Services, Alberta Health Services, Calgary, Alberta, Canada
| | - Deana Sabuda
- Pharmacy Services, Alberta Health Services, Calgary, Alberta, Canada
| | - Wrechelle Ocampo
- O’Brien Institute for Public Health, University of Calgary, Calgary, Alberta, Canada
- W21 Research and Innovation Centre, University of Calgary and Alberta Health Services, Calgary, Alberta, Canada
| | - Lauren Bresee
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- O’Brien Institute for Public Health, University of Calgary, Calgary, Alberta, Canada
| | - Blanda Chow
- Infection Prevention and Control, Alberta Health Services, Alberta, Canada
| | - Jared R. Fletcher
- Department of Health and Physical Education, Mount Royal University, Calgary, Alberta, Canada
| | - Elizabeth Henderson
- Infection Prevention and Control, Alberta Health Services, Alberta, Canada
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- O’Brien Institute for Public Health, University of Calgary, Calgary, Alberta, Canada
| | - Jaime Kaufman
- W21 Research and Innovation Centre, University of Calgary and Alberta Health Services, Calgary, Alberta, Canada
| | - Joseph Kim
- Infection Prevention and Control, Alberta Health Services, Alberta, Canada
- Department of Medicine, Cumming School of Medicine University of Calgary, Calgary, Alberta, Canada
| | - Maitreyi Raman
- Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Scott Kraft
- W21 Research and Innovation Centre, University of Calgary and Alberta Health Services, Calgary, Alberta, Canada
| | - Nicole C. Lamont
- W21 Research and Innovation Centre, University of Calgary and Alberta Health Services, Calgary, Alberta, Canada
| | - Oscar Larios
- Infection Prevention and Control, Alberta Health Services, Alberta, Canada
- Department of Medicine, Cumming School of Medicine University of Calgary, Calgary, Alberta, Canada
- Department of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary and Alberta Health Services, Calgary, Alberta, Canada
| | - Bayan Missaghi
- Infection Prevention and Control, Alberta Health Services, Alberta, Canada
- Department of Medicine, Cumming School of Medicine University of Calgary, Calgary, Alberta, Canada
- Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | - Jayna Holroyd-Leduc
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- O’Brien Institute for Public Health, University of Calgary, Calgary, Alberta, Canada
- Department of Medicine, Cumming School of Medicine University of Calgary, Calgary, Alberta, Canada
| | - Thomas Louie
- Infection Prevention and Control, Alberta Health Services, Alberta, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Medicine, Cumming School of Medicine University of Calgary, Calgary, Alberta, Canada
| | - John Conly
- Infection Prevention and Control, Alberta Health Services, Alberta, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- O’Brien Institute for Public Health, University of Calgary, Calgary, Alberta, Canada
- Department of Medicine, Cumming School of Medicine University of Calgary, Calgary, Alberta, Canada
- Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
- Department of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary and Alberta Health Services, Calgary, Alberta, Canada
| |
Collapse
|
40
|
Maldonado-Barrueco A, Moreno-Ramos F, Díaz-Pollán B, Loeches-Yagüe B, Rico-Nieto A, García-Rodríguez J, Ruiz-Carrascoso G. Increase of healthcare-onset Clostridioides difficile infection in adult population since SARS-CoV-2 pandemic: A retrospective cohort study in a tertiary care hospital from 2019 to 2022. Anaerobe 2024; 86:102836. [PMID: 38428802 DOI: 10.1016/j.anaerobe.2024.102836] [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: 10/02/2023] [Revised: 02/22/2024] [Accepted: 02/23/2024] [Indexed: 03/03/2024]
Abstract
OBJECTIVES The aim was to assess the impact of the SARS-CoV-2 pandemic on the prevalence, relative incidence (RI), incidence density (ID), ratio of rate incidence (RRI), rate of incidence density (RID), and relative risks (RR) of healthcare-onset Clostridioides difficile infection (HO-CDI) as well as its correlation with the antibiotic consumption. METHODS Demographic and analytical data of adult patients exhibiting diarrhoea and testing positive for C. difficile were systematically collected from a tertiary care hospital in Madrid (Spain). The periods analysed included: prepandemic (P0), first pandemic-year (P1), and second pandemic-year (P2). We compared global prevalence, RI of HO-CDI per 1,000-admissions, ID of HO-CDI per 10,000-patients-days, RRI, RID, and RR. Antibiotic consumption was obtained by number of defined daily dose per 100 patient-days. RESULTS In P0, the prevalence of HO-CDI was 7.4% (IC95%: 6.2-8.7); in P1, it increased to 8.7% (IC95%: 7.4-10.1) (p = 0.2), and in P2, it continued to increase to 9.2% (IC95%: 8-10.6) (p < 0.05). During P1, the RRI was 1.5 and RID was 1.4. However, during P2 there was an increase in RRI to 1.6 and RID to 1.6. The RR also reflected the increase in HO-CDI: at P1, the probability of developing HO-CDI was 1.5 times (IC95%: 1.2-1.9) higher than P0, while at P2, this probability increased to 1.6 times (IC95%: 1.3-2.1). There was an increase in prevalence, RI, ID, RR, RRI, and RID during the two postpandemic periods respect to the prepandemic period. During P2, this increase was greater than the P1. Meropenem showed a statistically significant difference increased consumption (p < 0.05) during the pandemic period. Oral vancomycin HO-CDI treatment showed an increase during the period of study (p > 0.05). CONCLUSIONS Implementation of infection control measures during the SARS-CoV-2 pandemic did not appear to alleviate the burden of HO-CDI. The escalation in HO-CDI cases did not exhibit a correlation with overall antibiotic consumption, except for meropenem.
Collapse
Affiliation(s)
- Alfredo Maldonado-Barrueco
- Clinical Microbiology and Parasitology Department, Hospital Universitario La Paz-Carlos III-Cantoblanco, Madrid, Spain.
| | - Francisco Moreno-Ramos
- Pharmacy Hospital Department, Hospital Universitario La Paz-Carlos III-Cantoblanco, Madrid, Spain
| | - Beatriz Díaz-Pollán
- Infectious Diseases Unit, Internal Medicine Department, Hospital Universitario La Paz-Carlos III-Cantoblanco, Madrid, Spain; CIBERINFEC (Centre for Biomedical Research Network on Infectious Diseases), Instituto de Salud Carlos III, Madrid, Spain
| | - Belén Loeches-Yagüe
- Clinical Microbiology and Parasitology Department, Hospital Universitario La Paz-Carlos III-Cantoblanco, Madrid, Spain; Infectious Diseases Unit, Internal Medicine Department, Hospital Universitario La Paz-Carlos III-Cantoblanco, Madrid, Spain; CIBERINFEC (Centre for Biomedical Research Network on Infectious Diseases), Instituto de Salud Carlos III, Madrid, Spain
| | - Alicia Rico-Nieto
- Clinical Microbiology and Parasitology Department, Hospital Universitario La Paz-Carlos III-Cantoblanco, Madrid, Spain; Infectious Diseases Unit, Internal Medicine Department, Hospital Universitario La Paz-Carlos III-Cantoblanco, Madrid, Spain; CIBERINFEC (Centre for Biomedical Research Network on Infectious Diseases), Instituto de Salud Carlos III, Madrid, Spain
| | - Julio García-Rodríguez
- Clinical Microbiology and Parasitology Department, Hospital Universitario La Paz-Carlos III-Cantoblanco, Madrid, Spain; CIBERINFEC (Centre for Biomedical Research Network on Infectious Diseases), Instituto de Salud Carlos III, Madrid, Spain
| | - Guillermo Ruiz-Carrascoso
- Clinical Microbiology and Parasitology Department, Hospital Universitario La Paz-Carlos III-Cantoblanco, Madrid, Spain; CIBERINFEC (Centre for Biomedical Research Network on Infectious Diseases), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
41
|
Watkin S, Yongblah F, Burton J, Hartley JC, Cloutman-Green E. Clostridioides difficile detection and infection in children: are they just small adults? J Med Microbiol 2024; 73. [PMID: 38526913 DOI: 10.1099/jmm.0.001816] [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: 03/27/2024] Open
Abstract
Clostridioides difficile is a well-recognized healthcare-associated pathogen, with its significance widely recognized in adult populations. Despite this, there is limited data on the significance of detection within paediatric populations, both for individual patient management and wider transmission risk-based considerations. High rates of colonization are understood to occur in infants, with increasing levels up to 11 months, and colonization rates similar to adults by 8 years old. Sources of C. difficile are ubiquitous, with detection in companion animals and food sources, as well as within the clinical and wider environment. Due to the close interactions that occur between children and the environment, it is understandable that increasing recognition is afforded to the community acquisition of C. difficile in children. Other risk factors for the detection of C. difficile in children are similar to those observed in adults, including prior hospitalization and underlying conditions affecting gut health and motility. Recent studies have shown rising awareness of the role of asymptomatic carriage of C. difficile in healthcare transmission. Prior to this, paediatric patient populations were less likely to be screened due to uncertainty regarding the significance of detection; however, this increased awareness has led to a review of possible carriage testing pathways. Despite this increased attention, C. difficile infection remains poorly defined in paediatric populations, with limited dedicated paediatric data sets making comparison challenging. This is further complicated by the fact that infection in children frequently self resolves without additional therapies. Due to this, C. difficile remains a management challenge in paediatric settings.
Collapse
Affiliation(s)
- Sam Watkin
- Department of Civil Environmental and Geomatic Engineering, Healthy Infrastructure Research Group, University College London, Chadwick Building, London, UK
| | - Francis Yongblah
- Great Ormond Street Hospital NHS Foundation Trust, Camelia Botnar Laboratories, Department of Microbiology, London, UK
| | - James Burton
- Great Ormond Street Hospital NHS Foundation Trust, Camelia Botnar Laboratories, Department of Microbiology, London, UK
| | - John C Hartley
- Great Ormond Street Hospital NHS Foundation Trust, Camelia Botnar Laboratories, Department of Microbiology, London, UK
| | - Elaine Cloutman-Green
- Department of Civil Environmental and Geomatic Engineering, Healthy Infrastructure Research Group, University College London, Chadwick Building, London, UK
- Great Ormond Street Hospital NHS Foundation Trust, Camelia Botnar Laboratories, Department of Microbiology, London, UK
| |
Collapse
|
42
|
Bachmann I, Behrmann O, Klingenberg-Ernst M, Rupnik M, Hufert FT, Dame G, Weidmann M. Rapid Isothermal Detection of Pathogenic Clostridioides difficile Using Recombinase Polymerase Amplification. Anal Chem 2024; 96:3267-3275. [PMID: 38358754 DOI: 10.1021/acs.analchem.3c02985] [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: 02/16/2024]
Abstract
Nosocomial-associated diarrhea due to Clostridioides difficile infection (CDI) is diagnosed after sample precultivation by the detection of the toxins in enzyme immunoassays or via toxin gene nucleic acid amplification. Rapid and direct diagnosis is important for targeted treatment to prevent severe cases and recurrence. We developed two singleplex and a one-pot duplex fluorescent 15 min isothermal recombinase polymerase amplification (RPA) assays targeting the toxin genes A and B (tcdA and tcdB). Furthermore, we adapted the singleplex RPA to a 3D-printed microreactor device. Analytical sensitivity was determined using a DNA standard and DNA extracts of 20 C. difficile strains with different toxinotypes. Nineteen clostridial and gastrointestinal bacteria strains were used to determine analytical specificity. Adaptation of singleplex assays to duplex assays in a 50 μL volume required optimized primer and probe concentrations. A volume reduction by one-fourth (12.4 μL) was established for the 3D-printed microreactor. Mixing of RPA was confirmed as essential for optimal analytical sensitivity. Detection limits (LOD) ranging from 119 to 1411 DNA molecules detected were similar in the duplex tube format and in the singleplex 3D-printed microreactor format. The duplex RPA allows the simultaneous detection of both toxins important for the timely and reliable diagnosis of CDI. The 3D-printed reaction chamber can be developed into a microfluidic lab-on-a-chip system use at the point of care.
Collapse
Affiliation(s)
- Iris Bachmann
- Institute of Microbiology and Virology, Brandenburg Medical School Theodor Fontane, Universitätsplatz 1, 01968 Senftenberg, Germany
| | - Ole Behrmann
- Institute of Microbiology and Virology, Brandenburg Medical School Theodor Fontane, Universitätsplatz 1, 01968 Senftenberg, Germany
| | | | - Maja Rupnik
- Center for Medical Microbiology, Department for Microbiological Research, National Laboratory for Health, Environment and Food, Prvomajska ulica 1, 2000 Maribor, Slovenia
- Faculty of Medicine, Maribor, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
| | - Frank T Hufert
- Institute of Microbiology and Virology, Brandenburg Medical School Theodor Fontane, Universitätsplatz 1, 01968 Senftenberg, Germany
- Department of Virology, University Medical Center, Kreuzbergring 57, 37075 Göttingen, Germany
- Brandenburg University of Technology Cottbus - Senftenberg, Universitätsplatz 1, 01968 Senftenberg, Germany
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology Cottbus - Senftenberg, the Brandenburg Medical School Theodor Fontane and the University of Potsdam, Karl-Liebknecht-Str. 24-25, 14476 Potsdam, Germany
| | - Gregory Dame
- Institute of Microbiology and Virology, Brandenburg Medical School Theodor Fontane, Universitätsplatz 1, 01968 Senftenberg, Germany
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology Cottbus - Senftenberg, the Brandenburg Medical School Theodor Fontane and the University of Potsdam, Karl-Liebknecht-Str. 24-25, 14476 Potsdam, Germany
| | - Manfred Weidmann
- Institute of Microbiology and Virology, Brandenburg Medical School Theodor Fontane, Universitätsplatz 1, 01968 Senftenberg, Germany
- Department of Virology, University Medical Center, Kreuzbergring 57, 37075 Göttingen, Germany
| |
Collapse
|
43
|
Shen Y, Lin S, You P, Chen Y, Luo Y, Song X, Chen Y, Jin D. Rapid discrimination between clinical Clostridioides difficile infection and colonization by quantitative detection of TcdB toxin using a real-time cell analysis system. Front Microbiol 2024; 15:1348892. [PMID: 38322317 PMCID: PMC10844495 DOI: 10.3389/fmicb.2024.1348892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 01/08/2024] [Indexed: 02/08/2024] Open
Abstract
Objectives It is important to accurately discriminate between clinical Clostridioides difficile infection (CDI) and colonization (CDC) for effective antimicrobial treatment. Methods In this study, 37 stool samples were collected from 17 CDC and 20 CDI cases, and each sample were tested in parallel through the real-time cell analysis (RTCA) system, real-time PCR assay (PCR), and enzyme-linked immunosorbent assay (ELISA). Results RTCA-measured functional and toxical C. difficile toxin B (TcdB) concentrations in the CDI group (302.58 ± 119.15 ng/mL) were significantly higher than those in the CDC group (18.15 ± 11.81 ng/mL) (p = 0.0008). Conversely, ELISA results revealed no significant disparities in TcdB concentrations between the CDC (26.21 ± 3.57 ng/mL) and the CDI group (17.07 ± 3.10 ng/mL) (p = 0.064). PCR results indicated no significant differences in tcdB gene copies between the CDC (774.54 ± 357.89 copies/μL) and the CDI group (4,667.69 ± 3,069.87 copies/μL) (p = 0.407). Additionally, the functional and toxical TcdB concentrations secreted from C. difficile isolates were measured by the RTCA. The results from the CDC (490.00 ± 133.29 ng/mL) and the CDI group (439.82 ± 114.66 ng/mL) showed no significant difference (p = 0.448). Notably, RTCA-measured functional and toxical TcdB concentration was significantly decreased when mixed with pooled CDC samples supernatant (p = 0.030). Conclusion This study explored the novel application of the RTCA assay in effectively discerning clinical CDI from CDC cases.
Collapse
Affiliation(s)
- Yuhang Shen
- School of Laboratory Medicine, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Biomarkers and In Vitro Diagnosis Translation of Zhejiang Province, Hangzhou, China
- Institute of Ageing Research, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China
| | - Shan Lin
- School of Laboratory Medicine, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Biomarkers and In Vitro Diagnosis Translation of Zhejiang Province, Hangzhou, China
- TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin, China
| | - Peijun You
- School of Laboratory Medicine, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Biomarkers and In Vitro Diagnosis Translation of Zhejiang Province, Hangzhou, China
| | - Yu Chen
- School of Laboratory Medicine, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Biomarkers and In Vitro Diagnosis Translation of Zhejiang Province, Hangzhou, China
| | - Yun Luo
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Xiaojun Song
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Yunbo Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Dazhi Jin
- School of Laboratory Medicine, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Biomarkers and In Vitro Diagnosis Translation of Zhejiang Province, Hangzhou, China
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People’s Hospital, Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
44
|
Gurung B, Stricklin M, Wang S. Gut Microbiota-Gut Metabolites and Clostridioides difficile Infection: Approaching Sustainable Solutions for Therapy. Metabolites 2024; 14:74. [PMID: 38276309 PMCID: PMC10819375 DOI: 10.3390/metabo14010074] [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: 12/23/2023] [Revised: 01/06/2024] [Accepted: 01/19/2024] [Indexed: 01/27/2024] Open
Abstract
Clostridioides difficile (C. difficile) infection (CDI) is the most common hospital-acquired infection. With the combination of a high rate of antibiotic resistance and recurrence, it has proven to be a debilitating public health threat. Current treatments for CDI include antibiotics and fecal microbiota transplantation, which contribute to recurrent CDIs and potential risks. Therefore, there is an ongoing need to develop new preventative treatment strategies for CDI. Notably, gut microbiota dysbiosis is the primary risk factor for CDI and provides a promising target for developing novel CDI therapy approaches. Along with gut microbiota dysbiosis, a reduction in important gut metabolites like secondary bile acids and short-chain fatty acids (SCFAs) were also seen in patients suffering from CDI. In this review study, we investigated the roles and mechanisms of gut microbiota and gut microbiota-derived gut metabolites, especially secondary bile acids and SCFAs in CDI pathogenesis. Moreover, specific signatures of gut microbiota and gut metabolites, as well as different factors that can modulate the gut microbiota, were also discussed, indicating that gut microbiota modulators like probiotics and prebiotics can be a potential therapeutic strategy for CDI as they can help restore gut microbiota and produce gut metabolites necessary for a healthy gut. The understanding of the associations between gut microbiota-gut metabolites and CDI will allow for developing precise and sustainable approaches, distinct from antibiotics and fecal transplant, for mitigating CDI and other gut microbiota dysbiosis-related diseases.
Collapse
Affiliation(s)
- Bijay Gurung
- Department of Biomedical Sciences, Ohio University Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA; (B.G.); (M.S.)
- Infectious and Tropical Disease Institute, Ohio University, Athens, OH 45701, USA
- Interdisciplinary Molecular and Cellular Biology Program, Ohio University, Athens, OH 45701, USA
| | - Maranda Stricklin
- Department of Biomedical Sciences, Ohio University Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA; (B.G.); (M.S.)
- Infectious and Tropical Disease Institute, Ohio University, Athens, OH 45701, USA
| | - Shaohua Wang
- Department of Biomedical Sciences, Ohio University Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA; (B.G.); (M.S.)
- Infectious and Tropical Disease Institute, Ohio University, Athens, OH 45701, USA
| |
Collapse
|
45
|
Cersosimo LM, Worley JN, Bry L. Approaching pathogenic Clostridia from a One Health perspective. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.08.574718. [PMID: 38260382 PMCID: PMC10802438 DOI: 10.1101/2024.01.08.574718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Spore-forming pathogens have a unique capacity to thrive in diverse environments, and with temporal persistence afforded through their ability to sporulate. These behaviors require a One Health approach to identify critical reservoirs and outbreak-associated transmission chains, given their capacity to freely move across soils, waterways, foodstuffs, and as commensals or infecting pathogens in human and veterinary populations. Among anaerobic spore-formers, genomic resources for pathogens including C. botulinum, C. difficile, and C. perfringens enable our capacity to identify common and unique factors that support their persistence in diverse reservoirs and capacity to cause disease. Publicly available genomic resources for spore-forming pathogens at NCBI's Pathogen Detection program aid outbreak investigations and longitudinal monitoring in national and international programs in public health and food safety, as well as for local healthcare systems. These tools also enable research to derive new knowledge regarding disease pathogenesis, and to inform strategies in disease prevention and treatment. As global community resources, the continued sharing of strain genomic data and phenotypes further enhances international resources and means to develop impactful applications. We present examples showing use of these resources in surveillance, including capacity to assess linkages among clinical, environmental, and foodborne reservoirs and to further research investigations into factors promoting their persistence and virulence in different settings.
Collapse
Affiliation(s)
- Laura M. Cersosimo
- Massachusetts Host-Microbiome Center, Dept. Pathology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA
| | - Jay N. Worley
- Massachusetts Host-Microbiome Center, Dept. Pathology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA
- National Center for Biotechnology Information, NIH, Bethesda, MD
| | - Lynn Bry
- Massachusetts Host-Microbiome Center, Dept. Pathology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA
- Clinical Microbiology Laboratory, Dept. Pathology, Brigham & Women's Hospital, Boston, MA
| |
Collapse
|
46
|
Carr A, Baliga NS, Diener C, Gibbons SM. Personalized Clostridioides difficile engraftment risk prediction and probiotic therapy assessment in the human gut. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.28.538771. [PMID: 37162960 PMCID: PMC10168307 DOI: 10.1101/2023.04.28.538771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Clostridioides difficile colonizes up to 30-40% of community-dwelling adults without causing disease. C. difficile infections (CDIs) are the leading cause of antibiotic-associated diarrhea in the U.S. and typically develop in individuals following disruption of the gut microbiota due to antibiotic or chemotherapy treatments. Further treatment of CDI with antibiotics is not always effective and can lead to antibiotic resistance and recurrent infections (rCDI). The most effective treatment for rCDI is the reestablishment of an intact microbiota via fecal microbiota transplants (FMTs). However, the success of FMTs has been difficult to generalize because the microbial interactions that prevent engraftment and facilitate the successful clearance of C. difficile are still only partially understood. Here we show how microbial community-scale metabolic models (MCMMs) accurately predicted known instances of C. difficile colonization susceptibility or resistance in vitro and in vivo. MCMMs provide detailed mechanistic insights into the ecological interactions that govern C. difficile engraftment, like cross-feeding or competition involving metabolites like succinate, trehalose, and ornithine, which differ from person to person. Indeed, three distinct C. difficile metabolic niches emerge from our MCMMs, two associated with positive growth rates and one that represents non-growth, which are consistently observed across 15,204 individuals from five independent cohorts. Finally, we show how MCMMs can predict personalized engraftment and C. difficile growth suppression for a probiotic cocktail (VE303) designed to replace FMTs for the treatment rCDI. Overall, this powerful modeling approach predicts personalized C. difficile engraftment risk and can be leveraged to assess probiotic treatment efficacy. MCMMs could be extended to understand the mechanistic underpinnings of personalized engraftment of other opportunistic bacterial pathogens, beneficial probiotic organisms, or more complex microbial consortia.
Collapse
Affiliation(s)
- Alex Carr
- Institute for Systems Biology, Seattle, WA, USA
- Molecular Engineering Program, University of Washington, Seattle, WA, USA
| | - Nitin S. Baliga
- Institute for Systems Biology, Seattle, WA, USA
- Molecular Engineering Program, University of Washington, Seattle, WA, USA
- Departments of Biology and Microbiology, University of Washington, Seattle, WA, USA
- Lawrence Berkeley National Lab, Berkeley, CA, USA
| | - Christian Diener
- Institute for Systems Biology, Seattle, WA, USA
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Graz, Austria
| | - Sean M. Gibbons
- Institute for Systems Biology, Seattle, WA, USA
- Molecular Engineering Program, University of Washington, Seattle, WA, USA
- Departments of Bioengineering and Genome Sciences, University of Washington, Seattle, WA, USA
- eScience Institute, University of Washington, Seattle, WA, USA
| |
Collapse
|
47
|
van Prehn J, Crobach MJT, Baktash A, Duszenko N, Kuijper EJ. Diagnostic Guidance for C. difficile Infections. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1435:33-56. [PMID: 38175470 DOI: 10.1007/978-3-031-42108-2_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Diagnosis of Clostridioides difficile infection (CDI) can be challenging. First of all, there has been debate on which of the two reference assays, cell cytotoxicity neutralization assay (CCNA) or toxigenic culture (TC), should be considered the gold standard for CDI detection. Although the CCNA suffers most from suboptimal storage conditions and subsequent toxin degradation, TC is reported to falsely increase CDI detection rates as it cannot differentiate CDI patients from patients asymptomatically colonised by toxigenic C. difficile. Several rapid assays are available for CDI detection and fall into three broad categories: (1) enzyme immunoassays for glutamate dehydrogenase, (2) enzyme immunoassays or single-molecule array assays for toxins A/B and (3) nucleic acid amplification tests detecting toxin genes. All three categories have their own limitations, being suboptimal specificity and/or sensitivity or the inability to discern colonised patients from CDI patients. In light of these limitations, multi-step algorithmic testing has been advocated by international guidelines (IDSA/SHEA and ESCMID) in order to optimize diagnostic accuracy. As a result, a survey performed in 2018-2019 in Europe revealed that most of all hospital sites reported using more than one test to diagnose CDI. CDI incidence rates are also influenced by sample selection criteria, as several studies have shown that if not all unformed stool samples are tested for CDI, many cases may be missed due to an absence of clinical suspicion. Since methods for diagnosing CDI remain imperfect, there has been a growing interest in alternative testing strategies like faecal microbiota biomarkers, immune modulating interleukins, cytokines and imaging methods. At the moment, these alternative methods might play an adjunctive role, but they are not suitable to replace conventional CDI testing strategies.
Collapse
Affiliation(s)
- Joffrey van Prehn
- Department of Medical Microbiology, Leiden University Centre for Infectious Diseases (LU-CID), Leiden University Medical Centre, Leiden, The Netherlands.
- ESCMID Study Group for C. difficile (ESGCD) and Study Group for Host and Microbiota Interaction (ESGHAMI), Basel, Switzerland.
| | - Monique J T Crobach
- Department of Medical Microbiology, Leiden University Centre for Infectious Diseases (LU-CID), Leiden University Medical Centre, Leiden, The Netherlands
| | - Amoe Baktash
- Department of Medical Microbiology, Leiden University Centre for Infectious Diseases (LU-CID), Leiden University Medical Centre, Leiden, The Netherlands
| | - Nikolas Duszenko
- Department of Medical Microbiology, Leiden University Centre for Infectious Diseases (LU-CID), Leiden University Medical Centre, Leiden, The Netherlands
| | - Ed J Kuijper
- Department of Medical Microbiology, Leiden University Centre for Infectious Diseases (LU-CID), Leiden University Medical Centre, Leiden, The Netherlands
- ESCMID Study Group for C. difficile (ESGCD) and Study Group for Host and Microbiota Interaction (ESGHAMI), Basel, Switzerland
| |
Collapse
|
48
|
Levy EI, Dinleyici M, Dinleyici E, Vandenplas Y. Clostridioides difficile Infections: Prevention and Treatment Strategies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1449:175-186. [PMID: 39060738 DOI: 10.1007/978-3-031-58572-2_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
Clostridioides difficile is the most common causative agent of antibiotic-associated diarrhea. This spore forming, obligate anaerobic, gram-positive bacillus is becoming responsible for an increasing number of infections worldwide, both in community and in hospital settings, whose severity can vary widely from an asymptomatic infection to a lethal disease. While discontinuation of antimicrobial agents and antibiotic treatment of the infection remain the cornerstone of therapy, more recent fecal microbiota transplantation has also been valid as a therapy. The use of probiotics, especially Saccharomyces boulardii CNCM I-745 have become valid forms of prevention therapy. Although there are studies in adults with microbiota-targeted new generation therapies and Clostridium difficile vaccines, there are no data in the paediatric age group yet.
Collapse
Affiliation(s)
- Elvira Ingrid Levy
- Department of Pediatrics, C.H.U. Saint-Pieter, Free University of Brussels, Brussels, Belgium
| | - Meltem Dinleyici
- Eskisehir Osmangazi University Faculty of Medicine, Department of Social Pediatrics, Eskisehir, Turkey
| | - Ener Dinleyici
- Department of Pediatrics, Eskisehir Osmangazi University School of Medicine, Eskisehir, Turkey
| | - Yvan Vandenplas
- Vrije Universiteit Brussel (VUB), UZ Brussel, KidZ Health Castle, Brussels, Belgium.
| |
Collapse
|
49
|
Campidelli C, Bruxelle JF, Collignon A, Péchiné S. Immunization Strategies Against Clostridioides difficile. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1435:117-150. [PMID: 38175474 DOI: 10.1007/978-3-031-42108-2_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Clostridioides difficile (C. difficile) infection (CDI) is an important healthcare but also a community-associated disease. CDI is considered a public health threat and an economic burden. A major problem is the high rate of recurrences. Besides classical antibiotic treatments, new therapeutic strategies are needed to prevent infection, to treat patients, and to prevent recurrences. If fecal transplantation has been recommended to treat recurrences, another key approach is to elicit immunity against C. difficile and its virulence factors. Here, after a summary concerning the virulence factors, the host immune response against C. difficile, and its role in the outcome of disease, we review the different approaches of passive immunotherapies and vaccines developed against CDI. Passive immunization strategies are designed in function of the target antigen, the antibody-based product, and its administration route. Similarly, for active immunization strategies, vaccine antigens can target toxins or surface proteins, and immunization can be performed by parenteral or mucosal routes. For passive immunization and vaccination as well, we first present immunization assays performed in animal models and second in humans and associated clinical trials. The different studies are presented according to the mode of administration either parenteral or mucosal and the target antigens and either toxins or colonization factors.
Collapse
Affiliation(s)
- Camille Campidelli
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Jean-François Bruxelle
- CIRI-Centre International de Recherche en Infectiologie, Université de Lyon, Université Claude Bernard Lyon 1, Inserm U1111, CNRS UMR5308, ENS Lyon, Lyon, France
| | - Anne Collignon
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Severine Péchiné
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France.
| |
Collapse
|
50
|
Raj N, Agarwal J, Singh V, Sen M, Das A. Healthcare-associated Diarrhea due to Clostridioides difficile in Patients Attending a Tertiary Care Teaching Hospital of North India. Euroasian J Hepatogastroenterol 2024; 14:60-64. [PMID: 39022194 PMCID: PMC11249909 DOI: 10.5005/jp-journals-10018-1429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 04/15/2024] [Indexed: 07/20/2024] Open
Abstract
Background Healthcare-associated diarrhea (HCAD) is diarrhea that develops at least after 3 days of hospitalization, with the most common infectious cause being Clostridioides difficile. Over the last decade, there has been a remarkable growth in the frequency and severity of C. difficile infection (CDI), making it one of the most prevalent healthcare-associated infections. This study aimed to analyze the prevalence and risk factors associated with CDI. Materials and methods A total of 107 patients with clinical suspicion of having HCAD were included in this study. Enzyme-linked fluorescent assay (ELFA) technique-based glutamate dehydrogenase (GDH) and toxin A/B assay were used as per the European Society of Clinical Microbiology and Infectious Diseases (ESCMID) for diagnosing CDI. The details about associated comorbidities were retrieved from the hospital information system records. The presence of risk factors was noted. Risk factors associated with CDI were looked for. Results Out of the 107 stool samples received in the microbiology laboratory from patients with suspected HCAD eight (7.6%) samples were positive for CDI. The most frequent comorbidity observed in these patients was renal illness (acute or chronic kidney disease). In this study, a total of 7/8 cases were on multiple antibiotics most common being carbapenem. Conclusion The 6-year prevalence of CDI observed in this study was found to be 7.6% risk factors, associated with CDI were kidney disease, diabetes mellitus, malignancy, and exposure to broad-spectrum antibiotics. How to cite this article Raj N, Agarwal J, Singh V, et al. Healthcare-associated Diarrhea due to Clostridioides difficile in Patients Attending a Tertiary Care Teaching Hospital of North India. Euroasian J Hepato-Gastroenterol 2024;14(1):60-64.
Collapse
Affiliation(s)
- Nikhil Raj
- Department of Microbiology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Jyotsna Agarwal
- Department of Microbiology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Vikramjeet Singh
- Department of Microbiology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Manodeep Sen
- Department of Microbiology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Anupam Das
- Department of Microbiology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| |
Collapse
|